Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biotechnol J ; 14(3): e1800020, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29802765

RESUMEN

Cationic bolaamphiphile polymers had been previously studied as efficient delivery system for the delivery of proteins with relatively low toxicity. Here, the authors investigate the use of a protein delivery system based on a cationic bolaamphiphile to sensitize cancer cells toward apoptosis-inducing drugs as a novel approach for cancer therapy. The authors demonstrates the efficacy of the system by two strategies. The first strategy involves delivery of a survivin antibody to inhibit survivin activity. Sensitization of MCF-7 cells to doxorubicin is observed by survivin inhibition by antibodies. The IC50 of doxorubicin is reduced ≈2.5-fold after delivery of survivin antibodies to breast cancer cells and induction of apoptosis is shown by Western blotting with apoptosis specific antibodies. In a second approach, functional wild type p53 is delivered into p53-null liver cancer (Hep3B) cells, sensitizing the cells toward the p53 pathway drug, Nutlin. Nutlin reduced the viability of Hep3B cells by ≈42% at 15 µM concentration, demonstrating the effectiveness of p53 delivery. The expression of p21, a downstream target of p53 further confirmed the functional status of the delivered protein. In conclusion. The successful delivery of apoptosis inducing proteins and sensitization of cancer cells via cationic bolaamphiphile polymer represents a promising system for cancer therapeutics.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Cationes/farmacología , Furanos/farmacología , Piridonas/farmacología , Línea Celular , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Doxorrubicina/farmacología , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Células MCF-7 , Transducción de Señal/efectos de los fármacos , Survivin/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
2.
Adv Healthc Mater ; 5(6): 696-701, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26792539

RESUMEN

In highly proliferative cancer cells, energy is predominantly produced by a high rate of glycolysis, followed by lactic acid fermentation, despite the availability of oxygen - an observation known as the Warburg effect. As a consequence, cells employing this glycolytic pathway require high uptake of glucose and increased metabolic rates to maintain their proliferation. It has been hypothesized that by blocking glucose uptake using modified glucose molecules, apoptosis in the cancer cells can be induced. In this study, it has been showed that several poly(ethylene glycol) (PEG)-modified glucose compounds could reduce cell proliferation in various cancer cell lines by a phenomenon that blocked the availability of the glucose transporters and reduced AKT1 (serine/threonine-specific protein kinase) activation. Xenograft cancer models that are intravenously administered with glucose-conjugated branched PEG (GBrP) daily for 14 d show little tumor development, as compared to the control group without GBrP treatment. The toxicological effects and the pharmacokinetics of the PEGylated glucose are studied in rodents. The PEGylated glucose exerts no systemic toxicity at 40 mg kg(-1) dosage. However, doses above 80 mg kg(-1) show dose-dependent toxicity in all the organs analyzed. The present results suggest PEGylated glucose as a promising "metabolic therapy" approach for the treatment of cancer.


Asunto(s)
Antineoplásicos/química , Glucosa/química , Polietilenglicoles/química , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Colorantes Fluorescentes/química , Glucosa/farmacología , Glucosa/uso terapéutico , Transportador de Glucosa de Tipo 1/antagonistas & inhibidores , Transportador de Glucosa de Tipo 1/metabolismo , Células Hep G2 , Humanos , Células MCF-7 , Ratones , Ratones SCID , Microscopía Fluorescente , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trasplante Heterólogo
3.
Biomaterials ; 35(26): 7479-87, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24912819

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most commonly diagnosed lethal cancers in the world. We previously showed two imidazolium salts (IBN-1 and IBN-9) with a moderate efficacy for HCC. Here we report a more potent imidazolium compound IBN-65 (1-benzyl-2-phenyl-3-(4-isopropyl)-benzyl-imidazolium chloride) and the associated mechanisms of action in a mouse model of HCC. The IC50 of this compound in various liver cancer cell lines was around 5 µm. IBN-65 dose-dependently arrested cell cycle at G1 phase and was associated with the down-regulation of the cyclin-dependent kinase-4, -6, cyclin D1, and cyclin E. In addition, IBN-65 induced apoptosis by down-regulating Survivin, Bcl-2 and up-regulating Bax, leading to sequential activation of Caspase-3, Caspase-9 and the cleavage of poly(ADP-ribose) polymerase (PARP). Dysregulation of the epidermal growth factor receptor (EGFR) signaling network has been frequently reported in HCC. We found that IBN-65 displayed a profound inhibitory effect on the EGFR/Raf/MEK/ERK signaling at the phosphorylation level. In Huh7 or Hep3B cells, pretreatment with IBN-65 attenuated EGF-induced phosphorylation of both EGFR and the downstream p44/42 MAPK. A siRNA knockdown of EGFR also proved that IBN-65 induced apoptosis mostly through inhibiting downstream EGFR pathway signaling, much less at the receptor level. Infrequent administration of IBN-65 (i.p., 5 mg/kg once weekly for four weeks) to mice bearing the Huh7 cells significantly reduced the tumor volume by 65% without affecting the body weight. Critically, many of the anti-tumor signaling features observed in the HCC cell lines were recaptured in the xenografted tissues. Thus, the metal-free imidazolium compound IBN-65 could be a potential candidate towards therapeutic development for HCC.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Imidazoles/química , Imidazoles/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Receptores ErbB/metabolismo , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Terapia Molecular Dirigida , Transducción de Señal/efectos de los fármacos
4.
Kidney Int ; 83(4): 593-603, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23389418

RESUMEN

Renal cells are used in basic research, disease models, tissue engineering, drug screening, and in vitro toxicology. In order to provide a reliable source of human renal cells, we developed a protocol for the differentiation of human embryonic stem cells into renal epithelial cells. The differentiated stem cells expressed markers characteristic of renal proximal tubular cells and their precursors, whereas markers of other renal cell types were not expressed or expressed at low levels. Marker expression patterns of these differentiated stem cells and in vitro cultivated primary human renal proximal tubular cells were comparable. The differentiated stem cells showed morphological and functional characteristics of renal proximal tubular cells, and generated tubular structures in vitro and in vivo. In addition, the differentiated stem cells contributed in organ cultures for the formation of simple epithelia in the kidney cortex. Bioreactor experiments showed that these cells retained their functional characteristics under conditions as applied in bioartificial kidneys. Thus, our results show that human embryonic stem cells can differentiate into renal proximal tubular-like cells. Our approach would provide a source for human renal proximal tubular cells that are not affected by problems associated with immortalized cell lines or primary cells.


Asunto(s)
Órganos Bioartificiales , Diferenciación Celular , Células Madre Embrionarias/fisiología , Células Epiteliales/fisiología , Túbulos Renales Proximales/fisiología , Ingeniería de Tejidos , Activinas/farmacología , Animales , Biomarcadores/metabolismo , Reactores Biológicos , Proteína Morfogenética Ósea 2/farmacología , Proteína Morfogenética Ósea 7/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Forma de la Célula , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/trasplante , Regulación del Desarrollo de la Expresión Génica , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/trasplante , Ratones , Ratones SCID , Técnicas de Cultivo de Órganos , Factores de Tiempo , Ingeniería de Tejidos/métodos , Tretinoina/farmacología
5.
Lab Invest ; 91(5): 744-51, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21339744

RESUMEN

Imidazolium salts (IMSs) are precursors to N-heterocyclic carbenes (NHCs), which are routinely used as ligands or organo-catalysts in synthetic chemistry. We recently identified several IMSs as anti-fibrotic agents in liver fibrosis, which often has a consequence in the oncogenesis of hepatocellular carcinoma (HCC). Here, we investigate the potential anti-tumor property of three IMSs (named IBN-1, IBN-9, and DPIM) in HCC cell lines and in a xenograft mouse model. Our results showed that both IBN-1 and IBN-9 significantly inhibited the cell proliferation and arrested HCC cells in the G1-phase, whereas DPIM did not have any anti-tumor activity. When tested in a Huh7 HCC xenograft mouse model, IBN-1 reduced the tumor volume by 31% (P<0.05), however accompanied by a 9% loss in body weight (P<0.005), suggesting a general toxicity. In contrast, IBN-9 significantly reduced the tumor volume by 45% (P<0.05) and 60% (P<0.01) at doses of 0.6 and 1.5 g/l in drinking water, respectively, without any loss in body weight. Our in vitro and in vivo data suggested that IBN-1 and IBN-9 inhibited the growth of HCC by suppressing the expression of Survivin and Cyclin-dependent kinases. The current study provides a proof of concept for using the metal-free IMSs to develop novel anti-cancer agents.


Asunto(s)
Carcinoma Hepatocelular/patología , División Celular/efectos de los fármacos , Imidazoles/farmacología , Neoplasias Hepáticas Experimentales/patología , Animales , Western Blotting , Ciclo Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Modelos Animales de Enfermedad , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Represoras/metabolismo , Sales (Química) , Survivin
6.
Cancer Lett ; 254(2): 217-26, 2007 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-17449172

RESUMEN

Adenovirus-mediated mda-7 (Ad-mda7) gene transfer has been shown to induce apoptosis in various human cancer cells while sparing normal cells. Vitamin E succinate (VES) is also known to exhibit antitumor activity against a number of human cancer cell lines. We hypothesized that a combination of the two agents would produce an enhanced antitumor effect in MDAH2774 human ovarian cancer cells. Treatment of MDAH2774 cells with Ad-mda7 plus VES resulted in enhanced antitumor activity that involved the activation of two apoptotic pathways. Activation of the extrinsic pathway was demonstrated by increased cell-surface Fas expression and cleavage of Bid and caspase-8. Activation of the intrinsic pathway was demonstrated by disruption of mitochondrial potential; and activation of downstream capase-9 and caspase-3 via cytochrome C release. In contrast, the combination of Ad-mda7 plus VES did not show any antitumor activity against normal fibroblasts, indicating selective tumor cell killing. Our in vitro results provide a basis for further preclinical testing of Ad-mda7 plus VES as a potential cancer treatment strategy.


Asunto(s)
Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Interleucinas/farmacología , Neoplasias Ováricas/patología , Receptores Virales/fisiología , Vitamina E/análogos & derivados , Adenoviridae/fisiología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Enterovirus/fisiología , Femenino , Humanos , Potenciales de la Membrana , Membranas Mitocondriales/fisiología , Tocoferoles , Vitamina E/farmacología
7.
Mol Cancer Ther ; 6(4): 1440-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17431123

RESUMEN

Nuclear factor-kappaB (NF-kappaB) activation promotes cell survival and growth. Reports show that chemotherapeutic agents and cytokines that are used for cancer therapy activate NF-kappaB expression in tumor cells and its suppression enhanced the antitumor activity. We hypothesized that adenovirus-mediated overexpression of melanoma differentiation-associated gene-7/interleukin-24 (Ad-mda7/IL-24) induces NF-kappaB expression and that inhibition of this expression results in enhanced tumor cell killing. Treatment of human lung tumor (H1299 and A549) cells with Ad-mda7 resulted in NF-kappaB activation in a dose- and time-dependent manner before activation of cell death pathways. To establish that inhibition of Ad-mda7-mediated NF-kappaB activation results in enhanced tumor cell killing, H1299 cells that overexpress the dominant-negative I kappa B alpha (dnI kappa B alpha) were treated with Ad-mda7 in vitro. An enhanced growth arrest and apoptosis was observed in Ad-mda7-treated H1299-dnI kappa B alpha compared with H1299-Neo cells. This Ad-mda7-mediated enhanced killing of H1299-dnI kappa B alpha cells involved cleavage of mitogen-activated protein kinase kinase kinase 1 (MEKK1) and caspase-3 in a feedback loop mechanism. The inhibition of MEKK1 or caspase-3 cleavage in H1299-dnI kappa B alpha cells resulted in reduced Ad-mda7-mediated cell killing. In vivo, the treatment of H1299-dnI kappa B alpha s.c. tumors with Ad-mda7 resulted in increased drug sensitivity and delayed the tumor growth rate compared with Ad-mda7-treated H1299-Neo tumors. Molecular analysis of Ad-mda7-treated H1299-dnI kappa B alpha tumors showed increased MEKK1 cleavage and activation of caspase-3 compared with Ad-mda7-treated H1299-Neo tumors. Our findings thus showed that the NF-kappaB activation induced by Ad-mda7 treatment of lung cancer cells is an intrinsic survival mechanism and that the inhibition of this NF-kappaB expression results in enhanced tumor cell killing.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Interleucinas/genética , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , FN-kappa B/antagonistas & inhibidores , Animales , Caspasa 3/metabolismo , Muerte Celular , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Activación Enzimática , Humanos , Proteínas I-kappa B/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Desnudos , Inhibidor NF-kappaB alfa , Factores de Tiempo
8.
Mol Cancer ; 6: 11, 2007 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-17274815

RESUMEN

BACKGROUND: Previous studies showed that the human melanoma differentiation-associated gene-7 (mda-7), also known as interleukin-24 (IL-24), has potent antitumor activity against human and murine cancer cells. However, the majority of these studies were limited to in vitro testing. In the present study, we investigated the antitumor activity of mda-7/IL-24 against human ovarian cancer cells both in vitro and in vivo. RESULTS: In vitro, treatment of ovarian cancer cells with an adenoviral vector carrying the mda-7 gene (Ad-mda7) resulted in inhibition of cell proliferation and induction of cell cycle arrest, leading to apoptosis. We did not observe inhibitory activity in Ad-mda7-treated normal cells. In vivo, treatment of subcutaneous tumor xenografts with Ad-mda7 resulted in significant tumor growth inhibition when compared with that in control groups (p < 0.001). Molecular analysis of ovarian tumor tissue lysates treated with Ad-mda7 showed that MDA-7 protein expression was associated with activation of the caspase cascade. CONCLUSION: Our results show that treatment of ovarian cancer cells with mda-7/IL-24 results in growth suppression both in vitro and in vivo.


Asunto(s)
Terapia Genética , Interleucinas/metabolismo , Interleucinas/uso terapéutico , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Adenoviridae/genética , Animales , Apoptosis , Biomarcadores , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Interleucinas/genética , Ratones , Ratones Endogámicos BALB C , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Curr Gene Ther ; 6(1): 73-91, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16475947

RESUMEN

Despite recent advances in treatment strategies, the overall 5-year survival rate for patients with common epithelial cancers is poor largely because of the difficulty in treating metastatic cancers. Therefore, therapeutic agents are urgently needed that can effectively inhibit both primary epithelial tumors and their metastases. One such agent that has shown promise in preclinical studies is the tumor suppressor/cytokine, melanoma differentiation associated gene-7 also known as interleukin-24 (mda-7/IL-24). Preclinical studies from our and other laboratories have shown that overexpression of MDA-7/IL-24 causes a strong tumor- suppressive effect in many human cancer cells but spares normal cells. This gene therapy also enhances the tumor-suppressive activity of radiotherapy and chemotherapy. Secreted MDA-7 protein that is glycosylated also has been shown to have potent antiangiogenic activity both in vitro and in vivo. Studies examining the immune properties of mda-7 have shown that MDA-7/IL-24 unlike the related IL-10, functions as a Th1 cytokine. Recently, an MDA-7 protein-mediated "bystander effect" on tumor cells has been documented. Building on these findings we successfully completed a Phase I clinical trial of adenovirus-based mda-7 cancer therapy that confirmed the safety of this gene therapy. Phase II trials evaluating the efficacy of mda-7-based gene therapy are warranted. The outcome of such ongoing mda-7-based gene therapy trials will allow us to better understand this therapy's clinical utility.


Asunto(s)
Terapia Genética , Interleucinas/genética , Neoplasias/terapia , Adyuvantes Inmunológicos/genética , Ensayos Clínicos como Asunto/tendencias , Terapia Combinada , Evaluación Preclínica de Medicamentos/tendencias , Terapia Genética/métodos , Humanos , Interleucinas/inmunología , Neoplasias/genética , Neovascularización Patológica/genética
10.
Cancer Res ; 65(8): 3017-24, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15833826

RESUMEN

The tumor-suppressive activity of melanoma differentiation-associated gene-7 (mda-7), also known as interleukin 24 (IL-24), has been shown in a spectrum of human cancer cells in vitro and in vivo. However, mechanisms responsible for antitumor activity of mda-7 in human ovarian cancer cells have not been identified. We investigated the therapeutic activity and underlying mechanisms of adenovirus-mediated mda-7 gene (Ad-mda7) transfer in human ovarian cancer cells. Ad-mda7 treatment resulted in overexpression of MDA-7/IL-24 protein in both ovarian cancer and normal ovarian epithelial cells. However, Ad-mda7 significantly (P = 0.001) inhibited cell proliferation and induced apoptosis only in tumor cells and not in normal cells. Studies addressing the mechanism of action of Ad-mda7-induced tumor cell apoptosis revealed early activation of the transcription factors c-Jun and activating transcription factor 2, which in turn stimulated the transcription of an immediate downstream target, the death-inducer Fas ligand (FasL), and its cognate receptor Fas. Associated with the activation of Fas-FasL was the activation of nuclear factor kappaB and induction of Fas-associated factor 1, Fas-associated death domain, and caspase-8. Promoter-based reporter gene analyses showed that Ad-mda7 specifically activated the Fas promoter. Inhibition of Fas using small interfering RNA resulted in a significant decrease in Ad-mda7-mediated tumor cell death. Additionally, blocking of FasL with NOK-1 antibody abrogated Ad-mda7-mediated apoptosis. Collectively, these results show that Ad-mda7-mediated killing of human ovarian cancer cells involves activation of the Fas-FasL signaling pathway, a heretofore unrecognized mediator of MDA-7 apoptosis induction.


Asunto(s)
Interleucinas/genética , Glicoproteínas de Membrana/fisiología , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Receptor fas/fisiología , Adenoviridae/genética , Apoptosis/genética , Apoptosis/fisiología , Ciclo Celular/genética , Ciclo Celular/fisiología , Línea Celular Tumoral , Proliferación Celular , Proteína Ligando Fas , Femenino , Técnicas de Transferencia de Gen , Genes Supresores de Tumor , Terapia Genética/métodos , Humanos , Interleucinas/biosíntesis , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Receptor fas/biosíntesis , Receptor fas/genética
11.
Mol Cancer Ther ; 4(2): 291-304, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15713900

RESUMEN

Several studies have shown antitumor activities of the melanoma differentiation-associated gene 7 (mda-7) and the nonsteroidal anti-inflammatory drug sulindac when used as a monotherapies against a wide variety of human cancers. However, the combined effects of mda-7 and sulindac have not previously been tested. Therefore, we tested the antitumor activity of an adenoviral vector expressing mda-7 (Ad-mda7) in combination with sulindac against non-small cell lung cancer cells in vitro and in vivo. When treated with Ad-mda7 in combination with sulindac, human lung cancer cells (A549 and H1299) underwent growth suppression resulting in apoptosis. The growth inhibition induced by Ad-mda7 in combination with sulindac was significantly greater than that observed with Ad-mda7 or sulindac alone. Furthermore, the degree of growth inhibition induced using this combination was dose-dependent for sulindac. Treatment with Ad-mda7 in combination with sulindac had no growth inhibitory effects on human normal lung (CCD-16) fibroblasts. We then investigated the mechanism by which sulindac enhances Ad-mda7-mediated apoptosis. Sulindac increased expression of ectopic MDA-7 protein in tumor cells, thereby increasing the expression of downstream effectors RNA-dependent protein kinase, p38MAPK, caspase-9, and caspase-3 and enhancing apoptosis of non-small cell lung cancer cells. Pulse-chase experiments showed that the increased expression of MDA-7 protein in sulindac-treated cells was due to increased half-life of the MDA-7 protein. Finally, treatment of human lung tumor xenografts in nude mice with Ad-mda7 plus sulindac significantly suppressed growth (P = 0.001) compared with Ad-mda7 or sulindac alone. Our results show for the first time that combined treatment with Ad-mda7 plus sulindac enhances growth inhibition and apoptosis of human lung cancer cells. The increased antitumor activity observed with the combination treatment is a result of increased half-life of MDA-7 protein. Regulation of protein turnover is a heretofore-unrecognized mechanism of this nonsteroidal anti-inflammatory drug.


Asunto(s)
Adenoviridae/genética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Interleucinas/genética , Neoplasias Pulmonares/metabolismo , Sulindac/farmacología , Animales , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Genes Supresores de Tumor , Vectores Genéticos/genética , Humanos , Interleucinas/metabolismo , Ratones , Ratones Desnudos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Transducción de Señal/genética , Transducción Genética
12.
Cancer Gene Ther ; 12(3): 238-47, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15578066

RESUMEN

We have previously reported that overexpression of the melanoma differentiation-associated gene -7 (mda-7) using a replication-defective adenovirus (Ad-mda7), results in tumor-specific growth suppression and induction of apoptosis in wide variety of cancer cells. In the present study, we investigated the antitumor activity of Ad-mda7 and the underlying mechanism in human prostate cancer cells and normal prostate epithelial cells. Overexpression of MDA-7 induced significant (P=.001) suppression of cell growth and apoptosis in prostate cancer cells (DU 145, LNCaP, and PC-3). In normal prostate epithelial cells (PrEC) some degree of growth inhibition but not apoptosis was observed. However, the inhibitory effects in normal cells were less compared to tumor cells. Growth inhibitory effects were mediated by the intracellular and not by extracellular MDA-7 protein. Molecular effectors that are involved in Ad-mda7-mediated tumor killing included activation of the caspase cascade, and the induction of G2 phase cell cycle arrest through the inhibition of Cdc25C pathway. These results demonstrate the mechanisms by which Ad-mda7 exerts its antitumor activity in human prostate cancer cells. The antitumor activity combined with previously reported antiangiogenic and proimmune properties of Ad-mda7 can serve as a potential therapeutic agent for treatment of primary and disseminated prostate cancer.


Asunto(s)
Apoptosis/genética , Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Terapia Genética/métodos , Interleucinas/genética , Neoplasias de la Próstata/terapia , Adenoviridae , Análisis de Varianza , Anexina A5/metabolismo , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular , Citometría de Flujo , Técnicas de Transferencia de Gen , Genes Supresores de Tumor , Vectores Genéticos , Humanos , Immunoblotting , Interleucinas/farmacología , Masculino , Células Tumorales Cultivadas
13.
Technol Cancer Res Treat ; 3(6): 647-57, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15560723

RESUMEN

Cancer gene therapy for the treatment of lung cancer has shown promise in the laboratory and in Phase I/II clinical trials. However, it is currently limited to treating localized tumors due to host-immunity against the gene delivery vector and the transgene. Therefore, there is a tremendous effort to develop and test alternate gene delivery vectors that are efficient, non-immunogenic, and applicable for systemic therapy. One such gene delivery vehicle is the non-viral vector, DOTAP:cholesterol (DOTAP:Chol) nanoparticle. Preclinical studies from our laboratory has shown that DOTAP:Chol. nanoparticles are effective systemic gene delivery vectors that efficiently deliver tumor-suppressor genes to disseminated lung tumors. Based on our findings we have recently initiated a Phase-I trial for systemic treatment of lung cancer using a novel tumor suppressor gene, FUS1. Although DOTAP:Chol. nanoparticles complexed to DNA (DNA-nanoparticles) are efficient vectors for systemic therapy, induction of an inflammatory response in a dose-dependent fashion has also been observed thereby limiting its use. A better understanding of the underlying mechanism for DNA-nanoparticles-mediated inflammatory response will allow us to develop strategies to suppress inflammation and expand the therapeutic window in treating human cancer. In the present study we conducted experiments examining the mechanism of nanoparticle-mediated inflammatory response in vitro and in vivo. We demonstrate that systemic administration of DNA-nanoparticles induced multiple signaling molecules both in vitro and in vivo that are associated with inflammation. Use of small molecule inhibitors against the signaling molecules resulted in their suppression and thereby reduced inflammation without affecting transgene expression. Our results provide a rationale to use small molecule inhibitors to suppress nanoparticle-mediated inflammation when administered systemically. Further development and testing will allow us to incorporate this strategy into future clinical trials that is based on systemic non-viral vector gene therapy.


Asunto(s)
Terapia Genética/efectos adversos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Nanoestructuras/efectos adversos , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Línea Celular , Dinoprostona/metabolismo , Femenino , Expresión Génica , Humanos , Inflamación/genética , Inflamación/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Ratones , Naproxeno/farmacología , Naproxeno/uso terapéutico , Proteína FUS de Unión a ARN , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Transgenes/genética
14.
Cancer Gene Ther ; 11(11): 733-9, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15486560

RESUMEN

Lung cancer is one of the leading causes of death in the world. The underlying cause for lung cancer has been attributed to various factors that include alteration and mutation in the tumor suppressor genes. Restoration of normal function of the tumor suppressor gene is a potential therapeutic strategy. Recent studies have identified a group of candidate tumor suppressor genes on human chromosome 3p21.3 that are frequently deleted in human lung and breast cancers. Among the various genes identified in the 3p21.3 region, we tested the antitumor activity of the FUS1 gene in two human non-small-cell lung cancer (NSCLC) xenografts in vivo. Intratumoral administration of FUS1 gene complexed to DOTAP:cholesterol (DOTAP:Chol) liposome into subcutaneous H1299 and A549 lung tumor xenograft resulted in significant (P = .02) inhibition of tumor growth. Furthermore, intravenous injections of DOTAP:Chol-FUS1 complex into mice bearing experimental A549 lung metastasis demonstrated significant (P = .001) decrease in the number of metastatic tumor nodules. Finally, lung tumor-bearing animals when treated with DOTAP:Chol-FUS1 complex demonstrate prolonged survival (median survival time: 80 days, P = .01) compared to control animals. This result demonstrates the potent tumor suppressive activity of the FUS1 gene and is a promising therapeutic agent for treatment of primary and disseminated human lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Cromosomas Humanos Par 3 , Genes Supresores de Tumor , Terapia Genética , Vectores Genéticos , Neoplasias Pulmonares/terapia , Proteínas Supresoras de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Liposomas , Neoplasias Pulmonares/patología , Células Tumorales Cultivadas
15.
Mol Ther ; 9(4): 510-8, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15093181

RESUMEN

We have previously observed the suppression of lung tumor growth in response to overexpression of melanoma differentiation-associated gene-7 (MDA-7)/interleukin-24 (IL-24; approved gene symbol IL24) in vitro and in vivo. MDA-7/IL-24 exerts its tumor-suppressive effects by multiple mechanisms, including the activation of the caspase cascade and the inhibition of angiogenesis. In this study, we used an adenoviral vector (Ad-mda7) to examine the effect of the ectopic production of MDA-7/IL-24 on cell migration and invasion by human non-small-cell lung carcinoma cells. Lung tumor cells (H1299 and A549) treated in vitro with Ad-mda7 migrated and invaded less than cells treated with phosphate-buffered saline (PBS) or Ad-Luc (vector control). MDA-7/IL-24 inhibited migration and invasion by down-regulating the production of phosphatidylinositol 3-kinase/protein kinase B, focal adhesion kinase, and matrix metalloproteinase-2 and -9 relative to PBS and Ad-Luc. Furthermore, tumor cells treated with Ad-mda7 ex vivo or with DOTAP:Chol-mda7 complex in vivo formed significantly fewer tumors in an experimental lung metastasis model. These results show that MDA-7/IL-24 inhibits invasion and migration by lung cancer cells by down-regulating proteins associated with these processes, resulting in reduced metastasis. Thus, Ad-mda7 should be considered a therapeutic agent that can inhibit primary tumor growth and prevent metastasis.


Asunto(s)
Técnicas de Transferencia de Gen , Interleucinas/biosíntesis , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Adenoviridae/genética , Caspasas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Regulación hacia Abajo , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Gelatina/química , Genes Supresores de Tumor , Terapia Genética/métodos , Humanos , Immunoblotting , Luciferasas/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia/prevención & control , Neovascularización Patológica , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Fase de Descanso del Ciclo Celular , Factores de Tiempo
16.
Cancer Gene Ther ; 10(11): 803-13, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14605666

RESUMEN

The tumor suppressor phosphatase and tensin homologue deleted from chromosome 10 (PTEN) gene is a negative regulator of the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt/PKB) signaling pathway. Overexpression of PTEN in cancer cells results in cell-cycle arrest and cell death through inhibition of PI3K. Caffeine, a xanthine analogue, is well known to enhance the cytocidal and growth-inhibitory effects of DNA-damaging agents such as radiation, UV light, and anticancer agents on tumor cells by abrogating DNA-damage checkpoints through inhibition of ataxia-telangiectasia-mutated (ATM), and ATM and Rad3-related (ATR) kinase activity. In this study, we demonstrate that treatment with a combination of adenovirus-mediated transfer of PTEN (Ad-PTEN) and caffeine synergistically suppressed cell growth and induced apoptosis in colorectal cancer cells but not in normal colorectal fibroblast cells. This synergistic effect was induced through abrogation of G(2)/M arrest, downregulation of the Akt pathway, and modulation of the p44/42MAPK pathway. Thus, combined treatment with Ad-PTEN and caffeine is a potential therapy for colorectal cancer.


Asunto(s)
Adenoviridae/genética , Cafeína/farmacología , Neoplasias Colorrectales/terapia , Terapia Genética/métodos , Proteínas Tirosina Fosfatasas/genética , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Colon/citología , Colon/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/radioterapia , Sinergismo Farmacológico , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fase G2/efectos de los fármacos , Fase G2/genética , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Mitosis/efectos de los fármacos , Mitosis/genética , Fosfohidrolasa PTEN , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Fosfatasas/farmacología , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Radiación Ionizante , Valores de Referencia , Transducción de Señal , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA