Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
PLoS One ; 9(2): e87979, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24505341

RESUMO

Metformin, the most widely prescribed drug for treatment of type 2 diabetes, has been shown to exert significant anticancer effects. Hyperthermia has been known to kill cancer cells and enhance the efficacy of various anti-cancer drugs and radiotherapy. We investigated the combined effects of metformin and hyperthermia against MCF-7 and MDA-MB-231 human breast cancer cell, and MIA PaCa-2 human pancreatic cancer cells. Incubation of breast cancer cells with 0.5-10 mM metformin for 48 h caused significant clonogenic cell death. Culturing breast cancer cells with 30 µM metformin, clinically relevant plasma concentration of metformin, significantly reduced the survival of cancer cells. Importantly, metformin was preferentially cytotoxic to CD44(high)/CD24(low) cells of MCF-7 cells and, CD44(high)/CD24(high) cells of MIA PaCa-2 cells, which are known to be cancer stem cells (CSCs) of MCF-7 cells and MIA PaCa-2 cells, respectively. Heating at 42°C for 1 h was slightly toxic to both cancer cells and CSCs, and it markedly enhanced the efficacy of metformin to kill cancer cells and CSCs. Metformin has been reported to activate AMPK, thereby suppressing mTOR, which plays an important role for protein synthesis, cell cycle progression, and cell survival. For the first time, we show that hyperthermia activates AMPK and inactivates mTOR and its downstream effector S6K. Furthermore, hyperthermia potentiated the effect of metformin to activate AMPK and inactivate mTOR and S6K. Cell proliferation was markedly suppressed by metformin or combination of metformin and hyperthermia, which could be attributed to activation of AMPK leading to inactivation of mTOR. It is conclude that the effects of metformin against cancer cells including CSCs can be markedly enhanced by hyperthermia.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Hipertermia Induzida , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Células-Tronco Neoplásicas/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Proteínas de Neoplasias/biossíntese , Células-Tronco Neoplásicas/patologia
2.
Int J Hyperthermia ; 27(7): 698-707, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21992562

RESUMO

PURPOSE: We investigated the use of hyperthermia to improve the anti-cancer efficacy of doxorubicin (DOX)-loaded mesoporous silica nanocontainer Si-SS-CD-PEG. The hypothesis was that heat stimulates glutathione-mediated degradation of cyclodextrin gatekeeper, thereby causing the release of DOX from the carrier and DOX-induced cell death. MATERIALS AND METHODS: The release of DOX from DOX-loaded Si-SS-CD-PEG suspended in PBS containing glutathione (GSH) was studied by assessing the changes in DOX fluorescence intensity. The effect of heating at 42°C on the release of DOX from the intracellular carriers was determined with confocal microscopy. The extents of clonogenic and apoptotic cell death caused by DOX-loaded Si-SS-CD-PEG were determined. RESULTS: The release of DOX from DOX-loaded Si-SS-CD-PEG in PBS occurred only when GSH presented in the suspension, and heating at 42°C slightly increased the release of DOX from the carriers. Heating significantly elevated the GSH content in A549 cells and increased the release of DOX from the internalised carriers. Heating the cancer cells treated with the carriers at 42°C markedly increased the clonogenic death and apoptosis. The GSH content in A549 cells was greater than that in L-132 cells, and A549 cells were far more sensitive than L-132 cells to DOX-loaded Si-SS-CD-PEG at both 37°C and 42°C. CONCLUSIONS: Hyperthermia increased the GSH-mediated release of DOX from DOX-loaded Si-SS-CD-PEG. Furthermore, hyperthermia markedly elevated the GSH content in cancer cells, thereby increasing the release of DOX from the internalised carriers and potentiating the DOX-induced clonogenic and apoptotic cell death.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Hipertermia Induzida , Neoplasias Pulmonares/tratamento farmacológico , Nanoestruturas/administração & dosagem , Adenocarcinoma de Pulmão , Linhagem Celular Tumoral , Ciclodextrinas/administração & dosagem , Portadores de Fármacos , Glutationa/metabolismo , Humanos , Polietilenoglicóis/administração & dosagem , Succinimidas/administração & dosagem
3.
Int J Hyperthermia ; 27(1): 53-62, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21070139

RESUMO

PURPOSE: The anti-cancer effect of ß-lapachone (ß-lap) is positively related to the cellular activity of NAD(P)H:quinone oxidoreductase (NQO1). Heat shock has been reported to elevate cellular NQO1. The effect of heating on the NQO1 expression in human osteosarcoma cells (HOS) and the response of the cells to the combined treatment with ß-lap and hyperthermia was investigated. MATERIALS AND METHODS: The effects of ß-lap alone, hyperthermia alone and in combination to cause clonogenic death and apoptosis in HOS cells were elucidated. The effect of heating on the NQO1 expression was evaluated with western blot analysis. The effect of ß-lap on the cell cycle distribution was elucidated with flow cytometry and to cause DNA damage was determined by assessing the γH2AX foci formation. RESULTS: Treatment of HOS cells with ß-lap at 42°C was markedly more effective than that at 37°C in causing clonogenic cell death. Heating caused a long-lasting up-regulation of NQO1 in the cells, and sensitised the cells to ß-lap. The γH2AX foci formation was increased immediately after ß-lap treatment and preheating increased the ß-lap-induced γH2AX foci formation. CONCLUSIONS: The sensitivity of HOS cells to ß-lap was increased not only during heating but also after heating as demonstrated by the increase in the clonogenic cell death and γH2AX foci formation. The increase in ß-lap sensitivity after heating appeared to be due to the heat-induced elevation of NQO1 activity.


Assuntos
Hipertermia Induzida , Naftoquinonas/uso terapêutico , Osteossarcoma/terapia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Histonas/biossíntese , Histonas/efeitos dos fármacos , Humanos , NAD(P)H Desidrogenase (Quinona)/biossíntese , Osteossarcoma/tratamento farmacológico
4.
Lung Cancer ; 68(2): 278-82, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19596483

RESUMO

PURPOSE: To investigate the clinical significance of NQO1 C609T polymorphisms to treatment outcome after postoperative radiation therapy in completely resected non-small cell lung cancer (NSCLC). METHODS: One hundred and sixteen, Korean-ethnic, patients who were treated with surgery and postoperative radiation therapy from February 2000 to September 2005 in Asan Medical Center (Seoul, South Korea) were analyzed. All patients received 5040cGy radiation to surgical stump, mediastinum and ipsilateral supraclavicular node. NQO1 C609T polymorphisms were examined from a peripheral blood sample in all patients. Three types of NQO1 C609T polymorphisms were designated as C/C, C/T and T/T. Chest computed tomography was routinely checked after radiation therapy at every 3 or 6 months and locoregional tumor control, distant metastasis and survival by NQO1 C609T genotype were analyzed. RESULTS: The proportion of NQO1 C609T polymorphisms was 27.6% for C/C, 53.4% for C/T and 19.0% for T/T. Most patients were men and had squamous cell carcinoma or adenocarcinoma. Median age of patients was 61 years. Major failure pattern was distant metastasis and 13 (11.2%) patients showed locoregional recurrence within the field of previous radiation therapy. Crude locoregional recurrence rate was significantly different by NQO1 genotype; 6.3% in C/C, 8.1% in C/T, and 27.3% in T/T (p=0.03), but distant metastasis was not different. Median follow-up time was 49.2 months (range: 3.4-103.5 months). Locoregional recurrence-free survival (LRRFS) was affected in T/T genotype compared with C/C or C/T genotype (p=0.01, Kaplan-Meier method), but distant metastasis-free survival (DMFS) or overall survival (OS) was not different by NQO1 genotype in univariate analysis. NQO1 genotype was also a significant factor for LRRFS (p=0.01) in multivariate analysis. Radiation-induced pneumonitis or esophagitis was tolerable in all patients and the difference by NQO1 genotype was not observed. CONCLUSIONS: NQO1 C609T polymorphisms could be a predictive factor for the locoregional tumor control after postoperative radiation therapy in completely resected NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/terapia , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/terapia , NAD(P)H Desidrogenase (Quinona)/genética , Pneumonectomia , Polimorfismo Genético , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/fisiopatologia , Análise Mutacional de DNA , Intervalo Livre de Doença , Feminino , Estudos de Associação Genética , Genótipo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/fisiopatologia , Masculino , NAD(P)H Desidrogenase (Quinona)/metabolismo , Metástase Neoplásica , Recidiva Local de Neoplasia , Cuidados Pós-Operatórios , Prognóstico , Radiografia Torácica , Radioterapia Adjuvante , República da Coreia , Resultado do Tratamento
5.
Int J Hyperthermia ; 25(6): 477-87, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19657853

RESUMO

NAD(P)H:quinone oxidoreductase (NQO1) mediates cell death caused by the novel anti-cancer drug beta-lapachone (beta-lap). Therefore, beta-lap sensitivity of cells is positively related to the level of cellular NQO1. Heat shock up-regulates NQO1 expression in cancer cells, thereby enhancing the clonogenic cell death caused by beta-lap. The mechanisms by which heat shock elevates NQO1 expression were investigated in the present study using human A549 lung cancer cells and human MDA-MB-231 breast cancer cells. When MDA-MB-231(NQO1+) cells stably transfected with NQO1 were heated at 42 degrees C for 1 h the expression of NQO1 and the sensitivity of the cells to beta-lap progressively increased during the 24-48 h post-heating period. Heating increased NQO1 transcription by cis-acting elements such as xenobiotic response element and antioxidant response element located in the NQO1 gene promoter region. The turnover of NQO1 protein in heated cells was much slower than in unheated cells. NQO1 and heat shock protein 70 (Hsp70) co-precipitated and co-localised in cells before and after heating, demonstrating the close association of these two proteins in the cells. These results suggest that NQO1 is stabilised by the Hsp70 molecular chaperone. It is concluded that the prolonged increase in NQO1 expression after heat shock is due to increased NQO1 transcription, and also increased Hsp70-mediated NQO1 stabilisation.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Proteínas de Choque Térmico HSP70/fisiologia , Resposta ao Choque Térmico , NAD(P)H Desidrogenase (Quinona)/biossíntese , Naftoquinonas/farmacologia , Morte Celular , Linhagem Celular Tumoral , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Hipertermia Induzida
6.
J Control Release ; 140(1): 55-60, 2009 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-19616596

RESUMO

Immunoliposomes directed by monoclonal antibodies are promising vehicles for tumor targeted drug delivery. Development of a long-circulating formulation of pH-sensitive liposomes (PSLs) with epidermal growth factor receptor (EGFR) antibody attached was designed and tested using A549 cells and BALB/c-nu/nu mouse tumor model. PSL formulation was prepared using small unilamellar vesicles of DOPE and CHEMS (6:4 molar ratio) by REV method. The average size and zeta-potential of the formulation measured by dynamic laser-light scattering were approximately 146+/-43.9 nm (PDI=0.09+/-0.02) and -1.77+/-0.03 mV, respectively. A549 cells were xenotransplanted into BALB/c-nu/nu mice and various formulations of gemcitabine (gem), such as in its free form, PSLs or Ab-PSLs, were injected intravenously via a tail vein. The rate of tumor volume increment in Ab-PSLs with gem-treated group was remarkably slower than that of other drug-treated group. The tumor from Ab-PSLs with gem 160 mg/kg-injected group exhibited a markedly lowest account of PCNA labeled cells and had highest TUNEL-positive cells among tested. This suggests that treatment of Ab-PSLs with gem resulted in an increased apoptosis of tumor cells, leading to tumor growth inhibition. These results demonstrate that PSLs provide an efficient and targeted delivery of gemcitabine and may represent a useful new treatment approach for tumors which overexpress the EGFR.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Desoxicitidina/análogos & derivados , Receptores ErbB/metabolismo , Lipossomos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Receptores ErbB/imunologia , Humanos , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas/métodos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Tamanho da Partícula , Antígeno Nuclear de Célula em Proliferação/metabolismo , Distribuição Aleatória , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Gencitabina
7.
J Control Release ; 139(3): 239-45, 2009 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-19619590

RESUMO

Effective delivery of radiosensitizer to target tumor cells, causing preferentially increased tumor cytotoxicity, while simultaneously minimizing damage to healthy cells around the tumor, is an ideal strategy for the improvement of radiotherapeutic efficacy against human cancer. We aimed to enhance radiotherapeutic efficacy by using biocompatible gold nanoparticles (AuNP) as a vehicle for systemic delivery of ss-lapachone (lap). Lap is a novel anticancer agent displaying potent cytotoxicity against cancer cells expressing NAD(P)H:quinone oxidoreductase-1 enzyme (NQO1). Although lap is expected to be a very promising radiosensitizer, its poor solubility and non-specific distribution obstruct preclinical evaluation and clinical application. In this study, the property of AuNPs carrying lap (AuNPs/lap) for active-targeting tumor cells and improving in vivo radiotherapeutic efficacy was evaluated. Murine monoclonal anti-EGFR antibody was conjugated to the AuNPs/lap as a ligand for active targeting. The active tumor-targeting property of AuNPs/lap conjugating anti-EGFR antibody was validated in vitro experiments using cell lines expressing EGFR at different levels. In mice bearing xenograft human tumors, the intravenous injection of AuNPs/lap exhibited highly enhanced radiotherapeutic efficacy. AuNPs/lap offers a new modality for improvement of radiotherapeutic efficacy and feasibility of further clinical application for human cancer treatment.


Assuntos
Portadores de Fármacos , Ouro/química , Neoplasias Pulmonares/radioterapia , Nanopartículas Metálicas , Naftoquinonas/administração & dosagem , Radiossensibilizantes/administração & dosagem , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Transporte Biológico , Linhagem Celular Tumoral , Sobrevivência Celular , Química Farmacêutica , Composição de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Receptores ErbB/metabolismo , Humanos , Injeções Intravenosas , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Naftoquinonas/química , Radiossensibilizantes/química , Reprodutibilidade dos Testes , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Int J Hyperthermia ; 24(2): 161-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18283592

RESUMO

PURPOSE: Bio-reduction/activation of anti-cancer drug beta-lapachone (beta-lap) is mediated by NAD(P)H: Quinone oxidoreductase (NQO1). We investigated the feasibility of using mild temperature hyperthermia to increase the anti-cancer effect of beta-lap by up-regulating NQO1 expression. METHODS: NQO1 expression in FSaII fibrosarcoma of C3H mice and A549 human lung cancer cells was evaluated with Western blot analysis and immunostaining of cells at different times after water-bath heating. Clonogenic cell survival method was used to determine the sensitivity of cells to heating, beta-lap, and in combination. The growth of FSaII tumors in the right hind legs of C3H mice was studied after heating the tumors at 42 degrees C for 1 h with water bath, an i.p. injection of beta-lap to host mice or an i.p. injection of beta-lap 24 h after heating the tumors. RESULTS: Heating at 42 degrees C for 1 h significantly increased the expression of NQO1 in the cancer cells with a maximum increase occurring 8-24 h after heating. The sensitivity of cancer cells to beta-lap treatment progressively increased until 24 h after heating most likely due to the increase in NQO1 expression. Heating the FSaII tumors at 42 degrees C for 1 h and treating the host mice with an i.p. injection of 50 mg/kg beta-lap 24 h after the tumor heating was far more effective than heating alone or beta-lap treatment alone to suppress the tumor growth. CONCLUSION: Mild temperature heat shock elevates the NQO1 expression in cancer cells, which in turn markedly increases the sensitivity of the cells to the bioreductive drug beta-lap in vitro and in vivo.


Assuntos
Antineoplásicos/farmacologia , Hipertermia Induzida/métodos , NAD(P)H Desidrogenase (Quinona)/metabolismo , NADPH Desidrogenase/metabolismo , Naftoquinonas/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/patologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Terapia Neoadjuvante/métodos , Temperatura
9.
Clin Cancer Res ; 11(24 Pt 1): 8866-71, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16361576

RESUMO

PURPOSE: The purpose of the present study was to evaluate the efficacy of mild hyperthermia to potentiate the anticancer effects of beta-lapachone (3,4-dihydro-2,2-dimethyl-2H-naphthol[1,2-b]pyran-5,6-dione) by up-regulating NAD(P)H:quinone oxidoreductase (NQO1) in cancer cells. EXPERIMENTAL DESIGN: Effects of beta-lapachone alone or in combination with mild heating on the clonogenic survival of FSaII fibrosarcoma cells of C3H mice and A549 human lung tumor cells in vitro was determined. Effects of heating on the NQO1 level in the cancer cells in vitro were assessed using Western blot analysis for NQO1 expression, biochemical determination of NQO1 activity, and immunofluorescence microscopy for NQO1 expression. Growth of FSaII tumors in the hind legs of C3H mice was determined after treating the host mice with i.p. injection of 45 mg/kg beta-lapachone followed by heating the tumors at 42 degrees C for 1 hour every other day for four times. RESULTS: Incubation of FSaII tumor cells and A549 tumor cells with beta-lapachone at 37 degrees C reduced clonogenic survival of the cells in dose-dependent and incubation time-dependent manner. NQO1 level in the cancer cells in vitro increased within 1 hour after heating at 42 degrees C for 1 hour and remained elevated for >72 hours. The clonogenic cell death caused by beta-lapachone increased in parallel with the increase in NQO1 levels in heated cells. Heating FSaII tumors in the legs of C3H mice enhanced the effect of i.p.-injected beta-lapachone in suppressing tumor growth. CONCLUSION: We observed for the first time that mild heat shock up-regulates NQO1 in tumor cells. The heat-induced up-regulation of NQO1 enhanced the anticancer effects of beta-lapachone in vitro and in vivo.


Assuntos
Antineoplásicos/uso terapêutico , Hipertermia Induzida , NAD(P)H Desidrogenase (Quinona)/metabolismo , Naftoquinonas/uso terapêutico , Neoplasias/terapia , Animais , Morte Celular , Linhagem Celular Tumoral , Terapia Combinada , Dicumarol/uso terapêutico , Inibidores Enzimáticos/farmacologia , Humanos , Camundongos , NAD(P)H Desidrogenase (Quinona)/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Regulação para Cima
10.
Int J Radiat Oncol Biol Phys ; 57(3): 813-9, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14529788

RESUMO

PURPOSE: Perillyl alcohol (POH) (4-isopropenyl-cyclohexenecarbinol) is a member of the monoterpenes, which are present in various fruits and vegetables. POH has been demonstrated to be cytotoxic against a variety of experimental cancer cells in vitro and in vivo. Phase I clinical trials have indicated that POH may be useful for human tumor treatment. The purpose of our study was to reveal whether the anticancer effect of POH could be enhanced by hyperthermia. METHODS AND MATERIALS: SCK mammary carcinoma cells of A/J mice were used. The effects of POH or hyperthermia alone were studied by incubating the cells during exponential growth phase in culture with 0.25-1.0 mM of POH at 37 degrees C for varying lengths of time or heating cells at 41-43 degrees C for varying lengths of time. The combined effect of POH and hyperthermia was investigated by heating the cells with 1 mM of POH at 41-43 degrees C for varying lengths of time. The effects of the treatments were evaluated using the clonogenic cell survival assay and three types of apoptosis assays. RESULTS: An incubation of SCK cells with 1 mM of POH at 37 degrees C for 60 min or hyperthermia at 43 degrees C for 1 h decreased clonogenic cell survival to 40% and 60%, respectively. When the cells were heated at 43 degrees C for 1 h in the presence of 1 mM of POH, clonogenic cell survival decreased to 0.2%, indicating that hyperthermia potentiated the effect of POH to cause clonogenic cell death. Hyperthermia also markedly increased the degree of POH-induced apoptosis. CONCLUSION: Hyperthermia synergistically potentiates the cytotoxicity of naturally occurring POH against cancer cells.


Assuntos
Antineoplásicos/uso terapêutico , Hipertermia Induzida , Neoplasias Mamárias Experimentais/terapia , Monoterpenos/uso terapêutico , Animais , Apoptose , Terapia Combinada , Sinergismo Farmacológico , Feminino , Marcação In Situ das Extremidades Cortadas , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos A , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA