Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
ACS Appl Bio Mater ; 7(1): 154-167, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38088856

RESUMEN

The drug delivery system for transporting anticancer agents to targeted tissues in the body is a challenging issue. In search of a suitable biocompatible carrier having controlled and sustained drug release properties of poorly soluble drugs, carbon nano-onions (CNOs) were loaded with an anticancer drug, bis-chloroethyl nitrosourea (BCNU/carmustine). CNOs being autofluorescent, drug-loaded functionalized CNOs (f-CNO-BCNU) can be detected in vivo. Transmission electron microscopy (TEM) and differential light scattering (DLS) techniques were used to analyze the sizes of these f-CNOs. The molecular study revealed that the f-CNO-BCNU readily and noncovalently binds with the folate receptors present on the cancer cell surface in excess. Computer modeling and molecular dynamics simulation followed by binding free energy calculation shows f-CNOs have -29.9 kcal/mol binding free energy, and it noncovalently binds the receptor FRα using loop dynamics of three essential loops present in the protein along with polar stabilization interactions provided by Asp55 and Glu86 residues present in the active site. The f-CNO effectively decreased cancer cell viability with a low IC50 value (the concentration that led to 50% killing of the cells). The cell-based Franz diffusion assay was performed to study the drug release profile. The f-CNO-BCNUs also decreased the mitochondrial membrane potential of U87 cells, increased reactive oxygen species release, and caused a loss of mitochondrial membrane integrity. The f-CNOs also increased the percentage of apoptotic cells observed by the Annexin V assay. Based on observed results, it can be concluded that the f-CNO-BCNU efficiently targets the cancer cells, enhances the bioavailability of carmustine, and can be used as a smart chemotherapeutic agent. This strategy offers better patient compliance and greater bioavailability of the drug.


Asunto(s)
Antineoplásicos , Glioblastoma , Humanos , Carmustina/farmacología , Carmustina/química , Glioblastoma/tratamiento farmacológico , Carbono/química , Preparaciones Farmacéuticas , Cebollas , Sistemas de Liberación de Medicamentos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
2.
J Nanosci Nanotechnol ; 21(12): 6196-6204, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34229821

RESUMEN

This study aimed to investigate the effects of DMSO@γ-Fe2O3 nanomagnetic fluid thermotherapy combined with the chemotherapy drug carmustine on cervical cancer cells under a certain intensity of alternating magnetic field. And the role of Mir-590-3P in the development and progression of cervical cancer. The optimal thermotherapy concentration of γ-Fe2O3 nanomaterials on cervical cancer cells was determined by in vitro heating. In addition, the MTT colorimetric method was used to evaluate the toxic effect of γ-Fe2O3 magnetic nanoparticles on cervical cancer cells, and the optimal therapeutic concentration of carbachol on cervical cancer cells was optimized (0.015 g · L-1). The cervical cancer cells were divided into control, γ-Fe2O3 hyperthermia, chemotherapy, and DMSO@γ-Fe2O3 combined chemotherapy groups. After 2 h exposure to hypothermic conditions, flow cytometry was used to assess cell apoptosis for each group. The heating effect of the γ-Fe2O3 magnetic nanomaterials was apparent. When the concentration of γ-Fe2O3 was ≥6 g· L-1, the temperature rise above 41 °C. γ-Fe2O3 is non-toxic to cervical cancer cells and has good biocompatibility. Taking the drug concentration of IC25 as the working concentration of this study, the working concentration of carmustine was 0.015 g · L-1. Both the 41 °C heat treatment and chemotherapy alone had a killing effect on glioma and cervical cancer cells (P < 0.05). Additionally, the combined inhibitory effect of DMSO@γ-Fe2O3 nanomagnetic fluid thermotherapy and drugs at this temperature was significantly stronger than that of thermotherapy and chemotherapy alone (P < 0.05). For the control, gamma-Fe2O3 hyperthermia, chemotherapy, and DMSO@γ-Fe2O3 combined chemotherapy groups, the apoptosis rates of the cervical cancer cells were 1.4%, 18.6%, 24.12%, and 38.97%, respectively. DMSO@γ-Fe2O3 nanomagnetic fluid thermotherapy combined with the chemotherapeutic drug carmustine exerted a noticeable toxic effect on the cervical cancer cells, and DMSO@γ-Fe2O3 significantly enhanced the killing effect of carmustine on cervical cancer cells.


Asunto(s)
Hipertermia Inducida , MicroARNs , Neoplasias del Cuello Uterino , Carmustina/farmacología , Dimetilsulfóxido/farmacología , Femenino , Compuestos Férricos , Humanos , Nanopartículas Magnéticas de Óxido de Hierro
3.
Biosci Rep ; 38(4)2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-29789400

RESUMEN

Carnosic acid (CA), a major polyphenolic diterpene present in Rosmarinus officinalis, has been reported to have multiple functions, including antitumor activity. The MTT assay, BrdU incorporation, wound healing, and colony formation were used to detect melanoma B16F10 cell growth and proliferation. Flow cytometry was used for cell cycle detection. p21 and p27 expression was detected by Western blotting. B16F10 cell xenograft model was established, and treated with CA, carmustine (BCNU), or lomustine (CCNU). The present study found that CA exhibits significant growth inhibition and cell cycle arrest in melanoma B16F10 cells. We also found that CA triggers cell cycle arrest at G0/G1 phase, and enhances p21 expression. Additionally, CA can enhance BCNU- and CCNU-mediated cytotoxicity and cell cycle arrest in B16F10 cells. Finally, we found that CA inhibits tumor growth, and reduces the values of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in vivo The present study study concluded that CA may be safe and useful as a novel chemotherapeutic agent.


Asunto(s)
Abietanos/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Antioxidantes/uso terapéutico , Carmustina/uso terapéutico , Lomustina/uso terapéutico , Melanoma Experimental/tratamiento farmacológico , Abietanos/farmacología , Animales , Antineoplásicos Alquilantes/farmacología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Carmustina/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Lomustina/farmacología , Masculino , Melanoma Experimental/patología , Ratones Endogámicos C57BL
4.
Biomaterials ; 32(27): 6523-32, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21645920

RESUMEN

The successful delivery of anti-cancer drugs relies on the simultaneous capability to actively target a specific location, a sufficient lifetime in the active form in the circulation, and traceability and quantification of drug distribution via in vivo medical imaging. Herein, a highly magnetic nanocarrier (HMNC) composed of an Fe(3)O(4) core and an aqueous-stable, self-doped poly[N-(1-one-butyric acid)]aniline (SPAnH) shell was chemically synthesized. This nanocarrier exhibited a high capacity for 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) drug loading. BCNU and o-(2-aminoethyl)polyethylene glycol (EPEG) were covalently immobilized on the surface of the HMNC to form a self-protecting magnetic nanomedicine (i.e., SPMNM) that could simultaneously provide low reticuloendothelial system uptake, high active-targeting, and in vivo magnetic resonance imaging (MRI) traceability. Meanwhile, the SPMNM was found to reduce the phagocytosis by macrophages and reduce the hydrolysis rate of BCNU. The high magnetization (approximately 1.2-fold higher than Resovist) of the HMNC allowed efficient magnetic targeting to the tumor. The synergetic drug delivery approach provided approximately a 3.4-fold improvement of the drug's half-life (from 18 h to 62 h) and significantly prolonged the median survival rate in animals that received a low dose of BCNU, compared with those that received a high dose of free BCNU (63 days for those that received 4.5 mg BCNU/kg carried by the nanocarrier versus 50 days for those that received 13.5 mg of free-BCNU). This improvement could enhance the potential of magnetic targeting therapy in clinical applications of cancer treatments.


Asunto(s)
Carmustina/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Glioma/tratamiento farmacológico , Nanopartículas de Magnetita/química , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carmustina/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Simulación por Computador , Reactivos de Enlaces Cruzados/farmacología , ADN/metabolismo , Portadores de Fármacos/química , Estabilidad de Medicamentos , Semivida , Humanos , Nanopartículas de Magnetita/ultraestructura , Ratones , Imagen Molecular , Sistema Mononuclear Fagocítico/efectos de los fármacos , Temperatura , Resultado del Tratamiento
5.
NMR Biomed ; 24(5): 473-82, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21674650

RESUMEN

Early imaging or blood biomarkers of tumor response is needed to customize anti-tumor therapy on an individual basis. This study evaluates the sensitivity and relevance of five potential MRI biomarkers. Sixty nude rats were implanted with human glioma cells (U-87 MG) and randomized into three groups: one group received an anti-angiogenic treatment (Sorafenib), a second a cytotoxic drug [1,3-bis(2-chloroethyl)-1-nitrosourea, BCNU (Carmustine)] and a third no treatment. The tumor volume, apparent diffusion coefficient (ADC) of water, blood volume fraction (BVf), microvessel diameter (vessel size index, VSI) and vessel wall integrity (contrast enhancement, CE) were monitored before and during treatment. Sorafenib reduced tumor CE as early as 1 day after treatment onset. By 4 days after treatment onset, tumor BVf was reduced and tumor VSI was increased. By 14 days after treatment onset, ADC was increased and the tumor growth rate was reduced. With BCNU, ADC was increased and the tumor growth rate was reduced 14 days after treatment onset. Thus, the estimated MRI parameters were sensitive to treatment at different times after treatment onset and in a treatment-dependent manner. This study suggests that multiparametric MR monitoring could allow the assessment of new anti-tumor drugs and the optimization of combined therapies.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Glioma/tratamiento farmacológico , Glioma/patología , Imagen por Resonancia Magnética/métodos , Inhibidores de la Angiogénesis/farmacología , Animales , Bencenosulfonatos/farmacología , Bencenosulfonatos/uso terapéutico , Volumen Sanguíneo/efectos de los fármacos , Carmustina/farmacología , Carmustina/uso terapéutico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Glioma/irrigación sanguínea , Humanos , Masculino , Microvasos/efectos de los fármacos , Microvasos/patología , Modelos Biológicos , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Piridinas/farmacología , Piridinas/uso terapéutico , Ratas , Ratas Desnudas , Sorafenib , Coloración y Etiquetado , Análisis de Supervivencia
6.
BMC Complement Altern Med ; 6: 27, 2006 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-16914050

RESUMEN

BACKGROUND: Flavonoid-rich extracts from the mature roots of Scutellaria baicalensis have been shown to exhibit antiproliferative effects on various cancer cell lines. We assessed the ability of an ethanolic extract of S. baicalensis root to inhibit the proliferation of malignant glioma cells. METHODS: Cell lines derived from primary and recurrent brain tumors from the same patient and cells selected for resistance to the chemotherapeutic agent 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) were used to identify antiproliferative effects of this extract when used alone and in conjunction with BCNU. RESULTS AND DISCUSSION: Results indicated that Scutellaria baicalensis not only inhibits cellular growth in recurrent and drug resistant brain tumor cell lines, but also demonstrates an increased inhibitory effect when used in conjunction with BCNU. CONCLUSION: The results of this study support the efficacy of S. baicalensis as an anticancer agent for glioblastomas multiforme and a potential adjuvant treatment to current chemotherapeutic agents used in the treatment of both primary and recurrent GBMs. Further studies of the effects of individual flavonoids alone and in combination with each other and with currently used therapies are needed.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Fitoterapia/métodos , Extractos Vegetales/farmacología , Neoplasias Encefálicas/metabolismo , Carmustina/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimioterapia Combinada , Glioma/metabolismo , Humanos , Neuroglía/efectos de los fármacos , Raíces de Plantas , Scutellaria baicalensis , Resultado del Tratamiento , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
7.
Int J Oncol ; 27(4): 1131-40, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16142332

RESUMEN

2-Chloroethyl-3-sarcosinamide-1-nitrosourea (SarCNU) has been used to treat patients with advanced solid tumours. However, the molecular mechanisms are not well understood. In the present study, we report that SarCNU inhibited proliferation of human HK-1 and CNE-2 nasopharyngeal carcinoma (NPC) in vivo and in vitro. In vitro study showed that wild-type p53 HK-1 cells were 3-fold more sensitive to SarCNU than p53 mutant CNE-2 cells. G2/M arrest, reduction in p21(Cip1/Waf1) and inactivation of cellular cdc-2 activity were seen in both SarCNU-treated HK-1 and CNE-2 cells. Upregulation of p53, phosphorylated p53 at Ser15 and biochemical markers for apoptosis, such as cleaved caspase-3, cleaved caspase-7 and cleaved PARP, were observed in SarCNU-treated HK-1 but not CNE-2 cells. The levels of cyclin B1, Wee1 and phosphorylated cdc-2 but not total cdc-2 in HK-1 cells were significantly reduced by SarCNU treatment. In contrast to HK-1 cells, decrease in total cdc-2 but increase in phosphorylated cdc-2 at Tyr15, cyclin B1 and Wee1 was observed in CNE-2 cells treated with SarCNU. Introduction of mutant p53 into HK-1 cells resulted in growth enhancement in vivo and increased resistance to SarCNU-induced apoptosis in vitro. Furthermore, CNE-2 cells transfected with wild-type p53 became susceptible to SarCNU-induced apoptosis in vitro but not their growth rate in vivo. The data indicate that in NPC cells SarCNU-induced apoptosis was p53-dependent while SarCNU-induced G2/M arrest was mediated by altering the levels of cyclin B1-cdc-2 complex and phosphorylation of cdc-2 at Tyr15 resulting in inactivation of cellular cdc-2 activity. Our data suggest a potential use of SarCNU in the treatment of NPC.


Asunto(s)
Carcinoma/patología , Carmustina/análogos & derivados , Neoplasias Nasofaríngeas/patología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis , Western Blotting , Proteína Quinasa CDC2/metabolismo , Carmustina/farmacología , Caspasa 3 , Caspasa 7 , Caspasas/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , División Celular , Proliferación Celular , Supervivencia Celular , Ciclina B/metabolismo , Ciclina B1 , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Fase G2 , Genes p53 , Terapia Genética/métodos , Humanos , Inmunoprecipitación , Técnicas In Vitro , Masculino , Ratones , Ratones SCID , Mitosis , Mutación , Proteínas Nucleares/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Factores de Tiempo , Transfección , Tirosina/química , Regulación hacia Arriba
8.
J Gene Med ; 5(9): 737-47, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12950064

RESUMEN

BACKGROUND: Erythropoietic protoporphyria (EPP) is an inherited disease characterised by a ferrochelatase (FECH) deficiency, the latest enzyme of the heme biosynthetic pathway, leading to the accumulation of toxic protoporphyrin in the liver, bone marrow and spleen. We have previously shown that a successful gene therapy of a murine model of the disease was possible with lentiviral vectors even in the absence of preselection of corrected cells, but lethal irradiation of the recipient was necessary to obtain an efficient bone marrow engraftment. To overcome a preconditioning regimen, a selective growth advantage has to be conferred to the corrected cells. METHODS: We have developed a novel bicistronic lentiviral vector that contains the human alkylating drug resistance mutant O(6)-methylguanine DNA methyltransferase (MGMT G156A) and FECH cDNAs. We tested their capacity to protect hematopoietic cell lines efficiently from alkylating drug toxicity and correct enzymatic deficiency. RESULTS: EPP lymphoblastoid (LB) cell lines, K562 and cord-blood-derived CD34(+) cells were transduced at a low multiplicity of infection (MOI) with the bicistronic constructs. Resistance to O(6)-benzylguanine (BG)/N,N'-bis(2-chloroethyl)-N-nitrosourea (BCNU) was clearly shown in transduced cells, leading to the survival and expansion of provirus-containing cells. Corrected EPP LB cells were selectively amplified, leading to complete restoration of enzymatic activity and the absence of protoporphyrin accumulation. CONCLUSIONS: This study demonstrates that a lentiviral vector including therapeutic and G156A MGMT genes followed by BG/BCNU exposure can lead to a full metabolic correction of deficient cells. This vector might form the basis of new EPP mouse gene therapy protocols without a preconditioning regimen followed by in vivo selection of corrected hematopoietic stem cells.


Asunto(s)
Terapia Genética , Vectores Genéticos , Lentivirus/genética , O(6)-Metilguanina-ADN Metiltransferasa/genética , Porfiria Hepatoeritropoyética/terapia , Animales , Antígenos CD34/inmunología , Antineoplásicos/farmacología , Carmustina/farmacología , Línea Celular , ADN Complementario/genética , ADN Complementario/metabolismo , Resistencia a Antineoplásicos , Ferroquelatasa/genética , Ferroquelatasa/metabolismo , Regulación de la Expresión Génica , Regulación Viral de la Expresión Génica , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Mutación Puntual , Porfiria Hepatoeritropoyética/genética , Porfiria Hepatoeritropoyética/metabolismo , Regiones Promotoras Genéticas , Linfocitos T/inmunología , Factores de Tiempo , Transgenes
9.
J Altern Complement Med ; 8(5): 573-80, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12470438

RESUMEN

OBJECTIVE: To improve the poor efficacy (< 10%) of chemotherapy for patients with hormone-refractory prostate cancer, we investigated a possible cytotoxic effect of carmustine/beta-glucan combination on prostatic cancer PC-3 cells, focusing on a glutathione-dependent detoxifying enzyme, glyoxalase I (Gly-I). METHODS: Carmustine (BCNU) is an anticancer agent and a putative inhibitor of Gly-I, while beta-glucan is a unique, nontoxic polysaccharide extracted from maitake mushrooms. The cytotoxic effects of BCNU or other anticancer agents with beta-glucan on PC-3 cells were assessed by cell-viability testing and Gly-I activity was measured using the spectrophotometric method. RESULTS: BCNU, 5-fluorouracil (5-FU), and methotrexate (MTX) were capable of inducing approximately a 50% reduction in cell viability at 72 hours, while etoposide, cisplatin, and mitomycin C were all ineffective. Only the combination of BCNU (50 micro ;mol) and beta-glucan (60 micro g/mL) exhibited an enhanced cytotoxicity with an approximate 90% cell viability reduction, but little improvement was seen with any combinations of 5-FU, MTX, or beta-glucon. Gly-I assays revealed that such a profound (approximately 90%) cell death was accompanied by an approximate 80% reduction in Gly-I activity by 6 hours. CONCLUSION: This study demonstrates a sensitized cytotoxic effect of BCNU with beta-glucan in PC-3 cells, which was associated with a drastic (approximately 80%) inactivation of Gly-I. Therefore, the BCNU/beta-glucan combination may help to improve current treatment efficacy by targeting Gly-I, which appears to be critically involved in prostate cancer viability.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Carmustina/farmacología , Glucanos/farmacología , Lactoilglutatión Liasa/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/enzimología , beta-Glucanos , Agaricales , Andrógenos/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Fluorouracilo/farmacología , Humanos , Técnicas In Vitro , Lactoilglutatión Liasa/efectos de los fármacos , Masculino , Metotrexato/farmacología , Neoplasias de la Próstata/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
10.
Acta Oncol ; 40(4): 529-35, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11504314

RESUMEN

Increased expression of gamma-glutamyltransferase (GGT) has been detected in a range of human malignancies and is thought to be involved in neoplastic proliferation and treatment resistance. Since GGT expression and its role in malignant glioma biology remain largely unknown, we investigated this phenomenon by immunostaining 26 higher-grade human astrocytic gliomas (WHO grades III and IV) with a monoclonal anti-GGT-antibody (138H11). Further, human pancreatic GGT cDNA was used for liposome-mediated transfection of 9L gliosarcoma cells. GGT-expressing and control 9L cells were cultured in media containing different amounts of essential amino acids and/or cytotoxic agents. Cell viability was evaluated by microplate MTT assay. Immunohistochemical staining of tumor specimens demonstrated that GGT expression is a frequent feature of higher-grade human astrocytic gliomas, but not of normal brain tissue. Human tumors were strongly GGT-positive in 6 of 7 cases of grade III astrocytoma, and in 12 of 19 grade IV astrocytoma (glioblastoma multiforme, GBM) cases. In the cell culture model, 9L-GGT cells had a growth advantage over control cells in cysteine-deficient medium. but not in standard or glutamine-free medium. No significant difference in numbers of viable cells of either clone was found in media containing the alkylating drug BCNU (5-200 microg/ml). In conclusion, GGT is expressed in a high percentage of human WHO grade III astrocytomas and GBM, but not in normal brain tissue. This molecule seems to give neoplastic cells a moderate growth advantage under in vivo conditions.


Asunto(s)
Glioblastoma/enzimología , Proteínas de Neoplasias/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , gamma-Glutamiltransferasa/biosíntesis , Adulto , Anciano , Animales , Anticuerpos Monoclonales/inmunología , Antineoplásicos Alquilantes/farmacología , Encéfalo/enzimología , Carmustina/farmacología , División Celular , Células Clonales/efectos de los fármacos , Células Clonales/metabolismo , Células Clonales/patología , Medios de Cultivo/farmacología , Cisteína/farmacología , ADN Complementario/genética , Resistencia a Antineoplásicos , Inducción Enzimática , Femenino , Glioblastoma/genética , Gliosarcoma/patología , Glutamina/farmacología , Humanos , Liposomas , Masculino , Microscopía Inmunoelectrónica , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Ratas , Proteínas Recombinantes de Fusión/biosíntesis , Transfección , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/enzimología , gamma-Glutamiltransferasa/genética , gamma-Glutamiltransferasa/inmunología
11.
Free Radic Biol Med ; 30(9): 1008-18, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11316581

RESUMEN

Apoptosis was studied under conditions that mimic the steady state of H(2)O(2) in vivo. This is at variance with previous studies involving a bolus addition of H(2)O(2), a procedure that disrupts the cellular homeostasis. The results allowed us to define three phases for H(2)O(2)-induced apoptosis in Jurkat T-cells with reference to cytosolic steady state concentrations of H(2)O(2) [(H(2)O(2))(ss)]: (H(2)O(2))(ss) values below 0.7 microM elicited no effects; (H(2)O(2))(ss) approximately 0.7-3 microM induced apoptosis; and (H(2)O(2))(ss) > 3 microM yielded no additional apoptosis and a gradual shift towards necrosis as the mode of cell death were observed. H(2)O(2)-induced apoptosis was not affected by either BCNU, an inhibitor of glutathione reductase, or diamide, a compound that reacts both with low-molecular weight and protein thiols, or selenols. Glutathione depletion, accomplished by incubating cells either with buthionine sulfoximine or in cystine-free medium, rendered cells more sensitive to H(2)O(2)-induced apoptosis, but did not change the threshold and saturating concentrations of H(2)O(2) that induced apoptosis. Two unrelated metal chelators, desferrioxamine and dipyridyl, strongly protected against H(2)O(2)-induced apoptosis. It may be concluded that, under conditions of H(2)O(2) delivery that mimic in vivo situations, the oxidative event that triggers the induction of apoptosis by H(2)O(2) is a Fenton-type reaction and is independent of the thiol or selenium states of the cell.


Asunto(s)
Apoptosis/fisiología , Peróxido de Hidrógeno/metabolismo , Compuestos de Sulfhidrilo/metabolismo , 2,2'-Dipiridil/farmacología , Apoptosis/efectos de los fármacos , Carmustina/farmacología , Quelantes/farmacología , Deferoxamina/farmacología , Diamida/farmacología , Inhibidores Enzimáticos/farmacología , Radicales Libres/metabolismo , Glutatión/metabolismo , Glutatión Reductasa/antagonistas & inhibidores , Humanos , Células Jurkat , Necrosis , Oxidación-Reducción , Selenio/metabolismo
12.
Cancer Res ; 61(5): 2002-7, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11280759

RESUMEN

The effects of chemotherapy [25 mg/kg 1,3-bis(2-chloroethyl)-1-nitrosourea administered with a single i.p. injection] on cellular energetics by 31P nuclear magnetic resonance (NMR) spectroscopy, total tissue sodium by single-quantum (SQ) 23Na NMR spectroscopy, and intracellular sodium by triple-quantum-filtered (TQF) 23Na NMR spectroscopy were studied in the s.c. 9L glioma. Animals were studied by NMR 2 days before therapy and 1 and 5 days after therapy. Destructive chemical analysis was also performed 5 days after therapy to validate the origin of changes in SQ and TQF 23Na signals. One day after treatment, there was no significant difference between control and treated tumors in terms of tumor size or 23Na and 31P spectral data. Five days after therapy, treated tumors had 28 +/- 16% (P < 0.1) lower SQ 23Na signal intensity, 46 +/- 20% (P < 0.05) lower TQF 23Na signal intensity, 125 +/- 51% (P < 0.05) higher ATP:Pi ratio, 186 +/- 69% (P < 0.05) higher phosphocreatine:Pi ratio, and 0.17 +/- 0.06 pH units (P < 0.05) higher intracellular pH compared with control tumors. No significant differences in TQF 23Na relaxation times were seen between control and treated tumors at any time point. Destructive chemical analysis showed that the relative extracellular space of control and treated tumors was identical, but the treated tumors had 21 +/- 8% (P < 0.05) lower total tissue Na+ concentration and 60 +/- 24% (P < 0.05) lower intracellular Na+ concentration compared with the controls. The higher phosphocreatine:Pi and ATP:Pi ratios after 1,3-bis(2-chloroethyl)-1-nitrosourea treatment indicate improved bioenergetic status in the surviving tumor cells. The decrease in SQ and multiple-quantum-filtered 23Na signal intensity was largely attributable to a decrease in Na(i)+ because the treatment did not change the relative extracellular space. The improved energy metabolism could decrease the intracellular concentration of Na+ by increasing the activity of Na+-K+-ATPase and decreasing the activity of Na+/H+. Although both 23Na and 31P spectra were consistent with improved cellular metabolism in treated tumors, the 23Na methods may be better suited for monitoring response to therapy because of higher signal:noise ratio and ease of imaging the single 23Na resonance.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/metabolismo , Carmustina/farmacología , Glioma/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Fósforo/metabolismo , Sodio/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Concentración de Iones de Hidrógeno , Masculino , Trasplante de Neoplasias , Fosfocreatina/metabolismo , Ratas , Ratas Endogámicas F344
13.
Invest New Drugs ; 15(1): 39-48, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9195288

RESUMEN

TNP-740, minocycline, suramin and genistein have demonstrated antiangiogenic activity in various experimental systems. The effect of these agents alone and in two agent combinations on the number of intratumoral vessels and response to cytotoxic anticancer therapies was assessed in animals bearing the Lewis lung carcinoma. Treatment with each of the antiangiogenic agents alone and in two agent combinations decreased the number of intratumoral vessels visualized by CD31 or Factor VIII staining to 30% to 50% of the number in the untreated control tumors. In general, the antiangiogenic agents are more effective adjuvants to cytotoxic therapies when used as two agent combinations than as single agents. The most effective antiangiogenic combinations were: TNP-470/minocycline > TNP-470/genistein > TNP-470/suramin. The increases in the response of the primary tumor to cyclophosphamide, adriamycin, CDDP, BCNU, x-rays or 5-fluorouracil and the lung metastases occur to about the same level with the addition of antiangiogenic agents to the therapies. With the treatment combination TNP-470/minocycline/cyclophosphamide 40% of the animals were cured. The results of these studies indicate that antiangiogenic agents can be very useful additions to treatment regimens for solid tumors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/uso terapéutico , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/patología , Carmustina/farmacología , Carmustina/uso terapéutico , División Celular/efectos de los fármacos , Cisplatino/farmacología , Cisplatino/uso terapéutico , Ciclohexanos , Ciclofosfamida/farmacología , Ciclofosfamida/uso terapéutico , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Sinergismo Farmacológico , Genisteína , Isoflavonas/farmacología , Isoflavonas/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos C57BL , Minociclina/farmacología , Minociclina/uso terapéutico , Invasividad Neoplásica , Neovascularización Patológica/radioterapia , O-(Cloroacetilcarbamoil) Fumagilol , Distribución Aleatoria , Sesquiterpenos/farmacología , Sesquiterpenos/uso terapéutico , Suramina/farmacología , Suramina/uso terapéutico
14.
Int J Hyperthermia ; 11(3): 379-88, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7636324

RESUMEN

Chemosensitization by two nitroimidazoles (NIs), metronidazole (METRO) and misonidazole (MISO), of the anti-tumour effect of alkylating agents was studied at three different temperatures: room temperature (RT), 37 and 41.5 degrees C. Three alkylating agents, cyclophosphamide (CY), 1,3 bis(2-chloroethyl)-N-nitrosourea (BCNU) and melphalan (L-PAM) were tested when the tumours reached an average diameter of 4 mm. Tumours were 4th generation isotransplants of a spontaneous fibrosarcoma, FSa-II. Treatment at 37 or 41.5 degrees C was given by immersing the tumour-bearing foot for 60 min in a water bath set at these temperatures. The test agents were injected ip immediately before immersing the foot in the water bath, whereas METRO or MISO (2.5 mmol/kg) was given ip 30 min before the injection of a test agent. Following treatment the tumour growth (TG) time, i.e. the time required for one-half of treated tumours to reach 1000 mm3 after the treatment day, was studied. For CY, MISO was a better sensitizer than METRO at RT and 37 degrees C, but the magnitude of the chemosensitization by MISO and METRO became identical at 41.5 degrees C. Notably, the chemosensitization was substantially enhanced at 41.5 degrees C, whereas neither 41.5 degrees C-heat, NIs or combined NI and heat prolonged the TG time. Although no chemosensitization was observed for BCNU at RT, both METRO and MISO equally enhanced the effect of BCNU at 41.5 degrees C.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Hipoxia de la Célula/efectos de los fármacos , Hipertermia Inducida , Metronidazol/farmacología , Misonidazol/farmacología , Sarcoma Experimental/tratamiento farmacológico , Sarcoma Experimental/terapia , Animales , Carmustina/farmacología , Terapia Combinada , Ciclofosfamida/farmacología , Femenino , Melfalán/farmacología , Ratones , Ratones Endogámicos C3H , Sarcoma Experimental/metabolismo
15.
In Vivo ; 9(1): 11-8, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7669943

RESUMEN

Tissue oxygen tensions were measured in the rat 9L gliosarcoma under conditions of normal air breathing or carbogen breathing and after intravenous administration of a hemoglobin solution with air breathing or carbogen breathing. Administration of the hemoglobin decreased the level of hypoxia in the tumors. Treatment of the animals with the antiangiogenic combination of TNP-470 and minocycline also increased tumor oxygenation compared with untreated controls. Treatment with the antiangiogenic agents along with administration of the hemoglobin solution/carbogen breathing decreased the hypoxic fraction (% pO2 readings < or = 5 mmHg) from 71 % to 30%. Treatment of the tumor-bearing animals with BCNU or adriamycin modestly reduced hypoxia in the tumors, while treatment with fractionated radiation markedly increased hypoxia in the tumors. Tumor growth delay was used to assess the response of the subcutaneous tumor to the various treatment combinations. There was a strong correlation between increased therapeutic response and decreased tumor hypoxia. Tumor growth delay from BCNU increased from 5.3 days to 16.4 days with TNP-470/-minocycline/hemoglobin solution/carbogen. Similarly, the tumor growth delay from adriamycin increased from 3.9 days to 17.0 days with TNP-470/minocycline/hemoglobin solution/carbogen. Finally, the tumor growth delay from fractionated radiation increased from 4.8 days to 13.3 days with TNP-470/minocycline/hemoglobin solution/carbogen. When etanidazole was added to the complete radiation regimen, the tumor growth delay increased further to 20.5 days. These data show that the addition of non-toxic agents that increase tumor oxygenation to cytotoxic therapies can markedly increase therapeutic response.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Gliosarcoma/tratamiento farmacológico , Gliosarcoma/radioterapia , Hemoglobinas/uso terapéutico , Animales , Antibióticos Antineoplásicos/farmacología , Dióxido de Carbono , Carmustina/farmacología , Bovinos , Terapia Combinada , Ciclohexanos , Femenino , Humanos , Minociclina/farmacología , O-(Cloroacetilcarbamoil) Fumagilol , Oxígeno/metabolismo , Fármacos Sensibilizantes a Radiaciones , Ratas , Ratas Endogámicas F344 , Sesquiterpenos/farmacología , Células Tumorales Cultivadas/efectos de los fármacos
16.
Oncol Res ; 7(5): 237-43, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-8534929

RESUMEN

Although the antiangiogenic agent TNP-470 does not, in general, increase the cytotoxicity of anti-cancer therapies in cell culture, the antiangiogenic agents TNP-470 and minocycline individually and especially in combination have been shown to increase the tumor growth delay produced by several standard cytotoxic therapies in the Lewis lung carcinoma. In an effort to understand the mechanism by which the antiangiogenic agent combination TNP-470/minocycline potentiates the antitumor activity of cytotoxic therapeutic agents in vivo, the biodistribution of [14C]-cyclophosphamide and cis-diamminedichloroplatinum(II) was determined 6 h after cytotoxic drug administration in animals bearing Lewis lung carcinoma pretreated with TNP-470/minocycline and in animals without pretreatment. Higher levels of 14C and platinum were found in 9 tissues (including tumor) except blood in animals pretreated with TNP-470/minocycline. The increased drug levels in the tumors may be sufficient to account for the increased tumor growth delays observed previously. DNA alkaline elution of tumors from animals pretreated with TNP-470/minocycline showed increased DNA cross-linking by both cyclophosphamide and cis-diamminedichloroplatinum(II). The possible implications of these results are discussed.


Asunto(s)
Antineoplásicos/farmacocinética , Carcinoma Pulmonar de Lewis/metabolismo , Cisplatino/farmacocinética , Ciclofosfamida/farmacocinética , Minociclina/farmacología , Sesquiterpenos/farmacología , Animales , Antineoplásicos/farmacología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carmustina/farmacología , Hipoxia de la Célula , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Ciclohexanos , Ciclofosfamida/análogos & derivados , Ciclofosfamida/farmacología , ADN de Neoplasias/análisis , ADN de Neoplasias/efectos de los fármacos , Combinación de Medicamentos , Masculino , Melfalán/farmacología , Ratones , Ratones Endogámicos C57BL , O-(Cloroacetilcarbamoil) Fumagilol , Platino (Metal)/análisis , Células Tumorales Cultivadas
17.
J Pharmacol Exp Ther ; 270(3): 858-64, 1994 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7932197

RESUMEN

The dipyridyl herbicide diquat undergoes redox cycling in vivo resulting in superoxide generation. Diquat administration causes hepatic and renal toxicity in rodents. Selenium deficiency worsens this injury and lipid peroxidation is a prominent feature of the toxicity. However, there is limited data regarding the role of lipid peroxidation in diquat-induced toxicity in selenium-adequate animals. In addition, little is known about the effect of glutathione-modulating agents on diquat-induced toxicity and lipid peroxidation in vivo. F2-isoprostanes are novel prostanoids which, both free in plasma and esterified to phospholipids in tissues, are markers of lipid peroxidation in vivo. By using F2-isoprostane quantitation, we examined the effects of selenium deficiency and modulation of glutathione status with 1,3-bis (2-chloroethyl)-1-nitrosourea, phorone or buthionine sulfoximine on diquat-induced toxicity and lipid peroxidation. F2-isoprostanes increased 2- to 9-fold in plasma, liver, kidney and lung in selenium-adequate Fischer 344 rats with liver injury after receiving 100 mumol of diquat per kg. Selenium deficiency or modulation of glutathione status increased diquat toxicity. This was accompanied by 10- to 100-fold increases in plasma and kidney F2-isoprostane levels. Liver F2-isoprostanes were increased 2- to 5-fold. These studies suggest that glutathione, in addition to selenium, is an important defense against diquat-induced toxicity and lipid peroxidation.


Asunto(s)
Diquat/toxicidad , Glutatión/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Selenio/deficiencia , Animales , Antimetabolitos/farmacología , Ácidos Araquidónicos/metabolismo , Butionina Sulfoximina , Carmustina/farmacología , Cetonas/farmacología , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacología , Ratas , Ratas Endogámicas F344
18.
Neurosurgery ; 34(4): 657-64, 1994 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8008163

RESUMEN

Human glioblastoma (U-87MG) and canine glioma (canine brain tumor [CBT]) cell lines were tested in vitro for their therapeutic sensitivity to sequential treatment with differentiating agents and chemotherapy or hyperthermia. Both cell lines responded to the inducer combination dibutyryl adenosine-3',5'-cyclic monophosphate/sodium butyrate by the formation of cytoplasmic processes detectable within 7 hours and attained approximately 90% morphological differentiation within 2 days of exposure. The clonogenicity of CBT and U-87MG cells gradually decreased after 1 to 7 days of exposure to the inducer combination, but this treatment alone failed to kill the cells. After the removal of the inducers, both lines dedifferentiated and the rate of clonogenesis increased. 1,3-bis-(2-Chloroethyl)-1-nitrosourea administered to CBT and U-87MG cells before or after 3 days of treatment with inducers potentiated the antiproliferative effects of the differentiating agents. Cisplatin administered to U-87MG cells enhanced the antiproliferative effect of the differentiating agents to a greater extent when added before the inducers rather than after differentiation was stimulated. The sequential treatment of CBT cells with a 44 degrees C heat pulse for 30 minutes followed by differentiating agents produced an additive potentiation of cell killing, whereas the reverse sequence did not. Hyperthermia pretreatment at 44 degrees C for 15 minutes or at 42 degrees C for 30 minutes failed to enhance the antiproliferative effects of inducing agents.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/patología , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Glioma/patología , Hipertermia Inducida , Células Tumorales Cultivadas/efectos de los fármacos , Animales , Bucladesina/farmacología , Butiratos/farmacología , Ácido Butírico , Carmustina/farmacología , División Celular/efectos de los fármacos , Línea Celular , Cisplatino/farmacología , Perros , Relación Dosis-Respuesta a Droga , Humanos , Técnicas In Vitro , Células Tumorales Cultivadas/patología , Ensayo de Tumor de Célula Madre
19.
Anticancer Res ; 13(5A): 1549-56, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8239534

RESUMEN

Since several anticancer drugs are known to become more cytotoxic to cells in an acidic milieu, we have attempted to utilize the carbonic anhydrase inhibitor, Acetazolamide, to acidify the blood and tumor of C3H mice bearing the FSaIIC fibrosarcoma in order to sensitize tumor cells in vivo to CDDP, Melphalan, BCNU, SR4233 or PtCl4 (Fast Black)2 +/- hyperthermia. The direct cytotoxic interactions between the anticancer drugs and Acetazolamide were tested in FSaIIC cells in vitro with the monacidifying diuretic Chlorothrozide as a control. When cells were exposed to CDDP both diuretics protected against cytotoxicity in a dose dependent fashion. In contrast, cells exposed to Melphalan were minimally sensitized and those exposed to BCNU, SR4233, or PtCl4 (Fast Black)2 were essentially unaffected by the presence of the diuretic agents. Both diuretics were essentially non-toxic to cells in vitro, and, interestingly, both drugs markedly protected cells against hyperthermia under low pH conditions. In vitro, however, Acetzolamide produced a tumor growth delay of 2.3 days alone when given at 10 mg/kg i.p. once (the most effective dose) and produced additive growth delays with CDDP and Melphalan, but probably greater than additive delays with SR4233 and PtCl4 (Fast Black)2. When Acetazolamide was given daily for 5 days starting on the day the anticancer drugs were given once (day 7) essentially no further increase in tumor growth delay of nearly 16 days was observed versus only 4,6 days for the drug alone. When hyperthermia (43 degrees C min.) was delivered locally to the tumor after i.p. injection of the drugs, further growth delays were produced for every drug combination which probably were additive in extent. Blood and urine pH determinations revealed that a pH drop of 1 units occurred in the blood and a pH elevation of 1 to 21 units occurred in the urine 1 hr. after i.p. injection of Acetazolamide. These results indicate that this carbonic anhydrase inhibitor can add to the anticancer activity of the drugs tested. The mechanism may involve its ability to acidify the intratumoral environment, but other mechanisms can not be excluded.


Asunto(s)
Acetazolamida/farmacología , Antineoplásicos/farmacología , Clorotiazida/farmacología , Neoplasias/tratamiento farmacológico , Animales , Carmustina/farmacología , División Celular/efectos de los fármacos , Cisplatino/farmacología , Interacciones Farmacológicas , Ensayos de Selección de Medicamentos Antitumorales , Fibrosarcoma/patología , Concentración de Iones de Hidrógeno , Hipertermia Inducida , Melfalán/farmacología , Ratones , Neoplasias/sangre , Neoplasias/patología , Neoplasias/orina , Compuestos de Platino/farmacología , Temperatura , Tirapazamina , Triazinas/farmacología , Células Tumorales Cultivadas
20.
Biochem Pharmacol ; 42(4): 883-8, 1991 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-1867643

RESUMEN

The effects of diquat-induced redox cycling on the levels of cellular ascorbic acid and alpha-tocopherol were investigated in isolated rat hepatocytes. In untreated hepatocytes, the metabolism of 1 or 2 mM diquat resulted in the depletion of cellular ascorbic acid and glutathione, but not of alpha-tocopherol, in association with the induction of cell death during the experimental period. In 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) pretreated cells, 1 mM diquat induced cell death accompanied by glutathione was rapid (to 9% of controls by 15 min) and cell ascorbate was completely consumed by 2 hr of incubation. In contrast, cellular alpha-tocopherol levels were stable for the first 30 min, but were depleted in association with the onset of lipid peroxidation. Supplementation of 0.1 or 1.0 mM ascorbic acid in the incubation medium delayed the onset of diquat-induced alpha-tocopherol loss, lipid peroxidation and cytotoxicity. When the concentration of exogenous cellular ascorbic acid was consumed to below that of endogenous ascorbic acid, alpha-tocopherol loss and lipid peroxidation were initiated. The results indicate that untreated hepatocytes have an effective multicomponent antioxidant system against diquat-induced oxidative stress. However, when glutathione is depleted from hepatocytes by treatment with BCNU and diquat, ascorbic acid plays a vital role in maintaining cellular alpha-tocopherol levels and survival of the cell.


Asunto(s)
Ácido Ascórbico/metabolismo , Diquat/toxicidad , Hígado/metabolismo , Vitamina E/metabolismo , Animales , Ácido Ascórbico/farmacología , Ácido Ascórbico/fisiología , Carmustina/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Diquat/metabolismo , Glutatión/metabolismo , Hígado/citología , Hígado/efectos de los fármacos , Masculino , Malondialdehído/metabolismo , Oxidación-Reducción , Ratas , Ratas Endogámicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA