Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
J Natl Cancer Inst ; 89(23): 1789-96, 1997 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-9392620

RESUMO

BACKGROUND: Increased intracellular glutathione has long been associated with tumor cell resistance to various cytotoxic agents. An inhibitor of glutathione biosynthesis, L-S,R-buthionine sulfoximine (BSO), has been shown to enhance the cytotoxicity of chemotherapeutic agents in vitro and in vivo. We performed a phase I study of BSO administered with the anticancer drug melphalan to determine the combination's safety/tolerability and to determine clinically whether BSO produced the desired biochemical end point of glutathione depletion (<10% of pretreatment value). METHODS: Twenty-one patients with advanced cancers received an initial 30-minute infusion of BSO totaling 3.0 g/m2 and immediately received a continuous infusion of BSO on one of the following schedules: 1) 0.75 g/m2 per hour for 24 hours (four patients); 2) the same dose rate for 48 hours (four patients); 3) the same dose rate for 72 hours (10 patients); or 4) 1.5 g/m2 per hour for 48 hours (three patients). During week 1, the patients received BSO alone; during weeks 2 or 3, they received BSO plus melphalan (15 mg/m2); thereafter, the patients received BSO plus melphalan every 4 weeks. Glutathione concentrations in peripheral blood lymphocytes were determined for all patients; in 10 patients on three of the administration schedules, these measurements were made in multiple sections from tumor biopsy specimens taken before, during, and after continuous-infusion BSO. RESULTS: Continuous-infusion BSO alone produced minimal toxic effects, although BSO plus melphalan produced occasional severe myelosuppression (grade 4) and frequent low-grade nausea/vomiting (grade 1-2). This treatment also produced consistent, profound glutathione depletion (<10% of pretreatment value). The degree of glutathione depletion in peripheral lymphocytes was considerably less than that observed in tumor sections. CONCLUSIONS: Continuous-infusion BSO is relatively nontoxic and results in depletion of tumor glutathione.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Glutationa/efeitos dos fármacos , Glutationa/metabolismo , Neoplasias/tratamento farmacológico , Adulto , Idoso , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/farmacocinética , Antineoplásicos Alquilantes/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Butionina Sulfoximina/administração & dosagem , Butionina Sulfoximina/farmacocinética , Esquema de Medicação , Feminino , Glutationa/sangue , Humanos , Infusões Intravenosas , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Masculino , Melfalan/administração & dosagem , Pessoa de Meia-Idade , Neoplasias/metabolismo , Resultado do Tratamento
2.
Cancer Res ; 46(6): 2892-7, 1986 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-3698015

RESUMO

The radiation sensitizing agent misonidazole (MISO) was combined with 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU) for the treatment of Mer+ (IMR-90, HeLa, and HeLa-S3) and Mer- (EMT-6/Ro, VA-13, and HeLa-MR) cell lines under hypoxic conditions in vitro. The magnitude of enhancement achieved by the addition of MISO was calculated by comparison with survival curves obtained by treating each cell line with CCNU alone, under hypoxic conditions. As expected, the Mer+ cells were more resistant to CCNU treatment than were their Mer- counterparts. In the presence of 1.0 mM MISO the toxicity of CCNU was enhanced (dose enhancement factor, 1.4-1.6) in all three of the Mer- lines. However, the Mer+ lines were less responsive to chemopotentiation by MISO. The toxicity of CCNU toward two of the Mer+ lines, IMR-90 and HeLa, was not modified by the addition of MISO, while a slight enhancement (dose enhancement factor, 1.2) was observed in the HeLa-S3 line. Similar results were obtained with IMR-90 and VA-13 cells treated by postincubation in which aerobic CCNU treatment was followed by hypoxic exposure to MISO for up to 6 h. While no correlation was observed between Mer status and the hypoxic toxicity of MISO, the data suggest that a relationship might exist between chemopotentiation and MISO sensitivity when each phenotype is considered separately. These observations suggesting that tumor cells of the Mer+ phenotype may be less responsive to MISO chemopotentiation have significant implications for ongoing and planned clinical trials designed to evaluate the potential of chemopotentiation using CCNU and MISO since greater than 75% of human tumors are Mer+.


Assuntos
Lomustina/farmacologia , Metiltransferases/metabolismo , Misonidazol/farmacologia , Neoplasias/tratamento farmacológico , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Células HeLa , Humanos , Camundongos , Misonidazol/metabolismo , O(6)-Metilguanina-DNA Metiltransferase
3.
Cancer Res ; 44(10): 4409-13, 1984 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-6467202

RESUMO

In order to assess the effect of oxygen on chemopotentiation by misonidazole (MISO), EMT-6/Ro tumor cells were exposed in vitro to combinations of CCNU and 1.0 mM MISO in culture medium equilibrated at various oxygen concentrations. The effect of oxygen on MISO cytotoxicity was similarly determined and compared with the relationship obtained for chemosensitization. MISO cytotoxicity and chemopotentiation were both oxygen sensitive, being maximal under anoxic conditions. Furthermore, the pattern of oxygen sensitivity was virtually identical for the two activities. These results suggest that a similar metabolic pathway, i.e., the oxygen-sensitive reduction of MISO to the nitroradical anion by cellular nitroreductases, is involved in the mechanism of both activities. The data further indicate that chemopotentiation can be expressed in cells treated at intermediate oxygen tensions. The implications of these findings with respect to the magnitude of chemopotentiation in vivo and the enhancement of normal tissue damage in animals treated with MISO and chemotherapy agents is discussed.


Assuntos
Neoplasias Mamárias Experimentais/patologia , Misonidazol/toxicidade , Nitroimidazóis/toxicidade , Oxigênio/farmacologia , Aerobiose , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cinética , Lomustina/toxicidade , Camundongos
4.
Cancer Res ; 43(8): 3539-43, 1983 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-6861127

RESUMO

The effect of incubating KHT-iv tumor cells with 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) alone or in simultaneous combination with the radiation sensitizer misonidazole (MISO) was determined for aerobic and hypoxic incubations. Relative to aerobic exposures, the cytotoxicity of BCNU was significantly enhanced when KHT-iv cells were exposed under hypoxic conditions for 1 hr. This effect was apparently related to a trypsin effect and was absent in experiments with cells trypsinized immediately prior to treatment. In contrast to 1-hr exposures, treatment for 4 hr to the same total BCNU exposure doses resulted in equivalent cell killing under aerobic and hypoxic conditions. The addition of 3 mM MISO to the 1-hr treatment protocol did not significantly modify the toxicity of BCNU toward aerobic or hypoxic cultures of KHT-iv cells. However, the cell-killing efficiency of BCNU during a 4-hr hypoxic exposure was significantly enhanced by the addition of MISO doses as small as 0.5 mM. Addition of sensitizer did not modify the 4-hr aerobic toxicity of BCNU. The decay of BCNU was not significantly altered by MISO or by low oxygen tensions, effectively eliminating the role of altered pharmacokinetics in this example of in vitro chemosensitization. These results suggest that prolonged exposure times and hypoxia are prerequisites for the expression of chemosensitization when MISO is combined simultaneously with certain chemotherapeutic agents in vitro. By comparison to the doses of MISO used in preincubation experiments, it was possible to produce enhanced cytotoxicity by relatively small MISO doses.


Assuntos
Carmustina/uso terapêutico , Fibrossarcoma/tratamento farmacológico , Misonidazol/uso terapêutico , Nitroimidazóis/uso terapêutico , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Fibrossarcoma/patologia , Camundongos , Fatores de Tempo
5.
Cancer Res ; 43(10): 4709-13, 1983 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-6883329

RESUMO

In an attempt to evaluate whether the radiation sensitizer misonidazole (MISO) could enhance the responsiveness of chemoresistant tumors, MISO was combined with 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) and/or 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU) for the treatment of BALB/c X DBA/2 F1 (hereafter called CD2F1) and C3H/HeJ mice bearing nitrosourea-resistant L1210/BCNU or KHT/CCR tumors, respectively. To determine whether comparable degrees of enhancement could be achieved in sensitive and resistant tumor lines, the magnitude of chemosensitization produced by treating the resistant tumors with the MISO-nitrosourea combinations was compared to the chemopotentiation produced in similarly treated nitrosourea-sensitive tumor lines from which the resistant lines had been derived. As evidenced by increased cure probabilities, the addition of MISO [5.0 mmol/kg (1.0 mg/g)] significantly potentiated the response of the parental nitrosourea-sensitive L1210/0 tumor to a 20-mg/kg dose of CCNU. When combined with doses of CCNU lower than 20 mg/kg or with BCNU, MISO failed to significantly modify the response of the L1210/0 tumor. Significant chemosensitization also was evident when 2.5- and 1.25-mmol/kg doses of MISO were used in combination with CCNU at 20 mg/kg. The effectiveness of BCNU and CCNU against the nitrosourea-resistant L1210/BCNU tumor was not significantly improved by MISO (5.0 mmol/kg), even when the sensitizer was combined with doses of nitrosoureas approaching 10% lethal dose (60 days) concentrations. In contrast, the effectiveness of CCNU against the parental KHT and resistant KHT/CCR tumors, assessed using a tumor regrowth assay, was equally enhanced by simultaneous MISO treatment. Therefore, one cannot safely predict the extent of enhancement which might result from the addition of MISO to a chemotherapeutic regimen based solely on the responsiveness of the tumor to the chemotherapy drug(s) alone.


Assuntos
Carmustina/uso terapêutico , Lomustina/uso terapêutico , Misonidazol/uso terapêutico , Nitroimidazóis/uso terapêutico , Compostos de Nitrosoureia/uso terapêutico , Animais , Linhagem Celular , Resistência a Medicamentos , Sinergismo Farmacológico , Humanos , Leucemia L1210/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C3H , Sarcoma Experimental/tratamento farmacológico
6.
Cancer Res ; 48(5): 1086-90, 1988 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-3342391

RESUMO

Experiments were designed to determine whether heat treatment could sensitize nitrosourea-resistant human tumor cell lines expressing a repair system (O6-alkylguanine DNA alkyltransferase; Mer+) capable of removing monoadducts from the DNA of treated cells prior to the formation of lethal interstrand cross-links. Effects of temperatures compatible with systemic hyperthermia were of particular interest, and, consequently, the effect of 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU) exposure in vitro for 4 h at 37 degrees C was compared with that for 1 h at 41 degrees C followed by 3 h at 37 degrees C. CCNU toxicity was significantly enhanced by heat treatment in the Mer+ HT-29 human colon carcinoma, and in HeLa-S3 and HeLa-CCL2 cell lines [thermal enhancement factor (ratio of CCNU doses required to reduce cell survival to 0.001 at 37 degrees C and 41 degrees C) = 1.3-1.4]. Pharmacokinetic studies indicated that the effect of heat treatment on CCNU toxicity was not attributable to exposure to increased concentrations of reactive species, nor was the enhancement due to a direct effect of heat and/or drug on alkyltransferase activity. A similar enhancement of CCNU toxicity was also observed in a Mer- line, HeLa-MR (thermal enhancement factor = 1.3). Heat-sequencing experiments clearly demonstrate that heat and CCNU must be administered concurrently. Alkaline elution experiments were designed to examine DNA-DNA cross-link formation in Mer+ and Mer- cells exposed to CCNU at 37 degrees C and 41 degrees C, but quantitation of cross-link formation was not possible owing to the persistence of single strand breaks in the DNA of drug-treated cells. Nevertheless, collectively the data indicate that thermal enhancement of CCNU toxicity is independent of effects on alkytransferase activity and indicate that hyperthermia could provide an effective strategy for improving the nitrosourea response of resistant Mer+ tumors.


Assuntos
Reparo do DNA , Temperatura Alta , Lomustina/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , DNA/metabolismo , Resistência a Medicamentos , Humanos , Metiltransferases/análise , O(6)-Metilguanina-DNA Metiltransferase
7.
Cancer Res ; 55(21): 4771-5, 1995 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-7585502

RESUMO

Although glutathione (GSH) has long been implicated in resistance to certain common chemotherapeutic agents, including alkylating agents, platinum analogues, and doxorubicin, evidence establishing a direct role in the resistant phenotype has been lacking. We cotransfected COS cells with the cDNAs for the two subunits of gamma-glutamylcysteine synthetase (GCS), which catalyzes the rate-limiting step in the de novo synthesis of GSH and is itself up-regulated in some drug-resistant tumor cells. Transfection resulted in increased GCS activity and elevated GSH levels (up to 2.6-fold). Cotransfection with the two subunits greatly enhanced the synthetic efficiency of the heavy subunit. A direct correlation (P < 0.01) between intracellular GSH levels and the LD99 dose of melphalan was observed, signifying that elevation of the thiol secondary to GCS expression is sufficient to confer the resistance phenotype.


Assuntos
Antineoplásicos Alquilantes/farmacologia , DNA Complementar/genética , Glutamato-Cisteína Ligase/genética , Glutationa/biossíntese , Melfalan/farmacologia , Animais , Catálise , Linhagem Celular , Resistencia a Medicamentos Antineoplásicos/fisiologia , Humanos , Líquido Intracelular/metabolismo , Rim/citologia , Substâncias Macromoleculares , Transfecção
8.
Cancer Res ; 51(7): 1783-8, 1991 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-2004362

RESUMO

Intracellular glutathione (GSH) levels for seven mammalian cell lines (four human tumors, two rodent, one monkey) were determined by flow cytometry following staining with monochlorobimane (MBCl), and the results were compared with GSH levels measured by the Tietze assay. The mean fluorescence intensity for all but the two rodent lines did not correlate with GSH levels determined biochemically. Good agreement between the two assays was observed for the rodent lines following depletion of GSH by buthionine sulfoximine, but the level of GSH depletion achieved in the human and monkey lines was always underestimated by MBCl/flow cytometry. These discrepancies were not resolved by increasing stain concentration or staining time. Total glutathione S-transferase (GST) activity and GST isozyme profiles were determined for each of the cell lines. Western analysis with antibodies raised against rat Ya, Yb1, and Yc and human pi isozymes revealed that the rodent cell lines expressed abundant alpha (Ya, Yc subunits) and mu (Yb1 subunits) class isozymes. In contrast, GST-pi was the predominant isozyme detected in the human tumor cell lines and Cos-7 monkey cells. Michaelis-Menten analysis with purified GSTs from rat liver as well as purified human placental (pi) GST revealed that the conjugation of MBCl and GSH catalyzed by the alpha (1-1 and 2-2) and mu (3-3 and 3-4) class GST isozymes was approximately 10 and 80 times more efficient than was conjugation by the GST pi form, respectively. These data indicate that the GST-catalyzed conjugation of GSH and MBCl is isozyme dependent and that MBCl is a relatively poor substrate for the pi isozyme. As a consequence of this isozyme rate differential, the MBCl/flow cytometry technique for GSH quantitation must be applied cautiously, particularly with human tumor cells, many of which have been shown to have high GST-pi activity. Application to other cell types should also be made after careful characterization of GSH levels and GST isozyme composition and only after comparison with other independent assays of GSH concentration.


Assuntos
Citometria de Fluxo/métodos , Glutationa Transferase/análise , Glutationa/análise , Neoplasias/química , Pirazóis , Animais , Biomarcadores , Humanos , Células Tumorais Cultivadas/química
9.
Cancer Res ; 48(4): 798-801, 1988 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-3338077

RESUMO

The cytotoxicity of two series (A and B) of novel mixed-function compounds (NI-CENU) combining nitroimidazole (NI) and chloroethylnitrosourea (CENU) functions were examined in Mer- HeLa-MR and Mer+ HeLa-S3 cells. Series A compounds differed from those in Series B by having a hydroxypropyl as opposed to an ethyl group linking the imidazole ring and the nitrosoureido function. Four analogues, including the imidazole and the 2-, 4-, and 5-NO2 derivatives, were evaluated in each series. Cells were exposed to the various compounds for 4 h under aerobic and hypoxic conditions, and toxicity was assessed by clonogenic assay. Corresponding analogues in Series A and B were equally toxic to HeLa-MR cells. Preferential hypoxic toxicity was observed only with the 2-NO2 derivative in either series (I-278, Series A; I-282, Series B). For either compound a dose enhancement factor of 2.4 was observed for hypoxic exposures. The Mer+ HeLa-S3 cells were considerably more resistant to the NI-CENU than were their HeLa-MR counterparts. In further contrast to the HeLa-MR data, the Series B compounds were consistently more effective against the HeLa-S3 cells than were their corresponding Series A analogues. The enhanced effectiveness of the Series B compounds in HeLa-S3 cells may be related to the fact that these compounds express carbamoylating activity whereas Series A compounds lack this property. Again only I-278 and I-282 were preferentially toxic to hypoxic cells; however, the aerobic/hypoxic differential was dramatically reduced (dose enhancement factor = 1.3) as compared to that observed with the HeLa-MR cells. The enhanced hypoxic toxicity of the 2-NO2 NI-CENUs was not due to direct hypoxic toxicity of the nitro moiety but presumably is the result of enhancement of CENU toxicity (i.e., chemosensitization). The data suggest that much lower concentrations of NI may be required to observe chemosensitization when the NI and chemotherapeutic agent are administered as a single mixed-function compound.


Assuntos
Antineoplásicos/toxicidade , Etilnitrosoureia/análogos & derivados , Nitroimidazóis/toxicidade , Aerobiose , Anaerobiose , Sobrevivência Celular/efeitos dos fármacos , Resistência a Medicamentos , Etilnitrosoureia/toxicidade , Células HeLa/citologia , Células HeLa/efeitos dos fármacos , Humanos , Compostos de Nitrosoureia/toxicidade , Relação Estrutura-Atividade
10.
Cancer Res ; 54(12): 3196-201, 1994 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-8205540

RESUMO

The metabolisms of two standard electron acceptors and a series of bioreductive antitumor compounds by purified rat and human DT-diaphorases (DTD) were compared. DTD was purified from rat liver cytosol and from Escherichia coli in which rat liver or human lung tumor DTD complementary DNA was expressed. Km and kcat values for menadione and 2,6-dichlorophenolindophenol reduction were similar for the three enzyme preparations except that rat E. coli DTD had 2-3-fold higher kcat values for both menadione and 2,6-dichlorophenolindophenol and a 2-3-fold higher Km for menadione than either rat liver or human E. coli DTD. Reduction of the antitumor compounds was 1.9-4.9 times faster with rat E. coli DTD than with human E. coli DTD. The antitumor compounds were reduced in the following order by rat E. coli DTD: 2,5-dimethyl-3,6-diaziridinyl-1,4-benzoquinone > streptonigrin > mitomycin A > diaziquone > mitomycin C (MC) > 5-(aziridin-1-yl)-2,4-dinitrobenzamide. The order was the same for human E. coli DTD with one exception; diaziquone was reduced slightly faster than mitomycin A. Metabolism of doxorubicin could not be detected using rat or human E. coli DTD. MC-induced DNA cross-linking was also more efficient using rat E. coli DTD relative to human E. coli DTD. Metabolism of MC by rat and human E. coli DTD was also compared under aerobic and hypoxic conditions. Rates of reduction of MC and metabolite formation were similar under aerobic and hypoxic conditions, and the toxicity of MC to DTD-rich HT-29 cells was also similar in aerobic and hypoxic conditions. In contrast, the toxicity of MC to DTD-deficient BE cells was potentiated markedly under hypoxia. These data show that although small catalytic differences between rat and human E. coli DTD can be observed, compounds such as 2,5-dimethyl-3,6-diaziridinyl-1,4-benzoquinone and streptonigrin are still excellent substrates for the human enzyme and may be useful in the therapy of tumors high in DTD activity. In addition, metabolism of MC by rat and human E. coli DTD was similar in aerobic and hypoxic conditions; in agreement with these data, cytotoxicity of MC to a DTD-rich cell line was oxygen independent. Increased MC cytotoxicity under hypoxia appears to be mediated by enzymes other than DTD.


Assuntos
Antineoplásicos/metabolismo , NAD(P)H Desidrogenase (Quinona)/metabolismo , Aerobiose , Animais , Antineoplásicos/farmacocinética , Biotransformação , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/genética , Hipóxia Celular/fisiologia , Cromatografia Líquida de Alta Pressão , Dano ao DNA , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Humanos , Cinética , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Mitomicina/metabolismo , Mitomicina/farmacocinética , Mitomicina/toxicidade , NAD/metabolismo , Oxirredução , Ratos , Ratos Sprague-Dawley , Transfecção , Células Tumorais Cultivadas
11.
Cancer Res ; 48(13): 3576-80, 1988 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-3259904

RESUMO

Thermal radiosensitization was studied in two human T-cell acute lymphoblastic leukemia cell lines (JM and MOLT3) with regard to heat-irradiation sequence and heating duration. In MOLT3 thermal radiosensitization was maximal when 43.5 degrees C hyperthermia immediately preceded or followed irradiation; at 41.5 degrees C, radiosensitization was maximal with hyperthermia immediately before or up to 3 h after irradiation. In JM, enhancement of radiation killing was unexpectedly maximal when 41.5 or 43.5 degrees C hyperthermia preceded irradiation by 2 to 4 h. Thermal radiosensitization increased exponentially with increasing duration of heating at 41.5 degrees C for at least 3 h in MOLT3. In contrast, in JM, radiosensitization increased exponentially for 1.6 h but additional heating (up to 3 h net heating) had no appreciable further effect on radiation killing. For JM, repair of single and double stranded DNA breaks was investigated using alkaline and neutral elution techniques to determine whether the unusual results regarding heat-irradiation sequencing were related to effects of heat on repair of DNA damage. These studies were unable to detect significant differences in repair of single or double stranded DNA breaks between unheated control cells and cells heated at 41.5 degrees C for 1 h ending 4 h before irradiation. The direct cytotoxicity of hyperthermia was also studied in both cell lines.


Assuntos
Sobrevivência Celular/efeitos da radiação , Hipertermia Induzida , Células Tumorais Cultivadas/efeitos da radiação , Dano ao DNA , Reparo do DNA , Relação Dose-Resposta à Radiação , Raios gama , Humanos , Leucemia Linfoide , Tolerância a Radiação , Linfócitos T , Fatores de Tempo
12.
Cancer Res ; 52(18): 5115-8, 1992 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-1355406

RESUMO

The biochemical and molecular basis for the elevation of glutathione (GSH) levels commonly detected in many drug-resistant cells has not been elucidated. In a series of L-phenylalanine mustard-resistant human prostate carcinoma cell lines (DU-145), resistance was associated with elevated GSH levels, increased activity of gamma-glutamylcysteine synthetase (GCS), the rate-limiting enzyme in GSH biosynthesis, and a marked increase in the steady-state levels of GCS-specific transcripts (4.0 and 3.2 kilobases). Loss of the resistant phenotype was accompanied by a reduction in GSH and a return of GCS activity and transcript levels to values comparable to those detected in the drug-sensitive parent cells. These data strongly implicate up-regulation of GCS activity as an important mechanism in the evolution of drug resistance associated with increased levels of intracellular GSH. The results further suggest that the ability to synthesize GSH may be more indicative of resistance than steady-state GSH levels per se.


Assuntos
Glutamato-Cisteína Ligase/metabolismo , Glutationa/metabolismo , Melfalan/farmacologia , Neoplasias da Próstata/metabolismo , Resistência a Medicamentos , Expressão Gênica , Glutamato-Cisteína Ligase/genética , Humanos , Técnicas In Vitro , Masculino , Neoplasias da Próstata/genética , RNA Mensageiro/genética , RNA Neoplásico/genética , Células Tumorais Cultivadas
13.
Cancer Res ; 53(18): 4413-7, 1993 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-8364937

RESUMO

The anti-estrogen tamoxifen (TAM) is widely used in the therapy of human breast cancer. Shown to induce a G1 transition delay in vitro, the kinetic effects of TAM on breast carcinoma cells growing as tumor xenografts in nude mice have been less well characterized. In this study, we demonstrate a significant increase in the tumor potential doubling time (Tpot) and decrease in the labeling index (%LI) of estradiol (E2)-stimulated MCF-7 xenografts following TAM treatment or E2 deprivation. MCF-7 tumor pieces were transplanted s.c. into nude mice supplemented with Silastic capsules containing E2. After 2-4 weeks, animals were randomized to continued E2 treatment, E2 and TAM treatment, or E2 deprivation. At times ranging from 0 to 23 days after treatment, animals were given injections of bromodeoxyuridine and tumors excised for kinetic analysis. Using flow-cytometric techniques, the Tpot and %LI were estimated for all tumors. Seven independent experiments were performed and data pooled for statistical analysis. At the time of hormonal manipulation, E2-stimulated tumors had a volume doubling time of 5 days, a Tpot of 2.3 days, and a %LI of 23%. Continued E2 treatment resulted in only minimal changes in Tpot and %LI over the remainder of the observation period. Treatment with TAM resulted in a slowing of tumor growth (tumor doubling time, 12 days), a significant (P < 0.001) increase in Tpot to 6.6 days, and a decrease in %LI to 8% by 23 days posttreatment. E2 deprivation resulted in a cessation of tumor growth and similar changes in Tpot and %LI to 5.3 days and 10%, respectively (P < 0.001). In contrast to previous reports, these data demonstrate that TAM treatment and E2 deprivation both significantly decrease tumor cell proliferation in MCF-7 xenografts.


Assuntos
Bromodesoxiuridina/metabolismo , Neoplasias Mamárias Experimentais/patologia , Tamoxifeno/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Feminino , Citometria de Fluxo , Humanos , Camundongos , Transplante de Neoplasias , Transplante Heterólogo
14.
Cancer Res ; 51(4): 1099-104, 1991 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-1825475

RESUMO

SR-2508, a less lipophilic ane neurotoxic analogue of the nitroimidazole, misonidazole, has exhibited significant chemosensitization properties in preclinical studies with alkylating agents. A phase I trial was carried out to assess toxicity and possible pharmacological interactions of the combination of short infusions of SR-2508 and cyclophosphamide (CP). Patients were randomly assigned to receive either CP alone followed in 3 wk by CP + SR-2508, or CP + SR-2508 followed by CP alone. All additional courses were CP + SR-2508. The maximum tolerated dose of the combination was determined by dose escalation of SR-2508 while the dose of CP remained fixed, initially 1.0 g/m2, and then a second maximum tolerated dose was determined with CP at 1.6 g/m2. One hundred seventeen evaluated courses were administered to 39 patients, the majority of whom had received prior treatment. Somewhat unexpectedly, reversible grade 4 granulocytopenia was the dose-limiting toxicity occurring in four of five evaluable first combination courses at level 6 (SR-2508, 11.3 g/m2; CP, 1.0 g/m2), the initial maximum tolerated dose. SR-2508 enhanced CP-induced myelosuppression as exhibited by the significant difference (p less than 0.001) between the 27 paired courses (CP versus CP + SR-2508) for WBC nadirs over levels 1 to 6. The neurotoxicity encountered was similar to that seen in past clinical trials, being reversible, mild, and usually peripheral in nature. There was one treatment-related death (neutropenic sepsis) on study. No other significant toxicity was seen. SR-2508 exhibited linear pharmacokinetics over the dose range studied. The SR-2508 area under the concentration-time curve increased linearly with dose (r = 0.858; p less than 0.001). No other parameters were dose related. Neither drug appeared to affect the pharmacokinetics of the other, and CP pharmacokinetic values were consistent with those from prior studies. Due to the interaction noted between the two agents and the preclinical data suggesting preferential enhancement of antitumor efficacy under this combination, phase II study appears warranted.


Assuntos
Ciclofosfamida/administração & dosagem , Nitroimidazóis/administração & dosagem , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Medula Óssea/efeitos dos fármacos , Ciclofosfamida/farmacocinética , Ciclofosfamida/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Etanidazol , Feminino , Humanos , Injeções Intravenosas , Masculino , Pessoa de Meia-Idade , Nitroimidazóis/farmacocinética , Nitroimidazóis/toxicidade
15.
J Clin Oncol ; 12(1): 194-205, 1994 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8270977

RESUMO

PURPOSE: A phase I dose-escalation trial of intravenous (IV) L-buthionine-SR-sulfoximine (BSO) with melphalan (L-PAM) was performed to determine the toxicity and biologic activity of BSO, administered as a short infusion every 12 hours, and the combination of BSO plus L-PAM. PATIENTS AND METHODS: Twenty-eight patients with refractory malignancies received 30-minute infusions of BSO every 12 hours for 6 to 10 doses in week 1 followed in week 2 by either IV L-PAM (15 mg/m2) alone or BSO as in week 1 with L-PAM. Patients received the combination in week 5 (course no. 2) if they received L-PAM alone during week 2 and vice versa. BSO doses ranged from 1.5 g/m2 to 13.104 g/m2. RESULTS: The only toxicity observed with BSO infusions was occasional nausea/vomiting. Evaluation of 23 paired courses (L-PAM plus BSO v L-PAM) showed significantly (P < .001) greater leukopenia and thrombocytopenia with L-PAM plus BSO. No other significant toxicity was noted. Measurement of intracellular glutathione (GSH) levels in peripheral mononuclear cells (PBLs) of all patients receiving BSO showed a consistent, non-dose-dependent, linear decrease in GSH with repeated BSO doses. Maximal GSH depletion (40% of baseline values, absolute values 200 to 250 ng/10(6) PBLs) was noted after the sixth BSO dose; extended BSO dosing schedules beyond six total BSO doses did not further deplete GSH. Evaluation of gamma-glutamylcysteine synthetase (GCS) activity showed marked inhibition near the end of each infusion with near complete recovery of GCS activity before the next BSO dose. The pattern of GCS inhibition mirrored the plasma BSO concentrations with peak values (level 6, 4 to 8 mmol/L L,R+L,S BSO) observed at the end of the infusion with a rapid decrease in plasma concentrations with an estimated half-life (t1/2) of less than 2 hours. Differential elimination of the R+S stereoisomers was observed. Analysis of L-PAM pharmacokinetics showed marked interpatient variability and a significant decrease in total-body clearance (P = .01) and volume of distribution (P = .03) in courses with L-PAM plus BSO as compared with L-PAM alone. CONCLUSION: This study shows that BSO alone and in combination with L-PAM can be safely given to patients, but that a schedule of short infusions every 12 hours does not result in GSH depletion less than 30% of baseline values.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Glutationa/efeitos dos fármacos , Metionina Sulfoximina/análogos & derivados , Neoplasias/tratamento farmacológico , Adulto , Idoso , Antimetabólitos Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Butionina Sulfoximina , Feminino , Glutationa/metabolismo , Humanos , Infusões Intravenosas , Masculino , Melfalan/administração & dosagem , Metionina Sulfoximina/administração & dosagem , Metionina Sulfoximina/uso terapêutico , Pessoa de Meia-Idade , Neoplasias/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 21(11): 1846-51, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11701476

RESUMO

The lipid products derived from the cyclooxygenase pathway can have diverse and often contrasting effects on vascular cell function. Cyclopentenone prostaglandins (cyPGs), such as 15-deoxy-Delta(12,14)-prostaglandin-J(2) (15d-PGJ(2)), a peroxisome proliferator-activated receptor-gamma (PPARgamma) agonist, have been reported to cause endothelial cell apoptosis, yet in other cell types, cyPGs induce cytoprotective mediators, such as heat shock proteins, heme oxygenase-1, and glutathione (GSH). Herein, we show in human endothelial cells that low micromolar concentrations of 15d-PGJ(2) enhance GSH-dependent cytoprotection through the upregulation of glutamate-cysteine ligase, the rate-limiting enzyme of GSH synthesis, as well as GSH reductase. The effect of 15d-PGJ(2) on GSH synthesis is attributable to the cyPG structure but is independent of PPAR, inasmuch as the other cyPGs, but not PPARgamma or PPARalpha agonists, are able to increase GSH. The increase in cellular GSH is accompanied by abrogation of the proapoptotic effects of 4-hydroxynonenal, a product of lipid peroxidation present in atherosclerotic lesions. However, higher concentrations of 15d-PGJ(2) (10 micromol/L) cause endothelial cell apoptosis, which is further enhanced by depletion of cellular GSH by buthionine sulfoximine. We propose that the GSH-dependent cytoprotective pathways induced by 15d-PGJ(2) contribute to its antiatherogenic effects and that these pathways are distinct from those leading to apoptosis.


Assuntos
Apoptose , Endotélio Vascular/metabolismo , Glutationa/biossíntese , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacologia , Aldeídos/farmacologia , Apoptose/efeitos dos fármacos , Ácido Araquidônico/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Glutamato-Cisteína Ligase/biossíntese , Glutamato-Cisteína Ligase/genética , Glutationa/metabolismo , Humanos , Cinética , Prostaglandinas/farmacologia , RNA Mensageiro/biossíntese , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo
17.
Endocrinology ; 112(1): 172-7, 1983 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-6847815

RESUMO

We have previously reported a technique for the quantitative transplantation of monodispersed rat thyroid cells. In this study, the influence of the age and sexual status of the donor and recipient animals on the transplantation process has been examined using the number of cells required for the growth of 1 or more follicular units in 50% of the transplantation sites (follicular dose 50% or FD50) as the end point. The number of multifollicular clusters per 1000 inoculated cells was also explored. FD50 values in 5-week-old intact male recipients increased as the donor's age increased over the range of 4 weeks to 12 months. FD50 values in 5-week-old thyroidectomized male recipients were similar when donors were 4-8 weeks old, but were increased for cells from 52-week-old donors. The FD50 values of cells from 5-week-old donors was higher in 4-week-old intact recipients than in the 5- or 6-week-old recipients. In male recipients, the FD50 values were lower than in female recipients of 5 and 8 weeks of age. This sex-related effect was reduced by orchiectomy and ovariectomy. Generally, the frequency of multifollicular clusters per 1000 inoculated cells increased as the FD50 decreased. These results are discussed in the light of measurements of thyroidal DNA synthesis and TSH levels reported by others.


Assuntos
Envelhecimento , Caracteres Sexuais , Glândula Tireoide/transplante , Animais , Castração , Divisão Celular , Transformação Celular Neoplásica , Feminino , Masculino , Ratos , Glândula Tireoide/citologia
18.
FEBS Lett ; 448(2-3): 292-6, 1999 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-10218495

RESUMO

Nitric oxide protects cells from oxidative stress through a number of direct scavenging reactions with free radicals but the effects of nitric oxide on the regulation of antioxidant enzymes are only now emerging. Using bovine aortic endothelial cells as a model, we show that nitric oxide, at physiological rates of production (1-3 nM/s), is capable of inducing the synthesis of glutathione through a mechanism involving gamma-glutamylcysteine synthetase and gamma-glutamyl transpeptidase. This novel nitric oxide signalling pathway is cGMP-independent and we hypothesize that it makes an important contribution to the anti-atherosclerotic and antioxidant properties of nitric oxide.


Assuntos
Endotélio Vascular/metabolismo , Glutamato-Cisteína Ligase/fisiologia , Glutationa/biossíntese , Óxido Nítrico/metabolismo , gama-Glutamiltransferase/fisiologia , Animais , Bovinos , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Isoxazóis/farmacologia , Compostos Nitrosos/farmacologia , Fatores de Tempo
19.
Clin Exp Metastasis ; 2(1): 73-81, 1984.
Artigo em Inglês | MEDLINE | ID: mdl-6543692

RESUMO

Female C3H/HeJ mice were inoculated intravenously with KHT sarcoma cells. Once macroscopic colonies were established in the lungs, the thoraces of the animals were locally irradiated. Despite significant lung nodule regression following treatment, animals were observed to die with ovarian and renal metastases. By irradiating the lungs at various times after intravenous tumor cell injection, it was demonstrated that ovarian and renal metastases arose only from established lung colonies and were not a consequence of the initial cell inoculum. Incidence of metastases increased from 0 to 100 per cent when the time between cell inoculation and thoracic irradiation was increased from 4 to 16 days. Once established, ovarian and renal metastases grew with a doubling time of approximately 1-2 days. Metastatic tumors in the ovaries were found to be refractory to radiation therapy because of a large component of radiation-resistant hypoxic cells. Parallel experiments utilizing male C3H/HeJ mice demonstrated metastases only in the kidneys and these grew at a growth rate similar to that seen for renal metastases in female mice. This system may serve as a model for the study of factors influencing the dissemination of tumor cells to these anatomical sites and their response to treatment.


Assuntos
Neoplasias Pulmonares/patologia , Sarcoma Experimental/patologia , Animais , Feminino , Neoplasias Renais/secundário , Neoplasias Pulmonares/radioterapia , Masculino , Camundongos , Metástase Neoplásica , Neoplasias Ovarianas/secundário , Sarcoma Experimental/radioterapia , Fatores de Tempo , Raios X
20.
Int J Radiat Oncol Biol Phys ; 12(8): 1389-92, 1986 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3759561

RESUMO

Experiments were designed to measure cross-link formation following combined treatment of EMT-6/Ro tumor cells with 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU) and misonidazole (MISO) in vitro. To avoid MISO-induced glutathione (GSH) depletion, which might contribute to enhanced monoadduct formation by reducing the protective GSH pools, a post-incubation (i.e. treatment with CCNU for one hour in air followed by MISO treatment in hypoxia) protocol was adopted. Utilizing this treatment scheme, it was possible to significantly enhance CCNU toxicity by post-treating with MISO immediately after exposure to CCNU. Enhanced cross-link formation detected by alkaline elution, at this time, correlated well with the magnitude of cell-kill enhancement, thereby implicating enhanced cross-link formation in the mechanism of potentiation. However, if the cells were allowed to incubate for various intervals between CCNU and MISO treatments, the magnitude of potentiation progressively diminished. Beyond approximately 8-10 hours (corresponding to the time required for maximal cross-link formation after CCNU treatment), treatment with MISO was ineffective at potentiating CCNU cytotoxicity. These experiments suggest that chemopotentiation can be produced by treating with MISO after treatment with CCNU (post-incubation) and that enhanced cross-link formation is involved in the mechanism of MISO chemopotentiation of CCNU activity. The kinetic studies, using the post-incubation protocol, further suggest that the chemopotentiating effect of MISO is exerted subsequent to monoadduct formation and probably does not involve inhibition of DNA-DNA cross-link repair.


Assuntos
Antineoplásicos/farmacologia , Lomustina/farmacologia , Misonidazol/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Técnicas In Vitro , Neoplasias Experimentais/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA