Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell Biol ; 5(6): 1279-86, 1985 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-4033653

RESUMO

During the replication of chromatin, the syntheses of the histone protein and DNA components are closely coordinated but not totally linked. The interrelationships of total protein synthesis, histone protein synthesis, DNA synthesis, and mRNA levels have been investigated in Chinese hamster ovary cells subjected to several different types of inhibitors in several different temporal combinations. The results from these studies and results reported elsewhere can be brought together into a consistent framework which combines the idea of autoregulation of histone biosynthesis as originally proposed by W. B. Butler and G. C. Mueller (Biochim. Biophys. Acta 294:481-496, 1973] with the presence of basal histone synthesis and the effects of protein synthesis on DNA synthesis. The proposed framework obviates the difficulties of Butler and Mueller's model and may have wider application in understanding the control of cell growth.


Assuntos
Divisão Celular , Cromatina/biossíntese , Replicação do DNA , Histonas/biossíntese , Biossíntese de Proteínas , Animais , Linhagem Celular , Cricetinae , Cricetulus , Replicação do DNA/efeitos dos fármacos , Depressão Química , Feminino , Fibroblastos/metabolismo , Ovário , Biossíntese de Proteínas/efeitos dos fármacos , Fatores de Tempo
2.
Cancer Gene Ther ; 13(9): 886-95, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16645619

RESUMO

Administration of chemotherapy is often limited by myelosuppression. Expression of drug-resistance genes in hematopoietic cells has been proposed as a means to decrease the toxicity of cytotoxic agents. In this pilot study, we utilized a retroviral vector expressing methylguanine DNA methyltransferase (MGMT) to transduce hematopoietic progenitors, which were subsequently used in the setting of alkylator therapy (procarbazine, CCNU, vincristine (PCV)) for poor prognosis brain tumors. Granulocyte colony-stimulating factor (G-CSF)-mobilized peripheral blood progenitor cells were collected by apheresis and enriched for CD34+ expression. Nine subjects were infused with CD34+-enriched cells treated in a transduction procedure involving a 4-day exposure to cytokines with vector exposure on days 3 and 4. No major adverse event was related to the gene therapy procedure. Importantly, the engraftment kinetics of the treated product was similar to unmanipulated peripheral blood stem cells, suggesting that the ex vivo manipulation did not significantly reduce engrafting progenitor cell function. Gene-transduced cells were detected in all subjects. Although the level and duration was limited, patients receiving cells transduced using fibronectin 'preloaded' with virus supernatant appeared to show improved in vivo marking frequency. These findings demonstrate the feasibility and safety of utilizing MGMT-transduced CD34+ peripheral blood progenitor cells in the setting of chemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Vetores Genéticos/genética , Lomustina/uso terapêutico , O(6)-Metilguanina-DNA Metiltransferase/genética , Procarbazina/uso terapêutico , Vincristina/uso terapêutico , Adolescente , Adulto , Antígenos CD34/metabolismo , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/genética , Ensaio de Unidades Formadoras de Colônias , Primers do DNA , Feminino , Fibronectinas/metabolismo , Vetores Genéticos/metabolismo , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Lomustina/administração & dosagem , Masculino , Pessoa de Meia-Idade , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Transplante de Células-Tronco de Sangue Periférico/métodos , Projetos Piloto , Reação em Cadeia da Polimerase , Procarbazina/administração & dosagem , Retroviridae/genética , Transdução Genética/métodos , Vincristina/administração & dosagem
3.
Nucleic Acids Res ; 29(12): 2558-66, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11410664

RESUMO

DNA repair status plays a major role in mutagenesis, carcinogenesis and resistance to genotoxic agents. Because DNA repair processes involve multiple enzymatic steps, understanding cellular DNA repair status has required several assay procedures. We have developed a novel in vitro assay that allows quantitative measurement of alkylation repair via O(6)-methylguanine DNA methyltransferase (MGMT) and base excision repair (BER) involving methylpurine DNA glycosylase (MPG), human 8-oxoguanine DNA glycosylase (hOGG1) and yeast and human abasic endonuclease (APN1 and APE/ref-1, respectively) from a single cell extract. This approach involves preparation of cell extracts in a common buffer in which all of the DNA repair proteins are active and the use of fluorometrically labeled oligonucleotide substrates containing DNA lesions specific to each repair protein. This method enables methylation and BER capacities to be determined rapidly from a small amount of starting sample. In addition, the stability of the fluorometric oligonucleotides precludes the substrate variability caused by continual radiolabeling. In this report this technique was applied to human breast carcinoma MDA-MB231 cells overexpressing human MPG in order to assess whether up-regulation of the initial step in BER alters the activity of selected other BER (hOGG1 and APE/ref-1) or direct reversal (MGMT) repair activities.


Assuntos
Neoplasias da Mama/enzimologia , Carbono-Oxigênio Liases/metabolismo , Reparo do DNA/genética , Regulação Enzimológica da Expressão Gênica , N-Glicosil Hidrolases/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Alquilação , Neoplasias da Mama/genética , Soluções Tampão , Carbono-Oxigênio Liases/genética , DNA Glicosilases , Metilação de DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos) , DNA-Formamidopirimidina Glicosilase , Desoxirribonuclease IV (Fago T4-Induzido) , Fluorescência , Regulação Neoplásica da Expressão Gênica , Humanos , Células K562 , N-Glicosil Hidrolases/genética , O(6)-Metilguanina-DNA Metiltransferase/genética , Oligonucleotídeos/genética , Sensibilidade e Especificidade , Transfecção , Células Tumorais Cultivadas , Regulação para Cima
4.
J Natl Cancer Inst ; 84(4): 256-60, 1992 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-1531148

RESUMO

BACKGROUND: Fewer than 20% of patients with nonhematologic malignancies treated with chloroethylnitrosoureas (CENUs) respond, but streptozocin (STZ), which depletes O6-methylguanine-DNA-methyltransferase (MGMT), has been shown to reverse resistance to CENUs in vitro. PURPOSE: The purpose of this phase I study was to determine (a) the maximum tolerated dose (MTD) of carmustine (BCNU), a CENU, plus a fixed dose of STZ; (b) the toxic effects of the drugs; and (c) the effects on peripheral blood mononuclear cells (PBMC). METHODS: A clinical phase I study of STZ followed by BCNU was designed to simulate conditions that produce maximal sensitization of CENU-resistant HT-29 cells in vitro. Patients received a 20-minute infusion of the MTD of STZ (2 g/m2) followed 1 hour later with a 60-minute infusion of BCNU (100, 125, 137.5, or 150 mg/m2). Treatment was repeated after 6 weeks. Twenty-four patients with advanced malignancies received 32 courses of therapy (range, 1-2 courses). RESULTS: The MTD of BCNU was 125 mg/m2. The dose-limiting toxic effect was thrombocytopenia occurring about 22 days after treatment, with recovery between days 28 and 35. Transient hypophosphatemia and proteinuria were common, and serum creatinine was elevated in 9% of the courses. Two patients who received therapy died--one due to pulmonary toxic effects and one due to hepatic toxic effects. Two patients with previously untreated carcinoid achieved partial response. In three patients, MGMT levels in PBMC were more than 85% depleted after STZ administration and more than 90% depleted after BCNU infusion. CONCLUSIONS: These results show that the magnitude of MGMT depletion by STZ in PBMC is in the range necessary to produce sensitivity to CENUs in resistant cell lines but also that, when BCNU is combined with STZ, the MTD of BCNU is about 50% that of BCNU as a single agent and that platelet count suppression occurs earlier. IMPLICATIONS: We plan to conduct phase II studies of STZ plus BCNU in tumor types with low response to CENUs. One of the major goals will be to demonstrate that depletion of MGMT is greater in tumor cells than in normal cells.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/tratamento farmacológico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carmustina/administração & dosagem , Esquema de Medicação , Avaliação de Medicamentos , Feminino , Humanos , Masculino , Metiltransferases/efeitos dos fármacos , Pessoa de Meia-Idade , Neoplasias/enzimologia , O(6)-Metilguanina-DNA Metiltransferase , Estreptozocina/administração & dosagem
5.
Cancer Res ; 50(1): 62-6, 1990 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-2104539

RESUMO

This study was initiated to determine if DNA-damaging chemotherapeutic agents can suppress the expression of oncogenes. The effects of three structurally related bifunctional alkylating agents on the steady state mRNA levels of c-myc, c-fos, N-ras, and beta-actin in the human colon carcinoma cell line Colo320HSR were examined. Colo320HSR has an amplified c-myc oncogene, which is highly overexpressed, and is assumed to be one of the transforming genes of this cell line. Two concentrations of mechlorethamine, L-phenylalanine mustard, and 4-hydroperoxycyclophosphamide, which produced 1 or 3 log cell kills were used to examine the effects of drug exposure on the expression of specific genes. Steady state mRNA levels were measured by Northern blot analysis. Following a 1-h drug exposure, RNA was isolated from cells at 0, 6, 12, and 24 h following drug removal. The agents used produced changes in the expression of specific genes, and all three did so in a similar fashion. Immediately following drug removal, the steady state expression of c-myc in treated cells was increased 2- to 3-fold compared to control. At 6 and 12 h following drug removal, c-myc levels were depressed 2.5- to 5-fold. By 24 h, c-myc expression approached, but remained below, control levels. Immediately following drug removal, c-fos levels were increased 3- to 4-fold, and from 6 to 24 h following drug removal, c-fos levels gradually return to, or fell below low basal levels. During the 24-h time course, drug treatment had little or no effect on the steady state levels of N-ras or beta-actin. These data support the hypothesis that alkylating agents may suppress the expression of specific transforming genes.


Assuntos
Expressão Gênica/efeitos dos fármacos , Mecloretamina/farmacologia , Melfalan/farmacologia , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Northern Blotting , Linhagem Celular , Neoplasias do Colo , Humanos , Cinética , Proteínas Proto-Oncogênicas c-fos , Proteínas Proto-Oncogênicas c-myc , RNA Mensageiro/genética , RNA Mensageiro/isolamento & purificação
6.
Cancer Res ; 38(10): 3379-84, 1978 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-150942

RESUMO

The DNA of V-79 Chinese hamster cells was examined by alkaline elution following treatment of cultures with eight different nitrosoureas. Drug incubations were performed under consistent biological conditions of equal toxicity and equal mutation induction at the hypoxanthineguanine phosphoribosyltransferase locus. The goals of this study were to determine whether DNA damage could be detected in cells treated with biologically relevant doses of nitrosoureas and to determine whether the type and number of observed DNA lesions could be correlated with the cytotoxic and mutagenic effects of the drugs. All of the compounds tested produced, to some degree, lesions that were observed as DNA strand breaks upon exposure of the DNA to alkali. The levels of DNA strand breaks and/or alkali-labile lesions were comparable for all of the drugs at the equimutagenic doses. DNA cross-linking was observed at both the equitoxic and the equimutagenic concentrations of the haloethylnitrosoureas, but cross-linking was not observed with methylnitrosourea or streptozotocin. Methylnitrosourea and streptozotocin required approximately 40 times the drug concentration to produce toxicity equal to the haloethylnitrosoureas. These data suggest that the ability to cross-link DNA confers increased cytotoxicity to the haloethylnitrosoureas.


Assuntos
DNA/metabolismo , Compostos de Nitrosoureia/farmacologia , Carmustina/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cianatos/uso terapêutico , Relação Dose-Resposta a Droga , Cloreto de Etil/análogos & derivados , Cloreto de Etil/uso terapêutico , Lomustina/farmacologia , Metilnitrosoureia/farmacologia , Mutação/efeitos dos fármacos , Compostos de Nitrosoureia/administração & dosagem , Estreptozocina/análogos & derivados , Estreptozocina/farmacologia , Fatores de Tempo
7.
Cancer Res ; 45(9): 4043-7, 1985 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-3896476

RESUMO

Utilizing the DNA alkaline elution technique we have compared qualitatively and quantitatively the DNA lesions produced in L1210 cells after a 2 h exposure to the antitumor agents, cis-(diammino) (1,1-cyclobutanedicarboxylato)-platinum(II) (CBDCA) and cis-diamminedichloroplatinum(II) (DDP). DNA-protein and DNA interstrand cross-links are formed in cells exposed to either CBDCA or DDP. However, in comparison to DDP peak levels of these lesions occur 6 to 12 h later in CBDCA treated cells. Cytotoxicity studies reveal that CBDCA is 45 times less potent than DDP to L1210 cells when compared on a molar basis. The decreased cytotoxicity of CBDCA and the 12 h delay in peak cross-linking when compared to DDP is interpreted as a decreased reactivity of the intact CBDCA towards the DNA. This decreased reactivity may be due in part to the presence of a stable bidentate dicarboxylate chelate ring structure of CBDCA resulting in a much slower rate of hydrolysis to the active form of the drug.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , DNA/metabolismo , Compostos Organoplatínicos/farmacologia , Animais , Carboplatina , Linhagem Celular , Cisplatino/metabolismo , Reagentes de Ligações Cruzadas/metabolismo , Leucemia L1210/patologia , Camundongos , Compostos Organoplatínicos/metabolismo
8.
Cancer Res ; 54(16): 4371-5, 1994 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8044784

RESUMO

We have recently suggested that optimal reversal of 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) resistance might require complete inactivation of the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) for at least 24 h following BCNU administration (22). This study was undertaken to further evaluate the functional importance of the regeneration rate of MGMT activity following O6-benzylguanine (BG), O6-benzyl-2'-deoxyguanosine (dBG), and streptozotocin (STZ) in determining the potentiation of BCNU cytotoxicity in the highly resistant colon carcinoma cell line HT-29. To this end, we measured the enhancement of BCNU cytotoxicity utilizing regimens which provided complete inhibition, with partial or complete recovery of MGMT activity by 24 h. We were able to modulate the recovery rate of MGMT activity following BG or dBG administration by repeated washing of cells with complete medium. Subsequent to equally inhibitory doses of BG (100 microM) or dBG (1.0 mM) treatment without washing, MGMT activity was completely inactivated for 24 h. However, MGMT activity recovered to control levels by 24 h when cells were treated with BG or dBG and washed 4 times with complete medium. This recovery was completely inhibited for 24 h by combining BG or dBG with 2.5 mM STZ. These differential repletion profiles produced disparate potentiation of BCNU cytotoxicity. The regimens which produced complete inactivation of MGMT for 24 h produced the greatest enhancement of BCNU cytotoxicity. BG or dBG (without a wash) potentiated BCNU cytotoxicity by approximately 3 logs of synergistic cell kill. When the recovery rate of MGMT activity was markedly enhanced via washing of cells, BG-BCNU or dBG-BCNU produced less than 1 log of synergistic cell kill. The addition of STZ to BG or dBG inhibited this temporal recovery for 24 h and potentiated BCNU cytotoxicity by approximately 4 logs. These data further demonstrate that extended depletion of MGMT is required for optimal reversal of BCNU resistance. Because a three-drug combination of BG-STZ-BCNU or dBG-STZ-BCNU consistently produced greater cytotoxicity than any two-drug regimen, clinical testing of these combinations is warranted. Additionally, our data suggest that the design of clinical regimens targeting the inactivation of MGMT and the reversal of BCNU resistance should consider the functional importance of extended depletion of MGMT in order to increase the possibility of antitumor responses.


Assuntos
Antineoplásicos/farmacologia , Carmustina/farmacologia , Desoxiguanosina/análogos & derivados , Guanina/análogos & derivados , Metiltransferases/metabolismo , Estreptozocina/farmacologia , Desoxiguanosina/farmacologia , Resistência a Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Guanina/farmacologia , Humanos , O(6)-Metilguanina-DNA Metiltransferase , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco
9.
Cancer Res ; 51(8): 1984-9, 1991 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-2009517

RESUMO

1-beta-D-Arabinofuranosylcytosine (ara-C) and hydroxyurea (HU) were investigated as potential DNA repair inhibitors with cis-diammine-1,1-cyclobutane dicarboxylatoplatinum(II) (CBDCA). HU plus ara-C, known inhibitors of DNA excision-repair, had previously been found to produce cytotoxic synergy and delayed removal of DNA interstrand cross-links with cis-diamminedichloroplatinum(II) (DDP). Since CBDCA and DDP share a common active intermediate, it should be possible to reproduce this interaction with CBDCA. However, the stable dicarboxylate chelate ring structure of CBDCA results in kinetics that differ significantly from those of DDP, due to slower hydrolysis to the active species. DNA adducts form more slowly, with interstand cross-links peaking approximately 12-h later and disappearing more gradually than in the case of DDP. It was therefore expected that a longer antimetabolite exposure might be required for repair inhibition with CBDCA. The 12-h exposure to HU plus ara-C previously found effective with DDP produced no cytotoxic synergy with a 2-h CBDCA exposure. Lengthening the antimetabolite treatment to 24 h resulted in approximately 1 log of synergistic toxicity, while a 24-h simultaneous exposure to HU, ara-C, and CBDCA resulted in 2 logs. Cells exposed to all three drugs showed a 2- to 3-fold greater level of interstrand cross-links after 36- to 48-h of incubation following drug removal, compared to CBDCA alone. Taken together, these findings suggest that HU plus ara-C modulates the repair of platinum-DNA adducts and establishes an effective in vitro schedule at clinically achievable concentrations for the use of those antimetabolites with CBDCA.


Assuntos
Carboplatina/antagonistas & inibidores , Carboplatina/toxicidade , Citarabina/farmacologia , Reparo do DNA/efeitos dos fármacos , DNA de Neoplasias/efeitos dos fármacos , Hidroxiureia/farmacologia , Ensaio Tumoral de Célula-Tronco , Animais , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , Sinergismo Farmacológico , Humanos , Camundongos , Fatores de Tempo
10.
Cancer Res ; 38(3): 802-8, 1978 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-626984

RESUMO

Two human colon tumor cell lines were examined for their responses to 1-(2-chloroethyl)-3-(4-methylcyclohexyl)-1-nitrosourea treatment when maintained as cultured cell lines and xenograft tumors in nude mice. One tumor line, HT, was resistant to 1-(2-chloroethyl)-3-(4-methylcyclohexyl)-1-nitrosourea treatment both in tissue culture and in vivo. The other tumor line, BE, was sensitive to 1-(2-chloroethyl)-3-(4-methylcyclohexyl)-1-nitrosourea treatment in vitro and in vivo. The DNA of tissue-cultured cells treated with 1-(2-Chloroethyl)-3-(4-methylcyclohexyl)-1-nitrosourea was examined by alkaline elution for DNA damage. 1-(2-Chloroethyl)-3-(4-methylcyclohexyl)-1-nitrosourea was found to produce DNA strand breaks and DNA cross-links in both cell types. The DNA cross-links appear to be completely repaired in the resistant HT line over the 48-hr period following drug removal, but in the sensitive BE line little or no cross-link repair was observed during this interval.


Assuntos
Neoplasias do Colo/metabolismo , Reparo do DNA , DNA de Neoplasias/metabolismo , Compostos de Nitrosoureia/farmacologia , Semustina/farmacologia , Animais , Linhagem Celular , Neoplasias do Colo/tratamento farmacológico , Resistência a Medicamentos , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/metabolismo , Transplante Heterólogo
11.
Cancer Res ; 45(4): 1674-9, 1985 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2983885

RESUMO

2-Chloroethyl (methylsulfonyl)methanesulfonate (CIEtSoSo) is of interest as a possible new chloroethylating agent because its simpler chemistry suggests that it will not generate the hydroxyethyl products which are produced by chloroethylnitrosoureas (CIEtNUs). The effects of CIEtSoSo on DNA were studied in IMR-90 and VA-13 human embryo cells by means of DNA alkaline elution analysis. IMR-90 are normal cells, whereas VA-13 cells are SV40 transformed and are deficient in DNA-guanine O6-alkyltransferase activity. The effects of CIEtSoSo were essentially the same as those of CIEtNUs in the following respects: DNA interstrand cross-links were produced in VA-13 cells but not in IMR-90 cells; the interstrand cross-links in VA-13 cells were formed after a delay of 6 to 12 h and appeared to undergo repair; DNA-protein cross-links were formed promptly in both cell types and appeared to be repaired; DNA strand breaks and alkali-labile lesions were produced and repaired, and differences were observed between the two cell types; and VA-13 cells were more sensitive than IMR-90 cells (dose modification factor, 5). The differential cytotoxicity against VA-13 cells was similar to that produced by noncarbamoylating CIEtNUs, and significantly larger than that produced by carbamoylating CIEtNUs. The results suggest that CIEtSoSo acts by chloroethylating guanine O6 positions in DNA.


Assuntos
Antineoplásicos/toxicidade , DNA/metabolismo , Mesilatos/toxicidade , Alquilação , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Reparo do DNA , Humanos , Concentração de Íons de Hidrogênio
12.
Cancer Res ; 44(5): 1767-71, 1984 May.
Artigo em Inglês | MEDLINE | ID: mdl-6713380

RESUMO

L1210 murine leukemia cells were treated in vitro with the novel antineoplastic agent 8-carbamoyl-3-(2-chloroethyl)imidazo[5,1-d] -1,2,3,5-tetrazin-4(3H)-one (M&B 39565), and its interaction with cellular DNA was assessed by alkaline elution. DNA interstrand cross-link and DNA-protein cross-link formation was quantified with regard to drug concentration and length of incubation time after a 2-hr incubation period with drug. Cytotoxicity, as measured by colony formation assays, and DNA damage caused by M&B 39565 were compared with those caused by a breakdown product of M&B 39565, 5-[3-(2-chloroethyl)triazen -1-yl]imidazole-4-carboxamide (MCTIC) and also with 1-(2-chloroethyl)-1-nitrosourea (CNU). Both MCTIC and CNU decompose to yield a 2-chloroethyldiazo species which is capable of alkylating DNA. At equimolar concentrations, all three drugs possessed similar in vitro cytotoxicities; at equitoxic concentrations, they produced similar levels of DNA interstrand cross-linking. The time course for cross-link formation was different for CNU when compared with MCTIC and M&B 39565, with peaks at 6 hr (CNU) and 9 hr (M&B 39565 and MCTIC). This study suggests that M&B 39565 is cytotoxic against L1210 leukemia cells as a consequence of DNA interstrand cross-link formation, probably via its breakdown product MCTIC.


Assuntos
DNA de Neoplasias/metabolismo , Imidazóis/toxicidade , Leucemia L1210/fisiopatologia , Compostos de Mostarda Nitrogenada , Animais , Divisão Celular/efeitos dos fármacos , Dacarbazina/análogos & derivados , Dacarbazina/toxicidade , Etilnitrosoureia/análogos & derivados , Etilnitrosoureia/toxicidade , Cinética , Camundongos , Proteínas de Neoplasias/metabolismo , Ligação Proteica
13.
Cancer Res ; 44(5): 1772-5, 1984 May.
Artigo em Inglês | MEDLINE | ID: mdl-6713381

RESUMO

Normal (IMR-90) and SV40-transformed (VA-13) human embryo cells were treated with 8-carbamoyl-3-(2-chloroethyl)imidazo[5,1-d] -1,2,3,5-tetrazin-4(3H)-one (M&B 39565), and the effects of the drug on cell viability and cellular DNA integrity were studied. The effects of M&B 39565 were compared with one of its potential decomposition products 5-[3-(2-chloroethyl)triazen-1 -yl]imidazole-4-carboxamide (MCTIC). M&B 39565 and MCTIC were 5- to 6-fold more toxic to VA-13 cells than to IMR-90 cells for drug concentrations which produced a 2-log cell kill, as measured by colony-forming assays. Using alkaline elution analysis, VA-13 cells exhibited concentration-dependent DNA interstrand cross-link formation. In IMR-90 cells, little or no interstrand cross-link formation was detected. The DNA interstrand cross-link formation in VA-13 cells was found to peak 12 hr after drug removal. A linear correlation between DNA interstrand cross-link formation and log cell kill was observed in VA-13 cells but not in IMR-90 cells. DNA-protein cross-link formation was found to be comparable in both cell lines for each drug, suggesting that drug penetration and intracellular drug reactivity were similar. Initial chemical decomposition studies suggest that both M&B 39565 and MCTIC may produce a chloroethyldiazo species. This species has been implicated in the formation of chloroethyl-DNA adducts which convert to DNA interstrand cross-links in mammalian cells treated with chloroethylnitrosoureas [Erickson et al., Nature (Lond.), 288: 727, 1980]. These data suggest that DNA interstrand cross-link formation may be a common mechanism for the in vitro cytotoxicity of M&B 39565 and MCTIC.


Assuntos
Transformação Celular Neoplásica , DNA/metabolismo , Imidazóis/toxicidade , Compostos de Mostarda Nitrogenada , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Dacarbazina/análogos & derivados , Dacarbazina/toxicidade , Embrião de Mamíferos , Humanos , Cinética
14.
Cancer Res ; 44(4): 1352-7, 1984 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-6704956

RESUMO

The possibility was examined that the carbamoylating activity of some chloroethylnitrosoureas could interfere with the activity of normal human cells to survive treatment with these drugs; 1-(2-chloroethyl)-3-(trans-4-hydroxycyclohexyl)-1-nitrosourea, which has strong carbamoylating activity, inhibited the rejoining of drug or X-ray-induced DNA strand breaks in IMR-90 cells, whereas the noncarbamoylating cis-2-hydroxy isomer had little or no effect; 1-(2-chloroethyl)-3-(trans-4-hydroxycyclohexyl)-1-nitrosourea was twice as potent as the cis-2-hydroxy isomer in reducing colony survival. The moderate or high carbamoylating drugs 1,3-bis(2-chloroethyl)-1-nitrosourea and 1-(2-chloroethyl)-3-(cyclohexyl)-1-nitrosourea had effects resembling those of 1-(2-chloroethyl)-3-(trans-4-hydroxycyclohexyl)-1-nitrosourea. The low carbamoylating drug 1-(2-chloroethyl)-3-(2,6-dioxo-1-piperidyl)-1-nitrosourea had effects resembling those of the cis-2-hydroxy isomer. 1-(2-chloroethyl)-1-nitrosourea, although a strong carbamoylator in chemical systems, behaved biologically as if it were a low carbamoylator. This can be rationalized on the basis of limited cellular uptake of cyanate ion. The results suggest that carbamoylation may inhibit the nucleotide excision repair of chloroethylnitrosourea-induced DNA damage that may be crucial to the ability of normal human cells to recover from the action of these drugs. Previous work has indicated that susceptible human tumor cells are sensitive to chloroethylnitrosoureas because of a lack of a DNA repair protein (guanine O6-alkyltransferase) that is not involved in nucleotide excision repair. On the basis of these findings and other evidence, further clinical trials of appropriate noncarbamoylating chloroethylnitrosoureas would be justified.


Assuntos
Carmustina/toxicidade , Reparo do DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Etilnitrosoureia/toxicidade , Lomustina/análogos & derivados , Compostos de Nitrosoureia/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , DNA/isolamento & purificação , Etilnitrosoureia/análogos & derivados , Feminino , Humanos , Lomustina/toxicidade , Pulmão/embriologia , Gravidez , Relação Estrutura-Atividade
15.
Cancer Res ; 38(3): 672-7, 1978 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-203388

RESUMO

The effects of 1,3-bis(2-chloroethyl)-1-nitrosourea on the rejoining of X-ray-induced DNA strand breaks were examined in normal human fibroblasts (WI-38) and a simian virus 40-transformed derivative (VA-13) with the use of alkaline sucrose sedimentation. 1,3-Bis(2-chloroethyl)-1-nitrosourea was capable of partially inhibiting repair of X-ray-produced DNA strand breaks in both cell types when the drug was added to the culture medium immediately after X-irradiation. However, when 1,3-bis(2-chloroethyl)-1-nitrosourea exposure preceded X-ray by 1 hr, DNA repair was inhibited to a much greater extent than it was when 1,3-bis(2-chloroethyl)-1-nitrosourea followed X-ray. The inhibition of DNA repair by 1,3-bis(2-chloroethyl)-1-nitrosourea appeared to be complete in the transformed VA-13 cells, while only partial inhibition of repair was observed in the normal WI-38 cells.


Assuntos
Carmustina/farmacologia , Transformação Celular Neoplásica , Reparo do DNA/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Reparo do DNA/efeitos da radiação , DNA de Neoplasias/efeitos da radiação , Vírus 40 dos Símios , Raios X
16.
Cancer Res ; 46(10): 4995-8, 1986 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2944576

RESUMO

The majority of human tumor cell lines are proficient in the repair of guanine O6-alkylations (designated Mer+) and are thus capable of preventing the cytotoxic effects of chloroethylating agents. It has been proposed that in these cells guanine O6-chloroethylations are rapidly removed by the enzyme O6-alkylguanine DNA alkyltransferase before the formation of DNA interstrand cross-links can occur. In this study pretreatment of four Met+ human cells (A2182 lung carcinoma, A375 melanoma, HT-29 colon carcinoma, and IMR-90 normal lung fibroblasts) with the DNA methylating agent streptozotocin apparently saturates the monoadduct repair system and allows mitozolomide to form interstrand cross-links in these cells. The inhibition of the alkyltransferase results in the continued presence of guanine O6-chloroethylations which then undergo a series of reactions that lead to DNA interstrand cross-link formation. As observed by colony forming assays, streptozotocin pretreatment causes a dramatic increase in the sensitivity of these four Mer+ cell lines to the cytotoxic effects of mitozolomide. These results indicate that a combination of streptozotocin pretreatment followed by mitozolomide may be useful in the treatment of human cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Metiltransferases/análise , Neoplasias/enzimologia , Compostos de Mostarda Nitrogenada/administração & dosagem , Estreptozocina/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular , Reparo do DNA/efeitos dos fármacos , Humanos , Neoplasias/tratamento farmacológico , O(6)-Metilguanina-DNA Metiltransferase , Fenótipo , Ensaio Tumoral de Célula-Tronco
17.
Cancer Res ; 53(18): 4281-6, 1993 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-8364924

RESUMO

This study was undertaken to ascertain the importance of prolonged depletion of O6-methylguanine DNA methyltransferase (MGMT) activity, following O6-benzylguanine (BG) and streptozotocin (STZ) exposure, in reversing 1,3 bis(2-chloroethyl)-1-nitrosourea (BCNU) resistance in vitro. We evaluated BCNU-induced cytotoxicity and measured the temporal recovery of MGMT activity in human colon carcinoma HT-29 cells following treatment with BG, STZ, or the combination of BG and STZ. The pretreatment regimens which provided the greatest potentiation of BCNU cytotoxicity were those exhibiting the greatest temporal inhibition of MGMT activity. The combination of BG (10 microM) and STZ (1.0 mM) produced sustained inhibition of MGMT activity through 24 h and potentiated BCNU cytotoxicity by at least one log greater than either agent alone. Similarly, BG (10-100 microM) produced marked reductions in MGMT activity and increased BCNU cytotoxicity in a dose-dependent fashion. A 100-microM dose of BG inhibited MGMT activity for 48 h and potentiated BCNU induced cell kill by 3 logs greater than BCNU alone. In addition, we observed that during the period of sustained inhibition of MGMT activity, no changes in the steady-state MGMT mRNA levels occurred. We conclude that prolonged inhibition of MGMT activity is an important determinant in reversing BCNU resistance and that chemotherapeutic regimens targeting the inactivation of MGMT activity should be optimized such that MGMT activity is depleted for at least 24 h following BCNU administration.


Assuntos
Carmustina/farmacologia , Guanina/análogos & derivados , Metiltransferases/metabolismo , Estreptozocina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Guanina/farmacologia , Humanos , Metiltransferases/genética , O(6)-Metilguanina-DNA Metiltransferase , RNA Mensageiro/análise , Células Tumorais Cultivadas
18.
Cancer Res ; 48(13): 3603-6, 1988 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-2837318

RESUMO

Exposure of HT29 cells in culture to O6-methylguanine is known to result in a reduction in O6-alkylguanine-DNA alkyltransferase (AGT) activity and an enhancement of sensitivity to the cytotoxic effects of chloroethylating agents. Since cytotoxicity of these agents may be mediated by the formation of interstrand cross-links, alkaline elution analysis was performed on HT29 cells treated with 1-(2-chloroethyl)-1-nitrosourea, 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea, and Clomesone [2-chloroethyl(methylsulfonyl)methanesulfonate] in the presence or absence of O6-methylguanine pretreatment to determine if the enhanced toxicity was due to an increase in the number of cross-links formed. Interstrand cross-linking by 1-(2-chloroethyl)-1-nitrosourea or 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea was increased by pretreatment with 0.4 mM O6-methylguanine for 24 h. Cross-linking by Clomesone was observed only in cells exposed to 0.4 mM O6-methylguanine for 24 h prior to administration of the drug and for 12 h after administration, suggesting that the resynthesis of the AGT may prevent the cross-linking by Clomesone. Complete recovery of AGT activity after reduction to 20 to 30% of the basal level upon treatment with 0.4 mM O6-methylguanine required between 8 h and 15 h in both HT29 cells and in Raji cells which were also sensitized to 1-(2-chloro-ethyl)-3-cyclohexyl-1-nitrosourea by exposure to O6-methylguanine. These data suggest that the enhancement of chloroethylnitrosourea toxicity after treatment with O6-methylguanine may be related to an increase in the number of DNA cross-links and that the relatively rapid rate of AGT recovery plays a role in prevention of cross-links resulting from Clomesone.


Assuntos
Reagentes de Ligações Cruzadas , Dano ao DNA , Etilnitrosoureia/análogos & derivados , Guanina/análogos & derivados , Mesilatos/metabolismo , Metiltransferases/metabolismo , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Reparo do DNA , Etilnitrosoureia/metabolismo , Etilnitrosoureia/toxicidade , Guanina/metabolismo , Humanos , Lomustina/metabolismo , Lomustina/toxicidade , Mesilatos/toxicidade , O(6)-Metilguanina-DNA Metiltransferase , Células Tumorais Cultivadas
19.
Cancer Res ; 47(17): 4538-43, 1987 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-3113720

RESUMO

Recent evidence from our laboratory and from others suggested that pretreatment with alpha-difluoromethylornithine (DFMO) sensitizes some human and rodent tumor cell lines to 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). Many human tumor cells are resistant to chloroethylnitrosourea-induced DNA interstrand cross-linking and cell kill due to their high levels of the DNA repair protein O6-alkylguanine DNA alkyltransferase. We therefore investigated DFMO-mediated sensitization to BCNU in BCNU-sensitive and -resistant cells. Colony formation assays were used to compare BCNU cytotoxicity in DFMO-pretreated and control cultures of two colon tumor lines, HT-29 cells, which have high alkyltransferase levels and thus are BCNU-resistant, and BE cells, which are deficient in this repair capacity and thus are BCNU-sensitive. Polyamine depletion significantly enhanced BCNU cytotoxicity only for the repair-proficient HT-29 cell line. BE cells were 40-fold more sensitive to BCNU than were HT-29 cultures. However, in BE cells, no effect of polyamine depletion was found on cellular response to BCNU treatment at 72 h after DFMO treatment. Reverse-phase high-performance liquid chromatography assays of polyamine concentrations in cell extracts verified that DFMO produced comparable degrees of polyamine depletion for both cell lines. DNA alkaline elution analysis was used to monitor BCNU-induced formation of DNA single strand breaks, DNA interstrand cross-links, and DNA-protein cross-links. Equal concentrations of BCNU produced similar levels of strand breaks and DNA-protein cross-links in DFMO-pretreated and control cultures for both cell lines. These data suggest that DNA in polyamine-deficient HT-29 and BE cells is not more accessible to BCNU than is DNA in controls. No DNA interstrand cross-links were detected in either DFMO-pretreated or control HT-29 cells after BCNU treatment. Further, in BE cells which accumulate BCNU-induced DNA interstrand cross-links, no increase in the measureable levels of cross-links resulted from polyamine deficiency. Our observations suggest that mechanisms other than increased DNA interstrand cross-link formation may be mediating the enhanced efficacy of BCNU in polyamine-deficient HT-29 cell cultures. Our findings may also imply that cellular targets for BCNU other than DNA damage may be responsible for DFMO-induced chemosensitization in the repair-proficient cells.


Assuntos
Carmustina/farmacologia , Dano ao DNA , DNA de Neoplasias/metabolismo , Poliaminas/análise , Adenocarcinoma/análise , Adenocarcinoma/patologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Eflornitina/farmacologia , Humanos
20.
Cancer Res ; 51(6): 1581-5, 1991 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-1825618

RESUMO

Treatment of chloroethylnitrosourea-resistant cells with streptozotocin (STZ) prior to bis-chloroethylnitrosourea (BCNU) exposure has been shown to result in a depletion of O6-methylguanine DNA methyltransferase (MGMT) activity, increased BCNU-induced interstrand cross-linking, and a 2-3 log enhancement of BCNU cytotoxicity in vitro. The current study was undertaken to define the kinetics of repletion of MGMT activity following the STZ/BCNU combination and to assess at the molecular level the effects of the combination on MGMT mRNA expression. Results demonstrate that MGMT activity can be depleted by greater than 90% relative to untreated controls using an optimized STZ/BCNU combination regimen and that greater than 50% depletion can be maintained for at least 24 h. This depletion appears to be independent of effects at the mRNA level because neither STZ alone nor the STZ/BCNU combination significantly altered steady state levels of MGMT mRNA. Cytotoxicity studies are consistent with MGMT repletion data and demonstrate that, as the interval between STZ and BCNU exposures increases, the degree of enhanced cytotoxicity induced by the combination relative to BCNU alone decreases. These results suggest that the enhanced cytotoxicity induced by the STZ/BCNU combination over BCNU treatment alone is favored by both the lack of induction of expression of MGMT mRNA and by slow reappearance of MGMT activity.


Assuntos
Carmustina/farmacologia , Metiltransferases/análise , RNA Mensageiro/análise , Estreptozocina/farmacologia , Combinação de Medicamentos , Metiltransferases/genética , O(6)-Metilguanina-DNA Metiltransferase , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA