Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Natl Cancer Inst ; 83(9): 632-7, 1991 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-2023281

RESUMO

The distribution of porfiromycin was studied in BALB/c mice bearing EMT6 mammary tumors. The levels of 3H in blood and most tissues peaked approximately 15 minutes after intraperitoneal injection of [3H]porfiromycin. The levels of radioactivity present in most of the tissues and in the tumors were similar at 4 hours and 24 hours after administration. Most of the normal tissues showed uniform, low grain densities when analyzed by autoradiography; the liver and the small intestine had the highest labeling densities. Only kidney, bladder, and tumor showed differential distributions of grains from [3H]porfiromycin. In the kidney, higher grain counts were found in cortex than in medullary regions; grains were uniformly distributed within each region. In the bladder, the highest labeling densities were found in regions near the lumen. Tumor regions that had some necrotic features or regions of necrosis that included some viable cells showed higher labeling intensities than healthy-looking tumor regions, probably because the abnormal microenvironments in these regions led to increased rates of activation of porfiromycin to electrophilic species. These findings show that porfiromycin can reach and be activated in tumor regions containing cells resistant to many chemotherapeutic agents and to x rays. The results also support the concept that agents such as porfiromycin can target cells in specific microenvironmental subpopulations of solid tumors.


Assuntos
Neoplasias Mamárias Experimentais/metabolismo , Porfiromicina/farmacocinética , Animais , Medula Óssea/metabolismo , Encéfalo/metabolismo , Injeções Intraperitoneais , Intestino Delgado/metabolismo , Rim/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Miocárdio/metabolismo , Distribuição Tecidual , Bexiga Urinária/metabolismo
2.
Cancer Res ; 47(21): 5654-7, 1987 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-3664473

RESUMO

Mitomycin C and its methylated analogue porfiromycin (Por) have significant potential as adjuncts to regimens presently used for treating solid tumors because of their preferential toxicity to cells existing in an hypoxic environment. An understanding of the factors producing the differential activity of these drugs under aerobic and hypoxic conditions would facilitate the development of new agents of this class. Previous studies have focused on the enzymes that reductively activate the mitomycins and on the interaction of these drugs with DNA; none of these studies has fully explained the differences in cytotoxicity observed under hypoxic and aerobic conditions. The present investigation demonstrates that the rate of Por uptake is directly correlated with cytotoxicity under both aerobic and hypoxic conditions. Uptake of Por into hypoxic cells is more rapid than into aerobic cells at equal drug concentrations. Hypoxic cells also accumulate drug in concentrations well in excess of those in the extracellular medium; this is apparently a reflection of drug sequestration in these cells. This sequestration of Por, which affects the rate and extent of uptake in hypoxic cells, does not take place in aerobic cells. The failure of aerobic cells to sequester drug is evidenced by the very rapid efflux of Por from these cells upon removal of extracellular Por and by the fact that aerobic cells attain a state of equilibrium between the intracellular and extracellular drug concentrations. The findings demonstrate that differences in the uptake and retention of Por are associated with the preferential toxicity of Por to hypoxic cells.


Assuntos
Mitomicinas/farmacocinética , Oxigênio/farmacologia , Porfiromicina/farmacocinética , Animais , Sobrevivência Celular/efeitos dos fármacos , DNA/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Porfiromicina/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos
3.
Cancer Res ; 51(19): 5275-83, 1991 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-1717144

RESUMO

Stable acquired resistance to etoposide (VP-16) or teniposide (VM-26) in HCT116 human colon carcinoma cells and A549 human lung adenocarcinoma cells, was previously obtained by weekly 1-h exposures to either drug (B. H. Long, Natl. Cancer Inst. Monogr., 4: 123-127, 1987). The purpose of this study was to identify possible mechanisms of resistance present in these cells by using human mdr1 and topoisomerase II DNA probes, antibodies to these gene products, and P4 phage unknotting assay for topoisomerase II activities. HCT116(VP)35 cells were 9-, 7-, and 6-fold resistant to VP-16, VM-26, and Adriamycin, respectively, and showed no cross-resistance to colchicine and actinomycin D. These cells had no differences in mdr1 gene, mdr1 mRNA, or P-glycoprotein levels but displayed decreased levels of topoisomerase II mRNA and enzyme activity without any alteration of drug sensitivity displayed by the enzyme. HCT116(VM)34 cells were 5-, 7-, and 21-fold resistant to VP-16, VM-26, and Adriamycin; were cross-resistant to colchicine (7-fold) and actinomycin D (18-fold); and possessed a 9-fold increase in mdr1 mRNA and increased P-glycoprotein without evidence of mdr1 gene amplification. No alterations in topoisomerase II gene or mRNA levels, enzyme activity, or drug sensitivity were observed. A549(VP)28 and A549(VM)28 cells were 8-fold resistant to VP-16 and VM-26 and 3-fold resistant to Adriamycin. Both lines were not cross-resistant to colchicine or actinomycin D but were hypersensitive to cis-platinum. No alterations in mdr1 gene, mdr1 mRNA, or P-glycoprotein levels, but lower topoisomerase II mRNA levels and decreased enzyme activities, were observed. Of the four acquired resistant cell lines, resistance is likely related to elevated mdr1 expression in one line and to decreased topoisomerase II expression in the other three lines.


Assuntos
Adenocarcinoma/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Etoposídeo/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Teniposídeo/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP , Sequência de Bases , Linhagem Celular , DNA/análise , DNA/efeitos dos fármacos , Dano ao DNA , Sondas de DNA , DNA Topoisomerases Tipo II/biossíntese , DNA Topoisomerases Tipo II/genética , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Resistência a Medicamentos/genética , Etoposídeo/farmacocinética , Humanos , Immunoblotting , Glicoproteínas de Membrana/biossíntese , Dados de Sequência Molecular , Proteínas de Neoplasias/biossíntese , Porfiromicina/farmacocinética , Porfiromicina/farmacologia , RNA/análise
4.
Cancer Chemother Pharmacol ; 31(1): 23-31, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1458556

RESUMO

A mitomycin C (MMC)- and porfiromycin (PFM)-resistant subline of the HCT 116 human colon-cancer cell line was isolated after repeated exposure of HCT 116 cells to increasing concentrations of MMC under aerobic conditions. The MMC-resistant subline (designated HCT 116-R30A) was 5 times more resistant than the parent cells to MMC and PFM under aerobic conditions. Both the MMC-resistant cells and the parent HCT 116 cells accumulated similar amounts of PFM by passive diffusion, but levels of macromolecule-bound PFM were about 50% lower in the resistant cell line, implying a decrease in PFM reductive activation in the resistant cells. The finding that microsomes from either sensitive or resistant cells showed an equal ability to reduce MMC and PFM indicated that the activity of NADPH cytochrome P-450 reductase (EC 1.6.2.4) was not changed in the resistant subline. Soluble extracts of HCT 116 cells reduced MMC and PFM more effectively at pH 6.1, and NADH and NADPH were utilized equally well as electron donors under both aerobic and anaerobic conditions. These data suggest that quinone reductase (EC 1.6.99.2; DT-diaphorase) in soluble extracts is responsible for the reduction of MMC. Quinone reductase activities in soluble extracts of HCT 116-R30A cells for the reduction of dichlorophenol indophenol (DCPIP) and menadione-cytochrome c at optimal pHs were decreased by 95% as compared with those obtained in parent cells. However, the MMC-reducing activity of HCT 116-R30A soluble extracts was only 50% lower than that of the parent cell extracts. The kinetic constants (Km, Vmax) found for quinone reductase in the two cell lines with respect to the substrates DCPIP and menadione differed. Two species of mRNA for quinone reductase (2.7 and 1.2 kb) were detected in both cell lines, and there was no detectable difference between parent and resistant cells in the steady-state level of either of these mRNA species. Furthermore, incubation with the quinone reductase inhibitor dicoumarol rendered HCT 116 cells more resistant to MMC. Alteration of the quinone reductase activity in HCT 116-R30A cells appears to be the mechanism responsible for their resistance to MMC and PFM.


Assuntos
Neoplasias do Colo/enzimologia , Mitomicina/farmacologia , NAD(P)H Desidrogenase (Quinona)/fisiologia , Porfiromicina/farmacologia , 2,6-Dicloroindofenol/metabolismo , Radioisótopos de Carbono , Extratos Celulares , Tamanho Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Resistência a Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Líquido Intracelular/metabolismo , Microssomos/metabolismo , Mitomicina/metabolismo , NAD(P)H Desidrogenase (Quinona)/genética , Oxirredução , Porfiromicina/farmacocinética , RNA Mensageiro/genética , Distribuição Tecidual , Transcrição Gênica/genética , Células Tumorais Cultivadas/efeitos dos fármacos , Vitamina K/metabolismo
5.
Adv Enzyme Regul ; 35: 117-30, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7572339

RESUMO

Hypoxic cells in solid tumors represent a therapeutically resistant population that limits the curability of many solid tumors by irradiation and by most chemotherapeutic agents. The oxygen deficit, however, creates an environment conducive to reductive processes; this results in a major exploitable difference between normal and neoplastic tissues. The mitomycin antibiotics can be reductively activated by a number of oxidoreductases, in a process required for the production of their therapeutic effects. Preferential activation of these drugs under hypoxia and greater toxicity to oxygen-deficient cells than to their oxygenated counterparts are obtained in most instances. The demonstration that mitomycin C and porfiromycin, used to kill the hypoxic fraction, in combination with irradiation, to eradicate the oxygenated portion of the tumor, produced enhanced cytodestructive effects on solid tumors in animals has led to the clinical evaluation of the mitomycins in combination with radiation therapy in patients with head and neck cancer. The findings from these clinical trials have demonstrated the value of directing a concerted therapeutic attack on the hypoxic fraction of solid tumors as an approach toward enhancing the curability of localized neoplasms by irradiation.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Hipóxia Celular , Neoplasias Mamárias Experimentais/patologia , Porfiromicina/farmacologia , Animais , Antibióticos Antineoplásicos/farmacocinética , Sobrevivência Celular/efeitos dos fármacos , Radicais Livres/metabolismo , Radical Hidroxila/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mitomicina/farmacocinética , Mitomicina/farmacologia , Estrutura Molecular , NADPH-Ferri-Hemoproteína Redutase , Oxirredutases/metabolismo , Consumo de Oxigênio , Porfiromicina/farmacocinética
6.
Adv Enzyme Regul ; 33: 3-17, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-8356915

RESUMO

The mitomycin antibiotics, because of their preferential toxicities for hypoxic cells, have significant potential as adjuncts to ionizing radiation in the treatment of solid tumors. To gain information on the mechanism by which these agents exert their cytotoxicities to hypoxic and aerobic cells, the effects of MC, POR and several of their analogs were studied in EMT6 mammary carcinoma cells. The rate of uptake of POR by these cells was directly correlated with the cytotoxicity produced by this agent under both hypoxia and aeration. At equivalent concentrations, uptake of POR into hypoxic cells was more rapid than into aerobic cells. Hypoxic cells also accumulated the antibiotic in concentrations well in excess of that present in the extracellular medium, presumably as a result of reductive activation and covalent binding of POR to cellular structures. Such activation and binding occur to a much lesser degree in aerated cells, resulting in the rapid efflux of POR from these cells when the antibiotic is removed from the extracellular environment. To gain information on the reaction of POR with DNA, mono- and bis-adducts formed in EMT6 cells exposed to this agent were measured. Three major adducts were formed. Two were mono-adducts consisting of deoxyguanosine linked at its N2-position to the C-1 of POR and of 10-decarbamoyl POR. The third was a bis-adduct in which POR was cross-linked to two deoxyguanosines at their N2-positions. More adducts were formed in hypoxia than in air, and more bis-adducts were present in hypoxic cells. Simultaneous exposure of cells to both POR and DIC reduced the total adduct level and a new unknown adduct was formed, primarily under hypoxia. Several mitomycins were evaluated for their capacity to kill EMT6 cells and to produce DNA cross-links in both hypoxia and aeration. The number of cross-links required to produce a given amount of cell kill was similar, regardless of the mitomycin employed or the degree of oxygenation. The findings support the concept that DNA is a critical target in the action of the mitomycins and that cross-linking of the DNA creates an important lesion for cytodestruction.


Assuntos
Mitomicinas/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Animais , Transporte Biológico Ativo , Morte Celular/efeitos dos fármacos , Hipóxia Celular , Reagentes de Ligações Cruzadas/farmacologia , Dano ao DNA , DNA de Neoplasias/efeitos dos fármacos , Camundongos , Mitomicinas/farmacocinética , Oxigênio/metabolismo , Porfiromicina/farmacocinética , Porfiromicina/farmacologia , Células Tumorais Cultivadas/metabolismo
7.
J Pharm Sci ; 89(2): 191-8, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10688748

RESUMO

The isolation and identification of the major metabolites of porfiromycin formed in the presence of a rat liver preparation under aerobic conditions were performed with high-performance liquid chromatography and electrospray ionization mass spectrometry. Porfiromycin was extensively metabolized by the rat liver preparation in an aqueous 0.1 M potassium phosphate buffer (pH 7.4) containing an NADPH generating system at 37 degrees C. A total of eight metabolites was identified as mitosene analogs. Of these, three primary metabolites are 2-methylamino-7-aminomitosene, 1,2-cis and 1,2-trans-1-hydroxy-2-methylamino-7-aminomitosene, which are consistent with those previously observed in hypoxia using purified rat liver NADPH-cytochrome c reductase. Interestingly, 2-methylamino-7-aminomitosene is a reactive metabolite, which undergoes further activation at the C-10 position by the loss of carbamic acid and then links with the 7-amino group of the primary metabolites to yield two dimeric adducts. In addition, three phosphate adducts, 10-decarbamoyl-2-methylamino-7-aminomitosene-10-phosphate, 1,2-cis and 1,2-trans-2-methylamino-7-aminomitosene-1-phosphate, were also identified in the incubation system. The configurations of the diastereoisomeric metabolites were determined with (1)HNMR and phosphatase digestion. On the basis of the metabolite profile, we propose in vitro metabolic pathways for porfiromycin. The findings provide direct evidence for understanding the reactive nature and hepatic metabolism of the drug currently in phase III clinical trials.


Assuntos
Antibióticos Antineoplásicos/metabolismo , Fígado/metabolismo , Porfiromicina/isolamento & purificação , Porfiromicina/metabolismo , Acetilcisteína/metabolismo , Animais , Antibióticos Antineoplásicos/isolamento & purificação , Biotransformação , Cromatografia Líquida de Alta Pressão , Cisteína/metabolismo , Fígado/química , Espectrometria de Massas , Ressonância Magnética Nuclear Biomolecular , Porfiromicina/farmacocinética , Ratos , Ratos Sprague-Dawley , Espectrofotometria Ultravioleta
8.
Br J Cancer ; 59(3): 341-6, 1989 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2467684

RESUMO

Two non-transformed human skin fibroblast strains, GM38 and 3437T, were found to be more sensitive to the bioreductive alkylating agents mitomycin C (MMC) and porfiromycin (PM) under hypoxic compared to aerobic conditions. One of these strains, 3437T, was 6-7 times more resistant to these agents under aerobic exposure conditions, but was identical in sensitivity to the normal strain, GM38, under hypoxic conditions. Aerobic 3437T cells demonstrated no increased resistance to cisplatin compared to the normal strain, arguing against enhanced ability to repair DNA interstrand cross-links as the underlying explanation for the mitomycin resistance. The aerobic resistance of 3437T was not altered by dicumarol, an inhibitor of the enzyme DT-diaphorase which is believed to be involved in aerobic activation of MMC and PM. Dicumarol did increase the resistance of GM38, but not to the same level of resistance demonstrated by 3437T. These results suggest that the aerobic MMC and PM resistance of 3437T may arise, in part, from a deficiency in DT-diaphorase activity. The identical sensitivities under hypoxic conditions indicate that drug activation pathways operative in the absence of oxygen are similar in both the normal and 3437T cells.


Assuntos
Mitomicinas/farmacocinética , Oxigênio/metabolismo , Pele/citologia , Biotransformação , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Dicumarol/farmacologia , Resistência a Medicamentos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Mitomicina , Mitomicinas/farmacologia , Porfiromicina/farmacocinética , Porfiromicina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA