Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Leukemia ; 20(10): 1809-18, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16932348

RESUMO

Among the topoisomerase (topo) II isozymes (alpha and beta), topo IIbeta has been suggested to regulate differentiation. In this study, we examined the role of topo IIbeta in all-trans retinoic acid (ATRA)-induced differentiation of myeloid leukemia cell lines. Inhibition of topo IIbeta activity or downregulation of protein expression enhanced ATRA-induced differentiation/growth arrest and apoptosis. ATRA-induced apoptosis in topo IIbeta-deficient cells involved activation of the caspase cascade and was rescued by ectopic expression of topo IIbeta. Gene expression profiling led to the identification of peroxiredoxin 2 (PRDX2) as a candidate gene that was downregulated in topo IIbeta-deficient cells. Reduced expression of PRDX2 validated at the mRNA and protein level, in topo IIbeta-deficient cells correlated with increased accumulation of reactive oxygen species (ROS) following ATRA-induced differentiation. Overexpression of PRDX2 in topo IIbeta-deficient cells led to reduced accumulation of ROS and partially reversed ATRA-induced apoptosis. These results support a role for topo IIbeta in survival of ATRA-differentiated myeloid leukemia cells. Reduced expression of topo IIbeta induces apoptosis in part by impairing the anti-oxidant capacity of the cell owing to downregulation of PRDX2. Thus, suppression of topo IIbeta and/or PRDX2 levels in myeloid leukemia cells provides a novel approach for improving ATRA-based differentiation therapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Leucemia Mieloide/metabolismo , Tretinoína/farmacologia , Apoptose/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , DNA Topoisomerases Tipo II/genética , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Dicetopiperazinas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Células HL-60 , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Leucemia Mieloide/patologia , Leucemia Mieloide/fisiopatologia , Peroxidases/genética , Peroxidases/metabolismo , Peroxirredoxinas , Piperazinas/farmacologia , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Inibidores da Topoisomerase II
2.
J Natl Cancer Inst ; 68(6): 1027-32, 1982 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-6953267

RESUMO

Adriamycin (ADR) (NSC-123127) uptake and retention in ADR-sensitive P388 leukemia (P388/S) and ADR-resistant P388 leukemia (p388/R) cells were compared by fluorometry and laser flow cytometry (FCM) and were correlated with cytotoxic effects. Drug levels in P388/R cells treated in vitro with ADR (1-10 micrograms/ml) were twofold to fourfold lower than were levels in similarly treated P388/S cells FCM analysis of P388/S and P388/R cells exposed in vitro to ADR showed qualitative and quantitative differences in ADR fluorescence profiles of drug-treated cells (1-5 micrograms/ml) but not of the isolated nuclei (0.5- 10 micrograms/ml). Drug-induced perturbations in cell cycle traverse and chromosome aberrations were seen in P388/S but not in P388/R cells treated with 0.5-5 micrograms ADR/ml in vitro or 4-8 mg ADR/kg in vivo. The role of FCM in rapidly comparing and quantitating cellular ADR fluorescence profiles of ADR-sensitive and ADR-resistant tumors was demonstrated.


Assuntos
Doxorrubicina/metabolismo , Leucemia P388/metabolismo , Leucemia Experimental/metabolismo , Animais , Transporte Biológico , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Núcleo Celular/análise , Aberrações Cromossômicas , Doxorrubicina/análise , Resistência a Medicamentos , Citometria de Fluxo , Fluorometria , Leucemia P388/análise , Camundongos
3.
Cancer Res ; 43(8): 3696-9, 1983 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-6861140

RESUMO

Resistance to the cytotoxic effects of daunomycin and Adriamycin (ADR) in sublines of Ehrlich ascites and P388 mouse tumors has been demonstrated to be due to reduced cellular accumulation and retention of drug. In this study, the effect of the calmodulin inhibitor trifluoperazine on the cellular accumulation, retention, and cytotoxic effects of ADR in ADR-sensitive (P388/S) and ADR-resistant (P388/R) P388 mouse leukemia cells was determined. In cells treated in suspension culture for 24 hr or for 1 hr followed by plating in soft agar, a noncytotoxic concentration of 4 microM trifluoperazine, enhanced the sensitivity to ADR 2- to 6-fold in P388/R but not in P388/S cells. A marked enhancement in cellular retention rather than accumulation of ADR in only P388/R cells was obtained with trifluoperazine treatment. This study suggests the possible novel use of phenothiazines to improve drug sensitivity of tumors resistant to ADR treatment.


Assuntos
Doxorrubicina/uso terapêutico , Leucemia P388/tratamento farmacológico , Leucemia Experimental/tratamento farmacológico , Trifluoperazina/uso terapêutico , Animais , Calmodulina/antagonistas & inibidores , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Resistência a Medicamentos , Sinergismo Farmacológico , Camundongos
4.
Cancer Res ; 40(11): 3895-900, 1980 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-6937236

RESUMO

We have used a laser flow cytometer to excite and quantitate the intracellular fluorescence of cells exposed in vitro and in vivo to various anthracyclines. In cells exposed to Adriamycin (ADR), intracellular drug fluorescence appeared slowly and reached a peak after 4 hr of incubation. Cells incubated with 10 micrograms/ml were 5 times more fluorescent than were cells incubated with 1 microgram/ml. Cells exposed to daunomycin were 2 to 4 times more fluorescent than were cells similarly exposed to ADR, and the intracellular appearance of daunomycin fluorescence was much more rapid. Cells exposed to N-trifluoroacetyladriamycin and carminomycin had higher amounts of intracellular fluorescence (2 to 4 times), and peak values were reached much more rapidly than in cells exposed to ADR. In cells exposed to rubidazone, fluorescence increased 2- to 4-fold with increased drug concentration and length of exposure. In contrast, nogalamycin fluorescence reached a peak after 60 min of incubation, and a 10-fold increase in drug concentration increased fluorescence only 2-fold. In animals given injections of ADR (4 mg/kg) and sacrificed after 3 hr, drug fluorescence could be detected in tumor and spleen cells. In contrast, fluorescence in heart nuclei was barely recognizable. However, incubation of isolated nuclei in ADR (1 microgram/ml) showed that bone marrow and heart nuclei had greater amounts of ADR fluorescence (2- to 3-fold) than did spleen or liver nuclei similarly treated. The use of laser flow cytometry for monitoring intracellular anthracycline transport, binding, and efflux is demonstrated.


Assuntos
Naftacenos/metabolismo , Antibióticos Antineoplásicos/metabolismo , Carrubicina/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Daunorrubicina/análogos & derivados , Daunorrubicina/metabolismo , Doxorrubicina/análogos & derivados , Doxorrubicina/metabolismo , Glicosídeos/metabolismo , Humanos , Lasers , Microscopia de Fluorescência , Nogalamicina/metabolismo , Espectrometria de Fluorescência
5.
Cancer Res ; 40(4): 1103-8, 1980 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7357541

RESUMO

Administration of 2,3-dihydro-1 H-imidazo[1,2-b]pyrazole (IMPY; NSC 51143), 250 to 500 mg/kg, in Day 5 L1210 and P388 (ascites) tumor-bearing mice did not consistently prolong the life span of tumor-bearing animals. Flow cytometry and autoradiographic studies showed that, after 12 to 18 hr of a single IMPY injection, both P388 and L1210 tumor cells were synchronized in S phase. In contrast, IMPY inhibited cellular proliferation in both bone marrow and duodenal crypts during the first 24 hr. and a recovery was detectable only after 36 hr, returning to pretherapy values by 72 hr. Preliminary data indicate that this differential response of normal versus tumor cells to IMPY can be exploited to maximize chemotherapeutic efficacy in scheduled chemotherapy with cycle-specific agents.


Assuntos
Divisão Celular/efeitos dos fármacos , Leucemia L1210/tratamento farmacológico , Leucemia P388/tratamento farmacológico , Leucemia Experimental/tratamento farmacológico , Pirazóis/farmacologia , Animais , Medula Óssea/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Duodeno/efeitos dos fármacos , Imidazóis/farmacologia , Interfase/efeitos dos fármacos , Leucemia L1210/metabolismo , Leucemia P388/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Fatores de Tempo
6.
Cancer Res ; 53(14): 3262-5, 1993 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-8100737

RESUMO

Tumor cell resistance to chemotherapeutic agents of diverse structure and mechanism of action is thought to be due to efflux of drug by P-glycoprotein, which is overexpressed in tumor cells with the multidrug-resistant phenotype. Agents generally associated with the multidrug-resistant phenotype include inhibitors of topoisomerase II, e.g., doxorubicin, etoposide, and the microtubule poisons such as vinblastine, vincristine (VCR), and taxol. The antiepileptic drug phenytoin (DPH), an inhibitor of tubulin polymerization, potentiates (P < 0.05) the cytotoxicity of the chemotherapeutically useful microtubule poison VCR in tumor cells with the wild-type or multidrug-resistant phenotype. Among agents associated with the multidrug-resistant phenotype, the modulation of cytotoxicity by DPH was selectively effective with the microtubule poison VCR but not the topoisomerase II inhibitor doxorubicin. The potentiation of vincristine cytotoxicity by DPH was not due to binding to P-glycoprotein or by increasing VCR accumulation. We thus propose a novel mechanism for the modulation of resistance based on evidence that DPH at noncytotoxic concentrations can selectively enhance the cytotoxic potential of vincristine without interfering with P-glycoprotein function. Thus, studies with phenytoin could assist in characterizing other molecular determinants of multidrug resistance and the design of trials to modulate drug efficacy.


Assuntos
Fenitoína/farmacologia , Vincristina/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP , Animais , Proteínas de Transporte/metabolismo , Doxorrubicina/farmacologia , Interações Medicamentosas , Resistência a Medicamentos/genética , Leucemia L1210/tratamento farmacológico , Leucemia L1210/metabolismo , Leucemia P388/tratamento farmacológico , Leucemia P388/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Fenótipo , Células Tumorais Cultivadas , Vincristina/farmacocinética
7.
Cancer Res ; 46(8): 4171-7, 1986 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3731084

RESUMO

B16-BL6 murine melanoma cells were selected for cytogenetic evaluation during the stepwise development of increasing resistance in vitro to the antitumor antibiotic, doxorubicin (DOX). Karyotypic studies demonstrated extensive heteroploidy with both numerical and structural abnormalities which were not present in the parental DOX-sensitive B16-BL6 cells. Trypsin-Giemsa banding revealed the presence of several marker chromosomes containing abnormally banding regions (ABRs) in the 44-fold B16-BL6 DOX-resistant subline. These ABRs appeared to be more homogeneously staining at the higher DOX concentrations. Length measurements (ABR index) in seven banded metaphases indicated a direct correlation with increasing DOX concentration. When the DOX-resistant cells were grown in drug-free medium for 1 yr, the drug-resistant phenotype gradually declined in parallel with the level of resistance and the ABR index. DOX-induced cytogenetic damage examined by sister chromatid exchange methodology in parental B16-BL6 cells indicated a linear sister chromatid exchange:DOX dose-response relationship. However, after continuous treatment of parental B16-BL6 cells with DOX (0.01 microgram/ml) for 30 days, sister chromatid exchange scores were found to return to base-line values. The B16-BL6 resistant cells demonstrated a cross-resistant phenotype with N-trifluoroacetyladriamycin-14-valerate, actinomycin D, and the Vinca alkaloids but not with 1-beta-D-arabinofuranosylcytosine. The results suggest that ABR-containing chromosomes in DOX-resistant sublines may represent cytogenetic alterations of specific amplified genes involved in the expression of DOX resistance. Further studies are required to identify and define the possible gene products and to correlate their relationship to the cytotoxic action of doxorubicin.


Assuntos
Aberrações Cromossômicas , Bandeamento Cromossômico , Doxorrubicina/farmacologia , Melanoma/genética , Animais , Células Cultivadas , Resistência a Medicamentos , Amplificação de Genes , Glicoproteínas/análise , Camundongos , Peso Molecular , Fenótipo , Troca de Cromátide Irmã
8.
Cancer Res ; 38(11 Pt 1): 3656-62, 1978 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-698925

RESUMO

Adriamycin (ADR) and N-trifluoroacetyladriamycin-14-valerate, respectively, inhibit and enhance the nuclear fluorescence of cells stained with propidium iodide for DNA per cell estimation by flow cytometry. In cells incubated with ADR, the reduction in fluorescence is gradually manifested due to the slow intracellular drug transport. In contrast the effect of N-trifluoroacetyladriamycin-14-valerate on propidium iodide nuclear fluorescence is seen within 5 min of incubation. The effect of ADR on propidium iodide nuclear fluorescence could be detected in vivo even after 24 hr of ADR administration.


Assuntos
Núcleo Celular/efeitos dos fármacos , DNA/análise , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Fenantridinas , Propídio , Animais , Núcleo Celular/análise , Núcleo Celular/metabolismo , Doxorrubicina/metabolismo , Humanos , Técnicas In Vitro , Linfócitos/análise , Linfócitos/efeitos dos fármacos , Masculino , Camundongos , Neoplasias Experimentais/análise , Fenantridinas/metabolismo , Propídio/metabolismo , Espectrometria de Fluorescência , Fatores de Tempo
9.
Cancer Res ; 44(11): 5056-61, 1984 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-6488165

RESUMO

Calmodulin inhibitors enhance cytotoxic effects of doxorubicin (DOX) in DOX-resistant (P388/DOX) P388 mouse leukemia cells by increasing cellular accumulation and retention of drug. In P388/DOX cells treated for 3 hr, cytotoxic effects (based on colony formation in soft agar) of daunorubicin (DAU) in the presence of trifluoperazine (TFP) were DAU concentration-dependent and enhanced 2- to 100-fold. Additionally, in the presence of TFP, on a molar basis, equitoxic doses of DAU were 4-fold lower than DOX for P388/DOX cells. However, in P388/DOX cells treated for 3 hr with other anthracyclines, except for a slight enhancement in the cytotoxic effects of aclacinomycin A (ACM) with TFP, colony formation in soft agar of cells treated with N-trifluoroacetyladriamycin-14-valerate (AD32) and N-trifluoroacetyladriamycin were similar in the absence and presence of TFP. In DOX-sensitive (P388/S) P388 mouse leukemia cells treated for 3 hr, some enhancement in the cytotoxic effects due to TFP were observed with DAU and DOX but not with ACM, AD32, or N-trifluoroacetyladriamycin. Although accumulation of ACM and AD32 in P388/S and P388/DOX cells was similar and unaffected by TFP, the retention of ACM but not AD32 was enhanced 1.5-fold only in TFP-treated P388/DOX cells. In contrast, DAU accumulation in P388/S cells was 4-fold higher than in similarly treated P388/DOX cells, and the 2- and 4-fold increase due to TFP in the accumulation and retention, respectively, of DAU in P388/DOX cells was not observed in P388/S cells. Results from this study indicate that in P388/DOX cells, the calmodulin inhibitor TFP is more effective with DAU than DOX, significantly less effective with ACM, and ineffective with AD32 and N-trifluoroacetyladriamycin.


Assuntos
Daunorrubicina/toxicidade , Doxorrubicina/toxicidade , Leucemia P388/patologia , Leucemia Experimental/patologia , Trifluoperazina/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Daunorrubicina/metabolismo , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Cinética , Camundongos
10.
Cancer Res ; 41(7): 2745-50, 1981 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-6972801

RESUMO

N-Trifluoroacetyladriamycin-14-valerate differs from Adriamycin in its rapid intracellular transport and lack of fluorescent binding to nuclei or chromosomes. Both of these anthracyclines cause inhibition in the incorporation of labeled precursors into nucleic acids, extensive chromosomal damage, and arrest of cell cycle traverse in G2. In human lymphoid cells, N-trifluoroacetyladriamycin-14-valerate, unlike Adriamycin, does not show cell cycle phase-specific or proliferation-related cytotoxic effects. In an L1210 soft-agar assay, both Adriamycin and N-trifluoroacetyladriamycin-14-valerate show no enhanced sensitivity of mid-S-phase cells to their cytotoxic action.


Assuntos
Aberrações Cromossômicas , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Timidina/metabolismo , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Interfase/efeitos dos fármacos , Cinética , Leucemia L1210/metabolismo , Leucemia L1210/patologia , Camundongos , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
11.
Cancer Res ; 47(13): 3464-8, 1987 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-2884031

RESUMO

Adriamycin (ADR)-resistant sublines of B16-BL6 mouse melanoma selected by exposure to increasing concentrations of ADR were characterized in vitro for growth properties and in vivo for tumorigenicity and pulmonary metastases. The progressively resistant sublines adapted to grow in the presence of 0.025, 0.05, 0.1, and 0.25 microgram/ml ADR in monolayer culture were found to be 5-, 10-, 20-, and 40-fold ADR-resistant, respectively, compared to the parental sensitive cells, using a soft-agar colony assay and continuous ADR treatment for 7 days. The doubling time in monolayer culture of the parent sensitive and progressively ADR-resistant sublines of B16-BL6 melanoma cells was approximately 16-18 h. Although the colony-forming efficiency in soft agar of parental sensitive cells was only 0.5-4%, the 5-, 10-, 20-, and 40-fold ADR-resistant sublines had colony-forming efficiencies of 15, 20, 30, and 77%, respectively. Tumorigenicity in C57BL/6 mice of progressively ADR-resistant sublines was similar to parental sensitive cells following s.c. and i.p. implantation of 10(5)-10(6) tumor cells. Experimental pulmonary metastases were significantly lower in ADR-resistant sublines with progressive resistance. Additionally, unlike the parental sensitive and 5-fold ADR-resistant B16-BL6 cells, the 10-, 20-, and 40-fold ADR-resistant sublines were spontaneously nonmetastatic. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunochemical detection of P-glycoprotein revealed the presence of a Mr 170,000 plasma membrane glycoprotein in the 40-fold ADR-resistant subline and its counterpart maintained for 1 year in ADR-free medium. Results from this study suggest that progressively ADR-resistant B16-BL6 mouse melanoma cells selected in vitro demonstrate a marked increase in colony formation in soft agar and a decrease in the ability to produce pulmonary metastases, without alterations in tumorigenicity.


Assuntos
Resistência a Medicamentos , Glicoproteínas/metabolismo , Melanoma Experimental/fisiopatologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP , Animais , Divisão Celular , Doxorrubicina/farmacologia , Neoplasias Pulmonares/secundário , Melanoma Experimental/patologia , Camundongos , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia
12.
Cancer Res ; 46(11): 5553-7, 1986 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-3756902

RESUMO

1,3,7-Trimethylxanthine "caffeine" (CAF) is reported to induce a differential effect on the cytotoxicity of the DNA intercalators actinomycin-D versus Adriamycin (ADR). In the present study the effect of caffeine and/or trifluoperazine in modulating cell cycle traverse, drug accumulation, and cytotoxicity of anthracyclines was evaluated. The survival in soft agar of P388 mouse leukemia cells treated with ADR (0.05-0.25 micrograms/ml) alone for 1 h was 1.2- to 3-fold lower when the cells were incubated for 24 h in drug-free medium versus medium supplemented with 2 mM CAF. In contrast, for P388 cells treated with ADR in the presence of 2 mM CAF for 1 h and subsequently incubated for 24 h in the absence or presence of 2 mM CAF, cell killed based on colony formation in soft agar was 2- to 20-fold lower than in ADR-treated cells never exposed to 2 mM CAF. In cells treated continuously for 24 h with ADR (0.01-0.05 micrograms/ml) or the DNA nonbinding ADR analogue N-trifluoro-acetyladriamycin-14-valerate (AD32) (0.05 and 0.1 micrograms/ml) the survival in soft agar was 3- to 20-fold higher in the presence versus the absence of 2 mM CAF. The decreased cytotoxicity in cells treated with ADR or AD32 in the presence of CAF was accompanied by a significant reduction in the accumulation of cells in G2. However, in cells treated with ADR or AD32 in the presence of 2 mM CAF plus 5 microM trifluoperazine the decreased G2 accumulation was not accompanied by a reduction in anthracycline cytotoxicity. The modulation by CAF of ADR and AD32 cytotoxicity did not correlate with decreased cellular ADR and AD32 accumulation. Results from this study indicate that CAF markedly reduces the cytotoxicity of ADR or AD32 and trifluoperazine circumvents the effects of CAF.


Assuntos
Cafeína/farmacologia , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Trifluoperazina/farmacologia , Animais , Doxorrubicina/metabolismo , Camundongos , Fatores de Tempo
13.
Cancer Res ; 40(3): 630-3, 1980 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7471083

RESUMO

1-beta-D-Arabinofuranosylcytosine (ara-C) was encapsulated in anionic multilamellar liposomes prepared with different lecithin: cholesterol (L:C) ratios. The chemotherapeutic activity of encapsulated ara-C was compared with comparable doses of ara-C in 0.85% saline solution (single- and multiple-dose schedules) in mice bearing L1210 (i.p.) leukemia. Maximum survival was obtained in animals given injections of ara-C (40 mg/kg) encapsulated in liposomes with a L:C ratio of 1:1. The effect of L:C ratio on survival was not pronounced in multiple-dose schedules. Multiple doses (every 4.5 hr for 3 separate injections) of 40 mg/kg with L:C ratios of 1:1 and 1:0.5 were toxic, resulting in 83 and 50% mortality, respectively, of mice by Day 7. This study shows that drug efflux and in vivo antitumor activity and toxicity of encapsulated ara-C is influenced by the cholesterol content of the liposomal lipid bilayer.


Assuntos
Colesterol/administração & dosagem , Citarabina/administração & dosagem , Leucemia L1210/tratamento farmacológico , Lipossomos/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Camundongos , Fosfatidilcolinas/administração & dosagem
14.
Cancer Res ; 42(4): 1384-9, 1982 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-6949640

RESUMO

Surface-exposed proteins of vinblastine-sensitive human lymphoid cell line of leukemic origin (CCRF-CEM) were examined by the lactoperoxidase-catalyzed iodination and two-dimensional polyacrylamide gel electrophoresis methods. Spots which comigrate with bovine brain tubulin and rabbit muscle actin were prominently labeled in the whole membrane but not in the high-speed supernatant fraction of the disrupted cells. Mild trypsinization of labeled cells removed the iodinated tubulin and actin without significantly affecting the protein staining pattern. Iodination of normal human lymphocytes resulted in no labeling of the tubulin or actin. The presence of surface-exposed tubulin in this leukemic cell line suggests a possible mechanism for their enhanced sensitivity to the cytotoxic action of vinblastine.


Assuntos
Leucemia Linfoide/análise , Proteínas de Membrana/análise , Tubulina (Proteína)/análise , Linhagem Celular , Humanos , Tripsina/farmacologia
15.
Cancer Res ; 51(22): 6133-7, 1991 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-1682042

RESUMO

The mechanism(s) through which combination treatment with Adriamycin and recombinant interleukin 2 (rIL2) affects murine renal cell carcinoma was investigated. A single dose of Adriamycin (ADM) administered 1 day after implantation of tumor cells s.c. delayed the formation of tumor and significantly extended the median survival time. About 40% of the treated mice remained tumor free for more than 6 months. Treating mice with a similar dose of ADM 12 to 16 days after implantation of tumor caused complete regression of established tumors within 10 to 12 days in more than 90% of the mice. However, after a transient tumor free period of 15 to 20 days recurrence of tumor was observed in all treated mice. In contrast, a regimen that included treating tumor bearing mice with a single dose of ADM followed by a daily i.p. injection of rIL2 5 x 10(4) units for 10 days delayed the recurrence of tumors and significantly prolonged the survival time compared to the median survival time of mice treated with ADM alone. About 20 to 30% tumor bearing mice remained tumor free for 4 months following treatment with ADM and rIL2. Treatment with rIL2 alone produced no antitumor response. In addition, rIL2 itself was not inhibitory for tumor cell growth nor did it modulate the cytotoxic response of renal cell carcinoma cells to ADM in vitro. Mice that were cured following treatment with ADM and rIL2 were resistant to a rechallenge with viable tumor cells, and cured mice also expressed a tumor specific T-cell mediated delayed hypersensitivity reaction. The immune cells that mediate tumor rejection were identified as Thy-1.2+ T-cells. Taken together, these results indicate that antitumor activity of combined Adriamycin/rIL2 treatment is at least partly attributable to the production of tumor specific immunity.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Doxorrubicina/administração & dosagem , Interleucina-2/administração & dosagem , Neoplasias Renais/tratamento farmacológico , Animais , Antígenos de Superfície/análise , Carcinoma de Células Renais/imunologia , Imunização , Neoplasias Renais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/administração & dosagem , Antígenos Thy-1
16.
Cancer Res ; 58(23): 5298-300, 1998 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9850052

RESUMO

Numerous chemotherapeutic agents act via stabilization of a topoisomerase (topo) II-DNA complex. HL-60/AMSA, a human leukemia cell line, is resistant to intercalator-mediated DNA complex formation and cytotoxicity. HL-60/AMSA contains a mutant form of topo IIalpha that was thought to explain this resistance. However, our present data show that expression of topo IIbeta RNA in HL-60/AMSA is only 10% of that in HL-60, and topo IIbeta protein levels are undetectable. Southern analysis of topo IIbeta shows no differences in gene dosage between the two cell lines but does show differences in the restriction patterns. These data suggest that decreased topo IIbeta expression may contribute to the intercalator resistance of HL-60/AMSA cells.


Assuntos
Amsacrina/farmacologia , Antineoplásicos/farmacologia , DNA Topoisomerases Tipo II , DNA Topoisomerases Tipo II/deficiência , DNA Topoisomerases Tipo II/genética , Isoenzimas/genética , Antígenos de Neoplasias , Northern Blotting , Southern Blotting , DNA Topoisomerases Tipo II/biossíntese , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA , Resistencia a Medicamentos Antineoplásicos , Células HL-60/efeitos dos fármacos , Células HL-60/enzimologia , Humanos , Immunoblotting , Isoenzimas/biossíntese , Isoenzimas/metabolismo , Mutação , Fenótipo , RNA Mensageiro/metabolismo
17.
J Clin Oncol ; 6(5): 880-8, 1988 May.
Artigo em Inglês | MEDLINE | ID: mdl-3367191

RESUMO

Drug resistance to chemotherapy agents such as doxorubicin appears to be an important cause of therapeutic failure in cancer treatment. Based on preclinical information demonstrating that the phenothiazine calmodulin-inhibitor trifluoperazine can enhance retention and cytotoxicity of doxorubicin in resistant cells, a phase I/II trial of the combination was performed to determine the maximally tolerated dose (MTD) of trifluoperazine that could be administered with doxorubicin. Patients with intrinsic (no previous response) and acquired (previous response with relapse) doxorubicin resistance were eligible. Doxorubicin was administered as a 96-hour continuous infusion (60 mg/m2) on days 2 through 5. Trifluoperazine was administered in divided doses orally on days 1 through 6, with dose escalation from 20 to 100 mg/d. Thirty-six patients were evaluable. The MTD of trifluoperazine was 60 mg/d, with dose-limiting toxicity being extrapyramidal side effects. No alteration of doxorubicin toxicity was observed. Seven of the 36 patients responded (one complete response [CR], six partial responses [PR]), with seven of 21 patients having acquired resistance, and zero of 15 with intrinsic resistance demonstrating responses. Doxorubicin plasma levels were not affected by trifluoperazine, and the maximal trifluoperazine plasma levels achieved were 129.83 ng/mL. This trial demonstrates the combination of trifluoperazine and doxorubicin is well tolerated, and the schedule recommended for phase II trials is doxorubicin, 60 mg/m2 (continuous infusion) days 2 through 5, and trifluoperazine, 15 mg four times per day orally days 1 through 6. Continued investigation of this combination is indicated for patients with acquired doxorubicin resistance.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Calmodulina/antagonistas & inibidores , Doxorrubicina/administração & dosagem , Neoplasias/tratamento farmacológico , Trifluoperazina/administração & dosagem , Adulto , Idoso , Doxorrubicina/sangue , Avaliação de Medicamentos , Resistência a Medicamentos/efeitos dos fármacos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Trifluoperazina/efeitos adversos , Trifluoperazina/metabolismo
18.
Clin Cancer Res ; 4(3): 635-42, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9533531

RESUMO

Because tamoxifen and all-trans-retinoic acid (ATRA) have additive antitumor effects in preclinical systems, we performed a Phase I/II clinical trial of this combination in patients with advanced breast cancer. Patients with potentially hormone-responsive advanced breast cancer were enrolled. All received 20 mg of tamoxifen by mouth daily. Consecutive cohorts of 3-6 patients were treated on odd-numbered weeks with ATRA at doses of 70, 110, 150, 190, or 230 mg/m2/day. Twenty-six patients were entered in this trial; 25 were evaluable. A dose of 230 mg/m2 ATRA produced unacceptable headache and dermatological toxicity, but doses < or = 190 mg/m2 were tolerable. Two of 7 patients with measurable disease responded. Seven of 18 patients with evaluable, nonmeasurable disease achieved disease stability for more than 6 months. Plasma AUCs on day 1 of successive weeks of treatment were stable over time. A nonsignificant decrease in serum insulin-like growth factor I levels was noted during treatment, but this trend was similar to that observed in three "control" patients treated with tamoxifen alone. When given with daily tamoxifen, the maximum tolerated dose of ATRA that could be given on alternate weeks was 190 mg/m2/day. This schedule of ATRA resulted in repeated periods of exposure to potentially therapeutic concentrations of ATRA. Declines in the serum insulin-like growth factor I concentrations observed in patients treated with tamoxifen and ATRA were similar to those observed in patients treated with tamoxifen alone. Objective responses were observed, some in patients who had previously progressed while receiving tamoxifen, suggesting that further studies would be of interest.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Tamoxifeno/uso terapêutico , Tretinoína/uso terapêutico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Biomarcadores/sangue , Neoplasias da Mama/patologia , Esquema de Medicação , Feminino , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Fator de Crescimento Insulin-Like I/análise , Pessoa de Meia-Idade , Metástase Neoplásica , Estadiamento de Neoplasias , Tamoxifeno/administração & dosagem , Tamoxifeno/efeitos adversos , Tretinoína/administração & dosagem , Tretinoína/efeitos adversos
19.
Clin Cancer Res ; 6(1): 78-89, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10656435

RESUMO

Exisulind (sulindac sulfone; FGN-1), a metabolite of sulindac without known effects on prostaglandin synthesis, can promote apoptosis and inhibit tumorigenesis in preclinical systems. We performed a Phase I trial of this compound in patients with familial adenomatous polyposis (FAP) to examine the tolerability and safety of this drug in the cancer chemoprevention setting. Six patients each were treated with exisulind at doses of 200, 300, and 400 mg p.o. twice a day. Reversible hepatic dysfunction was noted in four of six patients treated at the 400-mg p.o., twice-a-day dose level, but in only one to two of six patients treated at each of the lower dose levels. The serum half-life of exisulind was 6-9 h; little drug accumulation was noted over time. A nonsignificant trend toward increased apoptosis in polyps was noted at the maximum tolerated dose, but no decrease in polyp numbers or significant effects on cellular proliferation was noted. After treatment, polyps tended to display a "halo" appearance grossly and mucinous differentiation histologically. The maximum safe dose of exisulind is 300 mg p.o. twice a day in patients with subtotal colectomies. Reversible hepatic dysfunction limits further dose escalation. A decrease in polyp numbers could not be demonstrated, but the trend toward increased apoptosis at the MTD and the observation of mucinous change histologically suggest that further investigation of drugs of this class might be warranted.


Assuntos
Polipose Adenomatosa do Colo/tratamento farmacológico , Antineoplásicos/efeitos adversos , Sulindaco/análogos & derivados , Polipose Adenomatosa do Colo/patologia , Administração Oral , Adolescente , Adulto , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Apoptose , Feminino , Meia-Vida , Humanos , Marcação In Situ das Extremidades Cortadas , Antígeno Ki-67/análise , Masculino , Pessoa de Meia-Idade , Sulindaco/administração & dosagem , Sulindaco/efeitos adversos , Sulindaco/farmacocinética
20.
Clin Cancer Res ; 4(6): 1459-67, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9626463

RESUMO

Because topoisomerase (topo) I- and topo II-targeting agents exert their principal effects on the two major classes of enzymes involved in regulating DNA topology in the cell, there has been considerable interest in evaluating combinations of these classes of agents. In preclinical studies of inhibitors of topo I and topo II in combination, drug scheduling and sequencing have been critical determinants of antitumor activity, with a greater magnitude of cytotoxicity generally occurring when treatment with the topo I inhibitor precedes treatment with the topo II-targeting agent. The underlying mechanism that has been proposed to explain this schedule dependency is compensatory up-regulation of topo II and, therefore, enhanced cytotoxicity of topo II inhibitors in cells treated initially with topo I inhibitors. The feasibility of sequentially administering the topo I inhibitor topotecan (TPT) followed by the topo II inhibitor etoposide to patients with advanced solid malignancies was evaluated in this Phase I and translational laboratory study. Fifty patients with solid neoplasms were treated with TPT doses ranging from 0.17 to 1.05 mg/m2/day as a 72-h continuous (i.v.) infusion on days 1-3 followed by etoposide, 75 or 100 mg/m2/day as a 2-h i.v. infusion daily on days 8-10. The combined rate of severe neutropenia and thrombocytopenia was unacceptably high above the TPT (mg/m2/day)/etoposide (mg/m2/day) dose levels of 0.68/100 and 0.68/75 in minimally and heavily pretreated patients, respectively, and these dose levels are recommended for further disease-directed evaluations of TPT/etoposide on this administration schedule. Successive biopsies of accessible tumors were obtained for quantitation of topo I and II levels prior to and immediately after treatment with TPT and prior to and immediately after treatment with etoposide in seven patients. The results of these limited studies in tumors did not fully support the proposed mechanistic rationale favoring the development of this particular sequential TPT/etoposide regimen, because only two of the six patients' tumors in whom topo I was successively measured had either modest or substantial decrements in topo I levels following treatment with TPT, and the principal effect of interest, up-regulation of topo II following treatment with TPT, was clearly documented in the tumors of only one of six subjects in whom successive measurements of topo I were performed. Even in view of the notable objective antitumor activity in three subjects, including a complete response in a patient with colorectal carcinoma and partial responses in one patient each with non-small cell lung and gastric carcinomas, the toxicity and ancillary laboratory results do not provide substantial evidence that sequential treatment with TPT and etoposide might be more advantageous than either TPT or etoposide administered as a single agent.


Assuntos
Etoposídeo/efeitos adversos , Neoplasias/tratamento farmacológico , Inibidores da Topoisomerase I , Inibidores da Topoisomerase II , Topotecan/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biópsia , Relação Dose-Resposta a Droga , Esquema de Medicação , Etoposídeo/administração & dosagem , Etoposídeo/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/enzimologia , Neoplasias/patologia , Topotecan/administração & dosagem , Topotecan/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa