Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Case Rep Ophthalmol Med ; 2011: 854784, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22606479

RESUMO

Lisch nodules associated with Neurofibromatosis Type 1 (NF1) are usually multiple and bilateral in nature. Here, we report a 21-year-old healthy, Caucasian female who was diagnosed with multiple, unilateral Lisch nodules during routine eye examination. A thorough history and physical examination revealed no other signs of NF1. We diagnosed the rare occurrence of numerous, unilateral Lisch nodules in the absence of additional features of NF1 in our patient and provide a discussion concerning the differential diagnosis of Lisch nodules as well as the potential genetic explanation of this finding.

2.
Hum Mol Genet ; 5(8): 1149-53, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8842734

RESUMO

Mutations in the dystrophin gene are responsible for Duchenne and Becker muscular dystrophy (DMD/BMD). Studies of dystrophin expression and function have benefited from use of the mdx mouse, an animal model for DMD/BMD. Here we characterized mutations in three additional strains of mdx mice, the mdx2cv, mdx4cv and mdx5cv alleles. The mutation in the mdx2cv mouse was found to be a single base change in the splice acceptor sequence of dystrophin intron 42. This mutation leads to a complex pattern of aberrant splicing that generates multiple transcripts, none of which preserve the normal open reading frame. In the mdx5cv allele, the dystrophin mRNA contains a 53 bp deletion of sequences from exon 10. Analysis of the genomic DNA uncovered a single A to T transversion in exon 10. Although this base change does not alter the encoded amino acid, a new splice donor was created (GTGAG) that generates a frameshifting deletion in the processed mRNA. In the mdx4cv allele, direct sequencing revealed a C to T transition in exon 53, creating an ochre codon (CAA to TAA). The differential location of these mutations relative to the seven known dystrophin promoters results in a series of mdx mouse mutants that differ in their repertoire of isoform expression, such that these mice should be useful for studies of dystrophin expression and function. The mdx4cv and mdx5cv strains may be of additional use in gene transfer studies due to their low frequency of mutation reversion.


Assuntos
Distrofina/genética , Camundongos Endogâmicos mdx/genética , Distrofia Muscular Animal/genética , Mutação , Alelos , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Complementar/genética , Mutação da Fase de Leitura , Expressão Gênica , Camundongos , Dados de Sequência Molecular , Mutação Puntual , Reação em Cadeia da Polimerase , Splicing de RNA/genética
3.
Cancer Chemother Pharmacol ; 35(6): 471-82, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7533669

RESUMO

Adozelesin is a highly potent alkylating agent that undergoes binding in the minor groove of double-stranded DNA (ds-DNA) at A-T-rich sequences followed by covalent bonding with N-3 of adenine in preferred sequences. On the basis of its high-potency, broad-spectrum in vivo antitumor activity and its unique mechanism of action, adozelesin has entered clinical trial. We report herein the cytotoxicity for Chinese hamster ovary (CHO) cells of several agents, including antitumor drugs, combined with adozelesin. The additive, synergistic, or antagonistic nature of the combined drug effect was determined for most combinations using the median-effect principle. The results show that in experiments using DNA- and RNA-synthesis inhibitors, prior treatment with the DNA inhibitor aphidicolin did not affect the lethality of adozelesin. Therefore, ongoing DNA synthesis is not needed for adozelesin cytotoxicity. Combination with the RNA inhibitor cordycepin also did not affect adozelesin cytotoxicity. In experiments with alkylating agents, combinations of adozelesin with melphalan or cisplatin were usually additive or slightly synergistic. Adozelesin-tetraplatin combinations were synergistic at several different ratios of the two drugs, and depending on the schedule of exposure to drug. In experiments using methylxanthines, adozelesin combined synergistically with noncytotoxic doses of caffeine or pentoxifylline and resulted in several logs of increase in adozelesin cytotoxicity. In experiments with hypomethylating agents, adozelesin combined synergistically with 5-azacytidine (5-aza-CR) and 5-aza-2'-deoxycytidine (5-aza-2'-CdR). Combinations of adozelesin with tetraplatin or 5-aza-2'-CdR were also tested against B16 melanoma cells in vitro and were found to be additive and synergistic, respectively. The synergistic cytotoxicity to CHO cells of adozelesin combinations with tetraplatin, 5-aza-CR, or pentoxifylline was not due to increased adozelesin uptake or increased alkylation of DNA by adozelesin.


Assuntos
Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ácidos Cicloexanocarboxílicos/uso terapêutico , Indóis , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Afidicolina/administração & dosagem , Afidicolina/farmacologia , Azacitidina/administração & dosagem , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Benzofuranos , Células CHO , Cafeína/administração & dosagem , Cafeína/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Cricetinae , Cricetulus , Ácidos Cicloexanocarboxílicos/administração & dosagem , Ácidos Cicloexanocarboxílicos/metabolismo , Cicloexenos , DNA/efeitos dos fármacos , DNA/metabolismo , Decitabina , Desoxiadenosinas/administração & dosagem , Desoxiadenosinas/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Duocarmicinas , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Melfalan/administração & dosagem , Melfalan/farmacologia , Mutagênicos/administração & dosagem , Mutagênicos/farmacologia , Compostos Organoplatínicos/administração & dosagem , Compostos Organoplatínicos/farmacologia , Pentoxifilina/administração & dosagem , Pentoxifilina/farmacologia , Células Tumorais Cultivadas
4.
Cancer Chemother Pharmacol ; 34(1): 44-50, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7513620

RESUMO

Bizelesin (U-77779) is a highly potent bis-alkylating antitumor agent that is effective against several tumor systems in vitro and in vivo. V79 cells that were 125- to 250-fold resistant to bizelesin developed after constant exposure to gradually increasing concentrations of the drug. Resistant cells exhibited a multidrug-resistant phenotype and genotype as indicated by cross-resistant to several structurally and functionally unrelated drugs, e.g., colchicine, actinomycin D, and Adriamycin, and overexpression of mdr mRNA. Very low levels of cross-resistance to the alkylating agents cisplatin and melphalan were seen. Multidrug-resistant mouse leukemia (P388/Adriamycin-resistant) and human (KB/vinblastine-resistant) cells were also resistant to bizelesin. Bizelesin resistance was unstable and decreased when cells were grown in the absence of the drug. Resistant and sensitive cell lines had similar levels of glutathione, and bizelesin cytotoxicity for resistant cells was not markedly affected by treatment with buthionine sulfoximine. Cross-resistance between bizelesin and several of its analogs is reported.


Assuntos
Antineoplásicos/farmacologia , Indóis/farmacologia , Pulmão/citologia , Ureia/análogos & derivados , Alquilantes/farmacologia , Animais , Northern Blotting , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Resistência a Medicamentos/fisiologia , Duocarmicinas , Fibroblastos/efeitos dos fármacos , Genótipo , Humanos , Pulmão/efeitos dos fármacos , Camundongos , Fenótipo , RNA/análise , Células Tumorais Cultivadas , Ureia/farmacologia , Verapamil/farmacologia
5.
Cancer Res ; 53(6): 1354-9, 1993 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-8443816

RESUMO

Adozelesin is a highly potent alkylating agent which has entered clinical trials based on its unique mechanisms of action and broad-spectrum antitumor activity in vivo. V79 cells resistant to adozelesin (V79/AdoR) were not resistant to the alkylating agent cisplatin but showed the phenotypic and genotypic characteristics of multidrug resistance. Thus V79/AdoR was cross-resistant to several structurally and functionally unrelated drugs, resistance was reversed by verapamil, and the resistant cell line expressed mdr mRNA and p170 glycoprotein. Also, adozelesin uptake and the amount of drug alkylated to DNA was much lower in the resistant cell line as compared to the sensitive parent. However, even with the same amount of drug bound to DNA (10 fmol/micrograms DNA) the survival of V79/S approximately 15% survival) was much lower than that of V79/AdoR (approximately 80%). Therefore the resistance of V79/AdoR cannot be explained solely by the multidrug resistance mechanism (i.e., lower drug uptake and less drug alkylation to DNA), which suggests that multiple mechanisms may account for resistance to adozelesin. V79/AdoR showed different levels of cross-resistance to several adozelesin analogues. The analogues could be divided into 2 groups; those with very low partition coefficients (log P < 2 as compared to 2.74 for adozelesin) had low levels of cross-resistance, whereas analogues with higher partition coefficients (log P > 2.4) were cross-resistant to adozelesin.


Assuntos
Alquilantes/farmacologia , Antineoplásicos/farmacologia , Ácidos Cicloexanocarboxílicos/farmacologia , Resistência a Medicamentos , Indóis , Animais , Benzofuranos , Butionina Sulfoximina , Linhagem Celular , Cricetinae , Ácidos Cicloexanocarboxílicos/metabolismo , Cicloexenos , DNA/metabolismo , Duocarmicinas , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacologia , Verapamil/farmacologia
7.
Cancer Chemother Pharmacol ; 32(3): 235-42, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-8098996

RESUMO

Taxol is a clinically active anticancer drug, which exerts its cytotoxicity by the unique mechanism of polymerizing tubulin monomers into microtubules and stabilizing microtubules. Our studies with ovarian (hamster CHO and human A2780) cells showed that taxol is a phase-specific agent that is much more cytotoxic to mitotic cells than interphase cells. First, the dose-survival pattern of taxol resembled that of other phase-specific agents, in which cell-kill reached a plateau at a certain concentration. This suggests that the asynchronous cell population consists of a taxol-sensitive (presumably mitotic) fraction and a taxol-resistant fraction. Second, the cells were more responsive to increased exposure time than to increased dose above the plateau concentration. Third, in both asynchronous and synchronous cultures taxol was much more cytotoxic to mitotic than interphase (G1, S and G2) cells. Fourth, the taxol concentration needed to kill cells corresponded to the dose needed to block cells in mitosis. Although taxol blocked cells in mitosis, the mitotic block was of short duration. Cells escaped the mitotic block, without cytokinesis, and entered the next round of DNA synthesis to form multinucleated polyploid cells. Taxol was 15- to 25-fold more toxic to A2780 (human ovarian carcinoma) cells compared to CHO cells. This difference in sensitivity correlated with a higher intracellular taxol concentration in A2780 as compared to CHO as determined by either an ELISA assay or by [H3]-taxol uptake.


Assuntos
Ovário/efeitos dos fármacos , Paclitaxel/farmacologia , Animais , Células CHO , Morte Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Cricetinae , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Cinética , Mitose/efeitos dos fármacos , Ovário/citologia , Ovário/metabolismo , Paclitaxel/metabolismo , Células Tumorais Cultivadas
8.
Cancer Res ; 52(20): 5687-92, 1992 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-1394193

RESUMO

Adozelesin (U-73975) is an extremely potent cytotoxic agent which causes 90% lethality, after 2 h exposure in vitro, of Chinese hamster ovary and lung (CHO and V79), mouse melanoma (B16), and human ovarian carcinoma (A2780) cells at 0.33, 0.19, 0.2, and 0.025 ng/ml, respectively. Under similar conditions, Adriamycin and cisplatin had 90% lethality values in CHO cells of 150 ng/ml (= 249 nM) and 6800 ng/ml (= 2266 nM), respectively. The relative drug sensitivity of the cell lines (A2780 > V79, B16, CHO) was correlated to the relative amounts of [3H]adozelesin alkylated to DNA. The greater sensitivity of A2780 was due to (a) greater DNA alkylation at different drug doses and (b) greater intrinsic sensitivity of A2780 which resulted in greater cell kill at comparable DNA alkylation. Phase specific toxicity studies show that adozelesin was least lethal to CHO cells in mitosis and very early G1. Lethality increased as cells progressed through G1 and was maximal in late G1 and early S. Mitotic cells had lower drug uptake and correspondingly less drug binding to DNA than G1 or S-phase cells. However, based on the amount of drug alkylated per micrograms of DNA, cells in M, G1, and S were equally sensitive. Therefore, the lower sensitivity of M-phase cells was due to lower drug uptake. Adozelesin had three different effects on progression of CHO, V79, B16, and A2780 through the cell cycle: (a) slowed progression through S which resulted in significantly increasing the percentage of S-phase cells. This effect was transient; (b) cell progression was blocked in G2 for a long time period; (c) the response of the cell lines to the G2 block differed. CHO and V79 cells escaped G2 block by dividing and entered the diploid DNA cycle or did not undergo cytokinesis and became tetraploid. On the contrary, B16 and A2780 cells remained blocked in G2 and did not become tetraploid. Cell progression was inhibited in a similar manner when a synchronized population of M, G1, or S-phase cells were exposed to adozelesin.


Assuntos
Antineoplásicos/toxicidade , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ácidos Cicloexanocarboxílicos/toxicidade , DNA/efeitos dos fármacos , Inibidores do Crescimento/toxicidade , Indóis , Alquilação/efeitos dos fármacos , Animais , Benzofuranos , Células CHO/citologia , Células CHO/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Cricetinae , Cicloexenos , Duocarmicinas , Feminino , Fase G1/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Humanos , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos , Mitose/efeitos dos fármacos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fase S/efeitos dos fármacos , Fatores de Tempo , Células Tumorais Cultivadas
9.
Cancer Chemother Pharmacol ; 30(5): 348-54, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1505072

RESUMO

Adozelesin (U-73975) was highly cytotoxic to V79 cells in culture and was more cytotoxic than several clinically active antitumor drugs as determined in a human tumor-cloning assay. Phase-specificity studies showed that cells in the M+early G1 phase were most resistant to adozelesin and those in the late G1 + early S phase were most sensitive. Adozelesin transiently slowed cell progression through the S phase and then blocked cells in G2. Some cells escaped the G2 block and either divided or commenced a second round of DNA synthesis (without undergoing cytokinesis) to become tetraploid. Adozelesin inhibited DNA synthesis more than it did RNA or protein synthesis. However, the dose needed for inhibition of DNA synthesis was 10-fold that required for inhibition of L1210 cell growth. The observation that cell growth was inhibited at doses that did not cause significant inhibition of DNA synthesis and that cells were ultimately capable of completing two rounds of DNA synthesis in the presence of the drug suggests that adozelesin did not exert its cytotoxicity by significant inhibition of DNA synthesis. It is likely that adozelesin alkylates DNA at specific sites, which leads to transient inhibition of DNA synthesis and subsequent G2 blockade followed by a succession of events (polyploidy and unbalanced growth) that result in cell death.


Assuntos
Alquilantes/farmacologia , Antineoplásicos/farmacologia , Ácidos Cicloexanocarboxílicos/farmacologia , Indóis , Animais , Benzofuranos , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cicloexenos , DNA/biossíntese , Relação Dose-Resposta a Droga , Duocarmicinas , Humanos , Biossíntese de Proteínas , Fatores de Tempo , Células Tumorais Cultivadas/efeitos dos fármacos
10.
J Virol ; 64(9): 4232-41, 1990 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2117070

RESUMO

Cytolytic T-lymphocyte (CTL) activity specific for respiratory syncytial (RS) virus was investigated after intranasal infection of mice with RS virus, after intraperitoneal infection of mice with a recombinant vaccinia virus expressing the F glycoprotein, and after intramuscular vaccination of mice with Formalin-inactivated RS virus or a chimeric glycoprotein, FG, expressed from a recombinant baculovirus. Spleen cell cultures from mice previously infected with live RS virus or the F-protein recombinant vaccinia virus had significant CTL activity after one cycle of in vitro restimulation with RS virus, and lytic activity was derived from a major histocompatibility complex-restricted, Lyt2.2+ (CD8+) subset. CTL activity was not restimulated in spleen cells from mice that received either the Formalin-inactivated RS virus or the purified glycoprotein, FG. The protein target structures for recognition by murine CD8+ CTL were identified by using target cells infected with recombinant vaccinia viruses that individually express seven structural proteins of RS virus. Quantitation of cytolytic activity against cells expressing each target structure suggested that 22K was the major target protein for CD8+ CTL, equivalent to recognition of cells infected with RS virus, followed by intermediate recognition of F or N, slight recognition of P, and no recognition of G, SH, or M. Repeated stimulation of murine CTL with RS virus resulted in outgrowth of CD4+ CTL which, over time, became the exclusive subset in culture. Murine CD4+ CTL were highly cytolytic for RS virus-infected cells, but they did not recognize target cells infected with any of the recombinant vaccinia viruses expressing the seven RS virus structural proteins. Finally, the CTL response in peripheral blood mononuclear cells of adult human volunteers was investigated. The detection of significant levels of RS virus-specific cytolytic activity in these cells was dependent on at least two restimulations with RS virus in vitro, and cytolytic activity was derived primarily from the CD4+ subset.


Assuntos
Vírus Sinciciais Respiratórios/imunologia , Infecções por Respirovirus/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos CD/análise , Antígenos de Diferenciação de Linfócitos T/análise , Antígenos CD4/análise , Antígenos CD8 , Linhagem Celular , Citotoxicidade Imunológica , Humanos , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Fenótipo , Recombinação Genética , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Baço/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Proteínas Virais/genética , Proteínas Virais/isolamento & purificação
11.
Cancer Res ; 50(8): 2485-92, 1990 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-2317831

RESUMO

U-71,184 is a CC-1065 analogue which is highly cytotoxic in vitro and has a broad spectrum of antitumor activity in vivo. Against B16 cells, U-71,184 was 8-fold and 253-fold more potent than Actinomycin D and Adriamycin, respectively. U-71,184 killed 90% of B16 cells at 0.01 ng/ml levels of drug in the medium, which was equivalent to an intracellular concentration of about 8 pg/10(6) cell (= 2 x 10(-8) pmol/cell). A B16 cell line resistant to U-71,184 developed after 3 months of in vitro exposure to gradually increasing concentrations of the drug. The sensitive and resistant cell lines were cloned and a B16/R clone was selected which was 60 to 100 times more resistant to U-71,184 than the cloned sensitive parent (B16/S). Cells grown in the absence of U-71,184 for 2 months retained resistance to the drug. B16/R was slightly cross-resistant only to Adriamycin but not to Actinomycin D, vinblastine, or colchicine. Among alkylating agents, it was slightly cross-resistant to Melphalan but not to 1,3-bis(2-chloroethyl)-1-nitrosourea or cisplatin. B16/R did not overexpress mdr mRNA. Therefore, this cell line does not exhibit the multidrug-resistant phenotype. Most karyotypes of B16/R had a marker chromosome which carried an aberrantly staining region apparently containing repetitive replication of the same segment. Resistance can be partly accounted for by the approximately 10-fold lesser uptake of [3H]-U-71,184 in B16/R, as compared to B16/S. B16/R was cross-resistant in varying degrees to several other CC-1065 analogues. The ratio of the 50% lethal dose of U-71,184 for B16/R, as compared to B16/S, was about 60 (i.e., R/S = 60). In comparison, the following compounds had an R/S ratio of less than 20 (i.e., modest level of cross-resistance to U-71,184): U-68,819, U-73,975, U-75,500, U-75,559, and CC-1065. In contrast, the following compounds had an R/S ratio greater than 20 (i.e., highly cross-resistant to U-71,184): U-71,184 analogues U-71,185, U-73,903, and U-75,012; U-73,975 analogues U-75,613, U-75,032, and U-73,896; and CC-1065 enantiomer U-76,915. We cannot yet explain the difference in the level of cross-resistance between these compounds in vitro. B16/S and B16/R cells were tumorigenic in mice and B16/R was resistant to U-71,184 in vivo. There was no clear indication of cross-resistance of B16/R in vivo to Adriamycin, Actinomycin D, cisplatin, or Melphalan. However, U-73,975, a compound with modest cross-resistance in vitro, was significantly cross-resistant in vivo.


Assuntos
Antineoplásicos/farmacologia , Resistência a Medicamentos/genética , Indóis/farmacologia , Melanoma Experimental/genética , Animais , Transporte Biológico , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Indóis/metabolismo , Cariotipagem , Cinética , Camundongos , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
12.
Sel Cancer Ther ; 6(4): 153-66, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2151298

RESUMO

We have used both HPLC and flow cytometry to measure and compare the uptake of two anthracyclines, menogaril (MEN) and Adriamycin (ADR), in V79 Chinese hamster lung fibroblasts grown as monolayers and as 650 microns multicell spheroids. In order to compare intracellular drug accumulation in spheroid cells measured by the two methods, we converted mean channel fluorescence of the flow cytometer to drug uptake expressed as ng/10(6) cells by using a standard curve. The standard curve related the flow cytometric mean channel fluorescence, of monolayer cells exposed to either drug, to the intracellular drug accumulation determined by HPLC. This standard curve was then used to convert the mean channel fluorescence of cells from drug-exposed spheroids to ng/10(6) cells. Our results show that equal intracellular drug accumulation (determined by HPLC) in spheroids and monolayers does not result in equal cellular fluorescence emission (determined by flow cytometry) by these 2 cell populations. For example, monolayer cells with an intracellular MEN accumulation of 650 ng/10(6) cells, emit 40 units of fluorescence as measured by flow cytometry. However, spheroid cells with the same intracellular accumulation emit about 80 units of fluorescence. This results in the intracellular MEN uptake in spheroids measured by flow cytometry being as much as 2- to 3-fold higher than that measured by HPLC. Intracellular ADR accumulation measured by flow cytometry was also higher than that obtained by HPLC. In spite of the quantitative difference between the two methods, qualitatively both methods gave similar results. Thus, both techniques showed that at equal drug concentration in medium drug uptake in monolayers was much greater than in spheroids.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Antineoplásicos/farmacocinética , Doxorrubicina/farmacocinética , Nogalamicina/análogos & derivados , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Citometria de Fluxo , Menogaril , Modelos Biológicos , Nogalamicina/farmacocinética , Organoides/metabolismo
13.
Cancer Res ; 48(1): 109-16, 1988 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-2446739

RESUMO

CC-1065 is a very potent antitumor antibiotic which selectively binds in the minor groove of DNA with alkylation at N-3 of adenine. Since therapeutic doses of CC-1065 caused delayed deaths in mice, analogues were synthesized, some of which had significant antitumor activity. The effects of several of these analogues on inhibition of CHO cell survival, cell progression, and their phase-specific toxicity are reported. CC-1065, U-66,664, U-66,819, U-66,694, and U-71,184 all have a left hand segment with an intact cyclopropyl group but have different tail segments. Lethality of these compounds after 2 h drug exposure was in the following order (50% lethal dose in nM in parentheses): CC-1065 (0.06) greater than U-71,184 (1.3) greater than U-66,694 (3.2) greater than U-68,819 (171) greater than U-66,664 (greater than 1200). In general, these compounds did not inhibit progression from G1 to S but slowed progression through S and blocked cells in G2-M. The phase-specific toxicity of U-71,184 and U-66,694 was different from that of CC-1065. CC-1065 was most cytotoxic to cells in M and early G1 and toxicity decreased as cells entered late G1 and S. In contrast, U-66,694 and U-71,184 were most toxic to cells in late G1. The biochemical and cellular effects of U-71,184 were then studied in detail since it was the most active among these analogues. After a 2-h exposure to 3 ng/ml U-71,184, 90% cell kill or growth inhibition was observed whereas 100 ng/ml was needed for similar inhibition of DNA and RNA synthesis. This discrepancy between the doses suggested that inhibition of nucleic acid synthesis may not be causally related to lethality. Further studies showed that when drug was removed after 2 h exposure, DNA synthesis continued to be inhibited whereas RNA and protein synthesis reached levels higher than the control. Therefore, it is likely that at cytotoxic doses the low level of inhibition of DNA synthesis combined with the stimulation of RNA and protein synthesis leads to unbalanced growth and cell death.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Indóis/farmacologia , Leucomicinas/farmacologia , Animais , Células Cultivadas , Cricetinae , DNA/biossíntese , Relação Dose-Resposta a Droga , Duocarmicinas , Biossíntese de Proteínas , RNA/biossíntese , Relação Estrutura-Atividade
14.
J Cell Physiol ; 132(2): 237-45, 1987 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3624316

RESUMO

Mitotic cells selectively harvested after several h of colcemid treatment are routinely used to obtain synchronized cell cultures. DNA flow cytometry shows that when colcemid-treated B16 mitotic cells divide, they give rise to daughter cells in G1, some of which contain abnormal amounts of DNA. Two subpopulations appear to exist, one having a DNA content distribution expected of G1 cells, another having a mean DNA content about 0.8 of expected and an SD of DNA content more than 5 times expected. The effect was dependent on dose and duration of exposure to colcemid. Colcemid was more cytotoxic to cells in G2 + M than to G1 + S phase cells, and it slowed the progression of G1 cells to S. These effects of colcemid were much greater in aneuploid B16 melanoma cells than in pseudodiploid Chinese hamster ovary (CHO) cells.


Assuntos
Demecolcina/farmacologia , Melanoma/patologia , Mitose/efeitos dos fármacos , Aneuploidia , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Cricetinae , DNA/biossíntese , Diploide , Interfase/efeitos dos fármacos , Camundongos , Vimblastina/farmacologia
15.
Cancer Res ; 46(12 Pt 1): 6125-30, 1986 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2946401

RESUMO

Menogaril (7-R-O-methylnogarol) is an anthracycline which has significant antitumor activity in vivo and is in Phase II clinical trial. We report here the drug effect on growth and cell cycle progression of L1210 mouse leukemia cells in vitro and in vivo. At doses which inhibited the growth of L1210 cells in vitro, menogaril slowed the progression of cells through S phase and blocked cells in G2 + M. 7-R-O-Methyl-N-demethylnogarol, the major metabolite of menogaril had the same effects on cell progression in vitro. Menogaril effect on cell progression in vivo was studied with peritoneal L1210 ascites growing in CD2F1 mice. Early in infection, i.e., 3 days after inoculation of 10(5) L1210 cells, DNA histograms of cells from control and drug-treated mice showed only a G1 peak. This presumably represented host diploid G0-G1 cells which predominated in the peritoneal cavity and masked the histogram of L1210 cells. Later in infection, when about 10(8) or more cells were present in the ascites, L1210 cells predominated and DNA histograms were representative of L1210 cells. When menogaril was injected at this time, the cell cycle effects were similar to those seen in vitro. Therefore, the L1210 in vivo model can be used to study cell progression effects only late in infection (when L1210 cells predominate), and due consideration should be given to contamination of the L1210 cells with host G0-G1 cells.


Assuntos
Antineoplásicos/farmacologia , Daunorrubicina/análogos & derivados , Leucemia L1210/tratamento farmacológico , Nogalamicina/farmacologia , Animais , Antineoplásicos/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , DNA de Neoplasias/análise , Leucemia L1210/patologia , Masculino , Menogaril , Camundongos , Camundongos Endogâmicos , Nogalamicina/análogos & derivados , Nogalamicina/metabolismo
16.
Cancer Res ; 46(4 Pt 1): 1688-93, 1986 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3456266

RESUMO

Our interest in prostaglandins (PGs) as antitumor agents stemmed from the report of Bregman and Meyskens (Cancer Res., 43: 1642-1645, 1983) that PGA1, PGA2, and PGD2 inhibited colony formation by human melanoma cells obtained from fresh biopsies of melanoma patients. We tested several PGs and found that PGA1, PGA2, and PGD2 were the most cytotoxic to L1210 cells in culture. Therefore, we studied these PGs for their effects on growth, cell survival, and cell progression of murine (B16) and human (RPMI7932,SK Mel 28) melanoma cells in culture. Although the three PGs equally inhibited the growth of B16 cells, PGD2 was more inhibitory to RPMI 7932 or SK Mel 28 than PGA1 or PGA2. Similarly the three PGs were almost equally active in inhibiting colony formation by B16 cells. However, against human melanoma cells, PGD2 was much more active than PGA1, whereas PGA2 was inactive. Towards the end of our study, we obtained PGJ2 and found that it was as cytotoxic as PGD2 for L1210 cells but was more lethal for human melanoma cells. The primary effect of all three PGs was to block cell progression from G1 to S. At 2.5 micrograms of PGD2 per ml, the blockade of cells in G1 and normal progression through the other phases resulted in accumulation of 80-90% of the cells in G1. At this dose, there was no inhibition of DNA synthesis, and cells in S progressed apparently normally through S, until all cells were blocked in G1. DNA synthesis was inhibited at 5 micrograms/ml which slowed cell progression through S and accumulated cells in G1. The partial synchronization of cells in G1 may be useful in devising new combinations of PGD2 with antitumor drugs.


Assuntos
Melanoma/patologia , Prostaglandinas A/farmacologia , Prostaglandinas D/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , DNA de Neoplasias/biossíntese , Humanos , Camundongos , Proteínas de Neoplasias/biossíntese , Prostaglandina D2 , RNA Neoplásico/biossíntese
17.
Invest New Drugs ; 4(4): 315-23, 1986.
Artigo em Inglês | MEDLINE | ID: mdl-3473056

RESUMO

Prostaglandin D2 (PGD2) is lethal to murine and human melanoma cells at high doses, but synchronizes cells at G1 at non-toxic doses (2.5 or 5 micrograms/ml). We tested the lethality to B16 mouse melanoma cells of combinations of PGD2 with anticancer drugs. The drugs selected were mostly those used in treating human melanoma: actinomycin D, Bleomycin, BCNU, cis-platin, melphalan, 5-fluorouracil, and 1-beta-D-arabinofuranosylcytosine (ara-C). PGD2 was combined with the drugs according to 3 different protocols: An asynchronous culture was given a long term (24 hr) exposure simultaneously to PGD2 + drug. Combinations with Bleomycin, ara-C or melphalan were additive or slightly antagonistic whereas PGD2 plus actinomycin D was significantly antagonistic. Cells synchronized in G1 by 24 hr PGD2 exposure were then given a short-term (2 hr) treatment with PGD2 + drug. Combinations with cis-platin, Bleomycin, BCNU or 5-fluorouracil were additive or slightly antagonistic, whereas melphalan and actinomycin D combinations were significantly antagonistic. Cells were released from a PGD2-induced G1 block and were exposed to drug at different times during cell progression. Actinomycin D was antagonistic when added immediately after release from the G1 block, but was significantly synergistic when added 10 to 12 hr later. The effect of the combinations cannot be explained by available cell cycle or biochemical information. The antagonism between PGD2 and several of the drugs resembles the "cytoprotective" effect of PGD2 towards various noxious agents.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Melanoma/patologia , Prostaglandinas D/administração & dosagem , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Interfase/efeitos dos fármacos , Camundongos , Prostaglandina D2 , Prostaglandinas D/farmacologia
18.
Invest New Drugs ; 3(3): 233-44, 1985.
Artigo em Inglês | MEDLINE | ID: mdl-2933360

RESUMO

Menogarol is a new anthracycline undergoing phase I clinical trial. We report here the lethality after 2 hr exposure to 2 drug combinations of menogarol and several antitumor agents. A new statistical procedure was used to identify synergistic combinations. Most of these combinations were additive, except for menogarol plus melphalan, which was synergistic. Adriamycin plus melphalan was also synergistic. The menogarol-melphalan combination wa studied in detail with regard to the effect of dose and drug-schedule, lethality for exponential and plateau phase cells and effect on cell cycle progression. Although the combination was synergistic for exponential cells it was additive for plateau phase cells. The combination exerted a synergistic effect in inhibiting progression of cells through the cell cycle. After 2 hr menogarol exposure cells were blocked in G2 for about 12 hr following which the block was reversed. This reversal was inhibited when menogarol was combined with melphalan. The uptake of menogarol or melphalan was not changed in the presence of the other drug.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Daunorrubicina/análogos & derivados , Melanoma/patologia , Melfalan/farmacologia , Nogalamicina/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Melanoma/metabolismo , Melfalan/administração & dosagem , Menogaril , Camundongos , Nogalamicina/administração & dosagem , Nogalamicina/análogos & derivados
19.
Cancer Res ; 44(5): 1796-801, 1984 May.
Artigo em Inglês | MEDLINE | ID: mdl-6713383

RESUMO

Didemnins are a new class of cyclic depsipeptides in which didemnin A is the major component, didemnin B the minor component, and a trace of didemnin C is present. Didemnin B was more potent than was didemnin A against B16 melanoma and P388 leukemia in vivo, and B was also approximately 20 times more cytotoxic than was didemnin A in vitro. Therefore, didemnin B was studied in greater detail for its biochemical and cellular effects. Didemnin B inhibited the in vitro growth of B16 greater than L1210 greater than V-79 cells = human foreskin fibroblast = 9L greater than Chinese hamster ovary cells. Didemnin B was more lethal to exponentially growing B16 cells (50% lethal dose for a 2-hr exposure, 17.5 ng/ml) than to plateau-phase cells (50% lethal dose for a 2-hr exposure, 100 ng/ml). After a 24-hr exposure, the 50% lethal dose for exponential- and plateau-phase B16 cells was 8.8 and 59.6 ng/ml, respectively. Chinese hamster ovary cells were not killed even at 25,000 ng/ml. Mitotic cells were the least sensitive to didemnin B, and cells became more sensitive as they progressed into G1 and S phase. However, since cells in all phases were killed, didemnin B cannot be considered a phase-specific agent. Didemnin B inhibited the synthesis of protein more than that of DNA, with much less inhibition of RNA synthesis. Cell progression studies showed that high doses (300 ng/ml for 2 hr or 100 ng/ml for 24 hr) of didemnin B "froze" the cells in their respective phases with complete inhibition of cell progression or growth. At low doses (10 ng/ml for 2 hr or 3 ng/ml for 24 hr), the cells were blocked at the G1-S border thereby increasing the percentage of G1 cells and decreasing the percentage of S-phase cells. Cells continued to progress from S phase to G2 + M and from G2 + M to G1. The cytotoxicity to different cell lines and inhibition of macromolecule synthesis by didemnin A is also reported.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Depsipeptídeos , Leucemia L1210/fisiopatologia , Melanoma/fisiopatologia , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Feminino , Cinética , Pulmão , Camundongos , Ovário , Peptídeos Cíclicos/toxicidade , Ratos , Fatores de Tempo
20.
Cancer Res ; 43(9): 4227-32, 1983 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-6871861

RESUMO

CC-1065 is the most potent antitumor agent tested in our laboratory. It is lethal to B16 and CHO cells and to a variety of human tumors in the clonogenic assay at 1 ng/ml and is effective against L1210 leukemia and B16 melanoma in vivo at 1 to 50 micrograms/kg. CC-1065 inhibits DNA synthesis and binds to DNA in a nonintercalative manner in the minor groove. We report here the kinetics of inhibition of DNA synthesis and of cell progression and the phase-specific toxicity of the drug. To determine phase-specific toxicity, we started synchronous CHO cultures from mitotic cells harvested after Colcemid pretreatment. These cultures showed that mitotic cells were the most sensitive, and sensitivity decreased as the cells progressed through G1 to S and G2. Experiments with B16 and CHO mitotic cells harvested without Colcemid pretreatment also showed that mitotic cells were more sensitive than G1/S-phase cells. Cell progression studies showed that CC-1065 did not affect progression from mitosis to G1 or from G1 to S. Cells progressed slowly through S at low levels (1 ng/ml) of the drug but were blocked in S at 5 ng/ml. Cell progression from G2 to M was blocked by CC-1065. DNA synthesis in B16 cells was measured at different times after 2-hr exposure to CC-1065. The percentage of inhibition of DNA synthesis was minimum at 4 hr and maximum at 19 hr after drug exposure. Since B16 cell progression studies showed a marked change in percentage of S-phase cells during this time, the DNA synthesis rate was recalculated as cpm/S-phase cell. After this correction (i.e., expressing DNA synthesis as cpm/S-phase cell), the percentage of inhibition of DNA synthesis was minimum at 0 hr and gradually increased to maximum inhibition at 19 hr without the decrease seen previously at 4 hr.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Ciclo Celular/efeitos dos fármacos , Indóis , Leucomicinas/toxicidade , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Replicação do DNA/efeitos dos fármacos , Duocarmicinas , Feminino , Cinética , Mitose/efeitos dos fármacos , Ovário
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...