Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 261
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Chem Phys ; 157(5): 054303, 2022 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-35933193

RESUMEN

A semiclassical model describing the charge transfer collisions of C60 fullerene with different slow ions has been developed to analyze available observations. These data reveal multiple Breit-Wigner-like peaks in the cross sections, with subsequent peaks of reactive cross sections decreasing in magnitude. Calculations of charge transfer probabilities, quasi-resonant cross sections, and cross sections for reactive collisions have been performed using semiempirical interaction potentials between fullerenes and ion projectiles. All computations have been carried out with realistic wave functions for C60's valence electrons derived from the simplified jellium model. The quality of these electron wave functions has been successfully verified by comparing theoretical calculations and experimental data on the small angle cross sections of resonant C60+C60 + collisions. Using the semiempirical potentials to describe resonant scattering phenomena in C60 collisions with ions and Landau-Zener charge transfer theory, we calculated theoretical cross sections for various C60 charge transfer and fragmentation reactions which agree with experiments.

2.
J Clin Invest ; 72(6): 1937-47, 1983 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-6643681

RESUMEN

Vitamin B12-deficient and normal rats were loaded with methylmalonic (MMA) and ethylmalonic acids labeled with 13C in the carboxyl groups and with 2H in the alkyl groups. Significant fractions of the administered acids were excreted in both the B12-deficient and the normal animal, having undergone exchange of both their 13C-labeled carboxyl groups with endogenous 12C. The exchange of the alpha-1H of MMA in 2H2O at 25 degrees C and pH 7.5 was found by 1H-nuclear magnetic resonance to have a half-life of 28.3 min. These results show that a fraction of in vivo metabolism through the propionate-to-succinate pathway occurs via a shunt involving free MMA. The enzymes of this pathway are thought to utilize only coenzyme A (CoA) esters. To allow for the exchange of the second CoA-bound carboxyl group, we propose the deacylation of the once exchanged acid with spontaneous racemization (relative to the 13C-carboxyl group), followed by reacylation, thus exposing the labeled carboxyl to decarboxylation. The significance of this mechanism involving free MMA is that racemization of methylmalonyl (MM)-CoA may also occur without the intervention of MM-CoA racemase. A deficiency of this enzyme need not result in symptomatic methylmalonic aciduria.


Asunto(s)
Isomerasas/deficiencia , Malonatos/metabolismo , Ácido Metilmalónico/metabolismo , Racemasas y Epimerasas/deficiencia , Deficiencia de Vitamina B 12/metabolismo , Animales , Fenómenos Químicos , Química , Marcaje Isotópico , Masculino , Ácido Metilmalónico/orina , Ratas , Ratas Endogámicas , Estereoisomerismo , Deficiencia de Vitamina B 12/enzimología
3.
Cancer Res ; 42(10): 3911-7, 1982 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7104990

RESUMEN

Contrary to a recent suggestion, new and useful drugs for the treatment of human cancer continue to be developed although clinical trials today are more difficult and the chances for success are diminished. In the past, research at Southern Research Institute led to the development of two new classes of clinically useful anticancer agents, the nitrosoureas and the imidazole triazines. More recently, we have developed an interesting family of cytotoxic compounds, the haloadenine nucleosides, that are resistant to deamination and show biological activity comparable to the corresponding adenine nucleosides given in combination with 2'-deoxycoformycin, a potent inhibitor of adenosine deaminase. Among them, the 2-haloadenine arabinonucleosides and 2'-deoxyribonucleosides appear to have the necessary selectivity for neoplastic cells to be useful anticancer agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/síntesis química , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Humanos , Leucemia L1210/tratamiento farmacológico , Ratones , Relación Estructura-Actividad
4.
Cancer Res ; 44(8): 3286-90, 1984 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6611198

RESUMEN

9-beta-D-Arabinofuranosyl-2-fluoroadenine (2-F-ara-A) and 2-fluoro-2'-deoxyadenosine (2-FdAdo) were potent inhibitors of L1210 cell growth in culture. Even though these 2-fluoroadenine nucleosides are very poor substrates for adenosine deaminase, erythro-9-(2-hydroxyl-3-nonyl)adenine potentiated the growth-inhibitory properties of 2-FdAdo but not 2-F-ara-A in a synergistic manner. 2-FdAdo and 2-F-ara-A inhibited the conversion of [3H]cytidine to deoxycytidine nucleotides and incorporation into DNA, suggesting that ribonucleotide reductase was an intracellular site of action. 2-F-ara-A (6 microM) in combination with 2,3-dihydro-1H-pyrazole[2,3-a]imidazole gave synergistic inhibition of L1210 cell growth. At lower concentrations of 2-F-ara-A, the inhibition by this combination was only additive. The addition of Desferal to the combination of 2-F-ara-A plus 2,3-dihydro-1H-pyrazole[2,3-a]imidazole provided a strong synergistic combination. Similar results were obtained with combinations which included F-ara-A, hydroxyurea, and Desferal. The combinations of 2-FdAdo plus 2,3-dihydro-1H-pyrazole[2,3-a]imidazole or hydroxyurea gave strong synergistic inhibition of L1210 cell growth, even at the lowest concentration of 2-FdAdo (0.6 microM) studied. The presence of Desferal in the combination served to further potentiate the synergism.


Asunto(s)
Desoxiadenosinas/análogos & derivados , Hidroxiurea/toxicidad , Leucemia L1210/fisiopatología , Pirazoles/toxicidad , Vidarabina/análogos & derivados , Animales , Supervivencia Celular/efectos de los fármacos , Citidina/metabolismo , Deferoxamina/toxicidad , Desoxiadenosinas/toxicidad , Desoxicitidina/biosíntesis , Sinergismo Farmacológico , Cinética , Ratones , Vidarabina/toxicidad
5.
Cancer Res ; 40(10): 3610-5, 1980 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-6254636

RESUMEN

The 2-fluoro derivative of 9-beta-D-arabinofuranosyladenine (2-F-ara-A) and its soluble 5'-formate and 5'-phosphate derivatives were therapeutically effective against the parent leukemia L1210 (L1210/0). 2-F-ara-A and 9-beta-D-arabinofuranosyladenine 5'-formate were inactive aginst a 1-beta-D-arabinofuranosylcytosine-resistant subline (L1210/ara-C) that was deficient in deoxycytidine kinase. Deoxycytidine prevented 2-F-ara-A-induced inhibition of proliferation of L1210/0 cells in culture and alleviated 2-F-ara-a inhibition of DNA synthesis. After treatment of mice with 9-beta-D-arabinofuranosyladenine 5'-formate, intracellular levels of the 5'-triphosphate of 9-beta-D-arabinofuranosylfluoroadenine in leukemia cells were more than 10 times higher in L1210/0 cells than in L1210/ara-C cells. Similar results were obtained in this line of leukemia cells from mice treated with the 5'-monophosphate of 9-beta-D-arabinofuranosyl-2-fluoroadenine. Thus, L1210/ara-C cells deficient in deoxycytidine kinase activity were also deficient in capacity to phosphorylate 2-F-ara-A. Kinase activity from L1210/0 cells for deoxycytidine and for 2-F-ara-A coeluted from calcium phosphate cellulose and from diethylaminoethyl cellulose columns and had similar mobility on gel electrophoresis. Deoxyadenosine kinase was clearly separated from deoxycytidine kinase. Deoxycytidine competed with 2-F-ara-A for phosphorylation by the partially purified enzyme from L1210 cells. These results indicate that 2-F-ara-A is phosphorylated to the 5'-monophosphate by deoxycytidine kinase of leukemia L1210 cells.


Asunto(s)
Desoxicitidina Quinasa/metabolismo , Leucemia L1210/tratamiento farmacológico , Fosfotransferasas/metabolismo , Vidarabina/análogos & derivados , Animales , Línea Celular , Cromatografía DEAE-Celulosa , ADN/biosíntesis , Desoxicitidina Quinasa/aislamiento & purificación , Electroforesis en Gel de Poliacrilamida , Leucemia L1210/enzimología , Leucemia L1210/patología , Ratones , Mutación , Fosforilación Oxidativa , Vidarabina/metabolismo , Vidarabina/uso terapéutico
6.
Cancer Res ; 44(10): 4297-302, 1984 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-6467191

RESUMEN

Several nucleoside analogues, some of which are potent inactivators of isolated S-adenosylhomocysteinase (AdoHcyase), were tested with respect to their effect on intracellular AdoHcyase and S-adenosylhomocysteine (AdoHcy) catabolism in intact rat hepatocytes. Among the analogues tested, 9-beta-D-arabinofuranosyladenine and 9-beta-D-arabinofuranosyl-3-deazaadenine were the most potent inactivators of intracellular AdoHcyase. Compounds like 2-chloroadenosine and carbocyclic adenosine are extremely efficient inactivators of the isolated enzyme, but these nucleosides exerted only a limited effect on the enzyme in intact hepatocytes. Only a moderate effect was observed with 2-chloro-3-deazaadenosine and 2'-deoxyadenosine, and a high concentration of 5'-deoxy-5'-methylthioadenosine was required to inactivate the enzyme. There was a correlation between inactivation and accumulation of AdoHcy. Carbocyclic-3-deazaadenosine caused no inactivation of AdoHcyase in liver cells but caused a massive accumulation of AdoHcy, suggesting that this analogue functions as a reversible inhibitor of the enzyme. Residual enzyme activity was observed with all the nucleoside analogues tested. The intracellular enzyme inactivated in the presence of 2'-deoxyadenosine and 9-beta-D-arabinofuranosyladenine was readily recovered when the cells were transferred to fresh medium containing adenosine deaminase, but reactivation of the enzyme activity after treatment of the cells with 2-chloroadenosine and 5'-deoxy-5'-methylthioadenosine was also observed. The inactivation of intracellular enzyme induced by 2-chloro-3-deazaadenosine, 9-beta-D-arabinofuranosyl-3-deazaadenine, and carbocyclic adenosine was essentially irreversible under the conditions of the experiments. Factors determining the effect of nucleoside analogues on intracellular AdoHcyase are discussed.


Asunto(s)
Hidrolasas/metabolismo , Hígado/enzimología , Nucleósidos/farmacología , Adenosilhomocisteinasa , Animales , Hidrolasas/antagonistas & inhibidores , Técnicas In Vitro , Cinética , Ratas , S-Adenosilhomocisteína/análisis , S-Adenosilmetionina/análisis , Relación Estructura-Actividad , Vidarabina/análisis
7.
Cancer Res ; 48(20): 5686-91, 1988 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-3167828

RESUMEN

Studies are described examining a new class of 4-aminofolate analogues modified by an N to C conversion and alkyl substitution at the N-5 position of aminopterin and methotrexate. All of these analogues were equivalent to aminopterin and methotrexate as inhibitors of tumor cell dihydrofolate reductase (Ki = 3.49-5.16 pM). N to C conversion at the N-5 position of aminopterin reduced its influx (inferred from the change in Ki) 3-fold, but the same modification increased influx of methotrexate 2-3-fold in Sarcoma 180 cells. Alkylation (methyl or ethyl) of this position on 5-deazaaminopterin increased influx 3-fold, while a similar alteration of 5-deazamethotrexate increased influx 4-5-fold. Influx of the methotrexate analogues was increased a total of 14-fold as a result of these modifications. Similar differences among these analogues were observed for inhibition of Sarcoma 180 cell growth in culture. Inhibitory potency was in the ascending order methotrexate less than 5-deazamethotrexate less than 5-deazaaminopterin less than aminopterin less than 5-alkyl (methyl or ethyl) analogues of 5-deazaaminopterin and 5-deazamethotrexate (the ethyl analogues were 2-fold more inhibitory than the methyl analogues). All of the analogues examined were equivalent in regard to efflux from Sarcoma 180 cells. Differences in transport alone did not account for all of the increased inhibitory potency (up to 33-fold) of the 5-alkyl-5-deaza analogues compared to the parent compounds. The extent of polyglutamylation of 5-deazaaminopterin and 5-deazamethotrexate and their 5-alkyl derivatives in Sarcoma 180 cells was substantially less compared to aminopterin and equivalent to methotrexate. Transport inward of 5-deazaaminopterin in isolated crypt cell epithelium from mouse small intestine was 2-fold lower than aminopterin (influx Km = 14.2 +/- 2 microM), while influx of 5-deazamethotrexate was 2-fold greater than methotrexate (influx Km = 98.6 +/- 23). However, transport inward of all of the 5-alkyl derivatives of these 5-deaza analogues was intermediate [influx Km = 44.4 +/- 11 (SEM) to 49.8 +/- 12 microM] between values for aminopterin and methotrexate. These differences accounted, to some extent, for the reduced toxicity of the 5-alkyl-5-deazaaminopterin analogues compared to aminopterin and the increased toxicity of 5-methyl-5-deazamethotrexate compared to methotrexate. All of the 5-alkyl derivatives of aminopterin and methotrexate were more active in vivo than methotrexate against four murine tumor models.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Aminopterina/análogos & derivados , Metotrexato/análogos & derivados , Metotrexato/uso terapéutico , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Alquilación , Aminopterina/uso terapéutico , Animales , Composición de Medicamentos , Epitelio/metabolismo , Intestino Delgado/citología , Intestino Delgado/metabolismo , Cinética , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Ratones , Sarcoma 180/tratamiento farmacológico , Relación Estructura-Actividad
8.
Cancer Res ; 46(6): 2866-70, 1986 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-3698011

RESUMEN

Adenosine and many adenosine analogues exert toxicity to mammalian cells at the nucleoside level. The mechanism of action of these agents is controversial. Previous experiments suggested that adenosine toxicity could be mediated by the accumulation of S-adenosylhomocysteine (AdoHcy), a potent inhibitor of S-adenosylmethionine (AdoMet) dependent methylation reactions. To analyze this question genetically, adenosine resistant, adenosine kinase deficient mutant clones of a murine T-lymphoma cell line (R1.1) have been selected and analyzed. Compared to parental lymphoma cells, the adenosine resistant mutants had severalfold elevated levels of AdoMet and an increased AdoMet:AdoHcy ratio. The activity of methionine adenosyltransferase was also raised in the mutants. The mutant cells were cross-resistant to agents postulated to cause accretion of AdoHcy, formation of AdoHcy analogues, impairment of AdoMet synthesis, or direct interference with AdoMet dependent reactions. These included 3-deazaadenosine, carbocyclic adenosine, carbocyclic 3-deazaadenosine, formycin A, 8-azaadenosine, 5'-deoxy-5'-methylthiotubercidin, 5'-deoxy-5'-methylthioadenosine, 5'-deoxy-5'-S-isobutylthioadenosine, adenine, cycloleucine, L-ethionine, seleno-DL-ethionine, and (+/-)-2-aminobicyclo[2.1.1]hexane-2-carboxylic acid. These results suggest that diverse purine nucleoside and methionine analogues may block the growth of adenosine kinase deficient cells by interference with AdoMet synthesis and degradation. An increase in AdoMet pools can render mammalian cells cross-resistant to multiple drugs affecting this essential metabolic pathway.


Asunto(s)
Adenosina/farmacología , Linfoma/metabolismo , Metionina/farmacología , S-Adenosilmetionina/metabolismo , Animales , Células Cultivadas , Resistencia a Medicamentos , Homocisteína/farmacología , Cinética , Linfoma/tratamiento farmacológico , Metionina/análogos & derivados , Metionina Adenosiltransferasa/análisis , Ratones , Mutación , S-Adenosilhomocisteína/metabolismo
9.
Cancer Res ; 43(1): 97-103, 1983 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-6847787

RESUMEN

In studies using a rapid kinetic technique, evidence was derived for multiplicity of systems mediating [3H]adenosine transport in L1210 cells. A variety of approaches were used in discriminating between transport and kinase-mediated phosphorylation. Under these conditions, two systems mediating influx were delineated which exhibited high-affinity [Km = 13.9 +/- 2 (S.E.) microM] or low-affinity [Km = 199 +/- 27 microM] for [3H]-adenosine. Both systems exhibited high capacities, but that associated with the low-affinity system (V 37 degrees max = 263 +/- 43 nmol = 99.6 +/- 12 nmol sec/g, dry weight). The relative difference in affinity of these two systems during influx was also reflected in the values for influx Ki obtained with other nucleosides and nucleoside analogues. Influx of [3H]-adenosine by each mediated system was inhibited by 6-(2-hydroxy-5-nitrobenzyl)thioguanosine, a specific transport inhibitor, and by 9-beta-D-arabinofuranosylpurine-6(1H)thione which is not phosphorylated in L1210 cells. Influx kinetics were the same in L1210 cells, in adenosine triphosphate-depleted L1210 cells (L1210/ara-C/MMPR) which have substantially reduced ability for [3H]adenosine phosphorylation, and in the presence of 2'-deoxycoformycin, a potent inhibitor of adenosine deaminase. The same multiplicity in mediated influx of [3H]adenosine was shown at 0 degrees when transport became rate limiting to total uptake. The high-affinity system mediating [3H]adenosine influx was also elucidated in L1210 cell plasma membrane vesicles in the presence or absence of 2'-deoxycoformycin. Almost all of the natural nucleosides examined competed less effectively with [3H]adenosine for influx by the high-affinity system than by the low-affinity system. These results are discussed with respect to possible pharmacological implications.


Asunto(s)
Adenosina/metabolismo , Leucemia L1210/metabolismo , Nucleósidos de Purina/metabolismo , Animales , Transporte Biológico , Membrana Celular/metabolismo , Cinética , Ratones , Temperatura , Factores de Tiempo , Vidarabina/análogos & derivados , Vidarabina/metabolismo
10.
Cancer Res ; 42(3): 924-8, 1982 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7059991

RESUMEN

The M5076 murine "ovarian" tumor which is naturally refractive to methotrexate was found to be highly responsive to the lipophilic antifolate, metoprine. M5076 cells were markedly deficient in mediated entry of methotrexate. This was in contrast to the L1210 leukemia, a tumor highly responsive to methotrexate but poorly responsive to metoprine. Two L1210 leukemia sublines, with acquired resistance to methotrexate by virtue of a deficiency in mediated entry of drug similar to that seen for M5076 cells, were found to be collaterally sensitive to metoprine. The insensitivity to methotrexate of the M5076 tumor and the two L1210 sublines is associated with low saturability (high Km) and reduced capacity (low Vmax) for mediated influx of drug. 5-Methyltetrahydrofolate, the major circulating folate in blood but not metoprine, shares this mediated route for entry. Therefore, a relatively low level of accumulation of this natural folate in these methotrexate-resistant tumors, in the face of a metoprine-induced blockade at the level of dihydrofolate reductase, probably accounts for the high sensitivity of these tumors to this lipophilic agent. Evidence for this notion was derived during transport and growth experiments in vitro using 5-formyltetrahydrofolate as a model folate coenzyme. The value for influx Vmax of this folate compound in a transport-deficient methotrexate-resistant subline compared to the parental L1210 was reduced to the same extent as that shown for methotrexate. Growth of this resistant L1210 subline showed a greater requirement for this model compound than did the parental line. Also, the concentration necessary for 50% inhibition by metoprine in the presence of this reduced folate was lower in the resistant subline. Inhibition of each cell line by metoprine, on the other hand, was the same when folic acid was used as the folate source. The implications of these findings for the use of lipophilic antifolates as alternative therapy for some methotrexate-resistant tumors are discussed.


Asunto(s)
Neoplasias Experimentales/metabolismo , Pirimetamina/análogos & derivados , Animales , Transporte Biológico , Línea Celular , Resistencia a Medicamentos , Formiltetrahidrofolatos/metabolismo , Cinética , Leucemia L1210/tratamiento farmacológico , Leucemia L1210/metabolismo , Metotrexato/administración & dosificación , Ratones , Trasplante de Neoplasias , Neoplasias Experimentales/tratamiento farmacológico , Pirimetamina/farmacología , Tetrahidrofolato Deshidrogenasa/metabolismo
11.
Cancer Res ; 54(7): 1742-5, 1994 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-8137289

RESUMEN

6-Thio-2'-deoxyguanosine (T-dGuo) has been reported to be both phosphorylated by deoxycytidine kinase and converted to 6-thioguanine by purine nucleoside phosphorylase (PNP). Combination of T-dGuo with an inhibitor of PNP would be expected to generate the 5'-triphosphate of T-dGuo and limit or prevent the formation of 6-thioguanosine triphosphate. Because the incorporation of 6-thioguanine into DNA is believed to be primarily responsible for the antitumor activity of the thiopurines, this treatment might result in enhanced activity against certain tumors, particularly those of T-cell origin. We have evaluated the metabolic basis of this strategy by examining the effects of 9-benzyl-9-deazaguanine (BDG), a potent inhibitor of PNP, on the metabolism of T-dGuo in CEM cells. The concentration of T-dGuo required to inhibit cell growth by 50% was approximately 50-fold greater in the presence of 8.0 microM BDG than in its absence. As expected, the addition of BDG to cells treated with T-dGuo prevented the metabolism of T-dGuo to 6-thio-guanine-containing ribo-nucleotides, but, unexpectedly, no 6-thio-2'-deoxyguanosine 5'-triphosphate was detected. In cells treated with T-dGuo plus BDG, the major phosphorylated metabolite was T-dGMP. These results indicated that even in the absence of PNP activity, T-dGuo cannot be phosphorylated directly to 6-thio-2'-deoxyguanosine 5'-triphosphate due to the inability of guanylate kinase to utilize T-dGMP as a substrate.


Asunto(s)
Compuestos de Bencilo/farmacología , Desoxiguanosina/análogos & derivados , Guanina/análogos & derivados , Purina-Nucleósido Fosforilasa/antagonistas & inhibidores , Tionucleósidos/toxicidad , Biotransformación , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Desoxiguanosina/metabolismo , Desoxiguanosina/toxicidad , Relación Dosis-Respuesta a Droga , Guanina/farmacología , Humanos , Tionucleósidos/metabolismo
12.
Cancer Res ; 49(5): 1182-6, 1989 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-2537144

RESUMEN

Clomesone was evaluated for antitumor activity against a spectrum of animal tumor models. Clomesone exhibited significant antitumor activity against the murine L1210 leukemia implanted i.p., s.c., and intracerebrally (i.c.). Activity against s.c.-implanted tumor was largely independent of schedule and route of administration. Therapeutically optimal single-dose treatment (for tumored mice) was less toxic to nontumored mice than therapeutically optimal prolonged treatment. Clomesone also exhibited activity against other murine tumors (P388 leukemia, B16 melanoma, Lewis lung carcinoma, and M5076 sarcoma). It was active against P388 leukemia sublines resistant to cyclophosphamide, L-phenylalanine mustard, and cis-diamminedichloroplatinum(II). No activity was observed against a P388 subline resistant to N,N'-bis(2-chloroethyl)-N-nitrosourea or against Ridgway osteogenic sarcoma, a nitrosourea-resistant murine solid tumor. Clomesone is generally as effective as the chloroethylnitrosoureas against experimental tumor models. Since clomesone does not have the hydroxyethylating and carbamoylating activities of the chloroethylnitrosoureas (which do not appear to contribute to antitumor activity), it would likely be a more toxicologically selective compound. It may prove to be less carcinogenic than the chloroethylnitrosoureas, and it may contribute less target organ toxicity and less interference with the actions of other drugs when used in combinations.


Asunto(s)
Antineoplásicos/uso terapéutico , Mesilatos/uso terapéutico , Neoplasias Experimentales/tratamiento farmacológico , Animales , Daño del ADN , Resistencia a Medicamentos , Etilnitrosourea/análogos & derivados , Etilnitrosourea/farmacología , Mesilatos/administración & dosificación , Mesilatos/farmacología , Ratones , Ratones Endogámicos
13.
Cancer Res ; 45(11 Pt 2): 5958-63, 1985 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3863711

RESUMEN

A novel nucleoside, in the amount of 400 micrograms, was isolated from a 24-h collection of urine of a chronic myelogenous leukemia patient. On the basis of ultraviolet, nuclear magnetic resonance, and mass spectrometry and chromatography, its structure was established to be 7-beta-D-ribofuranosylhypoxanthine. The ultraviolet and mass spectral data and the thin layer chromatographic mobilities of the natural material were identical to those of a synthetic sample. High performance liquid chromatographic retention times and the coinjection high performance liquid chromatography of the natural material with the synthetic samples of the alpha and beta-anomers of 7-ribofuranosylhypoxanthines further confirmed the identity of the isolated material as 7-beta-D-ribofuranosylhypoxanthine.


Asunto(s)
Inosina/orina , Leucemia Mieloide/orina , Adulto , Cromatografía Líquida de Alta Presión , Cromatografía en Capa Delgada , Humanos , Espectroscopía de Resonancia Magnética , Masculino , Espectrometría de Masas , Espectrofotometría Ultravioleta , Vitamina B 12/sangre
14.
Cancer Res ; 51(9): 2386-94, 1991 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-1707752

RESUMEN

2-Chloro-9-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)-adenine (Cl-F-ara-A) has activity against the P388 tumor in mice on several different schedules. Biochemical studies with a chronic myelogenous leukemia cell line (K562) grown in cell culture have been done in order to better understand its mechanism of action. Cl-F-ara-A was a potent inhibitor of K562 cell growth. Only 5 nM inhibited K562 cell growth by 50% after 72 h of continuous incubation. The 5'-triphosphate of Cl-F-ara-A was detected by strong anion exchange chromatography of the acid-soluble extract of K562 cells incubated with Cl-F-ara-A. Competition studies with natural nucleosides suggested that deoxycytidine kinase was the enzyme responsible for the metabolism to the monophosphate. Incubation of K562 cells for 4 h with 50 nM Cl-F-ara-A inhibited the incorporation of [3H]thymidine into the DNA by 50%. Incubation with 0.1, 1, or 10 microM Cl-F-ara-A for 4 h depressed dATP, dCTP, and dGTP pools but did not affect TTP pools. Similar inhibition of deoxyribonucleoside triphosphate pools was seen after incubation with 2-chloro-2'-deoxyadenosine. Both Cl-F-ara-ATP and Cl-dATP potently inhibited the reduction of ADP to dADP in crude extracts of K562 cells (concentration producing 50% inhibition, 65 nM). The effect of Cl-F-ara-ATP on human DNA polymerases alpha, beta, and gamma isolated from K562 cells grown in culture was determined and compared with those of Cl-dATP and 9-beta-D-arabinofuranosyl-2-fluoroadenine triphosphate (F-ara-ATP). Cl-F-ara-ATP was a potent inhibitor of DNA polymerase alpha. Inhibition of DNA polymerase alpha was competitive with respect to dATP (Ki of 1 microM). The three analogue triphosphates were incorporated into the DNA by DNA polymerase alpha as efficiently as dATP. The incorporation of Cl-F-ara-AMP inhibited the further elongation of the DNA chain, similarly to that seen after the incorporation of F-ara-AMP. Extension of the DNA chain after the incorporation of Cl-dAMP was not inhibited as much as it was with either Cl-F-ara-AMP or F-ara-AMP. Cl-F-ara-ATP was not a potent inhibitor of DNA polymerase beta, DNA polymerase gamma, or DNA primase.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Arabinonucleósidos/farmacología , ADN/biosíntesis , Inhibidores de la Síntesis del Ácido Nucleico , Ribonucleótido Reductasas/antagonistas & inhibidores , Nucleótidos de Adenina , Adenosina Trifosfato/metabolismo , Arabinonucleósidos/metabolismo , División Celular/efectos de los fármacos , Clofarabina , Citidina Trifosfato/metabolismo , Desoxicitidina/metabolismo , Guanidina , Guanidinas/metabolismo , Humanos , ARN/biosíntesis , Células Tumorales Cultivadas
15.
Biochim Biophys Acta ; 881(2): 175-84, 1986 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-3955073

RESUMEN

5'-Bromoacetamido-5'-deoxythymidine (BAT), 5'-iodoacetamido-5'-deoxythymidine (IAT), 5'-chloroacetamido-5'-deoxythymidine (CAT) and [14C]BAT were synthesized and their interactions with thymidylate synthase purified from L1210 cells were investigated. The inhibitory effects of these compounds on thymidylate synthase were in the order BAT greater than IAT greater than CAT, which is in agreement with their cytotoxic effects in L1210 cells. In the presence of substrate during preincubation, the concentration required for 50% inhibition of the enzyme activity by these inhibitors was 4-8-fold higher than it was in the absence of dUMP. The I50 values for BAT were 1 X 10(-5) M and 1.2 X 10(-6) M in the presence and absence, respectively, of dUMP during preincubation. These results were in agreement with the observed inhibition of thymidylate synthase by BAT in intact L1210 cells. A Lineweaver-Burk plot revealed that BAT behaved as a competitive inhibitor. The Km for the enzyme was 9.2 microM, and the Ki determined for competitive inhibition by BAT was 5.4 microM. Formation of a tight, irreversible complex is inferred from the finding that BAT-inactivation of thymidylate synthase was not reversible on prolonged dialysis and that the enzyme-BAT complex was nondissociable by gel filtration through a Sephadex G-25 column or by TSK-125 column chromatography. Incubation of thymidylate synthase with BAT resulted in time-dependent, irreversible loss of enzyme activity by first-order kinetics. The rate constant for inactivation was 0.4 min-1, and the steady-state constant of inactivation, Ki, was estimated to be 6.6 microM. The 5'-haloacetamido-5'-deoxythymidines provide specific inhibitors of thymidylate synthase that may also serve as reagents for studying the enzyme mechanism.


Asunto(s)
Timidina/análogos & derivados , Timidilato Sintasa/antagonistas & inhibidores , Animales , Cromatografía en Gel , Cinética , Leucemia L1210/enzimología , Ratones , Timidina/farmacología
16.
Biochim Biophys Acta ; 862(1): 72-80, 1986 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-3768370

RESUMEN

We have examined the growth-inhibitory potency of several pteridines encapsulated in negatively charged liposomes, including methotrexate, methotrexate-gamma-methylamide, methotrexate-gamma-dimethylamide, methotrexate-alpha-aspartate, and a lipophilic methotrexate-phosphatidylethanolamine conjugate. The potency of encapsulated methotrexate is greater than the potency of the free drug for CV1-P cells, but not for other cell lines. The potency of methotrexate-gamma-methylamide and methotrexate-gamma-dimethylamide is only minimally improved by encapsulation. The potency of methotrexate-alpha-aspartate is increased by encapsulation. In addition, the lipophilic methotrexate derivative has demonstrable potency when incorporated in liposomes. We have also examined the potency of several pteridines under conditions where the cells are exposed to the drug for periods shorter than the entire growth assay. Reduction of the exposure time decreases the potency of both encapsulated and free drugs. However, the difference in potency between the encapsulated and free drug is increased, because the potency of the encapsulated drug is affected less. Consequently, encapsulated methotrexate-gamma-aspartate is 300-fold more potent than free drug, if CV1-P cells are exposed to drug for 4 h. Moreover, encapsulated methotrexate is more potent than free methotrexate for growth inhibition of L929 fibroblasts, if the term of exposure is less than 8 h. Potency is least affected by reduction of exposure length for the lipophilic methotrexate derivative.


Asunto(s)
Liposomas/administración & dosificación , Metotrexato/análogos & derivados , Metotrexato/farmacología , Pteridinas/farmacología , Línea Celular , Metotrexato/administración & dosificación , Fosfatidiletanolaminas/administración & dosificación , Pteridinas/administración & dosificación , Pteridinas/metabolismo , Relación Estructura-Actividad , Factores de Tiempo
17.
Biochim Biophys Acta ; 829(2): 150-5, 1985 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-3995047

RESUMEN

The irreversible inactivation of S-adenosylhomocysteine hydrolase purified from hamster and bovine liver by adenosine analogs substituted in the 5' and 2 positions has been investigated in detail. 5'-Cyano-5'-deoxyadenosine inactivates as potently as 9-beta-D-arabinofuranosyladenine (Ara-A). Substitution of the Ara-A at the 2 position by halogens or deleting N at the 3 position decreases its potency. Although weak, 2',3'-dideoxyadenosine can also inactivate the enzyme. The irreversible inactivation of the hydrolase in rat hepatocytes incubated with 2-chloroadenosine or 3-deaza-Ara-A could be demonstrated, concomitant with increases in 35S-labeled S-adenosylhomocysteine and S-adenosylmethionine in the hepatocytes.


Asunto(s)
Hidrolasas/antagonistas & inhibidores , Nucleósidos/farmacología , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosilhomocisteinasa , Animales , Bovinos , Cricetinae , Técnicas In Vitro , Cinética , Hígado/enzimología , Masculino , Ratas , Ratas Endogámicas , Vidarabina/análogos & derivados , Vidarabina/farmacología
18.
J Mol Biol ; 267(3): 584-94, 1997 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-9126840

RESUMEN

The active site of influenza virus neuraminidase (NA) is formed by 11 universally conserved residues. A guanidino group incorporated into two unrelated NA inhibitors was previously reported to occupy different negatively charged sites in the NA active site, A new inhibitor containing two guanidino groups was synthesized in order to utilize both sites in an attempt to acquire a combined increase in affinity. The X-ray crystal structures of the complexes show that the expected increase in affinity could not be achieved even though the added guanidino group binds to the negatively charged site as designed. This suggests that the ligand affinity to the target protein is contributed both from ligand-protein interactions and solvation/conformation energy of the ligand.


Asunto(s)
Guanidinas/farmacología , Hidroxibenzoatos/farmacología , Virus de la Influenza B/enzimología , Neuraminidasa/química , Sitios de Unión , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , Guanidinas/química , Humanos , Hidroxibenzoatos/química , Modelos Moleculares , Neuraminidasa/antagonistas & inhibidores , Agua/química
19.
Hum Gene Ther ; 8(14): 1637-44, 1997 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-9322865

RESUMEN

We have developed a new strategy for the gene therapy of cancer based on the activation of purine nucleoside analogs by transduced E. coli purine nucleoside phosphorylase (PNP, E.C. 2.4.2.1). The approach is designed to generate antimetabolites intracellularly that would be too toxic for systemic administration. To determine whether this strategy could be used to kill tumor cells without host toxicity, nude mice bearing human malignant D54MG glioma tumors expressing E. coli PNP (D54-PNP) were treated with either 6-methylpurine-2'-deoxyriboside (MeP-dR) or arabinofuranosyl-2-fluoroadenine monophosphate (F-araAMP, fludarabine, a precursor of F-araA). Both prodrugs exhibited significant antitumor activity against established D54-PNP tumors at doses that produced no discernible systemic toxicity. Significantly, MeP-dR was curative against this slow growing solid tumor after only 3 doses. The antitumor effects showed a dose dependence on both the amount of prodrug given and the level of E. coli PNP expression within tumor xenografts. These results indicated that a strategy using E. coli PNP to create highly toxic, membrane permeant compounds that kill both replicating and nonreplicating cells is feasible in vivo, further supporting development of this cancer gene therapy approach.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Terapia Genética/métodos , Glioma/tratamiento farmacológico , Profármacos/farmacología , Purina-Nucleósido Fosforilasa/fisiología , Animales , Antimetabolitos Antineoplásicos/toxicidad , Escherichia coli/enzimología , Escherichia coli/genética , Vectores Genéticos/genética , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Nucleósidos de Purina/uso terapéutico , Nucleósidos de Purina/toxicidad , Purina-Nucleósido Fosforilasa/genética , Retroviridae/genética , Fosfato de Vidarabina/análogos & derivados , Fosfato de Vidarabina/uso terapéutico , Fosfato de Vidarabina/toxicidad
20.
J Med Chem ; 21(1): 112-4, 1978 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-619139

RESUMEN

A synthesis of 1-deaza-6-thioguanosine (8) and 1-deaza-6-(methylthio)guanosine (9) from 2-amino-6-chloro-1-deazapurine (4) is described. The reaction of the N2-acetyl derivative of 4 with 2,3,5-tri-O-acetyl-D-ribofuranosyl chloride in the presence of Linde 4A molecular sieve gave the blocked nucleoside 6. Deacetylation of 6 gave the chloro nucleoside 7 which was treated at high temperature with hydrogen sulfide and methyl mercaptan to give 8 and 9, respectively. The structure of 7 was confirmed by 1H NMR and by conversion to the cyclonucleoside 14. Compound 4 gave a 79% increase in life span in the L1210 mouse leukemia screen.


Asunto(s)
Guanosina/análogos & derivados , Tionucleósidos/síntesis química , Animales , Antineoplásicos/síntesis química , División Celular/efectos de los fármacos , Células Cultivadas , Guanosina/síntesis química , Guanosina/farmacología , Guanosina/uso terapéutico , Leucemia L1210/tratamiento farmacológico , Ratones , Tionucleósidos/farmacología , Tionucleósidos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA