Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Cancer Res ; 36(11 Pt 1): 3917-23, 1976 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-975040

RESUMO

8-Azainosine (8-aza-HR) is of interest because of its activity against experimental tumors. Metabolic studies in cell cultures were performed with 8-aza-HR and with the structurally related nucleoside, 8-azaadenosine (9-beta-D-ribofuranosyl-8-azaadenine) (8-aza-AR), which has a lower degree of antitumor activity than does 8-aza-HR. In H. Ep. 2 cells and in Ca755 cells, both 14C-labeled nucleosides were metabolized to nucleotides of 8-azaadenine (8-aza-A) and 8-azaguanine (8-aza-G) and incorporated into polynucleotides as 8-aza-A and 8-aza-G. 8-Aza HR was incorporated primarily as 8-aza-G, whereas 8-aza-AR was incorporated about equally as 8-aza-A and 8-aza-G. In H. Ep. 2 cells, the extent of incorporation of 8-aza-HR as 8-aza-G was about one-half that found when [14C]-8-aza-G was the precursor. In the H. Ep. 2/FA/FAR cell line, 8-aza-AR and 8-aza-HR were metabolized similarly, in that both were incorporated into polynucleotides principally as 8-aza-G; apparently, in this cell line which is deficient in adenosine kinase and adenine phosphoribosyltransferase, 8-aza-AR is metabolized by conversion to 8-aza-HR. A cell line (H. Ep 2/8-aza HR), which was resistant to 8-aza-HR but sensitive to 8-aza-AR and which retained hypoxanthine (guanine)-phosphoribosyltransferase activity, metabolized 8-aza-HR to only a small extent. However, in this cell-line, 8-aza-AR was more extensively metabolized and was incorporated primarily as 8-aza-A. The failure of these cells to convert 8-aza-AR or 8-aza-HR to 8-aza-G indicates that the basis for resistance may be a change in the substrate specificities of the enzymes of guanosine monophosphate synthesis such that these cells no longer effectively convert 8-azainosine monophosphate to 8-azaguanosine monophosphate. 8-Aza-AR was a potent inhibitor of purine synthesis de novo, but 8-aza-HR, at concentrations much higher than the inhibitory concentration of 8-aza-AR, did not inhibit this process. In H. Ep. 2 cells, 8-aza-HR blocked the conversion of orotic acid to uridine nucleotides and caused an accumulation of orotidine. This inhibition of pyrimidine biosynthesis apparently does not contribute significantly to the cytotoxicity of 8-aza-HR because uridine provided no degree of reversal of its inhibition of the growth of cell cultures.


Assuntos
Adenosina/análogos & derivados , Inosina/análogos & derivados , Adenosina/metabolismo , Adenosina/farmacologia , Animais , Azaguanina/metabolismo , Linhagem Celular , Cromatografia em Camada Fina , Humanos , Técnicas In Vitro , Inosina/farmacologia , Camundongos , Ácido Orótico/metabolismo , Polinucleotídeos/biossíntese , Purinas/biossíntese , Nucleotídeos de Pirimidina/biossíntese , Timidina/metabolismo , Uridina/metabolismo
2.
Cancer Res ; 36(4): 1375-8, 1976 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-130972

RESUMO

The effects of 2-amino-1,3,4-thiadiazole [aminothiadiazole (NSC 4728)] on purine and pyrimidine ribonucleotide pools of L1210 ascites cells in vivo are presented and discussed as they relate to the site of action. Within 1 hr after administration of the drug, the levels of guanosine triphosphate, guanosine diphosphate, adenosine triphosphate, and adenosine diphosphate were reduced, whereas those of inosine monophosphate (IMP) and uridine triphosphate were increased. The most pronounced effects were the lowering of guanine ribonucleotide pools and the elevation of IMP. Aminothiadiazole produced a marked inhibition (approximately 95%) of the incorporation of [8-14C]inosine into guanine nucleotides, whereas only a slight inhibition (approximately 20%) of incorporation into adenine nucleotides was observed. These results suggest that the thiadiazole (or a metabolite thereof) inhibits the conversion of IMP to guanosine monophosphate; this conclusion is reinforced by the observation that mycophenolic acid, a known inhibitor of this conversion, produced effects on ribonucleotide pools similar to those produced by aminothiadiazole. Aminothiadiazole did not inhibit IMP dehydrogenase isolated from L1210 cells. The effects of the thiadiazole on nucleotide pools were prevented by simultaneous administration of nicotinamide. Since nicotinamide is known to prevent or reverse the antileukemic activity of aminothiadiazole, it is probable that the inhibition of synthesis of guanosine monophosphate is related to the antileukemic action of this agent.


Assuntos
Leucemia L1210/metabolismo , Ribonucleotídeos/metabolismo , Tiadiazóis/farmacologia , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Nucleotídeos de Guanina/metabolismo , Guanosina Trifosfato/metabolismo , IMP Desidrogenase/metabolismo , Inosina/metabolismo , Nucleotídeos de Inosina/metabolismo , Niacinamida/farmacologia , Tiadiazóis/antagonistas & inibidores , Nucleotídeos de Uracila/metabolismo
3.
Cancer Res ; 37(1): 182-7, 1977 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-187332

RESUMO

The synthesis and isolation of two derivatives of 2-amino-1,3,4-thialdiazole(aminothiadiazole) are described. The derivatives are a nicotinamide adenine dinucleotide (NAD) analog prepared by an exchange reaction with NAD in the presence of nicotineamide adenine dinucleotide glycohydrolase and a presumed aminothiadiazole mononucleotide prepared by treatment of the NAD analog with nucleotide pyrophosphatase. Both derivatives are potent inhibitors of inosine 5'-phosphate (IMP) dehydrogenase obtained from leukemia L1210 cells. The NAD analog is a pseudoir-reversible inhibitor of the enzyme, noncompetitive with either IMP or NAD. The aminothiadiazole mononucleotide has a K1 of about 0.1 muM, is competitive with IMP, and is uncompetitive with NAD: the inhibition appears to be reversible by Ackermann-Potter analysis. A metabolite of [5-14C]aminothiadiazole is formed in L1210 cells in vivo to a level of 0.3 nmole/10(9) cells. Retention volume of the metabolite on a high-pressure liquid chromatography system is the same as that of the aminothiadiazole mononucleotide prepared as described above. These results suggest that IMP dehydrogenase is the site of action for aminothiadiazole metabolites as was indicated by earlier observations. There is no evidence that the NAD analog is formed in vivo. Nicotinamide prevented formation of the mononucleotide in vivo. Therefore, since formation and cleavage of the NAD analog apparently are not the route to the thiadiazole nucleotide, some other pathway for the metabolism of nicotinamide may be involved such as the action of a phosphoribosyltransferase or the sequential action of a nucleoside phosphorylase and a nucleoside kinase.


Assuntos
IMP Desidrogenase/antagonistas & inibidores , Cetona Oxirredutases/antagonistas & inibidores , Leucemia L1210/metabolismo , NAD/análogos & derivados , Tiadiazóis/farmacologia , Animais , Ligação Competitiva , Técnicas In Vitro , Nucleotídeos de Inosina/metabolismo , Camundongos , NAD/metabolismo , NAD/farmacologia , Niacinamida/metabolismo , Niacinamida/farmacologia , Tiadiazóis/metabolismo
4.
Cancer Res ; 42(6): 2260-4, 1982 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7042080

RESUMO

9-beta-D-Arabinofuranosyl-2-fluoroadenine (2-F-ara-A), a derivative of 9-beta-D-arabinofuranosyladenine (ara-A) that is resistant to deamination, selectively inhibits DNA synthesis and has activity against mouse leukemia L1210 comparable to that of ara-A plus the adenosine deaminase inhibitor, 2'-deoxycoformycin. To determine if these two nucleosides have similar modes of action, comparisons were made of their effects and those of their triphosphates on enzymes known to be inhibited by ara-A or 9-beta-D-arabinofuranosyladenine 5'-triphosphate. 9-beta-D-Arabinofuranosyl-2-fluoroadenine 5'-triphosphate was more effective than 9-beta-D-arabinofuranosyladenine 5'-triphosphate in inhibiting the reduction of adenosine 5'-diphosphate and cytidine 5'-diphosphate by ribonucleotide reductase from HEp-2 cells or L1210 cells. DNA polymerase alpha from L1210 cells was equally sensitive to 9-beta-D-arabinofuranosyl-2-fluoroadenine 5'-triphosphate and 9-beta-D-arabinofuranosyladenine 5'-triphosphate, and DNA polymerase beta from L1210 cells was much less sensitive to both triphosphates. S-Adenosylhomocysteine hydrolase from L1210 cells was inactivated by 2-F-ara-A and ara-A, but higher concentrations of the fluoro derivative were required. These results are consistent with 2-F-ara-A and ara-A inhibition of DNA synthesis by inhibition of ribonucleotide reductase and DNA polymerase alpha.


Assuntos
Replicação do DNA/efeitos dos fármacos , Hidrolases/antagonistas & inibidores , Leucemia L1210/enzimologia , Inibidores da Síntese de Ácido Nucleico , Ribonucleotídeo Redutases/antagonistas & inibidores , Vidarabina/análogos & derivados , Vidarabina/farmacologia , Adenosil-Homocisteinase , Animais , DNA Polimerase I/antagonistas & inibidores , DNA Polimerase II/antagonistas & inibidores , Camundongos
5.
Cancer Res ; 54(7): 1742-5, 1994 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-8137289

RESUMO

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.


Assuntos
Compostos de Benzil/farmacologia , Desoxiguanosina/análogos & derivados , Guanina/análogos & derivados , Purina-Núcleosídeo Fosforilase/antagonistas & inibidores , Tionucleosídeos/toxicidade , Biotransformação , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Desoxiguanosina/metabolismo , Desoxiguanosina/toxicidade , Relação Dose-Resposta a Droga , Guanina/farmacologia , Humanos , Tionucleosídeos/metabolismo
6.
Cancer Res ; 51(9): 2386-94, 1991 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-1707752

RESUMO

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)


Assuntos
Arabinonucleosídeos/farmacologia , DNA/biossíntese , Inibidores da Síntese de Ácido Nucleico , Ribonucleotídeo Redutases/antagonistas & inibidores , Nucleotídeos de Adenina , Trifosfato de Adenosina/metabolismo , Arabinonucleosídeos/metabolismo , Divisão Celular/efeitos dos fármacos , Clofarabina , Citidina Trifosfato/metabolismo , Desoxicitidina/metabolismo , Guanidina , Guanidinas/metabolismo , Humanos , RNA/biossíntese , Células Tumorais Cultivadas
7.
Hum Gene Ther ; 8(14): 1637-44, 1997 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-9322865

RESUMO

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.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Terapia Genética/métodos , Glioma/tratamento farmacológico , Pró-Fármacos/farmacologia , Purina-Núcleosídeo Fosforilase/fisiologia , Animais , Antimetabólitos Antineoplásicos/toxicidade , Escherichia coli/enzimologia , Escherichia coli/genética , Vetores Genéticos/genética , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Nucleosídeos de Purina/uso terapêutico , Nucleosídeos de Purina/toxicidade , Purina-Núcleosídeo Fosforilase/genética , Retroviridae/genética , Fosfato de Vidarabina/análogos & derivados , Fosfato de Vidarabina/uso terapêutico , Fosfato de Vidarabina/toxicidade
8.
Endocrinology ; 101(1): 203-8, 1977 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-324755

RESUMO

Increasing levels of magnesium were found to cause a marked depression of glucosestimulated insulin secretion at fixed calcium levels, particularly at levels which bracketed the concentration of ultrafiltrable magnesium found in normal rat plasma (1.3 meq/l), i.e., increasing magnesium from 0.6 to 1.2 meq/l depressed secretion, and increasing magnesium from 1.2 to 2.4 meq/l resulted in a further depression. Paradoxically, when magnesium was omitted from the perfusing medium, insulin secretion was also depressed. The data strongly suggest that the calcium/magnesium ratio is a primary regulator of the insulin secretory process, since a relatively slight alteration of the physiologic ratio of calcium to magnesium (approximately 2.5) results in a marked alteration of total insulin secretion. In addition, small amounts of magnesium are necessary for optimum secretion, possibly reflecting the requirement for magnesium in several enzymatic processes. Thus, magnesium may play an important role in the regulation of insulin secretion by altering the sensitivity of the beta cells of the Islets of Langerhans to glucose.


Assuntos
Glucose/farmacologia , Insulina/metabolismo , Magnésio/farmacologia , Pâncreas/metabolismo , Animais , Cálcio/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Pâncreas/efeitos dos fármacos , Ratos
9.
J Med Chem ; 24(10): 1254-8, 1981 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-6948961

RESUMO

Previously, 8-deazafolic acid (17) was shown to be a potent inhibitor of the folate-dependent bacteria, Streptococcus faecium (ATCC 8043) and Lactobacillus casei (ATCC 7469), and to have activity against lymphoid leukemia L1210 in mice. To examine the 5,6,7,8-tetrahydro derivatives, a new synthesis of 17 was developed from 8-deaza-2,4-dichloro-6-methylpteridine. Treatment of the latter with aqueous base gave the corresponding pteridin-4(3H)-one, which was aminated with ammonia to give 8-deaza-6-methylpterin (9). Bromination of 9 gave mainly 8-deaza-6-(tribromomethyl)pterin, which on reaction with p-aminobenzoyl-L-glutamic acid resulted in the formation of the 9-oxo derivative of 17. In contrast, bromination of the 2-acetyl derivative of 9 gave mainly the corresponding 6-(bromomethyl)pterin, which was converted to 17 in 23% yield (from 9). Hydrogenation of 17 at atmospheric pressure and room temperature was unsuccessful either in a basic medium or formic acid. In trifluoroacetic acid, overreduction occurred to give a mixture containing 8-deaza-5,6,7,8-tetrahydro-6-methylpterin and the 5,6,7,8-tetrahydro derivative of 17. The latter was characterized by conversion to the methenyl analogue 21, which was also prepared by hydrogenation of the 10-formyl derivative of 17. Treatment of 21 with hydroxide gave 8-deaza-10-formyl-5,6,7,8-tetrahydrofolic acid. Compound 21 showed cytotoxicity to cultured H.Ep.-2 cells and was tested as an inhibitor of bovine dihydrofolic reductase. Lineweaver-Burk analysis indicated inhibition competitive with dihydrofolate.


Assuntos
Antimetabólitos Antineoplásicos/síntese química , Ácido Fólico/análogos & derivados , Tetra-Hidrofolatos/síntese química , Animais , Antimetabólitos Antineoplásicos/farmacologia , Bovinos , Células Cultivadas , Ácido Fólico/síntese química , Ácido Fólico/farmacologia , Antagonistas do Ácido Fólico , Camundongos , Tetra-Hidrofolatos/farmacologia
10.
J Med Chem ; 25(2): 161-6, 1982 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7057422

RESUMO

Reaction of 5,6,7,8-tetrahydrofolic acid (THF,7) with phosgene, thiophosgene, and cyanogen bromide gave the bridged derivatives, 5,10-(CO)-THF (8), 5,10-(CS)-THF (9), and 5,10-(C = NH)-THF (11), respectively. Catalytic hydrogenation of 10-(chloroacetyl)folic acid (2) gave 5,10-(CH2CO)-THF (12). A similar reaction with 10-(3-chloropropionyl)folic acid (3) gave 10-(ClCH2CH2CO)-THF (14) rather than 5,10-(CH2CH2CO)-THF (13). In the catalytic hydrogenation of 10-ethoxalylfolic acid (5), the initial product 10-(EtO2CCO)-THF (22) rearranged readily to give 5-(EtO2CCO)-THF (21). Acylation of THF with chloroacetyl chloride gave a N5,N10-diacylated product (18 or 19), which could not be converted to 5,10-COCH2)-THF (17). Reductive alkylation of THF with glyoxylic acid and 5-hydroxypentanal, respectively, gave 5-(HO2CCH2)-THF (24) and 5-[HO(CH2)5]-THF (25). Reductive dialkylation of THF with formaldehyde gave 5,10-(CH3)2-THF (27), whereas glyoxal gave 5,10-CH2CH2)-THF (10). Also, both folic acid and 5-(CHO)-THF were reductively alkylated with formaldehyde to give 10-methylfolic acid (6) and 5-(CHO)-10-(CH3)-THF (28), respectively. These compounds were tested as inhibitors of the enzymes involved in folate metabolism and for activity against lymphocytic leukemia P388 in mice.


Assuntos
Inibidores Enzimáticos/síntese química , Ácido Fólico/metabolismo , Tetra-Hidrofolatos/síntese química , Animais , Antineoplásicos/síntese química , Fenômenos Químicos , Química , Leucemia P388/tratamento farmacológico , Camundongos , Tetra-Hidrofolatos/farmacologia
11.
J Med Chem ; 29(10): 2069-74, 1986 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-3489838

RESUMO

The synthesis and characterization of 8-amino-6-fluoro-9-beta-D-ribofuranosyl-9H-purine (3a) are presented. This compound is a substrate for adenosine deaminase and adenosine kinase. In L1210 cells 3a is converted to 8-aminoinosine monophosphate (4b), apparently by the action of AMP deaminase on the monophosphate of 3a, as well as to the triphosphate derivative of 3a. Pentostatin was used to inhibit adenosine deaminase, and coformycin was used to inhibit AMP deaminase in experiments designed to delineate the metabolic fate of 3a. Pentostatin was without influence on the cytotoxicity of 3a, but coformycin potentiated the cytotoxicity. The potentiation was associated with an increased cellular concentration of phosphates of 3a and a decreased concentration of 4b.


Assuntos
Antineoplásicos/síntese química , Nucleosídeos de Purina/síntese química , AMP Desaminase/antagonistas & inibidores , Inibidores de Adenosina Desaminase , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Coformicina/análogos & derivados , Coformicina/farmacologia , Inosina Monofosfato/metabolismo , Cinética , Leucemia L1210/metabolismo , Pentostatina , Nucleosídeos de Purina/farmacologia
12.
J Med Chem ; 37(6): 739-44, 1994 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-8145223

RESUMO

Cyclopentadiene was converted in six steps to the key intermediate (+/-)-(1 alpha,2 beta,4 alpha)-4-amino-2-(benzyloxy)cyclopentanol (10), which in turn was converted to the carbocyclic nucleoside analogs 14 and 19 by standard procedures developed in these laboratories. Compounds 14 and 19 were then further converted to the target phosphonates 1b and 2b by modification of literature procedures. The phosphonate 1b was 40-fold more cytotoxic to HEp-2 cells than its parent, CDG, presumably after conversion to the diphosphoryl phosphonate.


Assuntos
Antineoplásicos/síntese química , Nucleotídeos Cíclicos/síntese química , Organofosfonatos/síntese química , Animais , Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Humanos , Leucemia L1210/tratamento farmacológico , Camundongos , Nucleotídeos Cíclicos/farmacologia , Organofosfonatos/farmacologia , Suínos , Células Tumorais Cultivadas/efeitos dos fármacos
13.
J Endocrinol ; 65(2): 245-51, 1975 May.
Artigo em Inglês | MEDLINE | ID: mdl-1151206

RESUMO

The hamster exhibits a biphasic pattern of insulin secretion; however, the dynamic response differs qualitatively from that of the rat in that there is a steady-state second release phase. A marked attenuation of insulin secretion as a result of hypophysectomy was observed after 3 weeks, but not after 2 weeks. This depression of insulin secretion was restored to near or above normal levels by bovine growth hormone, human growth hormone, and prolactin.


Assuntos
Cricetinae/fisiologia , Hormônio do Crescimento/fisiologia , Insulina/metabolismo , Pâncreas/metabolismo , Hipófise/fisiologia , Prolactina/fisiologia , Animais , Bovinos , Humanos , Hipofisectomia , Secreção de Insulina , Mesocricetus , Pâncreas/efeitos dos fármacos , Perfusão , Fatores de Tempo
14.
Biochem Pharmacol ; 37(7): 1233-44, 1988 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-3355597

RESUMO

3-Deazaadenine, 3-deazaadenosine, and the carbocyclic analog of 3-deazaadenosine produced similar effects on nucleotide pools of L1210 cells in culture: each caused an increase in IMP and a decrease in adenine nucleotides and had no effect on nucleotides of uracil and cytosine. Concentrations of 50-100 microM were required to produce these effects. Although 3-deazaadenosine and carbocyclic 3-deazaadenosine are known to be potent inhibitors of adenosylhomocysteine hydrolase, the effects on nucleotide pools apparently are not mediated via this inhibition because they are also produced by the base, 3-deazaadenine, and because the concentrations required are higher than those required to inhibit the hydrolase. Cells grown in the presence of 3-deazaadenine or 3-deazaadenosine contained phosphates of 3-deazaadenosine (the mono- and triphosphates were isolated); from cells grown in the presence of the carbocyclic analog of 3-deazaadenosine, the monophosphate was isolated, but evidence for the presence of the triphosphate was not obtained. A cell-free supernatant fraction from L1210 cells supplemented with ATP catalyzed the formation of monophosphates from 3-deazaadenosine or carbocyclic 3-deazaadenosine, and a cell-free supernatant fraction supplemented with 5-phosphoribosyl 1-pyrophosphate (PRPP) catalyzed the formation of 3-deaza-AMP from 3-deazaadenine. Adenosine kinase apparently was not solely responsible for the phosphorylation of the nucleosides because a cell line that lacked this enzyme converted 3-deazaadenosine to phosphates. No evidence was obtained that the effects on nucleotide pools resulted from a block of the IMP-AMP conversion, but the results could be rationalized as a consequence of increased AMP deaminase activity. This explanation is supported by two observations: (a) coformycin, an inhibitor of AMP deaminase, prevented the effects on nucleotide pools, and (b) 3-deazaadenine decreased the conversion of carbocyclic adenosine to carbocyclic ATP and increased its conversion to carbocyclic GTP. The latter conversion requires the action of AMP deaminase and the observed effects can be rationalized by a nucleoside analog-mediated increase in AMP deaminase activity. Because these effects on nucleotide pools are produced only by concentrations higher than those required to inhibit adenosylhomocysteine hydrolase, they may not contribute significantly to the biological effects of 3-deazaadenosine or carbocyclic 3-deazaadenosine.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
AMP Desaminase/fisiologia , Adenina/análogos & derivados , Antibacterianos/farmacologia , Nucleotídeo Desaminases/fisiologia , Nucleotídeos/análise , Tubercidina/análogos & derivados , Tubercidina/farmacologia , Adenina/metabolismo , Adenina/farmacologia , Adenosina Quinase/fisiologia , Alanina/análogos & derivados , Alanina/farmacologia , Aminoglicosídeos , Animais , Sobrevivência Celular/efeitos dos fármacos , Coformicina/farmacologia , Hipoxantina , Hipoxantinas/metabolismo , Camundongos , Fatores de Tempo , Tubercidina/metabolismo
15.
Biochem Pharmacol ; 33(2): 261-71, 1984 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-6142712

RESUMO

2-Amino-6-chloro-1-deazapurine is of interest as a purine analog with demonstrated in vivo activity against mouse leukemia L1210. That the active form of this agent is a nucleotide and that the nucleotide is formed by the action of hypoxanthine (guanine) phosphoribosyltransferase were shown by the facts that (a) L1210 cells deficient in hypoxanthine phosphoribosyltransferase were insensitive to the analog; (b) hypoxanthine, but not adenine, prevented the formation of the analog nucleotide by enzyme preparations containing activities of both hypoxanthine and adenine phosphoribosyltransferases; and (c) the cytotoxicity of the analog was prevented by hypoxanthine. The ribonucleoside of this analog was not toxic to cell cultures and hence is not phosphorylated or cleaved to the base. In intact HEp-2 cells and L1210 cells, the analog was metabolized to the nucleoside 5'-phosphate which accumulated to concentrations as high as 1000 nmoles/10(9) cells; no di- or triphosphates were detected. In HEp-2 cells, the analog reduced the pools of purine nucleotides with some accumulation of IMP. The toxicity of minimal inhibitory concentrations of the analog to HEp-2 cells could be prevented or reversed by 4(5)-amino-5(4)-imidazolecarboxamide (AIC); the toxicity of higher concentrations could be prevented or reversed by a combination of adenine and guanosine but not by AIC. The analog inhibited the incorporation of formate into purine nucleotides and into macromolecules at concentrations that had no effect on utilization of hypoxanthine; at higher concentrations the incorporation of hypoxanthine was inhibited. Low concentrations also inhibited the utilization of uridine and thymidine. The incorporation of hypoxanthine and AIC into guanine nucleotides, but not adenine nucleotides, was inhibited. These results indicate two sites of inhibition of the biosynthesis of purine nucleotides, the more sensitive one being on an early step of the pathway and the less sensitive one on the IMP-GMP conversion. That the blockade of de novo synthesis probably was at the site of feedback inhibition was indicated by the fact that the analog inhibited the accumulation of formylglycinamide ribonucleotide in azaserine-treated cells but did not inhibit the synthesis of 5'-phosphoribosyl 1-pyrophosphate. Comparative studies were performed with the related analog, 2-amino-6-chloropurine, which has been reported to produce a similar dual blockade of the purine pathway. This purine was less toxic than its 1-deaza analog; it produced a modest decrease in adenine nucleotides but increased pools of guanine nucleotides.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
2-Aminopurina/análogos & derivados , Adenina/análogos & derivados , Antineoplásicos/farmacologia , Leucemia L1210/metabolismo , 2-Aminopurina/metabolismo , 2-Aminopurina/farmacologia , AMP Desaminase/metabolismo , Adenina/farmacologia , Animais , Carcinoma de Células Escamosas , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Glicina/análogos & derivados , Glicina/biossíntese , Humanos , Hipoxantina , Hipoxantinas/farmacologia , IMP Desidrogenase/antagonistas & inibidores , Neoplasias Laríngeas , Substâncias Macromoleculares , Camundongos , Nucleotídeos/biossíntese , Ribonucleotídeos/biossíntese , Relação Estrutura-Atividade
16.
Biochem Pharmacol ; 34(8): 1293-304, 1985 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-2859858

RESUMO

The metabolism and metabolic effects of 2-azahypoxanthine and 2-azaadenosine were studied to elucidate the biochemical basis for their known cytotoxicities. 2-Azaadenosine is a known substrate for adenosine kinase. That 2-azahypoxanthine is a substrate for hypoxanthine (guanine) phosphoribosyltransferase is shown by the observations that, in cell-free fractions from HEp-2 cells supplemented with 5-phosphoribosyl-1-pyrophosphate, 2-azahypoxanthine inhibited the conversion of hypoxanthine to IMP but not the conversion of adenine to AMP, and hypoxanthine, but not adenine, inhibited the conversion of 2-azahypoxanthine to 2-azaIMP. [8-14C]2-Azahypoxanthine was synthesized from [8-14C]hypoxanthine via [2-14C]-4-amino-5-imidazolecarboxamide. In HEp-2 cells in culture, the principal metabolite of [8-14C]-2-azahypoxanthine was 2-azaATP; there was no detectable 14C in deoxynucleotides or in DNA or RNA fractions. 2-Azaadenosine was much more toxic than 2-azahypoxanthine, and, when used in the presence of an adenosine deaminase inhibitor, 2'-deoxycoformycin, was converted in HEp-2 cells to 2-azaATP in amounts that exceeded those of ATP in control cells. The pool of ATP was reduced by as much as 75% as 2-azaATP accumulated. In a short-term experiment (4 hr), 2-azaadenosine selectively reduced the pools of adenine nucleotides, whereas 2-azahypoxanthine reduced the pools of guanine nucleotides selectively. Both 2-azahypoxanthine and 2-azaadenosine inhibited the incorporation of formate into purine nucleotides and were without effect on the conversion of thymidine and uridine to nucleotides. 2-Azahypoxanthine inhibited the incorporation of thymidine into macro-molecules but not that of uridine or leucine; 2-azaadenosine inhibited the incorporation of all three of these precursors non-selectively. 2-AzaIMP inhibited IMP dehydrogenase competitively with IMP (Ki = 66 microM). The difference in effects of 2-azahypoxanthine and 2-azaadenosine perhaps may be due to the production, from 2-azahypoxanthine but not from 2-azaadenosine + 2'-deoxycoformycin, of 2-azaIMP, which inhibits synthesis of guanine nucleotides and thereby results in inhibition of DNA synthesis. Specific sites of action for 2-azaadenosine are yet undefined.


Assuntos
Adenosina/análogos & derivados , Antineoplásicos/metabolismo , Hipoxantinas/metabolismo , Adenosina/metabolismo , Adenosina/farmacologia , Animais , Antineoplásicos/farmacologia , Carcinoma de Células Escamosas , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Desoxirribonucleotídeos/biossíntese , Humanos , Hipoxantina Fosforribosiltransferase/metabolismo , Hipoxantinas/farmacologia , IMP Desidrogenase/antagonistas & inibidores , Neoplasias Laríngeas , Leucemia L1210 , Substâncias Macromoleculares , Camundongos , Polinucleotídeos/biossíntese , Ribonucleotídeos/biossíntese
17.
Biochem Pharmacol ; 55(10): 1673-81, 1998 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9634004

RESUMO

Activation of purine nucleoside analogs by Escherichia coli purine nucleoside phosphorylase (PNP) is being evaluated as a suicide gene therapy strategy for the treatment of cancer. Because the mechanisms of action of two toxic purine bases, 6-methylpurine (MeP) and 2-fluoroadenine (F-Ade), that are generated by this approach are poorly understood, mechanistic studies were initiated to learn how these compounds differ from agents that are being used currently. The concentration of F-Ade, MeP, or 5-fluorouracil required to inhibit CEM cell growth by 50% after a 4-hr incubation was 0.15, 9, or 120 microM, respectively. F-Ade and MeP were also toxic to quiescent MRC-5, CEM, and Balb 3T3 cells. Treatment of CEM, MRC-5, or Balb 3T3 cells with either F-Ade or MeP resulted in the inhibition of protein, RNA, and DNA syntheses. CEM cells converted F-Ade and MeP to F-ATP and MeP-ribonucleoside triphosphate (MeP-R-TP), respectively. The half-life for disappearance of HeP-ribonucleoside triphosphate from CEM cells was approximately 48 hr, whereas the half-lives of F-ATP and ATP were approximately 5 hr. Both MeP and F-Ade were incorporated into the RNA and DNA of CEM cells. These studies indicated that the mechanisms of action of F-Ade and MeP were quite different from those of other anticancer agents, and suggested that the generation of these agents in tumor cells by E. coli PNP could result in significant advantages over those generated by either herpes simplex virus thymidine kinase or E. coli cytosine deaminase. These advantages include a novel mechanism of action resulting in toxicity to nonproliferating and proliferating tumor cells and the high potency of these agents during short-term treatment.


Assuntos
Adenina/análogos & derivados , Purinas/metabolismo , Células 3T3 , Adenina/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Cicloeximida/farmacologia , DNA/efeitos dos fármacos , DNA/metabolismo , Fluoruracila/farmacologia , Humanos , Camundongos , Inibidores da Síntese de Ácido Nucleico/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , RNA/efeitos dos fármacos , RNA/metabolismo
18.
Biochem Pharmacol ; 40(7): 1515-22, 1990 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-2171531

RESUMO

The carbocyclic analog of 2'-deoxyguanosine [(+-)-2-amino-1,9-dihydro-9-[(1 alpha,3 beta,4 alpha)-3-hydroxy-4-(hydroxymethyl)cyclopentyl]-6H-purine-6-one] (2'-CDG) is highly active in cell culture against strains S148 and E377 of herpes simplex virus type 1 (HSV-1), both of which code for thymidine kinase, and much less active against strain BW10168 which is deficient in this enzyme activity. Antiviral activity is associated primarily with the D-enantiomer; the L-enantiomer has much lower but significant activity. The metabolism of racemic 2'-CDG and its D- and L-enantiomers was studied in uninfected HEp-2 cells and in HEp-2 cells infected with the S148 or BW10168 strains of HSV-1. Nucleotides were separated by HPLC, and their elution was monitored by spectrophotometry. The chromatograms of extracts of cells infected with the S148 strain and treated with (+/-)-2'-CDG or D-2'-CDG included a new peak which appeared in the triphosphate region. This peak, the area of which exceeded that of the GTP peak, was shown to be due to the triphosphate of 2'-CDG. The new peak was not observed by HPLC of extracts of uninfected cells treated with (+/-)-2'-CDG or either of its enantiomers, cells infected with the S148 strain and treated with L-2'-CDG, or cells infected with the BW10168 strain and treated with (+/-)-2'-CDG or either of its enantiomers. The results were similar when these studies were performed with uninfected Vero cells and with Vero cells infected with strain S148 of HSV-1. In experiments with D-[8-3H]-2'-CDG, small amounts of phosphates of 2'-CDG could also be detected in uninfected HEp-2 cells and in cells infected with the BW10168 strain of HSV-1. Thus, 2'-CDG apparently is a good substrate for the virus-coded kinase and a very poor substrate for cellular phosphorylating enzymes. The selective phosphorylation of 2'-CDG by the virus-specific kinase presumably is critical for its antiviral activity as it is for that of acyclovir and other acyclic derivatives of guanine.


Assuntos
Antivirais/metabolismo , Desoxiguanosina/análogos & derivados , Simplexvirus/enzimologia , Timidina Quinase/metabolismo , Animais , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Desoxiguanosina/metabolismo , Herpes Simples/metabolismo , Mutação , Nucleotídeos/isolamento & purificação , Fosforilação , Simplexvirus/genética , Timidina Quinase/genética
19.
Biochem Pharmacol ; 50(5): 687-95, 1995 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-7669072

RESUMO

4'-Thiothymidine (S-dThd) is a potent inhibitor of L1210 cell growth and is active against P388 leukemia in mice. Because of these activities and its novel structure, we have begun studies of its metabolism and metabolic actions in L1210 cells in order to understand its mechanism of cytotoxicity, S-dThd inhibited the incorporation of radiolabeled precursors into DNA, but did not inhibit the incorporation of either uridine or leucine into RNA or protein, respectively, which indicated that the mechanism of its toxicity was due to its inhibition of DNA synthesis. S-dThd did not decrease the concentration of any of the natural deoxynucleoside triphosphates, which indicated that its cytotoxicity was not due to the inhibition of ribonucleotide reductase. S-dThd was readily phosphorylated and used as a substrate for DNA synthesis. Because the rate of incorporation of S-dThd into DNA was 20% that of thymidine, it is likely that the mechanism of action of S-dThd is not due to inhibition of DNA polymerases by the 5'-triphosphate of S-dThd, but instead to its incorporation into the DNA and its subsequent disruption of some function of DNA.


Assuntos
Antineoplásicos/farmacologia , Tionucleosídeos/farmacologia , Timidina/análogos & derivados , Animais , Antineoplásicos/metabolismo , Divisão Celular/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , DNA de Neoplasias/biossíntese , DNA de Neoplasias/metabolismo , Leucemia L1210 , Camundongos , Especificidade por Substrato , Tionucleosídeos/metabolismo , Timidina/metabolismo , Timidina/farmacologia , Timidina Quinase/metabolismo , Células Tumorais Cultivadas
20.
Chest ; 90(2): 172-6, 1986 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3525021

RESUMO

Eleven adult men with sleep apnea underwent nocturnal polysomnography on two successive nights. The first study, done without NCPAP, served as the control. The second (treatment) was done with the application of 7.5 to 15 cm H2O nasal continuous positive airway pressure (NCPAP). A subjective sleepiness index (SSI) was noted upon awakening from each night of polygraphic recording. During the control night, the mean frequency of apnea episodes/sleep hr was 35.95 +/- 4.5 SE, and the mean duration was 28.68 +/- 2.7 sec. Mean frequency of disorder of breathing (DOB) episodes/sleep hr was 19.25 +/- 6.2 and mean duration of DOB episodes was 23.1 +/- 2.8 sec. During the treatment night, all obstructive apnea episodes were abolished. During the control night, the mean decrease in arterial oxygen saturation during obstructive apnea episodes was 11.2 +/- 1.9 percent and the mean lowest saturation was 67.6 +/- 4.0 percent. NCPAP eliminated arterial oxygen desaturation. While 44.5 +/- 5.7 percent of total sleep time was spent in either apnea or disordered breathing during the control night, NCPAP decreased this to 0.73 +/- 0.3 percent. In addition to the improvement in respiration during sleep, SSI decreased from a mean of 3.73 +/- 0.49 after the control night to 1.64 +/- 0.24 after treatment, reflecting an improvement in daytime hypersomnolence. We conclude that nasal CPAP is effective in eliminating obstructive apnea episodes, and results in a marked decrease in daytime hypersomnolence after one treatment night.


Assuntos
Distúrbios do Sono por Sonolência Excessiva/prevenção & controle , Respiração com Pressão Positiva , Síndromes da Apneia do Sono/terapia , Transtornos do Sono-Vigília/prevenção & controle , Adulto , Idoso , Distúrbios do Sono por Sonolência Excessiva/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Oxigênio/sangue , Testes de Função Respiratória , Síndromes da Apneia do Sono/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA