Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters










Publication year range
1.
Cancer Res ; 51(9): 2386-94, 1991 May 01.
Article in English | MEDLINE | ID: mdl-1707752

ABSTRACT

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)


Subject(s)
Arabinonucleosides/pharmacology , DNA/biosynthesis , Nucleic Acid Synthesis Inhibitors , Ribonucleotide Reductases/antagonists & inhibitors , Adenine Nucleotides , Adenosine Triphosphate/metabolism , Arabinonucleosides/metabolism , Cell Division/drug effects , Clofarabine , Cytidine Triphosphate/metabolism , Deoxycytidine/metabolism , Guanidine , Guanidines/metabolism , Humans , RNA/biosynthesis , Tumor Cells, Cultured
2.
Anal Biochem ; 183(2): 279-82, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2516416

ABSTRACT

We have developed a new assay for purine nucleoside phosphorylase which is based on the release of tritium when [2-3H]inosine is used as the substrate and the reaction is coupled with xanthine oxidase. After the reaction is terminated, residual [2-3H]inosine is adsorbed on charcoal and the supernatant solution is assayed for radioactivity by liquid scintillation spectrometry. The new method gave results indistinguishable from those obtained by spectrophotometric determination of uric acid produced by the phosphorylase-xanthine oxidase-coupled reaction or by radioassay of chromatographically isolated [8-14C]hypoxanthine when [8-14C]inosine was used as substrate. The new method is faster than those involving chromatographic isolation of products. In comparison with spectrophotometric methods, it not only requires less manual time, but it also has the advantage that it can be used to study inhibitors whose ultraviolet absorption might interfere with spectrophotometric determination of uric acid.


Subject(s)
Pentosyltransferases/analysis , Purine-Nucleoside Phosphorylase/analysis , Animals , Cattle , Dose-Response Relationship, Drug , Guanine/analogs & derivatives , Guanine/metabolism , Inosine/metabolism , Kinetics , Methods , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Tritium , Xanthine Oxidase/metabolism
3.
Biochem Pharmacol ; 37(7): 1233-44, 1988 Apr 01.
Article in English | MEDLINE | ID: mdl-3355597

ABSTRACT

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)


Subject(s)
AMP Deaminase/physiology , Adenine/analogs & derivatives , Anti-Bacterial Agents/pharmacology , Nucleotide Deaminases/physiology , Nucleotides/analysis , Tubercidin/analogs & derivatives , Tubercidin/pharmacology , Adenine/metabolism , Adenine/pharmacology , Adenosine Kinase/physiology , Alanine/analogs & derivatives , Alanine/pharmacology , Aminoglycosides , Animals , Cell Survival/drug effects , Coformycin/pharmacology , Hypoxanthine , Hypoxanthines/metabolism , Mice , Time Factors , Tubercidin/metabolism
4.
J Med Chem ; 31(1): 250-4, 1988 Jan.
Article in English | MEDLINE | ID: mdl-3336023

ABSTRACT

Several nitrosoureido nucleosides (3a, 3b, 5a, 7a, 7c, and 10a) designed as inhibitors of enzymes that metabolize pyrimidine nucleotides have been prepared and their chemical and biological properties studied. The methylnitrosoureas 3a and 3b were not significantly cytotoxic to H.Ep.-2 and L1210 cells in vitro but showed moderate activity in the P388 mouse leukemia screen (79% ILS for 3a and 56% ILS for 3b). The (chloroethyl)nitrosoureas 7a and 7c inhibited proliferation of L1210 cells, were cytotoxic to H.Ep.-2 cells, and demonstrated good activity against P388 in vivo (135% ILS with one 30-day survivor for 7a and 191% ILS with two 30-day survivors for 7c). Overnight exposure of L1210 cells to 7a and 7c resulted in cell enlargement accompanied by cell lysis. Macromolecular synthesis in enlarged cells, particularly RNA and protein synthesis, was markedly increased relative to that in untreated control cells. The half-lives of each of the nitrosoureas in pH 7 buffer was determined and compared with biological activity.


Subject(s)
Nitrosourea Compounds/chemical synthesis , Nucleosides/chemical synthesis , Nucleotides/biosynthesis , Animals , Carmustine/pharmacology , Cell Division/drug effects , DNA Replication/drug effects , Drug Stability , Indicators and Reagents , Leukemia L1210/metabolism , Lomustine/pharmacology , Mice , Neoplasm Proteins/biosynthesis , Nitrosourea Compounds/pharmacology , Nucleosides/pharmacology , Nucleotides/antagonists & inhibitors , Structure-Activity Relationship , Transcription, Genetic/drug effects
5.
Invest New Drugs ; 5(4): 345-51, 1987 Dec.
Article in English | MEDLINE | ID: mdl-3436739

ABSTRACT

Deoxyspergualin, the 15-deoxy derivative of the antibiotic spergualin, is a novel guanidino analog structurally related to spermine. Deoxyspergualin has significant activity in selected experimental tumor models, and clinical trials have been initiated. Described here are in vivo evaluations of the therapeutic activity of deoxyspergualin against murine leukemia lines specifically resistant to eight clinically useful antitumor drugs. These were P388 lines resistant to doxorubicin, vincristine, L-phenylalanine mustard, cisplatin, ara-C, and methotrexate and L1210 lines resistant to 5-FU, L-phenylalanine mustard, and cyclophosphamide. Sensitivity to deoxyspergualin was evaluated in parallel comparisons of each resistant leukemia to the sensitive line from which it had been derived. All experiments were repeated at least once for confirmation of results. Responses were quantitated in terms of the change in tumor cell numbers from the beginning of treatment to the end of treatment as estimated from the median survival times of dying mice. The results indicated that P388 leukemia resistant to cisplatin (P388/DDPt) was cross-resistant to deoxyspergualin. No cross-resistance was observed in leukemias resistant to doxorubicin, vincristine, ara-C, methotrexate, or cyclophosphamide. L1210 resistant to 5-FU (L1210/5-FU) was collaterally sensitive to deoxyspergualin. Although cross-resistance was also observed in P388/L-PAM, L1210/L-PAM retained sensitivity to deoxyspergualin. Total glutathione concentrations in P388/L-PAM and L1210/L-PAM provided no apparent explanation for this unexpected result. It may be tentatively concluded that resistance to cisplatin, L-PAM, or other DNA alkylators or cross-linkers may increase the potential for cross-resistance to deoxyspergualin. This conclusion requires verification with additional alkylating agents, with drug-resistant human tumor cell lines, and with prospective clinical studies.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Leukemia, Experimental/drug therapy , Animals , Drug Resistance , Female , Glutathione/metabolism , Guanidines/pharmacology , Leukemia L1210/drug therapy , Leukemia P388/drug therapy , Melphalan/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA
6.
J Med Chem ; 30(5): 927-30, 1987 May.
Article in English | MEDLINE | ID: mdl-3572980

ABSTRACT

5'-(Bromoacetamido)-2',5'-dideoxyuridine (3) and derivatives (8, 10, 12, and 14) substituted at the 5-position with bromo, iodo, fluoro, and ethyl groups have been synthesized as potential inhibitors of enzymes that metabolize pyrimidine nucleosides. Also prepared were 2',5'-dideoxyuridine derivatives (4-6) substituted at the 5'-position with 2-bromopropionamido, iodoacetamido, and 4-(fluorosulfonyl)benzamido groups. Compounds 3, 5, 8, 12, and 14 were examined for effect on macromolecular synthesis in L1210 leukemia cells in culture and compared with 5'-(bromoacetamido)-5'-deoxythymidine (1, BAT), a compound with demonstrated cytotoxicity and activity in vivo against P388 murine leukemia. Compounds 3, 8, 12, and 14 inhibited DNA synthesis without significant inhibition of RNA synthesis, and protein synthesis was affected less than DNA synthesis. Compounds 3, 5, 6, 8, 10, 12, and 14 were cytotoxic to H.Ep.-2 and L1210 cells in culture, and 3, 5, 8, and 12 showed activity in the P388 mouse leukemia screen.


Subject(s)
Deoxyuridine/analogs & derivatives , Nucleotides/biosynthesis , Animals , Chemical Phenomena , Chemistry , DNA/biosynthesis , Deoxyuridine/pharmacology , Deoxyuridine/therapeutic use , Leukemia L1210/drug therapy , Leukemia L1210/metabolism , Leukemia P388/drug therapy , Leukemia P388/metabolism , Mice , Structure-Activity Relationship , Thymidylate Synthase/antagonists & inhibitors
7.
Cancer Res ; 47(3): 700-6, 1987 Feb 01.
Article in English | MEDLINE | ID: mdl-3802076

ABSTRACT

Studies have examined transport and phosphorylation of 9-beta-D-arabinofuranosyl-2-fluoroadenine (F-Ara-A), a deaminase resistant adenosine analogue, as mechanisms that could mediate the observed therapeutic efficacy of this agent against murine tumor models. Earlier finds by Avramis and Plunkett (Cancer Res., 42: 2587-2591, 1982) showed markedly less accumulation in vivo of administered F-Ara-A as cytotoxic triphosphate in gastrointestinal mucosa and bone marrow compared to P388 cells. We have pursued the basis for this difference in vitro using L1210 ascites and proliferative epithelial cells (85-95% crypt cells) isolated from mouse small intestine as representative sample populations of drug-sensitive tumor and drug-limiting normal regenerative host tissue. Using a rapid sampling technique, linear initial rates of substrate uptake were established at 25 degrees C for radiolabeled F-Ara-A and adenosine at a concentration range of 1-1000 microM. The relationship between velocity of initial transport and substrate concentration is indicative of Michaelis-Menten saturation kinetics for both substrates. Competition studies between F-Ara-A and adenosine suggest a common route of entry for both substrates in crypt epithelial cells. Results from double-reciprocal analysis of the velocity versus concentration data are consistent with a simple carrier-mediated facilitated diffusion process with Km, V25max, and Ki values of 317 +/- 44 (SE) microM, 49 +/- 7 nmol/s/g dry weight, and 301 +/- 34 microM for F-Ara-A, and 264 +/- 14 microM, 44 +/- 5 nmol/s/g dry weight, and 225 +/- 44 microM for adenosine, respectively. The presence of a single low-affinity carrier in the proliferative epithelial cells contrasts sharply with the high affinity (Km, 68 +/- 14 microM; V25max, 48 +/- 4 nmol/s/g dry weight) and low-affinity (Km, 326 +/- 48 microM; V25max, 124 +/- 44 nmol/s/g dry weight) routes of entry documented for L1210 cells. This differential in transport kinetics conveys a 7- to 8-fold greater capacity to L1210 ascites compared with crypt epithelial cells for uptake of the antitumor agent F-Ara-A. At pharmacologically achievable concentrations of F-Ara-A and in view of this differential, influx of F-Ara-A would be more rate limiting to phosphorylation of F-Ara-A in epithelial cells than in L1210 cells. Metabolism studies with L1210 ascites and proliferative intestinal epithelial cells show that intracellular phosphorylation of F-Ara-A is also elevated in L1210 cells.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Intestine, Small/metabolism , Leukemia L1210/metabolism , Vidarabine/analogs & derivatives , Animals , Biological Transport , Epithelium/metabolism , Kinetics , Leukemia L1210/drug therapy , Mice , Mice, Inbred Strains , Organ Specificity , Phosphorylation , Tritium , Vidarabine/metabolism , Vidarabine/therapeutic use
8.
J Med Chem ; 30(2): 431-4, 1987 Feb.
Article in English | MEDLINE | ID: mdl-3027342

ABSTRACT

5-Amino-4-(diazoacetyl)-1-beta-D-ribofuranosylimidazole (15), 5-amino-4-(chloroacetyl)-1-beta-D-ribofuranosylimidazole (16), and a number of related imidazole ribonucleosides have been synthesized. Compounds 15 and 16 are cytotoxic to both H.Ep.-2 and L1210 leukemia cells in culture. The (diazoacetyl)imidazole 15 is also active against the P388 leukemia in mice.


Subject(s)
Aminoimidazole Carboxamide/chemical synthesis , Antineoplastic Agents/chemical synthesis , Imidazoles/chemical synthesis , Ribonucleosides/chemical synthesis , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/toxicity , Animals , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Survival/drug effects , Humans , Indicators and Reagents , Leukemia L1210/pathology , Liver Neoplasms/pathology , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Ribonucleosides/toxicity , Structure-Activity Relationship
9.
J Med Chem ; 29(6): 1052-6, 1986 Jun.
Article in English | MEDLINE | ID: mdl-2423689

ABSTRACT

Fourteen derivatives of thymidine substituted at the 5'-position with haloacetamido (2-4), 2- and 3-bromopropionamido (5 and 6), bromoacetoxy (7), O-mesylglycolamido (8), bromo- and chloro-N-methylacetamido (10 and 11), bromomethanesulfonamido (12), ethyloxamido (13), 4- and 3-(fluorosulfonyl)benzamido (14 and 15), and (phenoxycarbonyl)amino (16) groups have been synthesized and evaluated as potential inhibitors of enzymes that metabolize purine and pyrimidine nucleosides. Rates of reaction of these nucleosides with mercaptoethanol at pH 7 were compared and related to biological activity. Compounds 2, 3, and 7 were cytotoxic to H.Ep.-2 and L1210 cells in culture and 5'-(bromo- and iodoacetamido)-5'-deoxythymidine (2 and 3) showed good activity against P388 leukemia in mice.


Subject(s)
Nucleotides/biosynthesis , Thymidine/analogs & derivatives , Animals , Cells, Cultured , DNA/biosynthesis , Half-Life , Leukemia, Experimental/drug therapy , Mice , Nucleic Acid Synthesis Inhibitors , RNA/biosynthesis , Structure-Activity Relationship , Thymidine/metabolism , Thymidine/pharmacology
10.
Biochim Biophys Acta ; 881(2): 175-84, 1986 Apr 11.
Article in English | MEDLINE | ID: mdl-3955073

ABSTRACT

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.


Subject(s)
Thymidine/analogs & derivatives , Thymidylate Synthase/antagonists & inhibitors , Animals , Chromatography, Gel , Kinetics , Leukemia L1210/enzymology , Mice , Thymidine/pharmacology
11.
Mol Pharmacol ; 27(6): 666-75, 1985 Jun.
Article in English | MEDLINE | ID: mdl-2987661

ABSTRACT

In cell cultures treated with the carbocyclic analog of adenosine (C-Ado, (+/-)-aristeromycin), the utilization of hypoxanthine and guanine has been observed to be blocked. In an attempt to define the mechanism of this inhibition, we have reexamined the metabolism of C-Ado and its effects on the metabolism of guanine and hypoxanthine. In cultures of L1210 cells, C-Ado at a concentration of 25 microM inhibited the utilization of hypoxanthine and guanine for nucleotide synthesis by more than 90% but produced little or no inhibition of the utilization of these bases in cultures of L1210/MeMPR cells which lack adenosine kinase and cannot phosphorylate C-Ado. In cultures of mammalian cells (L1210, HEp-2, and colon-26 cells), C-Ado was converted to the triphosphate (as previously observed) and also to the triphosphate of the carbocyclic analog of guanosine. The presence of coformycin in the medium at a concentration sufficient to inhibit AMP deaminase almost completely prevented the formation of carbocyclic GTP; thus, the deamination of C-Ado monophosphate is essential for the formation of phosphates of carbocyclic guanosine. Since hypoxanthine (guanine) phosphoribosyltransferase is known to be subject to end product inhibition, it was considered likely that phosphates of carbocyclic guanosine or carbocyclic inosine, present in C-Ado-treated cells, were responsible for inhibition of utilization of hypoxanthine and guanine. The 5'-phosphates of the carbocyclic analogs of inosine and guanosine were synthesized and found to be effective inhibitors of the phosphoribosyltransferase. Carbocyclic GMP was a better inhibitor than carbocyclic IMP and was also superior to GMP and IMP; the concentration of C-GMP that produced a 50% inhibition of GMP formation was approximately 1 microM. It is probable that the presence of phosphates of carbocyclic guanosine accounts for the inhibition of utilization of hypoxanthine and guanine in C-Ado-treated cells.


Subject(s)
Adenosine/analogs & derivatives , Cyclic GMP/analogs & derivatives , Cyclic IMP/pharmacology , Guanine/metabolism , Hypoxanthine Phosphoribosyltransferase/antagonists & inhibitors , Hypoxanthines/metabolism , Inosine Nucleotides/pharmacology , Pentosyltransferases/antagonists & inhibitors , Adenosine/metabolism , Adenosine/pharmacology , Animals , Carcinoma, Squamous Cell , Cell Line , Coformycin/pharmacology , Colonic Neoplasms/metabolism , Cyclic GMP/pharmacology , Cyclic IMP/analogs & derivatives , Humans , Hypoxanthine , Kinetics , Leukemia L1210/metabolism , Mice , Ribonucleotides/metabolism
12.
Biochim Biophys Acta ; 805(1): 104-16, 1984 Sep 14.
Article in English | MEDLINE | ID: mdl-6477969

ABSTRACT

Perchloric acid extracts of radiation-induced fibrosarcoma (RIF-1) tumors grown in mice have been analyzed by multinuclear NMR spectroscopy and by various chromatographic methods. This analysis has permitted the unambiguous assignment of the 31P resonances observed in vivo to specific phosphorus-containing metabolites. The region of the in vivo spectra generally assigned to sugar phosphates has been found in RIF-1 tumors to contain primarily phosphorylethanolamine and phosphorylcholine rather than glycolytic intermediates. Phosphocreatine was observed in extracts of these tumor cells grown in culture as well as in the in vivo spectra, indicating that at least some of the phosphocreatine observed in vivo arises from the tumor itself and not from normal tissues. In the 31P-NMR spectra of the perchloric acid extract, resonances originating from purine and pyrimidine nucleoside di- and triphosphate were resolved. HPLC analyses of the nucleotide pool indicate that adenine derivatives were the most abundant components, but other nucleotides were present in significant amounts. The 1H and 13C resonance assignments of the majority of metabolites present in RIF-1 extracts have also been made. Of particular importance is the ability to observe lactate, the levels of which may provide a noninvasive measure of glycolysis in these cells in both the in vitro states. In addition, the aminosulfonic acid, taurine, was found in high levels in the tumor extracts.


Subject(s)
Fibrosarcoma/analysis , Neoplasms, Radiation-Induced/analysis , Animals , Autoanalysis , Cell Line , Female , Magnetic Resonance Spectroscopy/methods , Mice , Mice, Inbred C3H , NAD/analysis , Phosphates/analysis , Ribonucleotides/analysis
13.
J Med Chem ; 27(5): 680-4, 1984 May.
Article in English | MEDLINE | ID: mdl-6716405

ABSTRACT

Several halomethyl ketone derivatives of pyrimidine nucleosides have been prepared for evaluation as cytotoxic agents. The first series are 1-(8-halo-2,5,6,8- tetradeoxy -beta-D-erythro-oct-7 - ulofuranosyl )thymines (7-9), whereas the second type are halo derivatives of acetophenone (12-14 and 16). These compounds are cytotoxic, and one (13) showed activity against the P388 leukemia in vivo.


Subject(s)
Pyrimidine Nucleosides/chemical synthesis , Animals , Drug Evaluation, Preclinical , Indicators and Reagents , Leukemia L1210/drug therapy , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Pyrimidine Nucleosides/toxicity , Spectrophotometry, Ultraviolet , Structure-Activity Relationship
14.
Biochem Pharmacol ; 33(2): 261-71, 1984 Jan 15.
Article in English | MEDLINE | ID: mdl-6142712

ABSTRACT

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)


Subject(s)
2-Aminopurine/analogs & derivatives , Adenine/analogs & derivatives , Antineoplastic Agents/pharmacology , Leukemia L1210/metabolism , 2-Aminopurine/metabolism , 2-Aminopurine/pharmacology , AMP Deaminase/metabolism , Adenine/pharmacology , Animals , Carcinoma, Squamous Cell , Cell Line , Cell Survival/drug effects , Glycine/analogs & derivatives , Glycine/biosynthesis , Humans , Hypoxanthine , Hypoxanthines/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Laryngeal Neoplasms , Macromolecular Substances , Mice , Nucleotides/biosynthesis , Ribonucleotides/biosynthesis , Structure-Activity Relationship
15.
Biochemistry ; 22(3): 600-11, 1983 Feb 01.
Article in English | MEDLINE | ID: mdl-6301523

ABSTRACT

Purified adenosine kinase from L1210 cells displayed substrate inhibition by high concentrations of adenosine (Ado), ATP, and MgCl2. When incubated with ATP and MgCl2, the enzyme was phosphorylated, and the phosphorylated kinase transferred phosphate to adenosine in the absence of ATP and MgCl2. Substrate binding, isotope exchange, and kinetic studies suggested that the enzyme catalyzes the reaction by means of a two-site ping-pong mechanism with the phosphorylated enzyme as an obligatory intermediate. Among many possible pathways within this mechanism probably a random-bi ordered-bi route is the preferred sequence in which the two substrates, adenosine and MgATP, bind in a random order to form the ternary complex MgATP . E . Ado followed by the sequential dissociation of MgADP and AMP. Dissociation constants of various enzyme-substrate and enzyme-product complexes and the first-order rate constant of the rate-limiting step were estimated.


Subject(s)
Adenosine Kinase/metabolism , Leukemia L1210/enzymology , Models, Chemical , Phosphotransferases/metabolism , Adenosine , Adenosine Diphosphate/pharmacology , Adenosine Kinase/antagonists & inhibitors , Adenosine Monophosphate/pharmacology , Adenosine Triphosphate , Animals , In Vitro Techniques , Kinetics , Magnesium/pharmacology , Mice , Phosphorylation
17.
Biochem Pharmacol ; 31(11): 1975-8, 1982 Jun 01.
Article in English | MEDLINE | ID: mdl-7115418

ABSTRACT

Analysis of blood from a dog given a 400 mg/m2 dose of 9-beta-D-arabinofuranosyl-2-fluoroadenine (2-F-araA) led to the identification of parent drug and a major metabolite, 9-beta-D-arabinofuranosyl-2-fluorohypoxanthine. 2-Fluoroadenine, a toxic derivative of 2-F-araA, was not detected in blood within the limits of detection, suggesting that parent drug was absorbed and distributed without systemic exposure to this toxic derivative. Parent drug, 2-fluoroadenine, and 9-beta-D-arabinofuranosyl-2-fluorohypoxanthine were identified in urine of dog, monkey, and mouse.


Subject(s)
Antineoplastic Agents/metabolism , Antiviral Agents/metabolism , Vidarabine/analogs & derivatives , Animals , Chemical Phenomena , Chemistry , Dogs , Female , Leukemia L1210/drug therapy , Macaca mulatta , Male , Mice , Vidarabine/metabolism , Vidarabine/pharmacology
18.
Cancer Res ; 42(6): 2260-4, 1982 Jun.
Article in English | MEDLINE | ID: mdl-7042080

ABSTRACT

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.


Subject(s)
DNA Replication/drug effects , Hydrolases/antagonists & inhibitors , Leukemia L1210/enzymology , Nucleic Acid Synthesis Inhibitors , Ribonucleotide Reductases/antagonists & inhibitors , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Adenosylhomocysteinase , Animals , DNA Polymerase I/antagonists & inhibitors , DNA Polymerase II/antagonists & inhibitors , Mice
20.
Mol Pharmacol ; 21(2): 474-7, 1982 Mar.
Article in English | MEDLINE | ID: mdl-7048062

ABSTRACT

9-beta-D-Arabinofuranosyl-2-fluoroadenine (2-F-araA) inhibited the growth in vitro of HeLa cells by 50% at a concentration of 0.25 microM and depressed the replication of herpes simplex virus Types 1 and 2 by 99% at 25 microM. The analogue served as a substrate for cytoplasmic but not mitochondrial deoxycytidine (dCyd) kinase partially purified from human peripheral chronic lymphocytic leukemic blast cells. The Km values of dCyd and 2-F-araA for the cytoplasmic enzyme were 5 microM and 213 microM, respectively. However, at concentrations of 0.4 mM, the analogue was phosphorylated 2.9 times faster than dCyd. The 5'-triphosphate of 2-F-araA was examined for its biochemical effects on partially purified ribonucleotide reductase and highly purified DNA alpha- and beta-polymerases from HeLa cells. 2-F-araATP was a potent inhibitor of ribonucleotide reductase; the concentration required for 50% inhibition of ADP reduction (0.3 mM ADP; 5 mM GTP or dGTP) was 1 microM and for CDP reduction (0.15 mM CPD; 5 mM ATP) was 8.5 microM. Furthermore, 2-F-araATP was a competitive inhibition (Ki = 1.2 microM) with respect to dATP (Km = 3.8 microM) of DNA alpha-polymerase, whereas DNA beta-polymerase was relatively insensitive to the drug. The results suggest that the cytotoxic actions of 2-F-araA may be due, in part, to a "self-potentiating" inhibition of DNA synthesis. This is, by inhibiting the formation of competing dATP, 2-F-araATP may potentiate its inhibition of DNA synthesis.


Subject(s)
Arabinonucleotides/pharmacology , Nucleic Acid Synthesis Inhibitors , Ribonucleotide Reductases/antagonists & inhibitors , Vidarabine/analogs & derivatives , Antiviral Agents , Binding, Competitive , Cell Survival/drug effects , Deoxycytidine Kinase/metabolism , HeLa Cells/metabolism , Humans , Kinetics , Phosphorylation , Vidarabine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...