Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Transpl Immunol ; 23(4): 180-4, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20553870

ABSTRACT

We investigated the immunosuppressive effects of the dihydroortate dehydrogenase (DHODH) inhibitor compounds ABR-222417 and ABR-224050 from Active Biotech (Sweden). We verified the inhibitory effects of these compounds on the proliferation of CD4(+) and CD8(+) T-cells in vivo by using superantigen staphylococcus enterotoxin A (SEA)-mediated proliferation test. To evaluate their efficacy, the compounds were screened in a low-responder heart allograft transplantation model in rats [heart from Piebald Virol Glaxo (PVG) transplanted to Dark Agouti (DA)]. The immunosuppressive effects of the compounds were then investigated in a high-responder model (DA to PVG). Treatment with ABR-222417 (30 mg/kg) was more efficient than that with ABR-224050 (10 mg/kg), and the former provided a longer graft median survival time (MST, 29.5 days) than the latter (MST, 18.5 days). Furthermore, there was a marked increase in graft survival time (53 days) when low doses of ABR-222417 and cyclosporine A (CsA) were used in combination. No sign of tolerability problems was detected using this combination or when ABR-222417 was used singly at a higher dose. Furthermore, T-cell proliferation studies in vitro support that the anti proliferative effect of ABR-222417 is caused by inhibition of de novo pyrimidine synthesis, which is the consequence of DHODH inhibition. These results show that ABR-222417 had marked immunosuppressive effects on the heart allograft transplantation and that it exerts an even more powerful inhibitory effect on graft rejection when used in combination with CsA, with good tolerability.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Graft Survival/drug effects , Heart Transplantation , ortho-Aminobenzoates/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Cyclosporine/administration & dosage , Dihydroorotate Oxidase/antagonists & inhibitors , Drug Synergism , Drug Therapy, Combination , Graft Survival/immunology , Humans , Immunosuppression Therapy , Jurkat Cells , Male , Rats , Rats, Inbred Strains , ortho-Aminobenzoates/pharmacology
2.
Bioorg Med Chem Lett ; 15(21): 4854-7, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16143532

ABSTRACT

Novel DHODH inhibitors were developed based on a previously described series by introduction of heteroatoms into the cyclopentene ring and hydroxyl groups attached to it. Also, the hydrophobic biphenyl side chain was replaced with benzyloxy phenyl groups. Activities on human, rat, and mouse enzymes indicate a species specificity of these inhibitors.


Subject(s)
Cell Proliferation/drug effects , Cyclopentanes/chemical synthesis , Dihydroorotate Oxidase/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Immunosuppressive Agents/chemical synthesis , Amides , Animals , Cyclopentanes/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Immunosuppressive Agents/pharmacology , Inhibitory Concentration 50 , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Mice , Rats , Species Specificity , Structure-Activity Relationship
3.
J Med Chem ; 44(18): 2861-4, 2001 Aug 30.
Article in English | MEDLINE | ID: mdl-11520193

ABSTRACT

Dihydroorotate dehydrogenases (DHODs) catalyze the conversion of dihydroorotate to orotate in de novo pyrimidine biosynthesis. We have found that 3,4-dihydroxybenzoate and 3,5-dihydroxybenzoate are competitive inhibitors vs dihydroorotate with the prototypical family 1A DHOD from Lactococcus lactis. The dissociation constants of these compounds, determined by spectral titrations, were similar to the dissociation constant of orotate, the enzymatic reaction product, suggesting that hydroxybenzoates could be developed into useful drugs for treating infections by certain protozoan parasites.


Subject(s)
Benzoates/chemical synthesis , Dihydroorotate Oxidase/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Pyrimidines/chemical synthesis , Trypanocidal Agents/chemical synthesis , Benzoates/chemistry , Enzyme Inhibitors/chemistry , Ligands , Pyrimidines/chemistry , Structure-Activity Relationship , Trypanocidal Agents/chemistry
4.
Biochem Pharmacol ; 50(8): 1157-64, 1995 Oct 12.
Article in English | MEDLINE | ID: mdl-7488229

ABSTRACT

Penclomedine (PEN) is a synthetic pyridine derivative that has been selected for clinical development based on its activity against human and mouse breast tumors implanted in mice. Its mechanism of action was unclear, and we were interested in determining its mechanism of cytotoxicity in vitro and in vivo. We found chromosome breaks, gaps, and exchanges in P388 ascites cells from BD2F1 mice treated with 200 mg/kg PEN. Maximal observed damage occurred 24 hr after drug administration. Alkaline elution indicated only limited DNA strand breaks and interstrand cross-linking. In vitro, PEN (75 micrograms/mL) inhibited RNA and DNA syntheses almost completely. In addition, incubation of [14C]PEN with rat liver S-9 fraction in the presence of calf thymus DNA resulted in the stable transfer of radioactivity to DNA. Addition of butylated hydroxytoluene, a free radical scavenger, to the incubation mixture inhibited the binding of drug to DNA, implicating free radicals as the ultimate reactive species. These data suggest that PEN can be metabolized to free radical, DNA-reactive products, and that its cytotoxicity is due to chromosomal damage produced by monofunctional alkylation. As an alternate mechanism, the ability of PEN to inhibit cellular dihydroorotate dehydrogenase was explored. Although PEN is an inhibitor of this enzyme in cells in vivo, in vitro, and in isolated cell sonicates, HPLC analyses of ribonucleotide triphosphate pools in P388 cells showed that all triphosphates had increased, especially UTP. Addition of uridine to the cell culture failed to prevent PEN-mediated cytotoxicity, suggesting that inhibition of de novo pyrimidine biosynthesis was not likely to be an important mechanism of action of this drug. These data suggest that PEN is activated in cells to a free radical that binds DNA.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage , Picolines/pharmacology , Animals , Cell Division/drug effects , Cell Line/drug effects , Chromatography, High Pressure Liquid , DNA/metabolism , Dihydroorotate Oxidase/antagonists & inhibitors , Humans , Leukemia P388 , Malondialdehyde/analysis , Picolines/metabolism , Uridine/metabolism
6.
Antiviral Res ; 20(1): 71-81, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8384434

ABSTRACT

CL 306,293, a substituted quinoline carboxylic acid, is a potent inhibitor of dihydroorotic acid dehydrogenase, an enzyme essential for the biosynthesis of pyrimidines. In mammalian cell culture, the agent exhibits antiproliferative properties that can be reversed by the addition of uridine. CL 306,293 inhibits the development of the clinical disease in a murine model of immunodeficiency induced by a mixture of LP-BM5 retroviruses. In infected mice, the agent prevents the development of hypergammaglobulinemia, lymphadenopathy, splenomegaly and induction of an IL-2 deficiency. The CD4/CD8 ratio and the number of B cells in the lymph nodes are decreased if the infected animals are treated with CL 306,293. CL 306,293 was more efficacious and potent than 3'-azido-3'-deoxythymidine. The beneficial effects of CL 306,293 observed in this model are most probably related to its antiproliferative properties.


Subject(s)
Aminoquinolines/therapeutic use , Biphenyl Compounds/therapeutic use , Dihydroorotate Oxidase/antagonists & inhibitors , Murine Acquired Immunodeficiency Syndrome/drug therapy , AIDS-Related Complex , Aminoquinolines/toxicity , Animals , Antibodies, Viral/blood , B-Lymphocytes/physiology , Biphenyl Compounds/toxicity , CD4-CD8 Ratio , Cells, Cultured , Fibroblasts/drug effects , Hypergammaglobulinemia , Immunoglobulin M/analysis , Interleukin-2/deficiency , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/enzymology , Recurrence , Splenomegaly , Zidovudine/therapeutic use
7.
Biochem Pharmacol ; 43(5): 1025-30, 1992 Mar 03.
Article in English | MEDLINE | ID: mdl-1313236

ABSTRACT

To verify the assumption of a specific and potent drug action on de novo pyrimidine biosynthesis, isolated dihydroorotate dehydrogenase (DHO-DH) (EC 1.3.3.1) was exposed to Brequinar Sodium (6-fluoro-2-(2'-fluoro-1,1'-biphenyl-4-yl)-3-methyl-4-quinoline carboxylic acid sodium salt, NSC 368 390) (Brequinar). The membrane-bound DHO-DH was purified to apparent homogeneity (25,000-fold) from rat liver mitochondria in six steps via detergent extraction and subsequent chromatography using the dye ligand Matrex Gel Orange A. Using molecular mechanistic studies (MM2) this ligand was found to mimic closely the stereochemical conformation of Brequinar. SDS-PAGE revealed two protein bands for the purified enzyme with apparent molecular masses of 58 (major) and 68 kDa (minor). In vitro, two modes of action of the DHO-DH are possible: (i) acting as a dehydrogenase in the presence of ubiquinone as proximal electron acceptor and (ii) direct reaction with oxygen as oxidase. A novel assay for the measurement of the oxidase activity was adapted using leuco-dichlorofluorescein-diacetate. Inhibition experiments revealed a striking difference in the susceptibility of DHO-dehydrogenase/oxidase to Brequinar: apparent Ki = 6.09 +/- 0.05 (SD) nM (DHO; ubiquinone n = 10), but Ki = 3.10 +/- 0.09 (SD) mM (DHO; O2). Analyses of initial velocity experiments showed non-competitive inhibition of Brequinar with respect to the substrate dihydroorotic acid in both assays (dehydrogenase and oxidase). The inhibitory effect of the latter was compared to that of the competitive inhibitor 5-aza-dihydroorotate (apparent Ki = 15 +/- 0.25 (SD) microM). The present kinetic data on the action of the purified rodent DHO-DH with Brequinar and computer-aided analyses provide a better insight into the drug-enzyme interaction.


Subject(s)
Biphenyl Compounds/pharmacology , Dihydroorotate Oxidase/antagonists & inhibitors , Animals , Dihydroorotate Oxidase/isolation & purification , Kinetics , Mitochondria, Liver/enzymology , Models, Chemical , Models, Molecular , Rats , Structure-Activity Relationship
8.
Biochem Int ; 22(6): 939-49, 1990 Dec.
Article in English | MEDLINE | ID: mdl-1965281

ABSTRACT

Exposure of mouse L1210 leukemia cells to 25 microM brequinar for 4 h results in large accumulations of N-carbamyl-L-aspartate and L-dihydroorotate to cellular concentrations of 8.5 mM and 0.8 mM, respectively, while UTP and CTP decrease to 4% of their initial levels; incorporation of [14C]bicarbonate into nucleic acids (DNA and RNA) was decreased to 47%. These data provide direct evidence for inhibition of DHO dehydrogenase by brequinar in growing cells. Exposure of leukemia cells to 200 microM ciprofloxacin for 4 h did not affect de novo pyrimidine nucleotide biosynthesis or the incorporation of [14C]bicarbonate into nucleic acids but resulted in a general decrease in nucleoside triphosphates, with concomitant accumulation of nucleoside mono- and diphosphates (the adenylate energy charge decreased from 0.89 to 0.69), consistent with inhibition of the electron transport chain or uncoupling of oxidative phosphorylation.


Subject(s)
Biphenyl Compounds/pharmacology , Ciprofloxacin/pharmacology , Dihydroorotate Oxidase/antagonists & inhibitors , Nucleotides/biosynthesis , Animals , Cell Division/drug effects , Chromatography, High Pressure Liquid , DNA, Neoplasm/biosynthesis , Enzyme Induction , Leukemia L1210 , Mice , RNA, Neoplasm/biosynthesis , Tumor Cells, Cultured
9.
Cancer Res ; 50(15): 4644-9, 1990 Aug 01.
Article in English | MEDLINE | ID: mdl-2164443

ABSTRACT

Little is known about the in vivo effects of inhibition of the mitochondrial pyrimidine de novo synthesis enzyme dihydroorotic acid dehydrogenase (DHO-DH). In mice a new inhibitor of DHO-DH, Brequinar sodium (DUP-785, NSC 368390) depleted the plasma uridine concentration to 40% within 2 h, followed by a small rebound after 7-9 days. The drug was subsequently evaluated in a Phase I clinical trial, during which it was possible to follow its biochemical effects in 24 patients (27 courses). In addition to the measurement of plasma uridine concentrations, we also measured in lymphocytes of 9 patients (10 courses) the duration of DHO-DH inhibition. Brequinar sodium was administered every 3 weeks as an i.v. infusion at dose levels of 15-2250 mg/m2. The biochemical effects were studied following the first administration of the drug. In sonicated extracts of lymphocytes from 7 healthy volunteers the activity of DHO-DH varied from 2.0 to 3.9 nmol/h per 10(6) cells, while in the lymphocytes of 9 patients obtained immediately before treatment this value was between 0.5 and 4.8 nmol/h per 10(6) cells. Within 15 min of drug administration DHO-DH activity was not detectable and was still low up to 1 week later. Duration of the inhibition appeared to be related to the extent of clinical toxicity, e.g., myelosuppression, nausea, vomiting, diarrhea, and mucositis. Severe lymphopenia was observed in patients receiving Brequinar sodium at the maximum tolerated dose. At dose levels of greater than or equal to 600 mg/m2, uridine depletion (40-85%) was observed between 6 h and 4 days, followed by a rebound of 160-350% after 4-7 days. The extent of the depletion and of the accompanying rebound of uridine levels and the extent and duration of DHO-DH inhibition in the individual patients could be partially associated with drug toxicity in these patients. This is the first report describing biological effects of DHO-DH inhibition in humans in relation to the degree and duration of inhibition of this enzyme.


Subject(s)
Antineoplastic Agents/therapeutic use , Biphenyl Compounds/therapeutic use , Dihydroorotate Oxidase/antagonists & inhibitors , Neoplasms/drug therapy , Oxidoreductases/antagonists & inhibitors , Animals , Biphenyl Compounds/pharmacokinetics , Biphenyl Compounds/pharmacology , Dose-Response Relationship, Drug , Humans , Lymphocytes/enzymology , Mice , Mice, Inbred BALB C , Reference Values , Uridine/blood
10.
Biochem Pharmacol ; 39(1): 135-44, 1990 Jan 01.
Article in English | MEDLINE | ID: mdl-2153375

ABSTRACT

Brequinar sodium (DUP-785) is a potent inhibitor of the pyrimidine de novo enzyme, dihydroorotic acid dehydrogenase (DHO-DH). In order to determine whether in vitro data could be extrapolated to the in vivo situation we investigated antipyrimidine effects of DUP-785 in mice bearing colon cancer. Two tumor models were used, Colon 26 and Colon 38, resistant and moderately sensitive to DUP-785, respectively. DUP-785 at 50 mg/kg caused a depletion of plasma uridine in mice, and depleted tissue uridine levels in Colon 38 down to 10%, which was retained for several days; in Colon 26 the decrease was less and tissue uridine levels recovered rapidly. In livers of these mice no significant effect on uridine was observed. DUP-785 depleted UTP in bone marrow cells within 2 hr to 25% of control levels, after 4 days normal levels were found. In livers of both Balb-c mice (bearing Colon 26) and C57Bl/6 mice (bearing Colon 38) a small decrease of uridine nucleotide pools was found. In Colon 26 DUP-785 increased uridine nucleotide pools to 170% after 2 hr, at 1 day normal levels were observed, but after 2 days again an increase was found. In Colon 38 DUP-785 decreased the uridine nucleotide pool by 50% after 1 and 2 days. DUP-785 did not affect cytidine nucleotide pools of livers and of Colon 26 and Colon 38. The ratio between uridine nucleotides and cytidine nucleotides decreased from 2.2 to 0.90 in Colon 38, in the other tissues the decrease was less. DHO-DH was measured in bone marrow cells and Colon 26 and 38 before and after treatment. Basal levels of DHO-DH were 3 times higher in Colon 26 than in Colon 38. In treated tumors DHO-DH was initially inhibited by more than 90%, after 7 days enzyme activity in Colon 26 was 50% and in Colon 38 about 200% of basal levels. In bone marrow cells DHO-DH was also rapidly inhibited but recovered within 4 days. It is concluded that the retention of antipyrimidine effects of DUP-785 in Colon 38 were more pronounced than in Colon 26, which is in agreement with the better antitumor effect of DUP-785 in Colon 38.


Subject(s)
Antineoplastic Agents , Biphenyl Compounds/pharmacology , Bone Marrow/metabolism , Colonic Neoplasms/metabolism , Dihydroorotate Oxidase/antagonists & inhibitors , Liver/metabolism , Oxidoreductases/antagonists & inhibitors , Pyrimidine Nucleotides/metabolism , Animals , Biphenyl Compounds/therapeutic use , Bone Marrow/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Female , Liver/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Uridine/metabolism , Uridine Triphosphate/metabolism
11.
Cancer Lett ; 46(2): 123-7, 1989 Jul 15.
Article in English | MEDLINE | ID: mdl-2546667

ABSTRACT

The growth inhibitory effects of Brequinar Sodium (DUP-785; NSC 368390) in 7 different cell lines were related to growth rates and to the inhibition of dihydroorotic acid dehydrogenase (DHO-DH) activity. IC50 values were between 0.2 and 5.8 microM; the fastest growing cell line was least sensitive. Despite a large variation in sensitivity, basal activity of DHO-DH showed little variation (only 2-fold) between the different cell lines. Residual activity of DHO-DH in the presence of Brequinar Sodium varied 30-fold. Drug sensitivity correlated with this residual DHO-DH activity; DHO-DH activity was only slightly inhibited by Brequinar Sodium in the most resistant lines, and almost completely in the most sensitive.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Cell Division/drug effects , Dihydroorotate Oxidase/antagonists & inhibitors , Oxidoreductases/antagonists & inhibitors , Animals , Humans , Mice , Rats , Tumor Cells, Cultured
13.
Biochem Pharmacol ; 37(20): 3807-16, 1988 Oct 15.
Article in English | MEDLINE | ID: mdl-2847745

ABSTRACT

The activity of dihydroorotate dehydrogenase (DHO-dehase) has been reported to decrease both in vitro and in vivo in hepatocellular carcinomas. DHO-dehase, the fourth enzyme of the de novo pyrimidine biosynthetic pathway, is a mitochondrial enzyme which is both a potential rate-limiting reaction in the de novo pyrimidine biosynthetic pathway and a potential therapeutic target for tumor inhibitors. This paper reports results on a series of pyrimidine analogs of dihydroorotate (DHO) and orotic acid (OA) as inhibitors of DHO-dehase. The enzyme test results established that the intact amide and imide groups of the pyrimidine ring and the 6-carboxylic acid are required for significant enzyme inhibition. The testing of several functional groups similar in characteristics to that of the carboxylic acid, such as sulfonamide, tetrazole and phosphate, indicated that the carboxylic acid group is preferred by the enzyme. Using various 5-substituted OA and DHO derivatives, it was shown that there is a steric limitation of a methyl group at this position. The compound D,L-5-trans-methyl DHO (7) (Ki of 45 microM) was both an inhibitor and a weak substrate for the enzyme, demonstrating that mechanism-based enzyme inhibitors should be effective. The testing results further suggest that a negatively charged enzyme substituent may be present near the 5-position of the pyrimidine ring and that there may be an enzyme-substrate metal coordination site near the N-1 and carboxylic acid positions of the pyrimidine ring. The combined testing results were then used to define both conformational and steric substrate enzyme binding requirements from which a model was proposed for the binding of DHO and OA to the DHO-dehase active site.


Subject(s)
Dihydroorotate Oxidase/antagonists & inhibitors , Oxidoreductases/antagonists & inhibitors , Pyrimidines/pharmacology , Kinetics , Molecular Conformation , Structure-Activity Relationship
14.
Biochem Pharmacol ; 37(17): 3257-66, 1988 Sep 01.
Article in English | MEDLINE | ID: mdl-2840910

ABSTRACT

DUP 785 (NSC 368390; Brequinar sodium) is a new inhibitor of pyrimidine de novo biosynthesis with antitumor activity against several experimental tumors. DUP 785 inhibits the mitochondrial enzyme dihydroorotate dehydrogenase, blocking the conversion of dihydroorotate to orotate. We examined the influence of exposure time to DUP 785 on its growth-inhibitory effects in L1210 murine leukemia and WiDR human adenocarcinoma cells and the effects of pyrimidine (deoxy) nucleosides on reversal of growth-inhibition. The results were correlated with changes in intracellular pyrimidine nucleotide pools and cell cycle distribution. In L1210 cells, a continuous exposure to 25 microM DUP 785 up to 96 hr caused complete growth inhibition. A 2 hr exposure of cells to the drug did not affect growth. In WiDR cells, exposure to the drug for 1-24 hr, followed by cultivation in drug-free medium resulted in recovery of growth. However, cells exposed to the drug for 48 hr or longer were not able to resume growth when recultured in drug-free medium. Reversal studies were performed to know whether selective depletion of one of the pyrimidine (deoxy) nucleotides might be related to the growth-inhibitory effects of DUP 785. Neither thymidine, deoxycytidine alone, deoxycytidine plus tetrahydrouridine; nor cytidine plus tetrahydrouridine added after 24 hr were able to reverse cell growth inhibition induced by 25 microM DUP 785. However, uridine and cytidine alone reversed growth inhibition. UTP and CTP pools in L1210 cells decreased to about 30-40% of control levels after 4 hr of drug exposure, while dTTP and dCTP pools decreased to about 30% of control levels. There were no significant changes in purine nucleotide pools. In WiDR cells, UTP and CTP pools decreased rapidly after drug exposure and were substantially depleted after 24 hr. Reculture of cells in drug-free medium resulted in a significant recovery of UTP and CTP levels only for cells exposed to DUP 785 for 1-24 hr. For cells exposed to the drug for 48 and 72 hr recovery of nucleotide pools was minimal. In L1210 cells, a 12-hr exposure to the drug caused an accumulation of cells in the early S-phase. In WiDR cells, there was a clear accumulation of cells in the S-phase of the cell cycle after 24 hr drug exposure.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Biphenyl Compounds/pharmacology , Growth Inhibitors , Animals , Biphenyl Compounds/administration & dosage , Cell Cycle/drug effects , Cell Division/drug effects , Deoxyribonucleotides/metabolism , Dihydroorotate Oxidase/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Mice , Pyrimidine Nucleotides/metabolism , Ribonucleotides/metabolism , Time Factors , Tumor Cells, Cultured
15.
Invest New Drugs ; 5(3): 235-44, 1987.
Article in English | MEDLINE | ID: mdl-2822596

ABSTRACT

The mechanism of action of NSC 368390 (DUP-785, 6-fluoro-2-(2'-fluoro-1, 1'-biphenyl-4-yl)-3-methyl-4-quinoline carboxylic acid sodium salt) was studied using three different approaches. First, we studied growth inhibition by DUP-785 in L1210 leukemia cells and M5 melanoma cells. The concentrations causing 50% growth inhibition after 48 hr of culture were 5.8 and 0.6 microM, respectively. DUP-785 had to be present continuously throughout culture. Growth inhibition by 25 microM DUP-785 could be prevented by addition of 1 mM uridine or orotic acid to cultures of these cell lines; in M5 cells cytidine was also able to prevent growth inhibition. Dihydro-orotic acid (DHO) and carbamyl-aspartate were not able to prevent growth inhibition by DUP-785. Second, we studied accumulation of orotic acid and of orotidine induced by incubation with 1 microM pyrazofurin, an inhibitor of the orotate phosphoribosyl-transferase-orotidine-monophosphate decarboxylase complex. Addition of DUP-785 to the culture medium prevented the orotic acid accumulation. Furthermore, DUP-785 prevented accumulation of H14CO3- into orotic acid of pyrazofurin-treated L1210 cells. Third, we measured the effect of DUP-785 on DHO-dehydrogenase (DHO-DH), since the results indicated that this enzyme was affected by DUP-785. DHO-DH was assayed in isolated rat liver mitochondria. The Km for L-DHO was about 12 microM. DUP-785 appeared to be a potent inhibitor of DHO-DH with an apparent Ki of about 0.1 microM and an apparent Ki' of about 0.8 microM. The mode of inhibition appeared to be linear mixed type. After exposure of L1210 cells to 25 microM DUP-785 for 2 hr DHO-DH was almost completely inhibited. After suspension in fresh medium without drug, DHO-DH activity was recovered to about 60% after 24 hr. In conclusion, DUP-785 is a potent inhibitor of pyrimidine de novo biosynthesis, by inhibition of the mitochondrial enzyme DHO-DH.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Pyrimidines/biosynthesis , Amides , Animals , Dihydroorotate Oxidase/antagonists & inhibitors , Leukemia L1210/metabolism , Orotic Acid/metabolism , Pyrazoles , Ribonucleosides/pharmacology , Ribose , Tumor Cells, Cultured/metabolism
16.
Cancer Res ; 46(10): 5014-9, 1986 Oct.
Article in English | MEDLINE | ID: mdl-3019518

ABSTRACT

Exposure of cultured clone A human colon tumor cells to 25 to 75 microM of NSC 368390 [6-fluoro-2-(2'-fluoro-1,1'-biphenyl-4-yl)-3-methyl-4-quinolinecarbox yli c acid sodium salt, DuP 785] for 48 to 72 h resulted in a 99.9% cell kill as determined by clonogenic assay. Cells exposed to NSC 368390 became depleted in intracellular pools of uridine 5'-triphosphate and cytidine 5'-triphosphate. Both uridine 5'-triphosphate and cytidine 5'-triphosphate were decreased to 50% of levels in control cells at 3 h and were undetectable at 15 h after addition of 25 microM of NSC 368390 to the cultures. Similar effects were observed in L1210 leukemia cells. Addition of 0.1 mM of uridine or cytidine restored intracellular pools of uridine 5'-triphosphate and cytidine 5'-triphosphate to control levels and rescued clone A cells from NSC 368390 cytotoxicity. Addition of uridine circumvented NSC 368390 cytotoxicity in L1210 cells, but addition of cytidine did not. This result is consistent with the fact that L1210 cells lack cytidine deaminase and thus cannot form uridine or its anabolites from cytidine. These results indicated that NSC 368390 inhibits a step in the de novo biosynthetic pathway leading to uridine 5'-monophosphate. Therefore, the effects of NSC 368390 on the six enzymes that comprise the de novo pathway leading to the formation of uridine 5'-monophosphate were examined. The results showed that NSC 368390 was a potent inhibitor of dihydroorotate dehydrogenase, the fourth enzyme in the pathway; thus, this study demonstrates that NSC 368390 exerts its tumoricidal effect by inhibiting a step in de novo pyrimidine biosynthesis resulting in the depletion of critical precursors for RNA and DNA synthesis.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Pyrimidine Nucleotides/biosynthesis , Cell Line , Cell Survival/drug effects , Clone Cells , Cytidine/metabolism , Cytidine/pharmacology , Dihydroorotate Oxidase/antagonists & inhibitors , Humans , Uridine/metabolism , Uridine/pharmacology
17.
Biochem Med ; 34(1): 60-9, 1985 Aug.
Article in English | MEDLINE | ID: mdl-2996510

ABSTRACT

Seventy-one pyrimidine analogs have been tested as possible inhibitors of human spleen mitochondrial dihydroorotate dehydrogenase. Of these nine were demonstrated to be effective inhibitors of the enzymic activity. Two compounds, dihydro-5-azaorotate and 6-thiobarbiturate appeared to be specific inhibitors of the DHO-DHase. In addition, three compounds, 5-azaorotate, 5-bromoorotate, and barbiturate were also inhibitory against the two subsequent enzymes of the pathway, orotate phosphoribosyltransferase and orotidylate decarboxylase, so that they could act against three enzymes of the mammalian pyrimidine de novo biosynthetic pathway.


Subject(s)
Dihydroorotate Oxidase/antagonists & inhibitors , Oxidoreductases/antagonists & inhibitors , Pyrimidines/pharmacology , Spleen/enzymology , Barbiturates/pharmacology , Humans , In Vitro Techniques , Kinetics , Orotate Phosphoribosyltransferase/antagonists & inhibitors , Orotic Acid/pharmacology , Orotidine-5'-Phosphate Decarboxylase/antagonists & inhibitors , Saccharomyces cerevisiae/enzymology , Structure-Activity Relationship , Thiobarbiturates/pharmacology
18.
Biochem Med ; 34(1): 70-82, 1985 Aug.
Article in English | MEDLINE | ID: mdl-2996511

ABSTRACT

Human spleen dihydroorotate dehydrogenase is associated with the mitochondrial membrane and is linked to the respiratory chain via ubiquinone. The enzyme activity was unaffected by pyridine nucleotides. The product of the reaction, orotate, was a potent inhibitor. However, a range of other naturally occurring pyrimidines or purines had no significant effect on the activity. No evidence for the involvement of a complexed metal ion or for an active sulfhydryl group was obtained. Purification of the enzyme was achieved by preparation of an acetone powder and extraction with Triton X-100, followed by preparative polyacrylamide gel electrophoresis. Activity was observed by the addition of the artificial electron acceptors, ubiquinone 50 or PMS. Purification resulted in alteration of the pH optimum and of other kinetic characteristics. Two molecular-weight species, of molecular weight 88,000 and 98,000, were consistently observed. The properties of the human spleen enzyme were similar in principle to those for the rat liver enzyme. Differences in the mode of linkage to the respiratory chain for the mitochondrially bound enzyme, and in the characteristics of the purified enzyme, were observed.


Subject(s)
Dihydroorotate Oxidase/metabolism , Oxidoreductases/metabolism , Spleen/enzymology , Dihydroorotate Oxidase/antagonists & inhibitors , Dihydroorotate Oxidase/isolation & purification , Electron Transport , Electrophoresis, Polyacrylamide Gel , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Intracellular Membranes/enzymology , Mitochondria/enzymology , Purines/pharmacology , Pyrimidines/pharmacology , Thermodynamics , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology
19.
Biochem J ; 225(1): 189-94, 1985 Jan 01.
Article in English | MEDLINE | ID: mdl-2983662

ABSTRACT

1,1,1-Trifluoro-3-thenoylacetone was shown to cause complete inhibition of dihydroorotate oxidation in rat liver mitochondria as measured by orotate formation and the rate of dihydro-orotate-dependent reduction of 2,6-dichlorophenol-indophenol or cytochrome c. The inhibition by trifluorothenoylacetone was dose-dependent, and a concentration of 1 mM completely inhibited dihydro-orotate dehydrogenase activity. 1,10-Phenanthroline, another iron-chelating agent, also caused total inhibition of the liver enzyme. Whereas the iron chelators inhibited 100% of dihydro-orotate dehydrogenase activity in liver mitochondria, they inhibited only a maximum of 72% in the case of the brain enzyme. The inhibition by trifluorothenoylacetone was not prevented by addition of phenazine methosulphate or ubiquinone. Dihydro-orotate dehydrogenase-mediated generation of superoxide was abolished when the enzyme was fully inhibited by trifluorothenoylacetone or when the electron-transport system was blocked by antimycin A. These results suggest that the iron component(s) of dihydro-orotate dehydrogenase is of strategic importance for catalytic activity and transfer of reducing equivalents from the primary enzyme to the electron-transport chain. Furthermore, the study indicates that production of superoxide radicals during dihydro-orotate dehydrogenase-catalysed oxidation of dihydro-orotate may be at the cytochrome b-c1 segment of the electron-transport chain (as a consequence of autooxidation of ubisemiquinone) rather than at a site on the primary enzyme.


Subject(s)
Dihydroorotate Oxidase/antagonists & inhibitors , Mitochondria, Liver/enzymology , Oxidoreductases/antagonists & inhibitors , Thenoyltrifluoroacetone/pharmacology , Thiophenes/pharmacology , Animals , Antimycin A/pharmacology , Brain/drug effects , Brain/enzymology , Cytochrome c Group/metabolism , In Vitro Techniques , Male , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria, Liver/drug effects , Rats , Rats, Inbred Strains , Superoxide Dismutase/pharmacology , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...