Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters










Publication year range
1.
Can J Physiol Pharmacol ; 79(6): 443-70, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11430583

ABSTRACT

Endothelium-derived hyperpolarizing factor (EDHF) is defined as the non-nitric oxide (NO) and non-prostacyclin (PGI2) substance that mediates endothelium-dependent hyperpolarization (EDH) of vascular smooth muscle cells (VSMC). Although both NO and PGI2 have been demonstrated to hyperpolarize VSMC by cGMP- and cAMP-dependent mechanisms, respectively, and in the case of NO by cGMP-independent mechanisms, a considerable body of evidence suggests that an additional cellular mechanism must exist that mediates EDH. Despite intensive investigation, there is no agreement as to the nature of the cellular processes that mediates the non-NO/PGI2 mediated hyperpolarization. Epoxyeicosatrienoic acids (EET), an endogenous anandamide, a small increase in the extracellular concentration of K+, and electronic coupling via myoendothelial cell gap junctions have all been hypothesized as contributors to EDH. An attractive hypothesis is that EDH is mediated via both chemical and electrical transmissions, however, the contribution from chemical mediators versus electrical transmission varies in a tissue- and species-dependent manner, suggesting vessel-specific specialization. If this hypothesis proves to be correct then the potential exists for the development of vessel and organ-selective vasodilators. Because endothelium-dependent vasodilatation is dysfunctional in disease states (i.e., atherosclerosis), selective vasodilators may prove to be important therapeutic agents.


Subject(s)
Biological Factors/physiology , Nitric Oxide/physiology , Animals , Humans
2.
J Med Chem ; 44(12): 1993-2003, 2001 Jun 07.
Article in English | MEDLINE | ID: mdl-11384244

ABSTRACT

Classical and nonclassical isosteric C8-N9 bridged analogues of the multitargeted antifolate LY231514 were synthesized as inhibitors of thymidylate synthase (TS), dihydrofolate reductase (DHFR), and as antitumor and antiopportunistic infection agents. The syntheses of the analogues were accomplished by reductive amination of the appropriate anilines with 2-amino-4-oxo-5-cyanopyrrolo[2,3-d]pyrimidine (28) followed by saponification of the ethyl esters, for the classical analogue 6. The N9-methyl analogues were obtained from the N9-H precursors by reductive methylation. In general, the nonclassical compounds 7-17 were similar in potency to TMP against Toxoplasma gondii DHFR, with selectivity ratios greater than 38 and 21 for 11 and 16, respectively. These compounds were poor inhibitors of Pneumocystis carinii DHFR and rat liver DHFR. The nonclassical analogues were also inactive against TS. The classical analogue 6 was a marginal inhibitor of isolated human TS (IC50 = 46 microM) and of human DHFR (IC50 = 10 microM), however, it was a potent inhibitor of the growth of two human head and neck squamous cell carcinoma cell lines and of CCRF-CEM human lymphoblastic leukemia cells in culture and was similar to LY231514 against ZR-75-1 human breast carcinoma cell line. Evaluation of 6 against MTX-resistant sublines indicated that DHFR is not the major target of 6. Metabolite protection studies of the growth inhibitory activity of 6 suggest that TS is a major target of this drug and that polyglutamyl forms of 6 may serve as the intracellular TS inhibitors. These studies also suggest that 6 has a site of action in addition to sites in the folate pathway.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cell Division/drug effects , Folic Acid Antagonists/chemical synthesis , Glutamates/chemistry , Guanine/analogs & derivatives , Guanine/chemistry , Pyrimidines/chemical synthesis , Pyrroles/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Drug Design , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Glutamates/pharmacology , Guanine/pharmacology , Humans , Liver/enzymology , Models, Molecular , Molecular Conformation , Pemetrexed , Pneumocystis/enzymology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Rats , Recombinant Proteins/antagonists & inhibitors , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/metabolism , Thymidylate Synthase/antagonists & inhibitors , Toxoplasma , Tumor Cells, Cultured
3.
J Med Chem ; 43(21): 3837-51, 2000 Oct 19.
Article in English | MEDLINE | ID: mdl-11052789

ABSTRACT

A novel N-¿2-amino-4-methyl[(pyrrolo[2, 3-d]pyrimidin-5-yl)ethyl]benzoyl¿-L-glutamic acid (3a) was designed and synthesized as a potent dual inhibitor of thymidylate synthase (TS) and dihydrofolate reductase (DHFR) and as an antitumor agent. Compound 3b, the N7-benzylated analogue of 3a, was also synthesized as an antitumor agent. The synthesis of 3a was accomplished via a 12-step sequence which involved the synthesis of 2-amino-4-methylpyrrolo[2,3-d]pyrimidine (10) in 5 steps from 2-acetylbutyrolactone. Protection of the 2-amino group of 10 and regioselective iodination at the 5-position followed by palladium-catalyzed coupling afforded intermediate 14 which was converted to 3a by reduction and saponification. Similar synthetic methodology was used for 3b. X-ray crystal structure of the ternary complex of 3a, DHFR, and NADPH showed that the pyrrolo[2, 3-d]pyrimidine ring binds in a "2,4-diamino mode" in which the pyrrole nitrogen mimics the 4-amino moiety of 2,4-diaminopyrimidines. This is the first example of a classical pyrrolo[2,3-d]pyrimidine antifolate shown to have this alternate mode of binding to DHFR. Compounds 3a and 3b were more inhibitory than LY231514 against TS from Lactobacillus casei and Escherichia coli. Analogue 3a was also more inhibitory against DHFR from human, Toxoplasma gondii, and Pneumocystis carinii. Evaluation of 3a against methotrexate (MTX)-resistant cell lines with defined mechanisms indicated that cross-resistance of 3a was much lower than that of MTX. Metabolite protection studies and folylpoly-gamma-glutamate synthetase studies suggest that the antitumor activity of 3a against the growth of tumor cells in culture is a result of dual inhibition of TS and DHFR. Compound 3a inhibited the growth of CCRF-CEM and FaDu cells in culture at ED(50) values of 12.5 and 7.0 nM, respectively, and was more active against FaDu cells than MTX. In contrast, compound 3b was inactive against both cell lines. Compound 3a was evaluated in the National Cancer Institute in vitro preclinical antitumor screening program and afforded IG(50) values in the nanomolar range against a number of tumor cell lines.


Subject(s)
Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Folic Acid Antagonists/chemical synthesis , Glutamic Acid/chemical synthesis , Pyrimidines/chemical synthesis , Tetrahydrofolate Dehydrogenase/metabolism , Thymidylate Synthase/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Crystallography, X-Ray , Drug Design , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli/chemistry , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Glutamic Acid/analogs & derivatives , Glutamic Acid/chemistry , Glutamic Acid/pharmacology , Humans , Lacticaseibacillus casei/chemistry , Models, Molecular , Pneumocystis/chemistry , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/chemistry , Thymidylate Synthase/chemistry , Toxoplasma/chemistry , Tumor Cells, Cultured
4.
J Med Chem ; 43(16): 3125-33, 2000 Aug 10.
Article in English | MEDLINE | ID: mdl-10956221

ABSTRACT

N-[4-[1-methyl-2-(2,4-diaminofuro[2, 3-d]pyrimidin-5-yl)ethyl]benzoyl]-L-glutamic acid (5) and its C8-C9 conformationally restricted E- and Z-isomers (6 and 7) were designed and synthesized in order to investigate the effect of incorporating a methyl group at the C9 position and of conformational restriction at the C8-C9 bridge of N-[4-[2-(2,4-diaminofuro[2, 3-d]pyrimidin-5-yl)ethyl]benzoyl]-L-glutamic acid (1) with respect to dihydrofolate reductase (DHFR) inhibitory activity as well as antitumor activity. The compounds were synthesized by a Wittig reaction of 2,4-diamino-5-(chloromethyl)furo[2,3-d]pyrimidine with ethyl 4-acetylbenzoate followed by catalytic reduction, hydrolysis, and standard peptide coupling with diethyl L-glutamate. The biological results indicated that the addition of a 9-methyl group to the C8-C9 bridge, as in 5, increased recombinant human (rh) DHFR inhibitory potency (IC(50) = 0.42 microM) as well as the potency against the growth inhibition of tumor cells in culture (CCRF-CEM EC(50) = 29 nM, A253 EC(50) = 28.5 nM, and FaDu EC(50) = 17.5 nM) compared with the 9-desmethyl analogue 1. However, the conformationally restricted 4:1 Z/E mixture of 7 and 6 was less potent than 5 in both assays, and the pure E-isomer 6 was essentially inactive. These three classical analogues were also evaluated as inhibitors of Lactobacillus casei, Escherichia coli, and rat and rh thymidylate synthase (TS) and were found to be weak inhibitors. All three analogues 5-7 were good substrates for human folylpolyglutamate synthetase (FPGS). These data suggested that FPGS is relatively tolerant to different conformations in the bridge region. Further evaluation of the cytotoxicity of 5 and 7 in methotrexate (MTX)-resistant CCRF-CEM cell sublines suggested that polyglutamylation was crucial for their mechanism of action. Metabolite protection studies of 5 implicated DHFR as the primary intracellular target. Compound 5 showed GI(50) values in 10(-9)-10(-7) M range against more than 30 tumor cell lines in culture.


Subject(s)
Antimetabolites, Antineoplastic/chemical synthesis , Folic Acid Antagonists/chemical synthesis , Folic Acid/analogs & derivatives , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Cell Division/drug effects , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli/chemistry , Folic Acid/chemical synthesis , Folic Acid/chemistry , Folic Acid/pharmacology , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Humans , Lacticaseibacillus casei/chemistry , Rats , Recombinant Proteins/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/chemistry , Toxoplasma/chemistry , Tumor Cells, Cultured
5.
Cancer Res ; 60(13): 3514-21, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10910063

ABSTRACT

Cellular resistance to the antifolate methotrexate (MTX) is often caused by target amplification, uptake defects, or alterations in polyglutamylation. Here we have examined MTX cross-resistance in a human breast carcinoma cell line (MCF7/MX) selected in the presence of mitoxantrone, an anticancer agent associated with the multidrug resistance (MDR) phenotype. Examination of protein expression and enzyme activities showed that MCF7/MX cells displayed none of the classical mechanisms of MTX resistance. They did, however, exhibit an ATP-sensitive accumulation defect accompanied by reduced polyglutamylation. Although the kinetics of drug uptake was similar between parental and resistant cells, the resistant cells exhibited increased energy-dependent drug efflux. This suggested the involvement of an ATP-binding cassette (ABC) transporter. However, cells transfected with the breast cancer resistance protein (BCRP)-the ABC transporter known to be highly overexpressed in MCF7/MX cells and to confer mitoxantrone resistance (D. D. Ross et al., J. Natl. Cancer Inst. 91: 429-433, 1999)-were not MTX resistant, which suggested that this transporter is not involved in MTX cross-resistance. Moreover, members of the MRP protein family of transport proteins, which had previously been implicated in MTX resistance, were not found to be overexpressed in the MCF7/MX cells. Thus, our data suggest that a novel MTX-specific efflux pump may be involved in this unusual cross-resistance phenotype.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/toxicity , Drug Resistance, Multiple , Methotrexate/pharmacokinetics , Methotrexate/toxicity , Mitoxantrone/toxicity , Neoplasm Proteins , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Biological Transport , Breast Neoplasms , Cell Survival/drug effects , Female , Humans , Mitoxantrone/pharmacokinetics , Peptide Synthases/metabolism , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured
6.
J Biol Chem ; 275(17): 13012-6, 2000 Apr 28.
Article in English | MEDLINE | ID: mdl-10777604

ABSTRACT

Folylpolyglutamate synthetase (FPGS) activity in CCRF-CEM human leukemia cells was found in the cytosolic ( approximately 67% of total) and mitochondrial ( approximately 22%) fractions. A polyclonal antipeptide antibody (430Ab) to human FPGS specifically recognized distinct immunoreactive bands ( approximately 60 kDa) present in each subcellular fraction. Human cytosolic FPGS (hcFPGS) migrated more rapidly than mitochondrial FPGS (hmFPGS); their estimated difference in molecular mass was 1 kDa. The human K562 acute nonlymphocytic leukemia and the A253 and FaDu head and neck cancer cell lines also expressed the two FPGS isoforms, and the ratio of hcFPGS to hmFPGS protein in each cell line was similar. Since K562 and A253 cells are intrinsically resistant to pulse methotrexate (MTX) exposure relative to CCRF-CEM and FaDu cells, respectively, because of decreased MTX polyglutamate synthesis (despite having similar levels of total FPGS activity expression), these data suggest that the natural difference in drug sensitivity cannot be explained by compartmentalization of FPGS activity. Higher expression of hmFPGS relative to hcFPGS was observed in some sublines of CCRF-CEM with acquired MTX resistance suggesting that differential expression of the hmFPGS isoform may contribute to MTX resistance caused by decreased FPGS activity.


Subject(s)
Cytosol/enzymology , Mitochondria/enzymology , Peptide Synthases/chemistry , Animals , Blotting, Western , Cattle , Cell Line , Cell Nucleus/enzymology , Electrophoresis , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/chemistry , Methotrexate/pharmacology , Tumor Cells, Cultured
7.
J Pharmacol Exp Ther ; 293(2): 569-77, 2000 May.
Article in English | MEDLINE | ID: mdl-10773030

ABSTRACT

The flavoprotein inhibitor, diphenyleneiodonium (DPI), inhibits the action of glyceryl trinitrate (GTN) and the D-enantiomer of isoidide dinitrate (IIDN), but not the L-enantiomer (L-IIDN), in isolated rat aorta via inhibition of the bioactivation of these prodrugs. Paradoxically, a vascular NAD(P)H oxidase, which also is inhibited by DPI, has been proposed to generate superoxide that quenches nitric oxide (NO) produced during GTN biotransformation, and increased oxidase levels are proposed to contribute to the phenomenon of organic nitrate tolerance. We examined the effect of DPI on isolated rat aorta using an in vivo model of organic nitrate tolerance. The EC(50) values for GTN-, D-IIDN-, and L-IIDN-induced relaxation of aorta from GTN-tolerant rats were increased 4.5- to 7.5-fold. Treatment of blood vessels with DPI (0.3 microM) increased the EC(50) values for GTN and D-IIDN by the same magnitude in control and tolerant aortae, a result that would not be predicted if DPI and GTN tolerance affected common targets. The expression of NADPH-cytochrome P450 reductase (CPR) during in vivo tolerance was assessed by NADPH-dependent cytochrome c reductase activity of aortic microsomes, immunoblotting, and Northern analysis. By all three determinants, CPR expression was unchanged in aorta from GTN-tolerant rats. Superoxide dismutase-inhibitable NADPH-dependent cytochrome c reductase activity (a measure of superoxide generation) of tolerant rat aortic microsomes was not different from that of controls. Superoxide dismutase-inhibitable NADH-dependent cytochrome c reductase activity was detected only in microsomes from tolerant animals. DPI caused a modest increase in the sensitivity for relaxation by the NO donor DEA NONOate to an equal extent in tolerant and nontolerant tissues, whereas the superoxide scavenger, 4,5-dihydroxy-1,3-benzene disulfonic acid (Tiron), had no effect on the sensitivity for relaxation by GTN. These results would not be expected if tolerance-induced increases in superoxide were a causative factor for the reduced relaxation response in tolerance. We conclude that neither reduced flavoprotein-dependent metabolic activation of organic nitrates, such as that mediated by CPR, nor increased superoxide due to increased NAD(P)H oxidase activity can account for the development of in vivo tolerance to GTN.


Subject(s)
Enzyme Inhibitors/pharmacology , Flavoproteins/antagonists & inhibitors , Nitrates/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Onium Compounds/pharmacology , Vasodilator Agents/antagonists & inhibitors , Animals , Aorta, Thoracic/drug effects , Biotransformation , Blotting, Northern , Drug Tolerance , Glutathione/analogs & derivatives , Glutathione/pharmacology , Isosorbide Dinitrate/antagonists & inhibitors , Isosorbide Dinitrate/metabolism , Isosorbide Dinitrate/pharmacology , Male , Microsomes/drug effects , Microsomes/metabolism , Muscle, Smooth, Vascular/drug effects , NADPH-Ferrihemoprotein Reductase/metabolism , Nitrates/metabolism , Nitrates/pharmacology , Nitric Oxide Donors/pharmacology , Nitroglycerin/antagonists & inhibitors , Nitroglycerin/metabolism , Nitroglycerin/pharmacology , Nitroso Compounds/pharmacology , Prodrugs/metabolism , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , S-Nitrosoglutathione , Stereoisomerism , Superoxides/metabolism , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology
8.
Eur J Pharm Sci ; 10(3): 237-45, 2000 May.
Article in English | MEDLINE | ID: mdl-10767601

ABSTRACT

Lipophilic methotrexate (MTX)-lipoamino acid conjugates coupled with amide or ester linkages (1a-1r) were synthesised. The inhibitory activity of the conjugates was evaluated on bovine liver DHFR. The in vitro growth inhibitory effect against MTX-sensitive human lymphoblastoid CCRF-CEM cells and an MTX-resistant sub-line (CEM/MTX), which displays defective intracellular transport of MTX, was determined under short-term and continuous (120-h incubation) exposure conditions. The alpha, gamma, or alpha,gamma amide conjugates showed different activity in inhibiting the growth of parent cells. CEM/MTX cells were much less susceptible than CCRF-CEM cells to inhibition by alpha or alpha,gamma-substituted lipoamino acid conjugates, whereas both cell lines were almost equally sensitive to the MTX-gamma conjugates. Although less potent than MTX, they could partially circumvent the impaired transport system. These findings confirm that lipophilic MTX conjugates may be good lead compounds on the drug development for the treatment of some MTX-resistant tumors. Ester-type conjugates displayed an interesting activity against parent CCRF-CEM cells, although they were less potent against the transport-resistant sub-line. Stability studies on these molecules indicated that they are not degraded into MTX in the culture medium, thus suggesting that they are not able to over-cross cell resistance despite of their lipophilicity.


Subject(s)
Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Methotrexate/chemistry , Methotrexate/pharmacology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Cattle , Cell Division/drug effects , Chemical Phenomena , Chemistry, Physical , Excipients , Folic Acid Antagonists/administration & dosage , Humans , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/pathology , Lipids/chemistry , Liver/drug effects , Liver/enzymology , Mass Spectrometry , Methotrexate/administration & dosage , Spectrophotometry, Infrared , Tetrahydrofolate Dehydrogenase/metabolism , Tumor Cells, Cultured
9.
Int J Oncol ; 15(6): 1245-50, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10568835

ABSTRACT

Effects of lipophilic thymidylate synthase (TS) inhibitor AG337 on human head and neck squamous cell carcinoma (HNSCC) cell lines and CCRF-CEM human leukemia cells and sublines with acquired methotrexate (MTX) resistance were assayed using continuous or intermittent drug exposure. During 120-h continuous exposure, HNSCC cell lines A253 and FaDu are equally MTX sensitive (EC50 equals approximately 15nM); AG337 is less potent (EC50 approximately equals 1 microM). A253 is intrinsically resistant to 24-h intermittent MTX exposure (EC50 equals approximately 17 microM; FaDu, EC50 equals approximately 0.3 microM); both HNSCC cell lines are resistant to 24-h AG337 exposure (EC50 >100 microM). CCRF-CEM shows MTX (EC50 =14 nM) and AG337 (EC50 equals approximately 0.6 microM) sensitivity in continuous exposure similar to HNSCC; however, AG337 retains potency against CCRF-CEM cells in intermittent exposure (24-h, EC50 equals approximately 2 microM; 6-h, EC50 equals approximately 48 microM). The reduced folate leucovorin (LV) at > or = 0.1 microM fully protects from growth inhibition by continuous MTX exposure, but growth inhibition by AG337 is reversed only slightly by < or = 100 microM LV. Thymidine fully protects A253 and FaDu against growth inhibition by AG337, while hypoxanthine alone is without effect, suggesting inhibition is TS-specific. CCRF-CEM sublines with acquired MTX-resistance resulting from DHFR overexpression, defective MTX transport, or defective MTX polyglutamylation retain full sensitivity to AG337 in continuous exposure (all EC50 =0.4 microM). These data indicate that AG337 may be useful in therapy of tumors that have acquired resistance to MTX by most common mechanisms.


Subject(s)
Enzyme Inhibitors/pharmacology , Head and Neck Neoplasms/drug therapy , Quinazolines/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Cell Cycle/drug effects , Cell Division/drug effects , Culture Media/chemistry , Culture Media/pharmacology , Drug Resistance , Fetal Blood/metabolism , Head and Neck Neoplasms/pathology , Humans , Hypoxanthine/pharmacology , Inhibitory Concentration 50 , Leucovorin/pharmacology , Leukemia/drug therapy , Leukemia/pathology , Methotrexate/pharmacology , Thymidine/metabolism , Thymidine/pharmacology , Thymidine Phosphorylase/metabolism , Thymidine Phosphorylase/pharmacology , Tumor Cells, Cultured
10.
Clin Cancer Res ; 5(9): 2548-58, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499632

ABSTRACT

The aim of this study was to investigate the influence of folylpolyglutamyl synthetase (FPGS) activity on the cellular pharmacology of the classical antifolates raltitrexed and methotrexate (MTX) using two human leukemia cell lines, CCRF-CEM and CCRF-CEM:RC2Tomudex. Cell growth inhibition and drug-induced inhibition of de novo thymidylate and purine biosynthesis were used as measures of the cellular effects of the drugs. CCRF-CEM:RC2Tomudex cells had <11% of the FPGS activity of CCRF-CEM cells, whereas MTX uptake and TS activity were equivalent. In CCRF-CEM:RC2Tomudex cells, MTX polyglutamate formation was undetectable after exposure to 1 microM [3H]MTX for 24 h. After exposure to 0.1 microM raltitrexed, levels of total intracellular raltitrexed-derived material in CCRF-CEM:RC2Tomudex cells were 30- to 50-fold lower than in the CCRF-CEM cell line. CCRF-CEM: RC2Tomudex cells were >1000-fold resistant to raltitrexed and 6-fold resistant to lometrexol but sensitive to MTX and nolatrexed when exposed to these antifolates for 96 h. After 6 h of exposure, CCRF-CEM cells retained sensitivity to MTX and raltitrexed but were less sensitive to lometrexol-mediated growth inhibition. In contrast, CCRF-CEM: RC2Tomudex cells were markedly insensitive to raltitrexed, lometrexol, and to a lesser degree, MTX. Simultaneous measurement of de novo thymidylate and purine biosynthesis revealed 90% inhibition of TS activity by 100 nM MTX in both cell lines, whereas inhibition of de novo purine synthesis was only observed in CCRF-CEM cells, and only after exposure to 1000 nM MTX. Ten nM raltitrexed induced >90% inhibition of TS activity in CCRF-CEM cells, whereas in CCRF-CEM:RC2Tomudex cells, there was no evidence of inhibition after exposure to 1000 nM raltitrexed. These studies demonstrate that polyglutamation is a critical determinant of the cellular pharmacology of both raltitrexed and MTX, markedly influencing potency in the case of raltitrexed and locus of action in the case of MTX.


Subject(s)
Folic Acid Antagonists/pharmacology , Leukemia/drug therapy , Leukemia/metabolism , Methotrexate/pharmacology , Purines/antagonists & inhibitors , Quinazolines/pharmacology , Thiophenes/pharmacology , Thymidine Monophosphate/antagonists & inhibitors , Biological Transport , Cell Division/drug effects , Drug Screening Assays, Antitumor , Folic Acid Antagonists/metabolism , Folic Acid Antagonists/pharmacokinetics , Growth Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Leukemia/enzymology , Methotrexate/metabolism , Methotrexate/pharmacokinetics , Peptide Synthases/antagonists & inhibitors , Peptide Synthases/metabolism , Purines/biosynthesis , Quinazolines/metabolism , Quinazolines/pharmacokinetics , RNA, Messenger/metabolism , Thiophenes/metabolism , Thiophenes/pharmacokinetics , Thymidine Monophosphate/biosynthesis , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/metabolism , Tumor Cells, Cultured
11.
Clin Cancer Res ; 5(7): 1925-34, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10430100

ABSTRACT

We studied the antitumor activity and toxicity of ZD1694 (tomudex), a specific inhibitor of thymidylate synthase (TS), in nude mice bearing human head and neck squamous cell carcinoma A253 and FaDu xenografts. Mice were treated by single i.v. push (i.v. x 1), i.v. push once a week for 3 weeks (weekly x 3), and i.v. push once a day for 5 days (daily x 5), and the maximum tolerated doses (MTDs) of ZD1694 were 300 mg/kg, 60 mg/kg/week, and 30 mg/kg/day, respectively. ZD1694 was moderately active against both A253 and FaDu xenografts. Antitumor activity was schedule-dependent in both tumors: weekly x 3 > or = i.v. x 1 >> daily x 5. In contrast, the rank order of toxicity was daily x 5 >> weekly x 3 > or = i.v. x 1. ZD1694 at the MTD produced 20% complete tumor regression and 20% partial tumor regression (PR) with i.v. x 1 and weekly x 3 schedules and 12-day tumor growth delay with daily x 5 schedule against FaDu xenografts. No complete tumor regression was achieved with ZD1694 with any schedule against A253; a 20% PR, 40% PR, and 10-day tumor growth delay were observed with i.v. x 1, weekly x 3, and daily x 5 schedules, respectively. The data indicate that ZD1694 was slightly more effective against FaDu than against A253. Of interest and potential clinical importance was the observation that ZD1694 was still active at doses lower than the MTD (> or =1/3 MTD), which showed a high therapeutic index and wide safety margin. Study of ZD1694 compared with 5-fluorouracil and 5-fluoro-2'-deoxyuridine at the MTD revealed that the antitumor activity of ZD1694 was comparable with or superior to 5-fluorouracil and 5-fluoro-2'-deoxyuridine against both A253 and FaDu xenografts, with less toxicity. High plasma thymidine in mouse relative to human (approximately 1.3 microM and <0.1 microM, respectively) may complicate the study of antitumor activity and toxicity of TS inhibitors with human tumor xenografts grown in the mouse. To test this hypothesis, we preadministered methoxypolyethyleneglycol-conjugated thymidine phosphorylase (MPEG-TPase; 2500 units/kg/dose) to reduce mouse plasma thymidine, then treated with various doses of ZD1694 using the daily x 5 or i.v. x 1 schedules in the A253 tumor model. MPEG-TPase significantly increased the toxicity of ZD1694; the MTD of ZD1694 plus MPEG-TPase was reduced 3- and 10-fold compared with ZD1694 alone for i.v x 1 and daily x 5 schedules, respectively. However, preadministration of MPEG-TPase did not potentiate the antitumor activity of ZD1694 with either schedule. The data indicate that the study of TS inhibitors in rodent models may not be suitable for predicting a safe dose for clinical study. However, rodent models, particularly human tumor xenografts, are still useful models for evaluation of antitumor activity and schedule selection for TS inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Head and Neck Neoplasms/drug therapy , Quinazolines/therapeutic use , Thiophenes/therapeutic use , Thymidine/blood , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Disease Models, Animal , Drug Administration Schedule , Female , Floxuridine/therapeutic use , Floxuridine/toxicity , Fluorouracil/therapeutic use , Fluorouracil/toxicity , Head and Neck Neoplasms/blood , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Quinazolines/administration & dosage , Quinazolines/toxicity , Thiophenes/administration & dosage , Thiophenes/toxicity , Thymidine/pharmacology , Thymidine/physiology , Transplantation, Heterologous , Tumor Cells, Cultured
12.
Nat Neurosci ; 2(8): 706-12, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10412059

ABSTRACT

Ocular growth and refraction are regulated by visual processing in the retina. We identified candidate regulatory neurons by immunocytochemistry for immediate-early gene products, ZENK (zif268, Egr-1) and Fos, after appropriate visual stimulation. ZENK synthesis was enhanced by conditions that suppress ocular elongation (plus defocus, termination of form deprivation) and suppressed by conditions that enhance ocular elongation (minus defocus, form deprivation), particularly in glucagon-containing amacrine cells. Fos synthesis was enhanced by termination of visual deprivation, but not by defocus and not in glucagon-containing amacrine cells. We conclude that glucagon-containing amacrine cells respond differentially to the sign of defocus and may mediate lens-induced changes in ocular growth and refraction.


Subject(s)
Adaptation, Ocular , DNA-Binding Proteins/genetics , Gene Expression Regulation/radiation effects , Immediate-Early Proteins/genetics , Light , Retina/radiation effects , Transcription Factors/genetics , Animals , Chickens , Gene Expression Regulation/physiology , Immunohistochemistry , Male , Neurons/physiology , Neurons/radiation effects , Proto-Oncogene Proteins c-fos/genetics , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Retina/cytology , Retina/metabolism
13.
Br J Pharmacol ; 126(1): 61-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10051121

ABSTRACT

1. We have shown previously that the D- and L- enantiomers of isoidide dinitrate (D-IIDN and L-IIDN) exhibit a potency difference for relaxation and cyclic GMP accumulation in isolated rat aorta and that this is related to preferential biotransformation of the more potent enantiomer (D-IIDN). The objective of the current study was to examine the effect of the flavoprotein inhibitor, diphenyleneiodonium sulphate (DPI), on the enantioselectivity of IIDN action. 2. In isolated rat aortic strip preparations, exposure to 0.3 microM DPI resulted in a 3.6 fold increase in the EC50 value for D-IIDN-induced relaxation, but had no effect on L-IIDN-induced relaxation. 3. Incubation of aortic strips with 2 microM D- or L-IIDN for 5 min resulted in significantly more D-isoidide mononitrate formed (5.0 +/- 1.5 pmol mg protein(-1)) than L-isoidide mononitrate (2.1 +/- 0.7 pmol mg protein(-1)) and this difference was abolished by pretreatment of tissues with 0.3 microM DPI. DPI had no effect on glutathione S-transferase (GST) activity or GSH-dependent biotransformation of D- or L-IIDN in the 105,000 x g supernatant fraction of rat aorta. 4. Consistent with both the relaxation and biotransformation data, treatment of tissues with 0.3 microM DPI significantly inhibited D-IIDN-induced cyclic GMP accumulation, but had no effect on L-IIDN-induced cyclic GMP accumulation. 5. In the intact animal, 2 mg kg(-1) DPI significantly inhibited the pharmacokinetic and haemodynamic properties of D-IIDN, but had no effect L-IIDN. 6. These data suggest that the basis for the potency difference for relaxation by the two enantiomers is preferential biotransformation of D-IIDN to NO, by an enzyme that is inhibited by DPI. Given that DPI binds to and inhibits NADPH-cytochrome P450 reductase, the data are consistent with a role for the cytochromes P450-NADPH-cytochrome P450 reductase system in this enantioselective biotransformation process.


Subject(s)
Hypoglycemic Agents/pharmacology , Onium Compounds/pharmacology , Sugar Alcohols/pharmacology , Vasodilator Agents/pharmacology , Aged , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Biotransformation , Blood Pressure/drug effects , Cyclic GMP/metabolism , Glutathione Transferase/drug effects , Glutathione Transferase/metabolism , Hemodynamics/drug effects , Humans , In Vitro Techniques , Male , Muscle Relaxation/drug effects , Rats , Rats, Sprague-Dawley , Stereoisomerism , Sugar Alcohols/chemistry , Sugar Alcohols/pharmacokinetics , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacokinetics
14.
Oncol Res ; 10(4): 193-200, 1998.
Article in English | MEDLINE | ID: mdl-9778690

ABSTRACT

Synthesis of poly(gamma-glutamyl) metabolites of many antifolates, such as methotrexate (MTX), by folylpolyglutamate synthetase (FPGS) is often essential to their cytotoxic activity. FPGS expression in the MTX-sensitive human T-lymphoblastic leukemia cell line CCRF-CEM and a number of MTX-resistant sublines was previously investigated at the DNA, RNA, and activity levels. Using an FPGS peptide deduced from its cDNA sequence, a rabbit polyclonal antibody to FPGS has now been elicited, immunoaffinity purified, and used to quantitate FPGS protein expression by chemiluminescent Western immunoblot analysis. The antibody was used to determine the half-life of human FPGS protein (3.7 +/- 1.1 h) in parental CCRF-CEM cells. A subline resistant to MTX as a result of amplified dihydrofolate reductase expression shows no change in FPGS protein or activity relative to CCRF-CEM. An MTX transport-defective line, however, displays both higher FPGS protein and activity levels. For several sublines in which the only apparent mechanism of MTX resistance is decreased FPGS activity, the FPGS protein level is decreased proportionally. However, we previously showed that these sublines have the same gene copy number, restriction map, and mRNA size and levels as the parent. Evidently, in these MTX-resistant sublines the mRNA is poorly translated and/or the protein turns over more rapidly.


Subject(s)
Folic Acid Antagonists/pharmacology , Methotrexate/pharmacology , Peptide Synthases/biosynthesis , Amino Acid Sequence , Animals , Antibodies , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Half-Life , Humans , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/enzymology , Molecular Sequence Data , Peptide Fragments/immunology , Peptide Synthases/metabolism , Rabbits , Tumor Cells, Cultured
15.
Biochem Pharmacol ; 56(7): 881-93, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9774150

ABSTRACT

We reported previously that the flavoprotein inhibitor diphenyleneiodonium sulfate (DPI) irreversibly inhibited the metabolic activation of glyceryl trinitrate (GTN) in isolated aorta, possibly through inhibition of vascular NADPH-cytochrome P450 reductase (CPR). We report that the content of CPR represents 0.03 to 0.1% of aortic microsomal protein and that DPI caused a concentration- and time-dependent inhibition of purified cDNA-expressed rat liver CPR and of aortic and hepatic microsomal NADPH-cytochrome c reductase activity. Purified CPR incubated with NADPH and GTN under anaerobic, but not aerobic conditions formed the GTN metabolites glyceryl-1,3-dinitrate (1,3-GDN) and glyceryl-1,2-dinitrate (1,2-GDN). GTN biotransformation by purified CPR and by aortic and hepatic microsomes was inhibited > 90% after treatment with DPI and NADPH. DPI treatment also inhibited the production of activators of guanylyl cyclase formed by hepatic microsomes. We also tested the effect of DPI on the hemodynamic-pharmacokinetic properties of GTN in conscious rats. Pretreatment with DPI (2 mg/kg) significantly inhibited the blood pressure lowering effect of GTN and inhibited the initial appearance of 1,2-GDN (1-5 min) and the clearance of 1,3-GDN. These data suggest that the rapid initial formation of 1,2-GDN is related to mechanism-based GTN biotransformation and to enzyme systems sensitive to DPI inhibition. We conclude that vascular CPR is a site of action for the inhibition by DPI of the metabolic activation of GTN, and that vascular CPR is a novel site of GTN biotransformation that should be considered when investigating the mechanism of GTN action in vascular tissue.


Subject(s)
NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/metabolism , Nitroglycerin/pharmacokinetics , Onium Compounds/pharmacology , Animals , Aorta/enzymology , Biotransformation/drug effects , Hemodynamics/drug effects , Injections, Intravenous , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Mitochondria, Liver/drug effects , Mitochondria, Liver/enzymology , NADPH-Ferrihemoprotein Reductase , Nitric Oxide Synthase/antagonists & inhibitors , Onium Compounds/administration & dosage , Rats , Rats, Sprague-Dawley
16.
Arch Biochem Biophys ; 355(1): 109-18, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9647673

ABSTRACT

Folylpolyglutamate synthetase (FPGS) catalyzes anATP-dependent ligation reaction that results in the synthesis of poly(gamma-glutamate) metabolites of folates and some antifolates. We have synthesized and characterized the prototype of a new class of mechanism-based FPGS inhibitor in which a phosphonate moiety mimics the tetrahedral intermediate formed during the ligation reaction. This phosphonate, 4-amino-4-deoxy-10-methyl-pteroyl-L-glutamyl-gamma-[Psi¿P(O)(OH)-O¿] glutarate (4-NH2-10-CH3-Pte-L-Glu-gamma-[Psi¿P(O)(OH)-O¿]glutarate), is not a substrate for human FPGS, but is a linear, competitive inhibitor (Kis = 46 nM) with respect to methotrexate as the variable substrate. Inhibition is not time-dependent and preincubation of FPGS with this phosphonate does not increase the degree of inhibition, suggesting that it is not a slow, tight-binding inhibitor involving a time-dependent isomerization, EI --> EI*. Substructures containing the phosphonate moiety but lacking the pterin are much less inhibitory to FPGS, indicating that a significant portion of the inhibitor binding energy is derived from the pterin moiety, a feature also observed in substrate binding. 4-NH2-10-CH3-Pte-L-Glu-gamma-[Psi¿P(O)(OH)-O¿]glutarate is also an analog of a proposed tetrahedral intermediate in the reaction catalyzed by gamma-glutamyl hydrolase (gamma-GH), another enzyme of importance in controlling folate homeostasis in cells. This intermediate would arise from direct attack of H2O on the dipeptide, 4-NH2-10-CH3-Pte-L-Glu-gamma-L-Glu. The fact that 4-NH2-10-CH3-Pte-L-Glu-gamma-[Psi¿P(O)(OH)-O¿]glutarate is not an inhibitor of gamma-GH strongly suggests that hydrolysis of poly-gamma-glutamates catalyzed by gamma-GH does not involve the direct attack of water at the scissile amide bond. Methotrexate, its gamma-glutamyl dipeptide metabolite, and 4-NH2-10-CH3-Pte-L-Glu-gamma-[Psi¿P(O)(OH)-O¿]glutarate are equipotent as inhibitors of human dihydrofolate reductase (the primary target of methotrexate), but the phosphonate does not significantly inhibit another important folate-dependent enzyme, thymidylate synthase. Thus, the phosphonate moiety in this analog represents an important new lead in the development of FPGS inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Peptide Synthases/antagonists & inhibitors , Animals , Binding, Competitive , Cell Division/drug effects , Dipeptides/chemical synthesis , Dipeptides/chemistry , Dipeptides/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Humans , In Vitro Techniques , Kinetics , Molecular Structure , Peptide Synthases/chemistry , Peptide Synthases/metabolism , Rats , Thermodynamics , Tumor Cells, Cultured , gamma-Glutamyl Hydrolase/antagonists & inhibitors
17.
J Med Chem ; 41(9): 1409-16, 1998 Apr 23.
Article in English | MEDLINE | ID: mdl-9554874

ABSTRACT

The synthesis of seven 2,4-diamino-5,6,7,8-tetrahydro-7-substituted pyrido[4',3':4,5]furo[2,3-d]pyrimidines 1-6 are reported as nonclassical antifolate inhibitors of dihydrofolate reductase (DHFR) and compound 7 as a classical antifolate inhibitor of tumor cells in culture. The compounds were designed as conformationally restricted analogues of trimetrexate. The synthesis was accomplished from the cyclocondensation of 3-bromo-4-piperidone with 2, 4-diamino-6-hydroxypyrimidine to afford regiospecifically 2, 4-diamino-5,6,7,8-tetrahydropyrido[4',3':4,5]furo[2, 3-d]pyrimidine-7-hydrobromide (16). This in turn was alkylated with the appropriate benzyl halide to afford the target compounds 1-6. The classical antifolate 7 utilized 4-(chloromethyl)benzoyl-l-glutamic acid diethyl ester (17) instead of the benzyl halide for alkylation, followed by saponification to afford 7. Compounds 1-6 showed moderate inhibitory potency against DHFR from Pneumocystis carinii, Toxoplasma gondii, Mycobacterium avium, and rat liver. The classical analogue 7 was 88-fold more potent against M. avium DHFR than against rat liver DHFR. The classical analogue was also inhibitory against the growth of tumor cells, CCRF-CEM, and FaDu, in culture.


Subject(s)
Anti-Infective Agents/chemical synthesis , Antimetabolites, Antineoplastic/chemical synthesis , Folic Acid Antagonists/chemical synthesis , Glutamic Acid/analogs & derivatives , Pyrimidines/chemical synthesis , Tetrahydrofolate Dehydrogenase/metabolism , Animals , Anti-Bacterial Agents , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Cell Division/drug effects , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Glutamic Acid/chemical synthesis , Glutamic Acid/chemistry , Glutamic Acid/pharmacology , Humans , Liver/enzymology , Methotrexate/pharmacology , Mycobacterium avium/enzymology , Pneumocystis/enzymology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Structure-Activity Relationship , Toxoplasma/enzymology , Tumor Cells, Cultured
18.
Biochem Pharmacol ; 53(5): 689-96, 1997 Mar 07.
Article in English | MEDLINE | ID: mdl-9113088

ABSTRACT

Repeated (10x) exposure of HCT-8 human ileocecal carcinoma cells to 5-fluorouracil (5-FU) for 2 or 72 hr, both incubations in the continuous presence of 20 microM leucovorin (LV), yielded two stable modulation-resistant sublines, FL2h and FL72h. Although LV potentiated growth inhibition by 5-FU 2-fold in parental HCT-8 cells, it did not potentiate the effect of 5-FU in the FL2h or FL72h sublines. LV modulation of 5-fluorodeoxyuridine (5-FdUrd) was also reduced (FL72h) or eliminated (FL2h). In the FL2h and FL72h sublines, the level of thymidylate synthase (TS) protein and TS activity in cell extracts, TS activity in situ, the rate of cellular uptake and metabolism of LV, and the level of 5-FU incorporation into total cellular RNA were similar to those in parental HCT-8 cells. However, LV significantly (P < 0.01) potentiated the inhibition of TS activity in situ in HCT-8 cells at 24 hr after a 2-hr treatment with either 5-FU or 5-FdUrd, but had no such activity in the FL2h and FL72h sublines (P > 0.1). Resistance to modulation of 5-FU by LV was associated with the inability of LV to increase the formation of intracellular TS-FdUMP-methylenetetrahydrofolate ternary complexes, and these complexes dissociated more rapidly (T1/2 > 1.5- to 3-fold faster) in the presence of different concentrations of 5,10-methylenetetrahydropteroylpentaglutamate. Thus, decreased stability of ternary complexes appears to be the mechanism of acquired resistance to the LV modulation of fluoropyrimidine cytotoxicity, possibly due to mutation(s) of TS in these two modulation-resistant HCT-8 sublines.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Fluorouracil/pharmacology , Leucovorin/pharmacology , Drug Resistance , Floxuridine/pharmacology , Humans , Leucovorin/pharmacokinetics , Thymidylate Synthase/metabolism , Tumor Cells, Cultured
19.
Oncol Res ; 9(3): 139-47, 1997.
Article in English | MEDLINE | ID: mdl-9220499

ABSTRACT

Thymidylate synthase (TS) inhibitor effects on growth of human head and neck squamous cell carcinoma (HNSCC) cell lines and CCRF-CEM human leukemia cells and sublines with acquired methotrexate (2,4-diamino-10-methylpteroylglutamic acid) (MTX) resistance were studied. During 120-h treatment, HNSCC cell lines A253 and FaDu are equally sensitive to MTX, whereas the polyglutamylatable TS inhibitors ZD1694 and BW1843U89 are 5- to 35-fold more potent than MTX and the lipophilic AG331 is approximately 10(2)-fold less potent than MTX. A253 is intrinsically resistant to intermittent (24 h) MTX and BW1843U89 exposure (higher EC50 values and shallower slopes of concentration-response curves relative to FaDu); AG331 and ZD1694 largely overcome this intrinsic resistance to intermittent exposure. Thymidine (TdR) protects against growth inhibition by these inhibitors, confirming that TS is their target in HNSCC; at high AG331 levels, TdR only partially protects, implying that a second site of action exists. Growth inhibition of HNSCC by ZD1694 and BW1843U89 is protected by leucovorin (LV) at > or = 10(-7) and > 10(-3) M, respectively; 10(-4) M LV cannot protect HNSCC cells against AG331. Results similar to protection studies are obtained if LV addition is delayed < or = 24 h after ZD1694 or BW1843U89 exposure. CCRF-CEM sublines with acquired MTX resistance resulting from dihydrofolate reductase (DHFR) overexpression, defective MTX transport, or defective MTX polyglutamylation retain full sensitivity to AG331. Cells with defective MTX transport are highly cross-resistant to ZD1694 and BW1843U89, implicating the reduced folate/MTX carrier in their transport. Minor cross-resistance of the DHFR overexpressing line to ZD1694 and BW1843U89 is observed. A subline with highly defective MTX polyglutamylation is cross-resistant to 120-h exposure to ZD1694, but not to BW1843U89, suggesting a profound contribution of polyglutamylation to the mechanism of action of ZD1694.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/therapeutic use , Head and Neck Neoplasms/drug therapy , Leukemia/drug therapy , Thymidylate Synthase/antagonists & inhibitors , Antimetabolites, Antineoplastic/pharmacology , Cell Division/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Growth Inhibitors/pharmacology , Head and Neck Neoplasms/enzymology , Humans , Leucovorin/pharmacology , Leukemia/enzymology , Methotrexate/pharmacology , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...