Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Semin Oncol ; 26(2 Suppl 6): 48-54, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10598555

ABSTRACT

The mechanism of action of an antifolate may be investigated using a variety of experimental methods. These include experiments in a cell culture setting to observe possible protection against drug effects afforded by the end products of metabolic pathways, assessing the activity of purified target enzymes in the presence of the antifolate, and, finally, the measurement of drug effects on intracellular folate and nucleoside triphosphate pools. The current discussion is focused on studies using CCRF-CEM leukemia cells that were designed to compare and contrast mechanisms of action of the antifolates methotrexate, which is primarily a dihydrofolate reductase inhibitor, raltitrexed, a thymidylate synthase inhibitor, LY309887, a glycinamide ribonucleotide formyltransferase inhibitor, and MTA (multitargeted antifolate), which is a novel antifolate antimetabolite. The results of these studies support the hypothesis that MTA affects multiple enzymatic targets and has a distinct mechanism of action from methotrexate, raltitrexed, and LY309887.


Subject(s)
Acid Anhydride Hydrolases/drug effects , Antimetabolites, Antineoplastic/pharmacology , Deoxyribonucleotides/metabolism , Enzyme Inhibitors/pharmacology , Folic Acid Antagonists/pharmacology , Glutamates/pharmacology , Guanine/analogs & derivatives , Thymidylate Synthase/antagonists & inhibitors , Thymine Nucleotides/metabolism , Acid Anhydride Hydrolases/metabolism , Folic Acid/drug effects , Folic Acid/metabolism , Guanine/pharmacology , Humans , Methotrexate/pharmacology , Nucleoside-Diphosphate Kinase/metabolism , Nucleoside-Triphosphatase , Pemetrexed , Quinazolines/pharmacology , Ribonucleotides/metabolism , Tetrahydrofolates/pharmacology , Thiophenes/pharmacology , Tumor Cells, Cultured
3.
Semin Oncol ; 26(2 Suppl 6): 68-73, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10598558

ABSTRACT

Prior studies have indicated that MTA requires intracellular polyglutamation for optimal cytotoxic effect and that these polyglutamates potently inhibit several key enzymes of folate metabolism, including thymidylate synthase (TS), dihydrofolate reductase, and glycinamide ribonucleotide formyltransferase (GARFT). In the present studies, we have investigated the mechanistic basis for resistance to MTA in several human tumor cell lines. The cell lines were developed for resistance by the gradual exposure to stepwise (fivefold) increases in the concentration of MTA over a 5-month period. The degree of resistance was 140-fold for GC3 colon carcinoma, 117-fold for HCT-8 ileocecal carcinoma, and 729-fold for CCRF-CEM leukemia cells adapted to 2 micromol/L MTA. The lines had strong cross-resistance (>3,200-fold) to raltitrexed. Only modest resistance was noted for methotrexate and the GARFT inhibitor, LY309887. The cytotoxicity of MTA in wild-type cells was only partially alleviated by thymidine addition (5 micromol/L) and complete protection required the addition of both hypoxanthine (100 micromol/L) and thymidine. In contrast, thymidine alone totally lacked protective activity in the MTA-resistant lines. The cells either demonstrated a GARFT-like reversal pattern (complete protection by hypoxanthine) for GC3MTA or a dihydrofolate reductase-like reversal pattern (complete protection by the combination of hypoxanthine and thymidine) for HCT-8MTA and CCRF-CEM(MTA) cells. Cellular resistance was multifactorial and stable on removal of selective pressure. Only GC3MTA cells showed increased TS activity (approximately 40-fold). Accumulations of 3H-MTA at 24 hours in CCRF-CEM(MTA), HCT-8MTA, and GC3MTA cells were 2%, 6%, and 46% of wild-type values, respectively. We also evaluated the cytotoxic activity of MTA in MCF-7 breast carcinoma and H630 colon carcinoma cells selected for resistance to raltitrexed and 5-fluorouracil, respectively, via TS amplification (provided by Dr P.G. Johnston, Belfast, Ireland). These cells demonstrated more than 200-fold less resistance to MTA compared with raltitrexed and MTA-induced cytotoxicity was prevented by hypoxanthine. These studies suggest that in addition to TS modulation, secondary targets emerge during the development of MTA resistance.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Folic Acid Antagonists/pharmacology , Glutamates/pharmacology , Guanine/analogs & derivatives , Thymidylate Synthase/antagonists & inhibitors , Antimetabolites, Antineoplastic/metabolism , Biological Transport , Drug Screening Assays, Antitumor , Enzyme Inhibitors/metabolism , Folic Acid Antagonists/metabolism , Glutamates/metabolism , Guanine/metabolism , Guanine/pharmacology , Humans , Pemetrexed , Quinazolines/pharmacology , Thiophenes/pharmacology , Tumor Cells, Cultured
4.
Br J Cancer ; 78 Suppl 3: 27-34, 1998.
Article in English | MEDLINE | ID: mdl-9717988

ABSTRACT

LY231514 (N-[4-[2-(2-amino-3,4-dihydro-4-oxo-7H-pyrrolo[2,3-d]pyrimidin-5-yl)ethy l]-benzoyl]-L-glutamic acid) is a new folate-based antimetabolite currently in broad phase II clinical evaluation. Previous in vitro studies (C. Shih et al, CancerRes 57: 1116-1123, 1997) have suggested that LY231514 could be a multitargeted antifolate (MTA) capable of inhibiting thymidylate synthase (TS), dihydrofolate reductase (DHFR) and glycinamide ribonucleotide formyltransferase (GARFT). The present study compared LY231514 with methotrexate, raltitrexed and a glycinamide ribonucleotide formyltransferase inhibitor, LY309887, at 300, 100, 30 and 100 nM, respectively, for their effects on intracellular folate and at 100, 66, 20 and 30 nM respectively, for their effects on nucleoside triphosphate pools in CCRF-CEM cells. Methotrexate induced an accumulation of dihydrofolate species, together with a rapid depletion of ATP, GTP and all of the deoxynucleoside triphosphates. LY309887 caused an accumulation of 10-formyltetrahydrofolate, a rapid loss of ATP, GTP and dATP, but a slower loss in dCTP, dTTP and dGTP. Both LY231514 and raltitrexed had minimal effects on folate pools. In contrast, they caused rapid depletion of dTTP, dCTP and dGTP, but induced an accumulation of dATP at different rates, with raltitrexed doing so about 2.5 times faster. Most of the observed metabolic changes could be understood on the basis of current knowledge of folate and nucleotide metabolism. We concluded that LY231514 was distinct from methotrexate, LY309887 and raltitrexed based on their metabolic effects in CCRF-CEM cells, and that in this cell line the inhibitory effects of LY231514 were exerted primarily against the thymidylate cycle and secondarily against de novo purine biosynthesis.


Subject(s)
Antineoplastic Agents/pharmacology , Folic Acid/metabolism , Glutamates/pharmacology , Guanine/analogs & derivatives , Nucleotides/metabolism , Antimetabolites, Antineoplastic/pharmacology , Chromatography, High Pressure Liquid , Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacology , Guanine/pharmacology , Humans , Hydroxymethyl and Formyl Transferases/antagonists & inhibitors , Methotrexate/pharmacology , Pemetrexed , Phosphoribosylglycinamide Formyltransferase , Quinazolines/pharmacology , Reproducibility of Results , Tetrahydrofolate Dehydrogenase/drug effects , Tetrahydrofolate Dehydrogenase/metabolism , Tetrahydrofolates/pharmacology , Thiophenes/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Tumor Cells, Cultured
5.
Cancer Res ; 57(6): 1116-23, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9067281

ABSTRACT

N-[4-[2-(2-amino-3,4-dihydro-4-oxo-7H-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl ]-benzoyl]-L-glutamic acid (LY231514) is a novel pyrrolo[2,3-d]pyrimidine-based antifolate currently undergoing extensive Phase II clinical trials. Previous studies have established that LY231514 and its synthetic gamma-polyglutamates (glu3 and glu5) exert potent inhibition against thymidylate synthase (TS). We now report that LY231514 and its polyglutamates also markedly inhibit other key folate-requiring enzymes, including dihydrofolate reductase (DHFR) and glycinamide ribonucleotide formyltransferase (GARFT). For example, the Ki values of the pentaglutamate of LY231514 are 1.3, 7.2, and 65 nM for inhibition against TS, DHFR, and GARFT, respectively. In contrast, although a similar high level of inhibitory potency was observed for the parent monoglutamate against DHFR (7.0 nM), the inhibition constants (Ki) for the parent monoglutamate are significantly weaker for TS (109 nM) and GARFT (9,300 nM). The effects of LY231514 and its polyglutamates on aminoimidazole carboxamide ribonucleotide formyltransferase, 5,10-methylenetetrahydrofolate dehydrogenase, and 10-formyltetrahydrofolate synthetase were also evaluated. The end product reversal studies conducted in human cell lines further support the concept that multiple enzyme-inhibitory mechanisms are involved in cytotoxicity. The reversal pattern of LY231514 suggests that although TS may be a major site of action for LY231514 at concentrations near the IC50, higher concentrations can lead to inhibition of DHFR and/or other enzymes along the purine de novo pathway. Studies with mutant cell lines demonstrated that LY231514 requires polyglutamation and transport via the reduced folate carrier for cytotoxic potency. Therefore, our data suggest that LY231514 is a novel classical antifolate, the antitumor activity of which may result from simultaneous and multiple inhibition of several key folate-requiring enzymes via its polyglutamated metabolites.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Folic Acid Antagonists/pharmacology , Glutamates/pharmacology , Guanine/analogs & derivatives , Hydroxymethyl and Formyl Transferases , Tetrahydrofolate Dehydrogenase/drug effects , 5,10-Methylenetetrahydrofolate Reductase (FADH2) , Acyltransferases/antagonists & inhibitors , Aminohydrolases/antagonists & inhibitors , Formate-Tetrahydrofolate Ligase/antagonists & inhibitors , Glutamates/chemistry , Guanine/chemistry , Guanine/pharmacology , Humans , Methotrexate/pharmacology , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Methylenetetrahydrofolate Reductase (NADPH2) , Molecular Structure , Multienzyme Complexes/antagonists & inhibitors , Oxidoreductases/antagonists & inhibitors , Pemetrexed , Phosphoribosylaminoimidazolecarboxamide Formyltransferase , Phosphoribosylglycinamide Formyltransferase , Polyglutamic Acid/pharmacology , Quinazolines/pharmacology , Tetrahydrofolates/pharmacology , Thiophenes/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Tumor Cells, Cultured/drug effects
6.
J Med Chem ; 40(6): 1018-25, 1997 Mar 14.
Article in English | MEDLINE | ID: mdl-9083492

ABSTRACT

A series of sulfonimidamide analogs of the oncolytic diarylsulfonylureas was synthesized and evaluated for (1) in vitro cytotoxicity against CEM cells, (2) in vivo antitumor activity against subaxillary implanted 6C3HED lymphosarcoma, and (3) metabolic breakdown to the o-sulfate of p-chloroaniline. The separated enantiomers of one sulfonimidamide analog displayed very different activities in the in vivo screening model. In general, several analogs demonstrated excellent growth inhibitory activity in the 6C3HED model when dosed orally or intraperitoneally. A correlative structure-activity relationship to the oncolytic sulfonylureas was not apparent.


Subject(s)
Antineoplastic Agents/pharmacology , Phenylurea Compounds/pharmacology , Sulfonylurea Compounds/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Division/drug effects , Drug Screening Assays, Antitumor , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C3H , Molecular Structure , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/metabolism , Sarcoma, Experimental/drug therapy , Sulfonylurea Compounds/chemical synthesis , Sulfonylurea Compounds/chemistry , Sulfonylurea Compounds/metabolism , Tumor Cells, Cultured
7.
Cancer Res ; 56(10): 2331-5, 1996 May 15.
Article in English | MEDLINE | ID: mdl-8625328

ABSTRACT

Recent clinical trials with lometrexol [(6R)-5,10-dideazatetrahydrofolate] have revealed a level of toxicity in humans that was not predicted on the basis of previous in vivo preclinical studies. Because standard laboratory animal diets contain high levels of folic acid relative to human folate intake, the toxicity and therapeutic activity of lometrexol was studied in mice under conditions of restricted dietary folate intake. Remarkably, the lethality of this drug increased by three orders of magnitude in mildly folate-deficient mice, mimicking the unexpected toxicity seen in humans. Lometrexol had limited therapeutic activity in folate-deficient mice bearing the C3H mammary adenocarcinoma, compared with the substantial therapeutic index for treatment of this tumor in animals on standard diet. When folic acid was administered p.o. to mice that were mildly folate deficient, antitumor activity was again observed at nontoxic doses of lometrexol, and the range of lometrexol doses that allowed safe therapeutic use of this drug increased at higher dietary folate intake. At a fixed dose of lometrexol, the antitumor effects in animals were dependent on the level of dietary folate and went through a distinct optimum. Excessively high folate intake reversed the antitumor effects of lometrexol. Optimization of the folic acid content in the diet and of the lometrexol dosage are predicted to have substantial impact on the clinical activity of this class of drugs.


Subject(s)
Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/therapeutic use , Folic Acid Antagonists/therapeutic use , Folic Acid/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Tetrahydrofolates/therapeutic use , Administration, Oral , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Dogs , Drug Screening Assays, Antitumor , Drug Synergism , Female , Folic Acid/administration & dosage , Folic Acid/pharmacology , Folic Acid Antagonists/administration & dosage , Folic Acid Antagonists/pharmacology , Humans , Mice , Mice, Inbred C3H , Tetrahydrofolates/pharmacology
8.
Semin Oncol ; 22(4 Suppl 11): 54-60, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7481846

ABSTRACT

A series of over 70 difluoropurine analogs was synthesized by varying the C-2, 6 and 8 substituents about the purine ring system. After initial in vitro and in vivo screening, testing concentrated on the 2,6-diaminopurine analog (dFdAP) and the guanosine analog (dFdG). dFDAP appears to be a prodrug for dFdG. Both compounds significantly inhibited mammary tumor growth in mice, caused a moderate inhibition in ovarian and lymphosarcoma models, and demonstrated no activity in lung and melanoma models. This is a narrower spectrum of activity than that of gemcitabine (dFdC). The antitumor activity of dFdAP in human xenografts that are refractory to standard clinical agents was comparable or superior to that of gemcitabine. However, during the preliminary toxicology testing, dFdG was associated with several deaths caused by cardiac toxicity. Therefore, although dFdG is a potentially useful oncolytic, further investigation is required.


Subject(s)
2-Aminopurine/analogs & derivatives , Antineoplastic Agents/pharmacology , Deoxyadenosines/chemistry , Deoxyadenosines/pharmacology , Deoxyguanosine/analogs & derivatives , Guanosine/analogs & derivatives , Neoplasms, Experimental/drug therapy , 2-Aminopurine/chemistry , 2-Aminopurine/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Blood Cell Count/drug effects , Blood Pressure/drug effects , Deoxyadenosines/chemical synthesis , Deoxyguanosine/chemical synthesis , Deoxyguanosine/chemistry , Deoxyguanosine/pharmacology , Dogs , Drug Screening Assays, Antitumor , Female , Heart/drug effects , Humans , In Vitro Techniques , Infusions, Intravenous , Injections, Intraperitoneal , Liver/drug effects , Lymphoid Tissue/drug effects , Male , Mice , Ovarian Neoplasms/drug therapy , Structure-Activity Relationship , Testis/drug effects
9.
J Pharm Sci ; 82(7): 683-8, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8360840

ABSTRACT

The absorption and pharmacokinetics of sulofenur [N-(indan-5-sulfonyl)-N'-(4-chlorophenyl)urea, LY186641] and its major metabolites were examined in mice, rats, monkeys, and dogs. The compound is a diarylsulfonylurea currently being evaluated as an oncolytic agent in phase I and II trials. In all species, sulofenur was well absorbed after an oral dose, but over a prolonged period, and sulofenur exhibited a fairly long half-life of elimination from plasma. These values ranged from 6 h in rats up to 30, 110, and 200 h in mice, monkeys, and dogs, respectively, at doses (240-1000 mg/m2) within the range of those used in clinical trials. Experiments describing the high degree of binding of sulofenur to plasma proteins (consistently > 99%) help to explain these relatively long half-lives. There is, however, a large difference between these plasma half-lives in the species studied. Sulofenur was previously found to be extensively metabolized to products that are excreted primarily into the urine. In this study, its major metabolites, which are found mainly in the urine, were also minor components of the drug-related material (< 10% of the sulofenur concentrations) in the plasma of rats treated with sulofenur. The absorption, binding characteristics, and elimination of these major metabolites after their administration to rats were also compared with sulofenur.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antineoplastic Agents/pharmacokinetics , Sulfonylurea Compounds/pharmacokinetics , Animals , Blood Proteins/metabolism , Dogs , Female , Half-Life , Macaca mulatta , Mice , Mice, Inbred C3H , Protein Binding , Rats , Rats, Inbred F344 , Species Specificity
10.
Chem Res Toxicol ; 5(5): 667-73, 1992.
Article in English | MEDLINE | ID: mdl-1446007

ABSTRACT

The metabolic formation of p-chloroaniline from the oncolytic agent sulofenur [N-(5-indanesulfonyl)-N'-(4-chlorophenyl)urea, LY186641,] and from similar diaryl-substituted sulfonylureas, and its possible relevance to the compound's toxicity, was studied. In previous studies it was found that significant amounts of metabolites such as 2-amino-5-chlorophenyl sulfate (II), which is also a metabolite of p-chloroaniline, are formed from sulofenur in mice, rats, monkeys, and humans. The metabolism of N-(4-tolyl)-N'-(2-hydroxy-4-chlorophenyl)-urea (V) was studied, and V was not found to be an intermediate in the metabolic formation of II from the sulfonylurea N-(4-tolyl)-N'-(4-chlorophenyl)urea (LY181984, III). The amounts of this p-chloroaniline metabolite (II) formed in C3H mice from a series of diarylsulfonylureas were found to correlate with the compound's propensities to form methemoglobin, one notable toxicity of p-chloroaniline. This metabolism was also found to correlate with the structure of the arylsulfonyl moiety of the sulfonylurea. Other evidence supports the hypothesis that p-chloroaniline is directly formed by metabolism of sulfofenur and similar diarylsulfonylureas as well. Metabolic formation of p-chloroaniline thus appears to be a plausible explanation for the methemoglobinemia and anemia found to be dose-limiting toxicities of sulofenur in Phase I trials.


Subject(s)
Aniline Compounds/metabolism , Antineoplastic Agents/toxicity , Sulfonylurea Compounds/toxicity , Animals , Antineoplastic Agents/metabolism , Female , Magnetic Resonance Spectroscopy , Mass Spectrometry , Methemoglobin/analysis , Mice , Mice, Inbred C3H , Structure-Activity Relationship , Sulfonylurea Compounds/chemistry , Sulfonylurea Compounds/metabolism
11.
Invest New Drugs ; 8(3): 241-51, 1990 Aug.
Article in English | MEDLINE | ID: mdl-2272765

ABSTRACT

Data collection for transplantable solid tumors has been automated with electronic digital calipers and a balance which have been coupled through an RS-232 interface to a microcomputer. BASIC programs handle data entry, calculations and data storage. A "PROTOCOL" program accepts keyboard input of sample name, notebook number, submitter and dose along with necessary information on tumor system, and then initial animal weights for treatment groups are sent from balance to computer. Data is stored as an ASCII file on floppy disks, and protocol reports are printed. When the test is to be measured, a "MEASURE" program prompts the user for keyboard entry of toxic deaths in each group. Then the computer requests input of width and length of tumors for each animal. These tumor dimensions are sent to microcomputer by pressing a button on the calipers. When a group is completed, final animal weights are sent from balance to microcomputer. Then tumor weights and percent inhibition as compared to appropriate control groups are calculated, and the data is appended to the file for that test. A hard copy is generated as tumors are measured, and reports including percent inhibition can be printed immediately after a test is measured. The data as an ASCII file is transferred via modem to mainframe computer, where another program transfers the information to a database management program. These automated procedures for tumor measurement save time and lessen the chance for error by eliminating manual recording of solid tumor dimensions and subsequent reentry of this data for calculation.


Subject(s)
Drug Screening Assays, Antitumor/methods , Microcomputers , Neoplasm Staging/methods , Software , Animals , Computer Systems , Database Management Systems , Mice , Neoplasm Staging/instrumentation , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...