Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Res ; 48(24 Pt 1): 6986-91, 1988 Dec 15.
Article in English | MEDLINE | ID: mdl-2973370

ABSTRACT

The purpose of this study was to characterize the transport properties of trimetrexate in WI-L2 human lymphoblastoid cells and determine the mode of resistance that had developed in a subline, WI-L2/TMQ, that was grown in increasing concentrations of trimetrexate. WI-L2/TMQ cells were 62-fold resistant to trimetrexate and 68- and 96-fold cross-resistant to the other lipophilic antifolates metoprine and piritrexim (BW 301U). No cross-resistance was observed with vincristine or doxorubicin, and sensitivity was not increased with 5 micrograms/ml of verapamil, indicating that it was not a typical multidrug resistance phenotype. WI-L2/TMQ exhibited a 2-fold increase in dihydrofolate reductase; however, this did not contribute significantly to the observed resistance, since these cells retained full sensitivity to methotrexate. Nor were there any kinetic alterations in dihydrofolate reductase toward trimetrexate or differences in the levels of thymidylate synthase. The major difference between the sensitive and resistant cell line was a 50% decrease in the influx rate of WI-L2/TMQ cells which produced a corresponding decrease in cellular trimetrexate at the steady state. No difference in efflux rates was detected nor were there any differences in intracellular water or metabolism of trimetrexate. Additional characterization of trimetrexate transport in WI-L2 showed that influx was nonsaturable up to 5 mM extracellular trimetrexate, relatively insensitive to sodium azide, and exhibited a Q10 of 2.7. Influx was, however, inhibited in a dose-dependent manner by concentrations of p-chloromercuribenzylsulfonate above 10 microM. Efflux studies revealed a large nonexchangeable fraction of trimetrexate that was well above the dihydrofolate reductase binding capacity and varied depending on the initial level of cell-associated drug. The intracellular exchangeable trimetrexate concentration at the steady state was always several-fold higher than the extracellular concentration. Retention of trimetrexate appeared to be coupled to some component of energy metabolism, since the presence of sodium azide stimulated this process by 2- to 3-fold. The data suggest that trimetrexate enters cells by passive diffusion but then is distributed and concentrated within the cell through more complex mechanisms which may involve energy coupling, compartmentation, or binding to macromolecules or organelles, although some type of carrier-mediated process cannot be ruled out.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Folic Acid Antagonists/pharmacology , Lymphocytes/metabolism , Quinazolines/pharmacokinetics , Antimetabolites, Antineoplastic/pharmacology , Azides/pharmacology , Cell Line , Dose-Response Relationship, Drug , Drug Resistance , Humans , Lymphocytes/drug effects , Pyrimidines/pharmacology , Sodium Azide , Trimetrexate
2.
Biochem Pharmacol ; 37(21): 4063-8, 1988 Nov 01.
Article in English | MEDLINE | ID: mdl-2847752

ABSTRACT

Fostriecin is a new antitumor antibiotic which is being developed further as an anticancer agent based on its marked activity in murine leukemias. Its mechanism of action, however, has thus far remained unknown. The present study demonstrates that fostriecin inhibits the catalytic activity of partially purified type II topoisomerase from Ehrlich ascites carcinoma. Under the experimental conditions employed, fostriecin completely inhibited the enzyme at 100 microM. A general kinetic analysis showed that fostriecin inhibited topoisomerase in an uncompetitive manner with a Ki,app of 110 microM and produced kinetics that were distinctly different from those of VM-26 which exhibited noncompetitive inhibition. Fostriecin did not cause DNA strand breaks in L1210 cells, suggesting that it did not stabilize a cleavable complex as do other known inhibitors of this enzyme. Fostriecin, however, did partially inhibit DNA strand breaks produced by amsacrine. An analysis by flow cytometry showed that L1210 cells exposed to 5 microM fostriecin for 12 hr caused a block in the G2 phase of the cell cycle. These studies thus suggest that the mechanism by which fostriecin produces its antitumor effects may be through inhibition of topoisomerase II and that the type of inhibition is markedly different from existing antitumor agents which inhibit this enzyme.


Subject(s)
Topoisomerase II Inhibitors , Adenosine Triphosphate/metabolism , Alkenes/pharmacology , Animals , Cell Cycle/drug effects , Cell Line , DNA Damage , DNA, Mitochondrial/ultrastructure , In Vitro Techniques , Mice , Polyenes , Pyrones
3.
Mol Pharmacol ; 33(1): 84-92, 1988 Jan.
Article in English | MEDLINE | ID: mdl-2447481

ABSTRACT

Cl-958, PD 121373, and PD 114595 belong to a new class of DNA complexers, substituted 2H-[1]benzothiopyrano[4,3,2-cd] indazoles, and are being further developed as antitumor drugs based on their curative properties against murine solid tumor models. The biochemical effects of these drugs on L1210 leukemia cells and their interaction with DNA were studied and compared to clinically used intercalators. The benzothiopyranoindazoles bound to DNA with a relatively high affinity, having intrinsic association constants of between 3 and 4 x 10(5) M-1. Based on viscosity measurements, the mode of DNA binding appears to be through intercalation. Unwinding angles were calculated to be approximately 18 degrees. The benzothiopyranoindazoles were potent inhibitors of nucleic acid synthesis, reducing both DNA and RNA synthesis to the same extent at similar concentrations. Like other known intercalators, these compounds produced DNA single- and double-strand breaks in a time- and concentration-dependent manner in L1210 cells. Between one and two DNA strand breaks were formed per protein-strand crosslink. Repair of these DNA lesions after the drug was removed from the cells was either very slow or did not occur at all for at least 2 hr. Finally, since the high incidence of cardiotoxicity associated with the administration of anthracyclines has been related to the formation of reactive oxygen species, the ability of the benzothiopyranoindazoles to augment superoxide dismutase-sensitive oxygen consumption was observed in a rat liver microsomal system. These compounds produced less than 5% of the activity in this assay that doxorubicin produced.


Subject(s)
Antineoplastic Agents , DNA Damage , DNA , Indazoles/metabolism , Intercalating Agents , Pyrazoles/metabolism , Animals , DNA/biosynthesis , DNA Repair , Free Radicals , Indazoles/pharmacology , Leukemia L1210 , RNA/biosynthesis , Spectrum Analysis , Superoxides/metabolism , Tumor Cells, Cultured/drug effects
4.
Adv Enzyme Regul ; 26: 301-16, 1987.
Article in English | MEDLINE | ID: mdl-3673707

ABSTRACT

1. Guanine-7-oxide is a novel purine antibiotic produced by a Streptomyces species, ATCC 39364. 2. Guanine-7-oxide is cytotoxic to murine and human leukemia cells in vitro at sub-micromolar concentrations. Murine and human carcinoma cells are much less sensitive. 3. Guanine-7-oxide has significant in vivo antitumor activity, particularly against the intraperitoneal and subcutaneous L1210 leukemia systems. 4. Guanine-7-oxide, at highly cytotoxic concentrations, has little effect on biosynthesis of RNA and DNA. 5. There is preliminary evidence for an early effect of guanine-7-oxide on cellular protein synthesis. 6. Guanine, guanosine and hypoxanthine protect cells from the cytotoxicity of guanine-7-oxide. 7. Activation of guanine-7-oxide requires the presence of the enzyme hypoxanthine-guanine phosphoribosyltransferase in the target cells. 8. Cytotoxic concentrations of guanine-7-oxide do not cause depletion of cellular guanine nucleotides during a two hr incubation period. 9. Guanine-7-oxide is converted within mouse and human cells to a metabolite with chromatographic mobility corresponding to a ribonucleoside 5'-triphosphate.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Guanine/analogs & derivatives , Neoplasms, Experimental/drug therapy , Animals , Cell Line , Cell Survival/drug effects , Female , Guanine/therapeutic use , Humans , Leukemia L1210/metabolism , Leukemia, Experimental/drug therapy , Mice , Neoplasm Proteins/biosynthesis , Neoplasms, Experimental/pathology , Ribonucleotides/analysis
5.
Cancer Lett ; 31(3): 253-60, 1986 Jun.
Article in English | MEDLINE | ID: mdl-2941142

ABSTRACT

Several antifolate compounds were examined for their cytotoxic activity in a pleiotropically resistant P388 cell line (P388R). The sensitivity of P388R cells to methotrexate (MTX) and the lipophilic antifols, metoprine and methotrexate gamma-mono t-butyl ester (MTX-gamma-t-butyl ester) were comparable with that activity observed in the parental cell line (P388S). P388R cells were, however, resistant to 2 other lipophilic antifols, trimetrexate (TMQ) and BW 301U. The degree of resistance to TMQ and BW 301U was 22-fold and 15-fold, respectively and could be partially overcome by the calcium channel blocker, verapamil (VER) or the detergent Tween 80. Transport studies showed that net accumulation of trimetrexate was markedly reduced in P388R cells resulting in a steady-state level which was 25% of the sensitive line. This impaired uptake was reversed by 5 micrograms/ml VER which increased the steady-state to a level comparable to P388S. P388R also exhibited a 50% reduction in the unindirectional influx rate, however, this defect could not be reversed by VER. The resistance of P388R cells to TMQ and BW 301U and their potentiation by VER extends pleiotropic resistance to yet another class of drugs which have important clinical implications.


Subject(s)
Folic Acid Antagonists/therapeutic use , Pyrimidines/therapeutic use , Quinazolines/therapeutic use , Animals , Drug Resistance , Leukemia P388/drug therapy , Leukemia P388/pathology , Methotrexate/therapeutic use , Mice , Trimetrexate
6.
Biochem Pharmacol ; 34(19): 3499-508, 1985 Oct 01.
Article in English | MEDLINE | ID: mdl-2413861

ABSTRACT

CI-937 and CI-942 belong to a new class of DNA complexers, the anthra[1,9-cd]pyrazol-6(2H)-ones (anthrapyrazoles), and are being further developed as antitumor drugs based on their curative properties against murine solid tumour models. The biochemical effects of these agents were studied in L1210 leukemia in relation to other clinically used intercalators. After a 1-hr exposure, CI-937 and CI-942 reduced the cloning efficiency of L1210 cells by 50% at 3.0 X 10(-8) and 1.5 X 10(-7) M respectively. Based on an ethidium displacement assay, these drugs bound strongly to DNA, reducing the fluorescence of an ethidium-DNA complex by 50% at concentrations of 23 and 33 nM for CI-937 and CI-942 respectively. This was comparable to mitoxantrone at 15 nM, but much more potent than Amsacrine which required over 1.3 microM. A distinct property of the anthrapyrazoles was a much more potent inhibitory effect on whole cell DNA synthesis than on RNA synthesis. After L1210 cells were exposed to drug for 2 hr the concentration needed to inhibit DNA synthesis by 50% was 0.33 and 0.57 microM for CI-937 and CI-942, respectively, whereas 2.0 and 11.3 microM were required to inhibit RNA synthesis by the same extent. This was in contrast to Adriamycin and mitoxantrone which inhibited both activities equally at similar concentrations. It was apparent that the inhibition of these processes was not due to substrate depletion since intracellular ribonucleoside and deoxyribonucleoside triphosphates either remained constant or were elevated after a 2-hr exposure to 1 or 10 microM drug. A similar discriminatory effect was observed on DNA and RNA polymerase in permeabilized cells, and the inhibition of nucleic acid synthesis in this system could be reversed by exogenously added DNA. Since the high incidence of cardiotoxicity associated with the administration of anthracyclines has been related to the formation of reactive oxygen species, the ability of the anthrapyrazoles to augment superoxide dismutase sensitive oxygen consumption was observed in a rat liver microsomal system. CI-937 and CI-942 induced 5- and 10-fold less oxygen consumption than Adriamycin, producing rates of 12.4, 24.2 and 138.9 nmoles/min/mg microsomal protein, respectively, at a drug concentration of 0.5 mM.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Anthraquinones/therapeutic use , DNA/metabolism , Leukemia L1210/drug therapy , Nucleic Acids/biosynthesis , Pyrazoles/therapeutic use , Aminoacridines/therapeutic use , Amsacrine , Animals , Anthraquinones/metabolism , Colony-Forming Units Assay , DNA/biosynthesis , Doxorubicin/therapeutic use , Kinetics , Leukemia L1210/metabolism , Mice , Mitoxantrone , Nucleotides/metabolism , Oxygen Consumption , Pyrazoles/metabolism , RNA/biosynthesis
7.
Biochem Pharmacol ; 34(7): 1109-14, 1985 Apr 01.
Article in English | MEDLINE | ID: mdl-3985993

ABSTRACT

2-beta-D-Ribofuranosyl-4-selenazolecarboxamide (selenazofurin, CI-935), the selenium analog of tiazofurin (CI-909), was 3- to 10-fold more cytotoxic to murine or human tumor cells in vitro than tiazofurin and was also more active against P388 mouse leukemia in vivo. In vitro cytotoxicity could be reversed by guanosine or guanine but not by other purine nucleosides or bases. Three human tumor cell lines selected for selenazofurin or tiazofurin resistance showed cross resistance between selenazofurin and tiazofurin. Treatment with tiazofurin, selenazofurin, or mycophenolic acid decreased guanylate pools and caused an accumulation of IMP in WIL2 human lymphoma cells. The decrease in guanylate pools was accompanied by inhibition of RNA and DNA synthesis. The NAD analogs of tiazofurin and selenazofurin were inhibitors of L1210 IMP dehydrogenase (IMP:NAD oxidoreductase, EC 1.2.1.14), and both showed uncompetitive inhibition with respect to NAD having Kii values of 5.7 X 10(-8)M and 3.3 X 10(-8)M respectively.


Subject(s)
Antineoplastic Agents/pharmacology , Organoselenium Compounds , Ribavirin/pharmacology , Ribonucleosides/pharmacology , Selenium/pharmacology , Animals , Cells, Cultured , Humans , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Nucleic Acids/biosynthesis , Ribavirin/analogs & derivatives , Ribonucleotides/analysis
8.
Invest New Drugs ; 3(3): 223-31, 1985.
Article in English | MEDLINE | ID: mdl-4066218

ABSTRACT

3-Deazaguanine (dezaguanine, USAN; CI-908) is a new antipurine antimetabolite which is entering Phase I studies in the USA. This compound differs from guanine only in the substitution of a carbon for the 3-nitrogen of guanine. Dezaguanine has an unusual spectrum of activity against experimental rodent tumors; its activity against transplantable rodent leukemias is only modest, but it has significant activity against transplantable rodent solid tumors, particularly mammary adenocarcinomas. Mammary adenocarcinoma models against which this compound is active include slow and fast-growing tumors, hormone sensitive and hormone insensitive tumors, and the subrenal capsule implanted human breast cancer xenograft, MX-1. Dezaguanine must be converted to its nucleotides to be active. Dezaguanine nucleotides inhibit synthesis of guanine nucleotides, and can be incorporated into nucleic acids in place of guanine nucleotides; incorporation into DNA may be particularly important in the cytotoxicity of this compound. Addition of certain purines or purine nucleosides can prevent dezaguanine cytotoxicity in vitro. Preclinical studies suggest that dezaguanine does not undergo deamination to 3-deazaxanthine, and is not metabolized by xanthine oxidase. Therefore, this compound may not be subject to metabolic inactivation in vivo, and active metabolites may have a prolonged half-life. This concept is supported by the prolonged half-life of radiolabelled dezaguanine in rats. Finally, dezaguanine can cross the blood-brain barrier. In summary, the novel biochemical and experimental antitumor properties of dezaguanine indicate that this compound could have better activity against some human solid tumors than currently used purine antimetabolites.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Guanine/analogs & derivatives , Purines/antagonists & inhibitors , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Survival/drug effects , Cells, Cultured , DNA, Neoplasm/biosynthesis , Drug Evaluation , Guanine/pharmacology , Humans , Kinetics , Leukemia L1210/metabolism , Leukemia L1210/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Time Factors
9.
Cancer Res ; 44(8): 3366-70, 1984 Aug.
Article in English | MEDLINE | ID: mdl-6547636

ABSTRACT

Cl-920 is a structurally novel antitumor antibiotic which has activity against a wide spectrum of tumor cells in vitro and is curative in L1210 leukemia in vivo. Several lines of evidence indicate that this drug penetrates L1210 cells via the reduced folate carrier system. Reduced folates (100 microM) including leucovorin and 5-methyltetrahydrofolate completely protected L1210 cells from growth inhibition by Cl-920. Protective effects were not observed, however, with folic acid, a compound which is transported by a process distinct from that for reduced folates. Cl-920 was a potent inhibitor of methotrexate influx exhibiting a mixture of competitive and noncompetitive inhibition and having a Ki (slope) of 30.0 microM and a Ki (intercept) of 58.8 microM. The inhibition appeared to be irreversible since, after cells were preincubated with drug, the inhibitory effects persisted after cells were washed in drug-free media. The irreversibility could be eliminated, however, by dithiothreitol, suggesting that Cl-920 may interact with a thiol which is essential to this transport system. Cells made 71-fold resistant to Cl-920 by continuous exposure to increasing concentrations of this drug were 245-fold cross-resistant to methotrexate but were collaterally sensitive to the lipophilic antifolate trimetrexate and contained normal levels of dihydrofolate reductase. This mutant cell line had a severely impaired reduced folate carrier system exhibiting methotrexate influx rates of less than 1% of control cells. Finally, inhibition of methotrexate influx by a number of Cl-920 analogues showed that the intact lactone ring and the presence of the phosphate ester were required for maximum interaction with the carrier system and that the degree of inhibition correlated with relative antitumor potency. These observations are compatible with the concept that Cl-920 utilizes the folate carrier system and could be of fundamental importance for understanding the cytotoxicity and selectivity of Cl-920.


Subject(s)
Folic Acid/metabolism , Leukemia L1210/metabolism , Alkenes/metabolism , Animals , Biological Transport , Drug Resistance , Folic Acid/toxicity , Kinetics , Methotrexate/metabolism , Mice , Polyenes , Pyrones , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/metabolism
10.
Adv Enzyme Regul ; 22: 187-206, 1984.
Article in English | MEDLINE | ID: mdl-6236675

ABSTRACT

Trimetrexate is a novel lipophilic folate antagonist that causes growth inhibition, inhibition of nucleic acid biosynthesis, and cytotoxicity at nanomolar concentrations in tissue cultures. The potency of trimetrexate cytotoxicity against most cell lines is greater than that of methotrexate. Trimetrexate has antitumor activity in vivo in several murine leukemia and solid tumor systems, including tumors in which methotrexate is inactive. Antitumor activity was seen following oral, intravenous, or intraperitoneal administration. Trimetrexate causes a pronounced and early depression in incorporation of deoxyuridine into DNA. In tumor cell lines resistant to methotrexate because of a drug transport defect, trimetrexate retains activity. In many such cases the methotrexate-resistant tumors show collateral sensitivity to trimetrexate. In methotrexate-resistant cells with impaired drug transport, trimetrexate sensitivity was even more pronounced when cells were grown in folate-free medium supplemented with physiological levels of tetrahydrofolate cofactor. In the human tumor stem cell colony assay, trimetrexate, at concentrations achievable in vivo, gave activity against many human tumors, including samples that were unresponsive to methotrexate. Trimetrexate crosses the blood-brain barrier, and at very high doses may cause neurotoxicity. At conventional doses the primary toxic effects in mice are gastrointestinal. This toxicity is reversible at therapeutic doses. Unlike earlier lipophilic antifolates, trimetrexate has rapid plasma clearance (t1/2 in mice of 45 minutes). Trimetrexate is a tight-binding competitive inhibitor of dihydrofolate reductase. The Ki,slope for inhibition of the human enzyme was 4 X 10(-11) M. A dose-dependent decrease in cellular purine ribonucleotide pools is given by trimetrexate. Pyrimidine ribonucleotide pools tend to increase in treated cells. Trimetrexate caused a marked depression of cellular pools of dTTP and dGTP, and a lesser depression in dATP. Cytotoxicity of trimetrexate in vitro was prevented by leucovorin. Leucovorin also protected mice from trimetrexate toxicity. Thymidine protected cells from lethal effects of low concentrations of trimetrexate, but not from high concentrations. The combination of thymidine and hypoxanthine completely protected cells from low and high concentrations of trimetrexate. A new, stable and highly water-soluble formulation of trimetrexate has been developed. Because of the interesting biochemical and pharmacological properties of trimetrexate, and its experimental antitumor activity, clinical trials are planned.


Subject(s)
Folic Acid Antagonists/pharmacology , Quinazolines/pharmacology , Animals , Cell Line , Culture Techniques , Deoxyuridine/metabolism , Drug Resistance , Folic Acid/metabolism , Folic Acid Antagonists/metabolism , Humans , Kinetics , Leucovorin/pharmacology , Leukemia L1210/drug therapy , Leukemia L1210/metabolism , Lymphocytes/metabolism , Methotrexate/pharmacology , Mice , Mice, Inbred Strains , Quinazolines/metabolism , Ribonucleotides/metabolism , Thymidine/pharmacology , Trimetrexate
SELECTION OF CITATIONS
SEARCH DETAIL
...