Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Pharm Sci ; 90(11): 1859-67, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11745743

ABSTRACT

Membrane-water partitioning of inhibitors of acyl-coenzyme A:cholesterol acyltransferase (ACAT) governs the concentration of inhibitor that ACAT is exposed to and determines the corresponding extent of cholesterol esterification inhibition. Partitioning of the ACAT inhibitors CI-976, CL 277,082, and SaH 58-035 into rat liver microsomes containing ACAT was detected by shifts in the level of inhibition that were independent of inhibitor concentration but inversely dependent on microsome membrane concentration. The equilibrium distribution of the ACAT inhibitors between aqueous and membrane phases was derived directly from these data by application of a previously described method of linear analysis. The accuracy of membrane partitioning analysis based on kinetic data was verified for CI-976 by direct measurements of [14C]CI-976 partitioning into phospholipid membranes. The results show that the ACAT inhibitors are highly partitioned into membranes by factors exceeding 1 x 10(6). This result is consistent with the far greater influence of membrane content over aqueous volume on inhibitor activity. The results demonstrate that the size of the membrane phase in aqueous suspension must be taken into account to obtain accurate and reproducible kinetic characterizations of membrane-active molecules. Analyses of the membrane-dependent shifts in activity can be used to calculate the membrane-water partitioning coefficient of membrane-active molecules such as ACAT inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacokinetics , Membranes, Artificial , Phospholipids/pharmacokinetics , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Rats , Rats, Sprague-Dawley , Sterol O-Acyltransferase/antagonists & inhibitors , Sterol O-Acyltransferase/metabolism
2.
J Lipid Res ; 39(6): 1197-209, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9643351

ABSTRACT

Cholesterol absorption from bile acid micelles is suppressed by phosphatidylcholine (PC) in the micelles. The effects of micellar phospholipid composition on absorption, metabolism, and secretion of lipids were examined in Caco-2 cells incubated with micelles composed of taurocholic acid, cholesterol, oleic acid, monooleoylglycerol, and phospholipid. Significant amounts of all micelle lipids were absorbed from micelles lacking phospholipid. Cholesterol absorption was accompanied by cholesterol esterification and secretion. Micellar oleic acid was also absorbed and reesterified primarily into triacylglycerol which was also secreted. Lipid absorption and secretion from micelles containing lysophosphatidylcholine (LPC) were similar to that obtained with phospholipid-free micelles. LPC was also extensively absorbed. In contrast, incubations with PC-containing micelles resulted in large reductions in the absorption, esterification, and secretion of cholesterol without significant decreases in oleic acid absorption, conversion to acylated lipids, or triacylglycerol secretion. A relatively small reduction in monoacylglycerol absorption from PC-containing micelles was detected. Retinol absorption was not affected by micellar PC. Substitution of LPC for half or more of the PC reversed the PC-dependent decrease in cholesterol absorption. Pancreatic phospholipase A2 (pPLA2) enhanced cholesterol absorption from PC-containing micelles. The pPLA2-dependent increase in cholesterol absorption was inhibited by the pPLA2 inhibitor FPL 67047XX. The results indicate micellized cholesterol absorption by enterocytes is uniquely dependent on the elimination of micellar phosphatidylcholine and thus directly dependent on the lipolytic action of pancreatic phospholipase A2 (pPLA2). Consequently, pPLA2 inhibitors may be a new and novel class of cholesterol absorption inhibitors for therapeutic use.


Subject(s)
Cholesterol/metabolism , Intestinal Absorption/drug effects , Intestinal Mucosa/physiology , Phosphatidylcholines/metabolism , Phosphatidylcholines/pharmacology , Phospholipases A/pharmacology , Vitamin A/pharmacokinetics , Benzhydryl Compounds/pharmacology , Caco-2 Cells , Enzyme Inhibitors/pharmacology , Glycerides , Humans , Intestinal Mucosa/drug effects , Lysophosphatidylcholines/metabolism , Lysophosphatidylcholines/pharmacology , Micelles , Oleic Acid/metabolism , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Taurocholic Acid , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/pharmacology
3.
J Med Chem ; 41(5): 682-90, 1998 Feb 26.
Article in English | MEDLINE | ID: mdl-9513596

ABSTRACT

We prepared a series of alpha-substituted malonester amides that were evaluated for their ability to inhibit acyl-CoA:cholesterol O-acyl transferase activity in vitro and to lower plasma total cholesterol levels in a variety of cholesterol-fed animal models. Compounds of this series were also useful in examining the relationship between adrenal toxicity and ACAT inhibition. One compound from this series, 9f, was a potent inhibitor of ACAT in both the microsomal and cellular assays. It was also bioavailable as determined by both a bioassay and a HPLC-UV assay. This compound was evaluated in both guinea pig and dog models of adrenal toxicity and compared to tetrazole amide 15. In the most sensitive species, the dog, both of these compounds achieved good plasma levels; however, compound 9f caused adrenal necrosis, whereas compound 15 had no effect on the adrenal gland. This adds to the growing body of evidence that the adrenal toxicity observed with ACAT inhibitors may not be mechanism related.


Subject(s)
Adrenal Glands/drug effects , Adrenal Glands/pathology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Malonates/pharmacology , Malonates/toxicity , Phenylacetates/pharmacology , Phenylacetates/toxicity , Sterol O-Acyltransferase/antagonists & inhibitors , Amides/pharmacology , Amides/toxicity , Animals , Anticholesteremic Agents/chemical synthesis , Biological Availability , Cholesterol/blood , Cholesterol, Dietary/administration & dosage , Chromatography, High Pressure Liquid , Dogs , Enzyme Inhibitors/chemical synthesis , Female , Guinea Pigs , Male , Malonates/chemical synthesis , Mice , Microsomes, Liver/enzymology , Necrosis , Phenylacetates/chemical synthesis , Rats , Rats, Sprague-Dawley , Tetrazoles/pharmacology , Tetrazoles/toxicity
5.
Bioorg Med Chem ; 5(4): 739-47, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9158873

ABSTRACT

Our continued interest in developing novel, potent acyl-CoA:cholesterol acyltransferase (ACAT) inhibitors, and our discovery of several active series of disubstituted urea ACAT inhibitors, have led us to investigate a series of trisubstituted ureas that are structural hybrids of our disubstituted series and of a trisubstituted urea ACAT inhibitor series disclosed by scientists at Lederle. This investigation has led to the discovery of novel trisubstituted ureas, several of which inhibit ACAT in the nanomolar range and effectively lower total plasma cholesterol when administered as a diet admixture in an acute model of hypercholesterolemia in rats. One analogue (35) also lowered total cholesterol as efficaciously as CL 277,082 in our chronic hypercholesterolemic rat model. The most notable finding of this study is that the SAR of the trisubstituted ureas diverges from that seen in our previously disclosed disubstituted urea series. This series showed optimal activity with 2,4-difluoro and 2,4,6-trifluoro substitution on the urea N-phenyl, whereas the disubstituted series showed optimal activity with bulky 2,6-disubstitution on the phenyl ring.


Subject(s)
Anticholesteremic Agents/chemical synthesis , Sterol O-Acyltransferase/antagonists & inhibitors , Urea/analogs & derivatives , Administration, Oral , Animals , Anticholesteremic Agents/chemistry , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Benzene Derivatives/chemistry , Cholesterol/blood , Diet , Disease Models, Animal , Fluorides/chemistry , Hypercholesterolemia/drug therapy , Magnetic Resonance Spectroscopy , Rabbits , Rats , Structure-Activity Relationship , Urea/chemistry , Urea/pharmacology
7.
J Med Chem ; 39(22): 4382-95, 1996 Oct 25.
Article in English | MEDLINE | ID: mdl-8893833

ABSTRACT

A series of diaryl-substituted heterocyclic ureas was prepared, and their ability to inhibit acyl-CoA: cholesterol O-acyltransferase (ACAT) in vitro and to lower plasma total cholesterol in cholesterol-fed animal models in vivo was examined. N-(2,6-Diisopropylphenyl)-N'-tetrazole or isoxazole-substituted heterocyclic ureas proved optimal. A carbon chain of 11-14 carbons substituted 1,3 with respect to the amine provided the optimal side chain. Substitution of the alkyl chain generally lowered activity. Tetrazole urea 2i dosed at 3 mg/kg lowered plasma total cholesterol (TC) 67% in an acute, cholesterol-fed (C-fed) rat model of hypercholesterolemia and 47% in C-fed dogs. Tetrazole 2i, dosed at 10 mg/kg, also lowered TC 52% and raised HDL cholesterol 113% in rats with pre-established hypercholesterolemia.


Subject(s)
Anticholesteremic Agents/chemistry , Sterol O-Acyltransferase/antagonists & inhibitors , Urea/analogs & derivatives , Animals , Cholesterol, HDL/blood , Chromatography, High Pressure Liquid , Disease Models, Animal , Dogs , Female , Hypercholesterolemia/drug therapy , Male , Rats , Tetrazoles/chemistry
8.
J Med Chem ; 39(20): 3908-19, 1996 Sep 27.
Article in English | MEDLINE | ID: mdl-8831757

ABSTRACT

A series of heterocyclic amides were synthesized and evaluated as inhibitors of acyl-CoA: cholesterol O-acyltransferase (ACAT) in vitro and for cholesterol lowering in cholesterol-fed rats. Compounds were evaluated for cell-based macrophage ACAT inhibition, bioactivity, and adrenal toxicity. Candidates were selected for evaluation in cholesterol-fed dogs and, ultimately, the injured cholesterol-fed rabbit model of atherosclerosis. The heterocyclic amides potently inhibited rabbit liver ACAT (IC50's = 0.014-0.11 microM), and the majority of compounds significantly lowered plasma cholesterol (42-68%) in an acute cholesterol-fed rat model at 3 mg/kg. The most efficacious compounds in the rat were evaluated for bioactivity in vivo and arterial ACAT inhibition in a cell-based macrophage ACAT assay. Two highly bioactive analogs, (+/-)-2-(3-dodecylisoxazol-5-yl)-2-phenyl-N-(2,4,6-trimethoxypheny l) acetamide (13a) and (+/-)-2-(5-dodecylisoxazol-3-yl)-2-phenyl-N-(2,4,6-trimethoxypheny l) acetamide (16a), were selected for further study and were found to be nontoxic in a guinea pig model of adrenal toxicity. Compounds 13a and 16a lowered total cholesterol in the cholesterol-fed rat, rabbit, and dog models of pre-established hypercholesterolemia. Compound 13a in the injured cholesterol-fed rabbit model of atherosclerosis was effective in slowing the development of cholesteryl ester-rich thoracic aortic lesions, reducing lesion coverage by 53% at a dose of 1 mg/kg.


Subject(s)
Acetamides/chemical synthesis , Anticholesteremic Agents/chemical synthesis , Arteriosclerosis/drug therapy , Enzyme Inhibitors/chemical synthesis , Isoxazoles/chemical synthesis , Sterol O-Acyltransferase/antagonists & inhibitors , Acetamides/therapeutic use , Acetamides/toxicity , Adrenal Gland Diseases/chemically induced , Animals , Anticholesteremic Agents/therapeutic use , Cholesterol/blood , Dogs , Enzyme Inhibitors/therapeutic use , Guinea Pigs , Isoxazoles/therapeutic use , Isoxazoles/toxicity , Liver/enzymology , Male , Molecular Structure , Rabbits , Rats
9.
J Med Chem ; 39(12): 2354-66, 1996 Jun 07.
Article in English | MEDLINE | ID: mdl-8691430

ABSTRACT

A series of tetrazole amide derivatives of (+/-)-2-dodecyl-alpha-phenyl-N-(2,4,6-trimethoxyphenyl)-2H-tetrazole-5- acetamide (1) was prepared and evaluated for their ability to inhibit acyl-CoA: cholesterol O-acyltransferase (ACAT) in vitro and to lower plasma total cholesterol in vivo. For this series of compounds, our objective was to systematically replace substituents appended to the amide and tetrazole moieties of 1 with structurally diverse functionalities and assess the effect that these changes have on biological activity. The ensuing structure-activity relationship (SAR) studies identified aryl (7b) and heteroaryl (7f,g) replacements for 2,4,6-trimethoxyphenyl that potently inhibit liver microsomal and macrophage ACAT in vitro and exhibit good cholesterol lowering activity (56-66% decreases in plasma total cholesterol at 30 mg/kg), relative to 1, when compared in the acute rat model of hypercholesterolemia. Replacement of the alpha-phenyl moiety with electron-withdrawing substituents (13e-h), however, significantly reduced liver microsomal ACAT inhibitory activity (IC50 > 1 microM). This is in contrast to electron-donating substituents (13ij,m-q), which produce IC50 values ranging from 5 to 75 nM in the hepatic microsomal assay. For selected tetrazole amides (1, 7b, 13n,o), reversing the order of substituents appended to the 2- and 5-positions in the tetrazole ring (36a-d), in general, improved macrophage ACAT inhibitory activity and provided excellent cholesterol-lowering activity (ranging from 65% to 77% decreases in plasma total cholesterol at 30 mg/kg) in the acute rat screen. The most potent isomeric pair in this set of unsubstituted methylene derivatives (13n and 36a) caused adrenocortical cell degeneration in guinea pigs treated with these inhibitors. In contrast, adrenal glands taken from guinea pigs treated with the corresponding alpha-phenyl-substituted analogs (7b and 36c) were essentially unchanged compared to untreated controls. Subsequent evaluation of 7b and 36c in a rabbit bioassay showed that both compounds and/or their metabolities were present in plasma after oral dosing. Unlike 7b and 36c, compound 1 and related 2,4,6-trimethoxyanilides (13j, 30c,d) showed poor oral activity in the rabbit bioassay. Nevertheless, in cholesterol-fed rabbits, both systemically available (7b, 36c) and poorly absorbed inhibitors (1, 36d) were more effective in lowering plasma total cholesterol than the fatty acid amide CI-976.


Subject(s)
Anticholesteremic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Sterol O-Acyltransferase/antagonists & inhibitors , Tetrazoles/pharmacology , Animals , Anticholesteremic Agents/chemical synthesis , Anticholesteremic Agents/chemistry , Arteriosclerosis/prevention & control , Cholesterol/blood , Cholesterol, Dietary/pharmacokinetics , Drug Design , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Guinea Pigs , Hypercholesterolemia/chemically induced , Hypercholesterolemia/drug therapy , Macrophages/enzymology , Male , Microsomes, Liver/enzymology , Molecular Structure , Rabbits , Rats , Structure-Activity Relationship , Tetrazoles/chemical synthesis , Tetrazoles/chemistry
10.
J Med Chem ; 39(6): 1243-52, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8632431

ABSTRACT

Several series of acyl-CoA:cholesterol O-acyltransferase inhibitors were prepared by the stepwise addition of nitrogen, oxygen, and sulfur nucleophiles to N-chlorosulfonyl isocyanate. The (aminosulfonyl)ureas 3-44 were the most potent inhibitors in vitro, with several compounds having IC50 values < 1 microM. Although the other series of compounds were not as potent in vitro, many compounds did display good in vivo activity in cholesterol-fed rats. Several of the oxysulfonyl carbamates (including CI-999, 115) showed excellent lipid-lowering activity in the chronic in vivo screen, demonstrating significant cholesterol lowering in a pre-established hypercholesterolemic state.


Subject(s)
Enzyme Inhibitors/pharmacology , Isocyanates/pharmacology , Sterol O-Acyltransferase/antagonists & inhibitors , Animals , Rats , Structure-Activity Relationship
11.
J Med Chem ; 37(15): 2394-400, 1994 Jul 22.
Article in English | MEDLINE | ID: mdl-8057287

ABSTRACT

Five series of compounds (4-9) derived from N-(chlorocarbonyl) isocyanate have been synthesized and evaluated for their ability to inhibit the enzyme acyl-CoA:cholesterol O-acyltransferase and lower plasma cholesterol levels in cholesterol-fed rats. Structure-activity relationships indicate that the imino dicarboxylates (6 and 7) and the oxycarbonyl thiocarbamates (8) are the most potent and efficacious series. In these series, the combination of a 2,6-diisopropylphenyl group and an aliphatic alkyl group with a chain length between 6 and 14 carbon atoms gives good activity in vitro and in vivo. In addition, a hydrogen donor is required to maintain good in vitro activity, and the acidic proton on the central nitrogen in these series appears to be important for in vivo activity.


Subject(s)
Hypolipidemic Agents/pharmacology , Isocyanates/pharmacology , Sterol O-Acyltransferase/antagonists & inhibitors , Animals , Cholesterol/blood , Hypolipidemic Agents/chemistry , Isocyanates/chemistry , Rats , Structure-Activity Relationship
12.
J Med Chem ; 37(12): 1810-22, 1994 Jun 10.
Article in English | MEDLINE | ID: mdl-8021920

ABSTRACT

A series of disubstituted ureas containing amide or amine groups was prepared and evaluated for their ability to inhibit acyl-CoA:cholesterol O-acyl transferase in vitro and to lower plasma total cholesterol in a variety of cholesterol-fed rat models in vivo. The presence of polar or ionizable functionalities within this class of compounds may impart greater aqueous solubility to those compounds and thus allow improved transport to the enzyme location within the intestinal enterocyte. Compounds from this class exhibit good cholesterol lowering in a chronic cholesterol-fed rat model of hypercholesterolemia even when dosed in an aqueous vehicle. In general, the amine-containing compounds were more potent and efficacious than the amides in the acute rat model of hypercholesterolemia. Further structure-activity relationship studies showed that the preferred position of the amide/amine group was beta to the urea moiety and not alpha, and that in this series, the presence of a secondary amine (or amide) proton is required for good in vitro potency. One of these compounds, 9n(-), lowered plasma total cholesterol (-47%) and elevated high-density lipoprotein cholesterol (+256%) when dosed in an aqueous vehicle to rats with preestablished hypercholesterolemia.


Subject(s)
Amides/chemistry , Amines/chemistry , Anticholesteremic Agents/chemistry , Carbamates/chemistry , Sterol O-Acyltransferase/antagonists & inhibitors , Urea/chemistry , Animals , Anticholesteremic Agents/pharmacology , Disease Models, Animal , Rabbits , Rats , Structure-Activity Relationship
13.
J Med Chem ; 37(11): 1652-9, 1994 May 27.
Article in English | MEDLINE | ID: mdl-8201599

ABSTRACT

We recently described our initial structure-activity relationship (SAR) studies on a series of N-phenyl-N'-aralkyl- and N-phenyl-N'-(1-phenylcycloalkyl)ureas as inhibitors of acyl-CoA: cholesterol acyltransferase (ACAT). From this series of analogs, compound 1 (PD 129337) was identified as a potent inhibitor of ACAT with an IC50 value of 17 nM. It was also shown to dose-dependently lower plasma cholesterol in cholesterol-fed rats. However, further investigation led to the suggestion that this compound was poorly absorbed, due to a lack of efficacy when administered by gavage in an aqueous vehicle. To overcome this deficiency, we continued our SAR study on this novel series of ACAT inhibitors using an acute in vivo screen in which the compounds are administered to rats in an aqueous, CMC/Tween suspension vehicle. Modification of the N'-phenyl moiety by incorporating functional groups which were amenable to forming salts and/or polar groups to reduce lipophilicity led to the identification of several inhibitors which displayed excellent efficacy employing this protocol. Overall, substitution on the phenyl ring in the ortho, meta, or para positions led to inhibitors with only a slight decrease in potency in vitro compared to the parent unsubstituted compound. Bulkier groups in the para position tended to lower the ACAT inhibitory activity in vitro. Polar groups, such as carboxyl (33,34), lowered in vitro activity significantly, suggesting that polar-ionic interactions are disfavored for the enzyme activity. From this series, compound 28 was evaluated further in secondary in vivo screens. In a chronic cholesterol-fed rat model of hypercholesterolemia, compound 28 dose-dependently reduced nonHDL cholesterol and significantly elevated HDL cholesterol. It showed significantly greater aqueous solubility than the parent compound 1. However, it was shown to cause adrenal toxicity in guinea pigs. This led us to design a series of homologs (44-51) with increased basicity and lower lipophilicity. Some of these compounds were more potent ACAT inhibitors in vitro and demonstrated excellent hypocholesterolemic activity in vivo. Interestingly, compound 45, unlike 28, did not produce adrenal toxicity in guinea pigs and demonstrated excellent lipid-modulating activity in the chronic model of preestablished dyslipidemia in rats.


Subject(s)
Anticholesteremic Agents/chemical synthesis , Phenylurea Compounds/chemical synthesis , Sterol O-Acyltransferase/antagonists & inhibitors , Adrenal Gland Diseases/chemically induced , Animals , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/toxicity , Cholesterol/blood , Guinea Pigs , Male , Molecular Structure , Phenylurea Compounds/pharmacology , Phenylurea Compounds/toxicity , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
15.
J Med Chem ; 36(20): 2943-9, 1993 Oct 01.
Article in English | MEDLINE | ID: mdl-8411011

ABSTRACT

A study of structure-activity relationships of substituted beta-ketoamide ACAT inhibitors I and II was performed. The results of this study suggest that whereas the beta-keto group was tolerated with no loss in activity, beta-hydroxy and oxime moieties led to significantly reduced activity in vitro and in vivo. The most potent inhibitor from the acyclic series (I) (11, IC50 = 0.006 microM) contained a C-13 alkyl chain. This compound reduced plasma total cholesterol by 38% and 66% at 3 and 30 mg/kg, respectively, in cholesterol-fed rats. Dimethylation alpha to the anilide core (5) and subsequent N-methylation of the amide NH (6) decreased in vitro potency significantly. It was also found that high potency was retained with inhibitors which incorporated the carbonyl into a lactam ring (II).


Subject(s)
Anilides/chemistry , Anilides/pharmacology , Anticholesteremic Agents/chemistry , Anticholesteremic Agents/pharmacology , Ketones/chemistry , Sterol O-Acyltransferase/antagonists & inhibitors , Anilides/chemical synthesis , Animals , Anticholesteremic Agents/chemical synthesis , Cholesterol/blood , Intestines/enzymology , Male , Microsomes/enzymology , Rabbits , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
18.
Cancer Res ; 47(16): 4299-304, 1987 Aug 15.
Article in English | MEDLINE | ID: mdl-2440564

ABSTRACT

A series of 2-aminoalkyl-5-nitropyrazolo[3,4,5-kl]acridines (pyrazoloacridines) was evaluated in vitro for activity against a panel of human tumor cell lines of breast, colon, or lung origin. Several pyrazoloacridines were found to possess solid tumor selectivity relative to their activity against murine leukemia L1210 cells as well as human lymphoblastoid cells. The superior compounds in this regard were also found to exhibit excellent activity against primary human tumors in stem cell clonogenic assays. In addition, many of the compounds tested were found to be selectively cytotoxic to hypoxic relative to oxic HCT-8 colon adenocarcinoma cells, a property that may be a consequence of the potentially reducible 5-nitro function. A number of pyrazoloacridines were also found to exhibit potency against noncycling Chinese hamster ovary cells comparable to that observed against actively dividing cultures. Consistent with their favorable activity against nondividing cells, further testing of the pyrazoloacridines revealed that generally less drug is required to inhibit RNA synthesis than DNA synthesis in L1210 cells. Collectively these data indicate that the pyrazoloacridines represent a novel class of antitumor agents which warrant further preclinical evaluation for their potential clinical usefulness in the treatment of solid tumors.


Subject(s)
Acridines/pharmacology , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Pyrazoles/pharmacology , Cell Cycle , Cell Line , DNA/biosynthesis , Humans , Oxygen , RNA/biosynthesis , Structure-Activity Relationship , Tumor Stem Cell Assay
19.
J Antibiot (Tokyo) ; 40(1): 73-6, 1987 Jan.
Article in English | MEDLINE | ID: mdl-3558118

ABSTRACT

The antibiotic acetomycin was active in vitro against HCT-8 human colon adenocarcinoma cells (IC50, 1.5 microgram/ml) and L1210 murine leukemia cells (IC50, 2.2 micrograms/ml). Acetomycin also had marked activity in the human tumor stem cell assay, with a 33% overall response rate (less than or equal to 30% survival) against 49 primary tumors. However, acetomycin was inactive in four in vivo tumor assay systems (L1210 and P388 leukemias, B16 melanoma and the MX-1 mammary xenograft system). This lack of in vivo activity may result from metabolic inactivation of acetomycin.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Leukemia L1210/drug therapy , Adenocarcinoma , Animals , Cell Line , Cell Survival/drug effects , Colonic Neoplasms , Furans/pharmacology , Furans/therapeutic use , Humans , Mice , Tumor Stem Cell Assay
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...