Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Lipid Mediat ; 7(2): 115-34, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8400114

RESUMO

MK 287 (L-680,573), a tetrahydrofuran analog, potently inhibited [3H]C18-PAF binding to human platelet, polymorphonuclear leukocyte (PMN) and lung membranes with K1 values of 6.1 +/- 1.5, 3.2 +/- 0.7, and 5.49 +/- 2.3 nM, respectively. The inhibitory effects are stereospecific and competitive. The racemate, L-668,750 is less potent and the enantiomer, L-680,574 is 20-fold less potent than MK 287. Inhibition of the binding of [3H]C18-PAF to human PMN membranes by MK 287 was associated with the reduction of the affinity of the radioligand but not the number of the receptor sites. Binding of other radioligands (e.g., LTB4, LTC4, C5a, FMLP) to their specific receptors was unaltered at 1-10 microM MK 287. [3H]MK 287 bound to membranes from human platelets and PMNs: KD = 2.1 +/- 0.6 and 2.9 +/- 1.2 nM, respectively. When examined on isolated human cells, MK 287 potently and selectively inhibited PAF-induced aggregation of platelets in plasma (ED50 = 56 +/- 38 nM) or gel-filtered platelets (ED50 = 1.5 +/- 0.5 nM) and elastase release from PMNs (ED50 = 4.4 +/- 2.6 nM). In studies in vivo, MK 287 inhibited PAF-induced lethality in mice (ED50 = 0.8 mg/kg orally) and PAF-induced bronchoconstriction in guinea pigs (ED50 = 0.18 mg/kg intraduodenally and 0.19 mg/kg intravenously). Inhibition of PAF-induced bronchoconstriction was accompanied by parallel rightward shifts in concentration-response curves for PAF-induced platelet aggregation measured ex vivo.


Assuntos
Furanos/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Administração Oral , Animais , Ligação Competitiva , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Broncoconstrição/efeitos dos fármacos , Cobaias , Humanos , Cinética , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Membranas/efeitos dos fármacos , Membranas/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Fator de Ativação de Plaquetas/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Coelhos , Sensibilidade e Especificidade , Trítio
2.
Biochem J ; 289 ( Pt 3): 889-95, 1993 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8094614

RESUMO

The beta-lactones L-659,699 [(E,E)-11-[3-(hydroxymethyl)-4-oxo-2- oxetanyl]-3,5,7-trimethyl-2,4-undecadienoic acid) and its radioactive derivative 3H-L-668,411 (the 2,3-ditritiated methyl ester of L-659,699) inhibited a partially purified preparation of rat liver cytosolic 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) synthase with an IC50 of 0.1 microM. These compounds were also found to inhibit the incorporation of [14C]acetate into sterols in cultured Hep G2 cells with an IC50 of 3 microM. New kinetic evidence indicated that inhibition of the isolated enzyme was irreversible. In contrast, sterol biosynthesis in cultured Hep G2 cells was rapidly restored upon removal of the compound from the medium of inhibited cultures, suggesting reversibility of inhibition in the cells. Radioactivity was found to be associated with a single cytoplasmic protein by SDS/PAGE of the cytoplasm of Hep G2 cells after incubation of the cells with the inhibitor 3H-L-668,411. This protein was identified as cytoplasmic HMG-CoA synthase. Binding of the radioactive compound to the enzyme was decreased with time if the radioactive inhibitor was removed from the medium. Exposure of a gel containing the radioactive enzyme-inhibitor complex to neutral hydroxylamine also resulted in a loss of radioactivity from the gel. The purified rat liver enzyme reacted with the 3H-ligand to form a stable enzyme-inhibitor complex which could be isolated by h.p.l.c. Radioactivity was also subsequently lost from this complex when it was incubated with neutral hydroxylamine. Incorporation of [14C]acetate into cholesterol in mouse liver was inhibited in a reversible manner after oral administration of the beta-lactone inhibitor. These studies, as well as the kinetic evidence presented, suggest that the beta-lactone inhibitors acylate HMG-CoA synthase in a reaction which appears to be irreversible in vitro, but is easily reversed in cultured cells and in animals.


Assuntos
Colesterol/biossíntese , Ácidos Graxos Insaturados/farmacologia , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Lactonas/farmacologia , Acetatos/metabolismo , Animais , Células Cultivadas , Cicloeximida/farmacologia , Citoplasma/enzimologia , Ácidos Graxos Insaturados/metabolismo , Feminino , Hidroxilamina , Hidroxilaminas/farmacologia , Hidroximetilglutaril-CoA Sintase/efeitos dos fármacos , Hidroximetilglutaril-CoA Sintase/isolamento & purificação , Hidroximetilglutaril-CoA Sintase/metabolismo , Lactonas/metabolismo , Fígado/metabolismo , Camundongos , Ratos , Trometamina/farmacologia
3.
J Med Chem ; 35(21): 3813-21, 1992 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-1433193

RESUMO

Substitution of hydroxy and hydroxyalkyl functionality at C-7 of the hexahydronaphthalene nucleus of simvastatin has provided novel analogs. The synthetic strategy employed epoxidation or Lewis acid-catalyzed aldol reaction of the 8-keto silyl enol ether as a key reactive intermediate. These analogs were evaluated as potential hypocholesterolemic agents via initial determination of their ability to inhibit HMG-CoA reductase in vitro. Oral activity of these compounds was determined in an acute rat model and a three-week study in cholestyramine-primed dogs. Compounds were identified that possessed in vitro and in vivo activity comparable to that of simvastatin.


Assuntos
Anticolesterolemiantes/síntese química , Inibidores de Hidroximetilglutaril-CoA Redutases , Lovastatina/análogos & derivados , Administração Oral , Animais , Anticolesterolemiantes/farmacologia , Modelos Animais de Doenças , Cães , Hipercolesterolemia/tratamento farmacológico , Lovastatina/síntese química , Lovastatina/farmacologia , Lovastatina/uso terapêutico , Espectroscopia de Ressonância Magnética , Ratos , Sinvastatina , Relação Estrutura-Atividade
4.
J Med Chem ; 35(19): 3474-82, 1992 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-1404229

RESUMO

(-)-trans-(2S,5S)-2-[3-[(2-Oxopropyl)sulfonyl]-4-n-propoxy-5-(3- hydroxypropoxy)phenyl]-5-(3,4,5-trimethoxyphenyl)tetrahydrofuran (10) is one of the most potent platelet-activating factor (PAF) antagonists in vitro and in vivo developed to date. This diaryltetrahydrofuran derivative evolved from modifications of MK 0287 which has been evaluated in clinical studies for asthma. Two structural modifications of MK 0287 were made: (1) elaboration of the 3'-[(hydroxyethyl)sulfonyl] group to a beta-keto propylsulfonyl, and (2) replacement of the 5'-methyl ether by a 3-hydroxypropyl ether. Compound 10 potently and specifically inhibits the binding of [3H]-C18-PAF to human platelet membranes (Ki 1.85 nM) and PMN membranes (Ki 2.89 nM). In vivo, 10 inhibits PAF-induced plasma extravasation and elevated N-acetyl-beta-D-glucosaminidase (NAGA) levels in male rats with ED50 values of 60 micrograms/kg, po and 4 micrograms/kg, iv respectively, and inhibits PAF-induced bronchoconstriction in guinea pigs with an ED50 value of 15 micrograms/kg after intraduodenal administration. Compound 15, a water-soluble phosphate ester prodrug derivative of 10 is at least equipotent to 10 in the in vivo models. Compound 19S, the primary and major metabolite of 10 and 15, is equipotent in in vitro and in vivo models.


Assuntos
Furanos/síntese química , Fator de Ativação de Plaquetas/antagonistas & inibidores , Pró-Fármacos/síntese química , Sulfonas/síntese química , Administração Oral , Animais , Furanos/química , Furanos/farmacologia , Cobaias , Humanos , Masculino , Inibidores da Agregação Plaquetária/farmacologia , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Ratos , Solubilidade , Estereoisomerismo , Relação Estrutura-Atividade , Sulfonas/química , Sulfonas/farmacologia
5.
Diabetes ; 40(12): 1630-9, 1991 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1756903

RESUMO

We previously reported that dog diabetes results in hypercholesterolemia and the accumulation of a high-density lipoprotein (HDL) subclass, HDL1. Hypercholesterolemic diabetic rodents exhibit hyperphagia, intestinal hypertrophy, and increased intestinal cholesterol synthesis and absorption; intestinal 3-hydroxy-3-methylglutaryl (HMG) CoA reductase activity is increased, whereas hepatic activity is unchanged or reduced. To determine whether similar mechanisms operate in the hypercholesterolemic diabetic dog, we measured hepatic and intestinal cholesterologenesis. Streptozocin-alloxan-induced diabetic dogs allowed access to food ad libitum were hyperphagic and hypercholesterolemic (10.1 vs. 4.47 mM) but normotriglyceridemic. Plasma HDL1 concentrations were markedly increased. Differences in renal and hepatic function were not statistically significant, except serum alkaline phosphatase, which was elevated 4-fold (P = 0.0003). Urinary mevalonate, an index of whole-body cholesterol synthesis, was increased 6-fold. Intestinal and hepatic weights were both increased, and direct measurements showed crypt and villus thickening. The activity of HMG CoA reductase per gram organ weight was increased 1.7-fold in liver and 2.1-fold in intestine. Calculated whole-organ activity in intestine was nearly twice that in liver. These observations provide strong evidence that intestinal cholesterogenesis is involved in the pathogenesis of hypercholesterolemia in dog diabetes and support the conclusion that increased cholesterol synthesis plays a role in the hypercholesterolemia of diabetes.


Assuntos
Colesterol/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Hidroximetilglutaril-CoA Redutases/metabolismo , Hipercolesterolemia/fisiopatologia , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Lipoproteínas/sangue , Fígado/metabolismo , Animais , Peso Corporal , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Cães , Feminino , Hipercolesterolemia/etiologia , Insulina/uso terapêutico , Mucosa Intestinal/patologia , Intestino Delgado/patologia , Lipoproteínas/isolamento & purificação , Masculino , Ácido Mevalônico/urina , Modelos Biológicos , Tamanho do Órgão
6.
J Med Chem ; 34(8): 2474-7, 1991 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1875344

RESUMO

A general route for preparing side chain ether analogues of lovastatin is presented. These analogues proved to be weaker inhibitors of HMG-CoA reductase than the corresponding side chain ester analogues. Interestingly, inhibitory potency was enhanced markedly when the 4-fluoro group was incorporated in the aromatic moiety of the side chain benzyl group of 2d.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Lovastatina/análogos & derivados , Fenômenos Químicos , Química , Ésteres , Éteres , Lovastatina/síntese química , Lovastatina/farmacologia , Estrutura Molecular , Relação Estrutura-Atividade
7.
J Med Chem ; 34(8): 2489-95, 1991 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1875346

RESUMO

Modification of the hexahydronaphthalene ring 5-position in simvastatin 2a via oxygenation and oxa replacement afforded two series of derivatives which were evaluated in vitro for inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reductase and acutely in vivo for oral effectiveness as inhibitors of cholesterogenesis in the rat. Of the compounds selected for further biological evaluation, the 6 beta-methyl-5-oxa 10 and 5 alpha-hydroxy 16 derivatives of 3,4,4a,5-tetrahydro 2a, as well as, the 6 beta-epimer 14 of 16 proved orally active as hypocholesterolemic agents in cholestyramine-primed dogs. Subsequent acute oral metabolism studies in dogs demonstrated that compounds 14 and 16 evoke lower peak plasma drug activity and area-under-the-curve values than does compound 10 and led to the selection of 14 and 16 for toxicological evaluation.


Assuntos
Anticolesterolemiantes/síntese química , Inibidores de Hidroximetilglutaril-CoA Redutases , Lovastatina/análogos & derivados , Oxigênio , Acetatos/metabolismo , Animais , Anticolesterolemiantes/farmacocinética , Anticolesterolemiantes/farmacologia , Fenômenos Químicos , Química , Colesterol/biossíntese , Cães , Cinética , Lovastatina/síntese química , Lovastatina/química , Lovastatina/farmacocinética , Lovastatina/farmacologia , Masculino , Conformação Molecular , Estrutura Molecular , Ratos , Sinvastatina , Relação Estrutura-Atividade
8.
Fundam Appl Toxicol ; 16(2): 320-9, 1991 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2055362

RESUMO

L-645, 164, a potent inhibitor of hydroxymethylglutarylcoenzyme A (HMG-CoA) reductase, is a structurally unique, synthetic monofluorinated-biphenyl that was administered to beagle dogs at dosages of 2, 10, or 50 mg/kg/day for 14 weeks to evaluate its toxic potential. Previously tested HMG-CoA reductase inhibitors from this laboratory have either been semisynthetic or fermentation-derived products containing a hexahydronaphthalene ring structure (i.e., lovastatin and simvastatin). Administration of L-645, 164 produced a significant spectrum of lesions, some of which have been previously associated with compounds of this pharmacological class, while others were unique to this monofluorinated-biphenyl inhibitor. Subcapsular lenticular opacities were produced in six of eight of the dogs receiving 50 mg/kg/day of L-645, 164 within 8 weeks of dosing. One dog receiving this dosage level experienced increases in serum alanine aminotransferase activity to levels 10 times those in concurrent control dogs. Light and electron microscopy of a wedge biopsy obtained within 3 days of this transaminase elevation failed to reveal any significant changes and the elevation resolved spontaneously despite continued drug administration. Lesions of the optic nerve and acoustic-vestibular tract and trapezoid decussation were observed in several dogs receiving 50 mg/kg/day. In addition, similar changes were observed in the optic tract in several of the dogs receiving 50 mg/kg/day and in one dog receiving 2 mg/kg/day of L-645,164. These were unique to L-645,164 and have not been observed after the administration of other HMG-CoA reductase inhibitors in this laboratory. Optic tract changes were generally mild, consisting of small to medium vacuoles without apparent myelin loss. Lesions in the other areas ranged from very slight to prominent vacuolation. No clinical signs were observed. Peak plasma drug levels of L-645,164 at 50 mg/kg were greater than 5 micrograms/ml, about one order of magnitude greater than those attained after administration of pharmacologically equipotent doses of lovastatin and simvastatin. These findings support previous observations that HMG-CoA reductase inhibitors producing high plasma drug levels are associated with a significant degree of systemic toxicity. In addition, the drug-induced CNS lesions attributed to L-645,164 appear also to be related to its chemical structure since similar lesions have not been observed after the administration of other structurally unrelated HMG-CoA reductase inhibitors that produce high plasma drug concentrations and comparable degrees of serum cholesterol lowering.


Assuntos
Compostos de Bifenilo/toxicidade , Inibidores de Hidroximetilglutaril-CoA Redutases , Pironas/toxicidade , Animais , Compostos de Bifenilo/sangue , Encéfalo/patologia , Catarata/induzido quimicamente , Cães , Vesícula Biliar/efeitos dos fármacos , Vesícula Biliar/patologia , Hiperplasia/induzido quimicamente , Lipídeos/sangue , Fígado/efeitos dos fármacos , Estrutura Molecular , Mucosa/efeitos dos fármacos , Mucosa/patologia , Pironas/sangue , Dermatopatias/induzido quimicamente
9.
Eur J Clin Pharmacol ; 40 Suppl 1: S11-4, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-2044637

RESUMO

Lovastatin and simvastatin are potent competitive inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase. Key inhibit the synthesis of cholesterol in cultured HepG23 cells, rat hepatocytes and in rats. The primary target organ of cholesterol synthesis inhibition by lovastatin and simvastatin is the liver. Lovastating and simvastatin lower levels of plasma cholesterol in rats, dogs and rabbits by inhibition the endogenous cholesterol synthesis and induction of LDL receptor in the liver.


Assuntos
Anticolesterolemiantes , Colesterol/sangue , Lovastatina/análogos & derivados , Lovastatina/farmacologia , Animais , Células Cultivadas , Colesterol/biossíntese , Inibidores de Hidroximetilglutaril-CoA Redutases , Fígado/efeitos dos fármacos , Fígado/metabolismo , Receptores de LDL/biossíntese , Receptores de LDL/efeitos dos fármacos , Sinvastatina
10.
Atherosclerosis ; 84(1): 1-12, 1990 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2248616

RESUMO

Hypercholesterolemia and increased concentrations of an apolipoprotein E (apoE)-containing HDL subclass, high density lipoprotein1 (HDL1) have been observed in streptozocin-alloxan diabetic dogs consuming normal amounts of dietary cholesterol. The aim of this study was to characterize the response of HDL1 and its targeting ligand, apoE, to insulin and HMG-CoA reductase inhibitor treatment in pancreatectomized diabetic dogs. Following induction of diabetes, plasma total cholesterol, HDL1, and apoE concentrations were all increased. Urinary mevalonate excretion, an index of cholesterol synthesis in humans, was 6-fold that of nondiabetic controls. Lipoprotein fractionation by Pevikon block electrophoresis and gel filtration chromatography showed that the increased cholesterol and apoE were associated with alpha 2-migrating particles corresponding to HDL1. Insulin treatment, resulting in near normal fasting blood glucose concentrations in the group as a whole (average 5.1 mM, 92 mg/dl), led to variable reductions in apoE, total plasma cholesterol, and HDL1. Uncorrected dyslipidemia during intensified insulin treatment appeared to be related to failure to achieve euglycemia. Despite unremitting hyperglycemia, treatment with lovastatin resulted in pronounced decreases in plasma cholesterol, HDL1 and apoE to concentrations below those observed in nondiabetic animals. Mevalonate excretion also fell, but remained twice normal. Thus neither modality corrected all of the abnormalities in canine diabetic dyslipidemia. Since apoE-containing HDL1 may mediate cholesterol traffic between the periphery and the liver (reverse cholesterol transport), the present observations suggest that increased cholesterol synthesis is accompanied by parallel abnormalities in cholesterol flux through the reverse transport pathway in the canine model.


Assuntos
Apolipoproteínas B/sangue , Diabetes Mellitus Experimental/sangue , Insulina/farmacologia , Lipoproteínas HDL/sangue , Lovastatina/farmacologia , Ácido Mevalônico/urina , Animais , Glicemia/análise , Colesterol/sangue , Diabetes Mellitus Experimental/urina , Cães , Masculino , Pancreatectomia
11.
J Med Chem ; 33(2): 758-65, 1990 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2299642

RESUMO

A variety of trans-6-[2-(substituted-1-naphthyl)ethyl(or ethenyl)]-3,4,5,6-tetrahydro-4-hydroxy-2H-pyran-2-ones were prepared and, upon conversion to their 3,5-dihydroxy carboxylates, were found to have good inhibitory activity against the enzyme 3-hydroxy-3-methylglutaryl-coenzyme A reductase, the rate-determining enzyme in cholesterogenesis. The most active compounds are 2,4,6- and 2,4,7-trichloro derivatives and would be expected to display about the same potency as the standard compactin upon resolution.


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores de Hidroximetilglutaril-CoA Redutases , Naftalenos/síntese química , Piranos/síntese química , Fenômenos Químicos , Química , Físico-Química , Inibidores Enzimáticos/farmacologia , Humanos , Técnicas In Vitro , Naftalenos/metabolismo , Naftalenos/farmacologia , Piranos/metabolismo , Piranos/farmacologia , Relação Estrutura-Atividade
12.
Exp Eye Res ; 50(1): 65-78, 1990 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-2307197

RESUMO

The administration of high dosages of various hydroxymethylglutaryl-CoA (HMG-CoA) reductase inhibitors has resulted in the development of subcapsular lenticular opacities in dogs. While dogs receiving cataractogenic doses of HMG-CoA reductase inhibitors experienced profound decreases in circulating serum cholesterol concentrations (40-60% reductions in total serum cholesterol), a causal relationship between serum cholesterol lowering and cataractogenesis was not established. A strong relationship was demonstrated, however, between the systemic exposure to inhibitor (plasma drug levels) and the cataractogenic potential of the various compounds studied. Analysis of lenses from dogs chronically dosed with various HMG-CoA reductase inhibitors revealed the presence of low drug levels in the lens (less than 500 ng equivalents g-1), but no correlation was observed between the amount of drug associated with the lens after chronic treatment and cataract development. In addition, no abnormalities in cholesterol content or sterol composition were observed in clear and/or cataract containing lenses from dogs chronically dosed with HMG-CoA reductase inhibitors. The kinetics of drug appearance in the aqueous and lens cortex was assessed after doses of various HMG-CoA reductase inhibitors, and suggested somewhat higher but not statistically significant peak concentrations of inhibitor were achieved by compounds which produced a higher incidence of cataracts. These data have suggested that high doses of HMG-CoA reductase inhibitors may increase lenticular exposure to drug via the aqueous humor by producing a substantial systemic exposure to drug substance. This may result in an increased concentration of inhibitor in the outer cortical region of the lens where cholesterol synthesis is critical, thereby resulting in the development of opacities. The production of lenticular changes by a HMG-CoA reductase inhibitor of diverse chemical structure establishes, with reasonable assurance, that these lens changes are mechanism based (i.e. a product of the biochemical mechanism of action of this class of compounds). An extrapolation of these findings to patients receiving therapeutic dosages enables a favorable risk evaluation since the doses to be employed clinically are much lower and result in a far lower systemic exposure to drug substance.


Assuntos
Compostos de Bifenilo/toxicidade , Catarata/induzido quimicamente , Inibidores de Hidroximetilglutaril-CoA Redutases , Lovastatina/análogos & derivados , Piranos/toxicidade , Pironas/toxicidade , Animais , Anticolesterolemiantes/administração & dosagem , Anticolesterolemiantes/farmacocinética , Anticolesterolemiantes/toxicidade , Compostos de Bifenilo/administração & dosagem , Compostos de Bifenilo/farmacocinética , Colesterol/sangue , Cães , Lovastatina/administração & dosagem , Lovastatina/farmacocinética , Lovastatina/toxicidade , Pironas/administração & dosagem , Pironas/farmacocinética , Sinvastatina
13.
Cardiology ; 77 Suppl 4: 14-21, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2073667

RESUMO

The microsomal enzyme 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase is a key rate-controlling step early in the cholesterol biosynthetic pathway that catalyzes the conversion of HMG CoA to mevalonic acid. Since this enzyme plays a significant role in regulating cholesterol synthesis, it is a rational target for pharmacologic intervention. The first potent, specific inhibitor of HMG CoA was mevastatin (compactin, ML-236B), which was discovered in 1976 by Endo et al. [J Antibiot 1976:29:1346-1348]. Subsequently, lovastatin, a novel, more active fungal metabolite was isolated from a strain of Aspergillus terreus. Lovastatin, the first of this class of agents to be approved for clinical use, was chemically modified to form simvastatin. Simvastatin is superior to lovastatin in intrinsic inhibitory potency. Both are inactive lactone prodrugs that must be converted to their respective dihydroxy open-acid forms to elicit inhibitory activity. Pharmacologic characterization of lovastatin and simvastatin has demonstrated that these potent inhibitors of HMG CoA reductase specifically inhibit cholesterol synthesis in animal cells, as well as in vivo after oral administration of the agents. Oral administration of either lovastatin or simvastatin to dogs in the presence or absence of the bile acid sequestrant cholestyramine results in a marked, sustained lowering of plasma cholesterol. Associated with the cholesterol lowering is a decrease in urinary and plasma levels of mevalonic acid, the end product of the HMG CoA reductase reaction. The target organ for inhibitors of HMG CoA reductase is the liver, the primary site of cholesterol biosynthesis. Both lovastatin and simvastatin are preferentially extracted by this organ.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Colesterol/biossíntese , Inibidores de Hidroximetilglutaril-CoA Redutases , Lovastatina/análogos & derivados , Lovastatina/farmacologia , Animais , Células Cultivadas , Colesterol/sangue , Cães , Humanos , Fígado/metabolismo , Lovastatina/farmacocinética , Ratos , Sinvastatina , Distribuição Tecidual
14.
Cardiology ; 77 Suppl 4: 22-32, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2073669

RESUMO

Lovastatin and benzafibrate have proved effective in lowering low-density-lipoprotein (LDL) cholesterol and elevating high-density-lipoprotein (HDL) cholesterol. We compared their tolerability, safety, and effects on lipoproteins and urinary mevalonate excretion in a short-term study. Forty patients with primary hypercholesterolemia were enrolled in a single-blind randomized study with a diet/placebo period of 8 weeks and a treatment period of 12 weeks. Twenty patients received lovastatin (final average dose 70.5 mg/day), and 20 patients received bezafibrate 400 mg/day. LDL cholesterol was lowered by 35% (from 323 to 208 mg/dl) with lovastatin and by 8% (from 289 to 264 mg/dl) with benzafibrate. HDL cholesterol increased by 21 and 20% with lovastatin and benzafibrate, respectively. Twenty-four-hour urinary mevalonic acid output decreased by 37% during treatment with lovastatin and by 2% during treatment with bezafibrate. Thus, the lowering of cholesterol by lovastatin, but not by bezafibrate, can be attributed to inhibition of HMG CoA (3-hydroxy-3-methylglutaryl coenzyme A) reductase. Both lovastatin and bezafibrate are well tolerated.


Assuntos
Bezafibrato/uso terapêutico , Hipercolesterolemia/tratamento farmacológico , Lovastatina/uso terapêutico , Ácido Mevalônico/urina , Adulto , Idoso , Bezafibrato/efeitos adversos , Feminino , Humanos , Hipercolesterolemia/sangue , Hipercolesterolemia/urina , Lipídeos/sangue , Lovastatina/efeitos adversos , Masculino , Pessoa de Meia-Idade , Método Simples-Cego
15.
Am J Med ; 87(4A): 28S-38S, 1989 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-2679082

RESUMO

Simvastatin, a hydroxy-methylglutaryl-coenzyme A reductase inhibitor intended for use as a hypocholesterolemic agent, has undergone a thorough preclinical toxicology evaluation. This review describes preclinical toxicology findings associated with simvastatin administration in animals and provides the rationale for our conclusion that these changes are not indicative of potential human toxicity. Although it was not surprising to find that a potent inhibitor of this key biochemical pathway produces toxicity at high dosages in animals, none of the observed changes poses a significant risk to humans at clinical dosages. Many of the toxicities produced by high dosage levels of simvastatin in animals are directly related to the drug's biochemical mechanism of action and are the result of a profound, sustained inhibition of the target enzyme that is not anticipated at clinical dosages. Furthermore, several of the simvastatin-induced changes are species-specific responses to this agent and are not relevant to human risk assessment. Of the treatment-related changes reported for simvastatin, the development of cataracts in dogs has received considerable attention. The available data demonstrate a wide margin of safety in terms of dosage levels required to elicit this response as well as the plasma concentrations associated with the development of these ocular lesions. The data suggest that the development of lenticular opacities at clinical doses of simvastatin is highly improbable. Overall, simvastatin is highly improbable. Overall, simvastatin was well-tolerated by animals in preclinical toxicology studies, and no findings contraindicating its use in humans were identified.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Lovastatina/análogos & derivados , Animais , Catarata/induzido quimicamente , Cães , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Lovastatina/sangue , Lovastatina/toxicidade , Masculino , Necrose , Coelhos , Ratos , Sinvastatina , Especificidade da Espécie , Estômago/efeitos dos fármacos , Testículo/efeitos dos fármacos
16.
Biochem Biophys Res Commun ; 163(1): 548-53, 1989 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-2570576

RESUMO

The compound L-660, 631 (2-oxo-5-(1-hydroxy-2,4,6-heptatriynyl)-1,3-dioxolane-4 heptanoic acid), a natural product isolated from an Actinomycete culture, was found to inhibit rat liver cytosolic acetoacetyl-CoA thiolase, the first step in the cholesterol biosynthesis pathway, with an IC50 of 1.0 x 10(-8) M. The inhibitor had no effect on other sulfhydryl containing enzymes of lipid synthesis such as HMG-CoA synthase, HMG-CoA reductase, and fatty acid synthase. When tested in cultured human liver Hep G2 cells the compound inhibited the incorporation of 14C-acetate and 14C-octanoate into sterols 56% and 48% respectively at 3 x 10(-6) M with no effect on fatty acid synthesis. No noticeable effect was seen on fatty acid biosynthesis. This strongly suggests that the locus of inhibition of acetate incorporation into sterols found with this compound is the acetoacetyl-CoA thiolase step in the cholesterol biosynthesis pathway.


Assuntos
Acetil-CoA C-Acetiltransferase/antagonistas & inibidores , Acetiltransferases/antagonistas & inibidores , Dioxolanos/farmacologia , Dioxóis/farmacologia , Ácidos Heptanoicos/farmacologia , Fígado/enzimologia , Actinomycetales/análise , Animais , Colesterol/biossíntese , Citoplasma/enzimologia , Ácidos Graxos/biossíntese , Ratos
17.
Biochem Biophys Res Commun ; 158(3): 667-75, 1989 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-2493245

RESUMO

Tissue selectivity of lovastatin, simvastatin and pravastatin was determined in male rats. Peak levels of active drug were found in all tissues examined between 0.5 and 2 hours after oral administration. The area under the curve describing 24 hour exposure of the tissues to drug indicated that the drugs were preferentially concentrated in the liver. However, the concentration of pravastatin was approximately 50% that of either lovastatin or simvastatin in the liver and 3-6 times higher in peripheral tissues. These studies demonstrate that the hydrophobic prodrugs, lovastatin and simvastatin show greater selectivity than the hydrophilic agent pravastatin towards the liver which is the target organ for inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A reductase.


Assuntos
Ácidos Heptanoicos/farmacocinética , Lovastatina/análogos & derivados , Lovastatina/farmacocinética , Naftalenos/farmacocinética , Glândulas Suprarrenais/metabolismo , Animais , Mucosa Gástrica/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases , Rim/metabolismo , Fígado/metabolismo , Masculino , Pravastatina , Ratos , Sinvastatina , Baço/metabolismo , Testículo/metabolismo , Distribuição Tecidual
18.
J Pharmacol Exp Ther ; 248(2): 498-505, 1989 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2918466

RESUMO

Lovastatin, a specific inhibitor of the rate-limiting enzyme in cholesterol biosynthesis, HMG-CoA reductase, has been shown to be highly effective in lowering serum cholesterol in animals and humans and thus represents a promising approach to the treatment and prevention of cardiovascular disease. During the preclinical safety assessment of lovastatin, oral doses that were tolerated by dogs, rats and mice were found to be lethal to rabbits in subacute studies. Postmortem findings in rabbits consisted of centrilobular hepatic necrosis, frequently accompanied by renal tubular necrosis and occasionally gallbladder necrosis. The liver lesions were associated with up to 300-fold elevations in serum aspartate and alanine aminotransferase activities, whereas the kidney lesions resulted in accumulations of serum urea nitrogen and creatinine. The organ damage was preceded by a progressive decline in food consumption and loss of body weight. All histopathological and serum biochemical changes induced by lovastatin were completely prevented by coadministration of mevalonate, the product of the inhibited HMG-CoA reductase enzyme. In addition, administration of mevalonate after the onset of lovastatin-induced hepatotoxicity effectively reversed the toxicity despite continued drug treatment. These findings indicated that the toxicity of high doses of lovastatin to rabbits is a consequence of a highly exaggerated pharmacologic action in blocking mevalonate synthesis. However, supplementation of lovastatin-treated rabbits with oral doses of the major product of mevalonate metabolism, cholesterol, paradoxically enhanced the liver and kidney damage, which suggested that the toxicity of lovastatin stemmed from depletion of a nonsterol metabolite(s) of mevalonate critical for cell viability.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Lovastatina/toxicidade , Animais , Colesterol na Dieta/administração & dosagem , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Ácido Mevalônico/farmacologia , Coelhos , Especificidade da Espécie
19.
Am J Cardiol ; 62(15): 10J-15J, 1988 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-3055919

RESUMO

Cholesterol is a 27-carbon steroid that is an essential component of the cell membrane, the immediate precursor of steroid hormones, the substrate for the formation of bile acids, and is required for the assembly of very low density lipoprotein in the liver. Because as much as two-thirds of total body cholesterol in patients is of endogenous origin, an effective means to control cholesterogenesis may occur by inhibition of its biosynthesis. Cholesterol is biosynthesized in a series of more than 25 separate enzymatic reactions that initially involve the formation of 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA). Early attempts to pharmacologically block cholesterol synthesis focused only on steps later in the biosynthetic pathway and resulted in compounds with unacceptable toxicity. Recent research had identified that HMG CoA reductase is a key rate-limiting enzyme in this pathway and is responsible for the conversion of HMG CoA to mevalonate. Additional research with fungal metabolites identified a series of compounds with potent inhibiting properties for this target enzyme, from which lovastatin was selected for clinical development. A reduction in cholesterol synthesis by lovastatin has been subsequently confirmed in cell culture, animal studies and in humans. A resultant decrease in circulating total and low-density lipoprotein (LDL) cholesterol has also been demonstrated in animals and humans. Because hepatic LDL receptors are the major mechanism of LDL clearance from the circulation, further animal research has confirmed that these declines in cholesterol are accompanied by an increase in hepatic LDL receptor activity. Lovastatin effectively diminishes endogenous cholesterol synthesis providing useful therapeutic properties for patients with hypercholesterolemia.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Hipercolesterolemia/tratamento farmacológico , Lovastatina , Animais , Fenômenos Químicos , Química , Colesterol/biossíntese , Humanos , Fígado/metabolismo , Lovastatina/farmacologia , Lovastatina/uso terapêutico , Receptores de LDL/efeitos dos fármacos
20.
Am J Cardiol ; 62(15): 16J-27J, 1988 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-3055920

RESUMO

Administration of lovastatin to animals at high dosage levels produces a broad spectrum of toxicity. This toxicity is expected based on the critical nature of the target enzyme (HMG CoA reductase) and the magnitude of the dosage levels used. The information reviewed in this paper demonstrates that these adverse findings in animals do not predict significant risk in humans. The reason for this derives from the fact that all the available evidence suggests that the adverse effects observed are produced by an exaggeration of the desired biochemical effect of the drug at high dosage levels. The presence of clear and high no-effect doses for these toxic effects along with the fact that most of the changes observed are clearly mechanism-based (directly attributable to inhibition of mevalonate synthesis) indicate that it is unlikely that similar changes will be observed at the therapeutic dosage levels in humans. This hypothesis is supported by the extensive human safety experience described by Tobert in the following report.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Lovastatina/toxicidade , Animais , Catarata/induzido quimicamente , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Fígado/efeitos dos fármacos , Lovastatina/administração & dosagem , Neoplasias Experimentais/induzido quimicamente , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA