Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Drug Metab Dispos ; 49(8): 601-609, 2021 08.
Article in English | MEDLINE | ID: mdl-34011531

ABSTRACT

Ozanimod, recently approved for treating relapsing multiple sclerosis, produced a disproportionate, active, MAO B-catalyzed metabolite (CC112273) that showed remarkable interspecies differences and led to challenges in safety testing. This study explored the kinetics of CC112273 formation from its precursor RP101075. Incubations with human liver mitochondrial fractions revealed K Mapp, V max, and intrinsic clearance (Clint) for CC112273 formation to be 4.8 µM, 50.3 pmol/min/mg protein, and 12 µl/min/mg, respectively, whereas Michaelis-Menten constant (K M) with human recombinant MAO B was 1.1 µM. Studies with liver mitochondrial fractions from preclinical species led to K Mapp, V max, and Clint estimates of 3.0, 35, and 33 µM, 80.6, 114, 37.3 pmol/min/mg, and 27.2, 3.25, and 1.14 µl/min/mg in monkey, rat, and mouse, respectively, and revealed marked differences between rodents and primates, primarily attributable to differences in the K M Comparison of Clint estimates revealed monkey to be ∼2-fold more efficient and the mouse and rat to be 11- and 4-fold less efficient than humans in CC112273 formation. The influence of stereochemistry on MAO B-mediated oxidation was also investigated using the R-isomer of RP101075 (RP101074). This showed marked selectivity toward catalysis of the S-isomer (RP101075) only. Docking into MAO B crystal structure suggested that although both the isomers occupied its active site, only the orientation of RP101075 presented the C-H on the α-carbon that was ideal for the C-H bond cleavage, which is a requisite for oxidative deamination. These studies explain the basis for the observed interspecies differences in the metabolism of ozanimod as well as the substrate stereospecificity for formation of CC112273. SIGNIFICANCE STATEMENT: This study evaluates the enzymology and the species differences of the major circulating metabolite of ozanimod, CC112273. Additionally, the study also explores the influence of stereochemistry on MAO B-catalyzed reactions. The study is of significance to the DMD readers given that this oxidation is catalyzed by a non-cytochrome P450 enzyme, and that marked species difference and notable stereospecificity was observed in MAO B-catalyzed biotransformation when the indaneamine enantiomers were used as substrates.


Subject(s)
Indans/pharmacokinetics , Monoamine Oxidase/metabolism , Oxadiazoles/pharmacokinetics , Animals , Biotransformation , Deamination , Drug Evaluation, Preclinical , Haplorhini , Humans , Indans/blood , Metabolic Clearance Rate , Mice , Mitochondria, Liver/metabolism , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Oxadiazoles/blood , Oxidation-Reduction , Rats , Species Specificity , Sphingosine 1 Phosphate Receptor Modulators/blood , Sphingosine 1 Phosphate Receptor Modulators/pharmacokinetics , Stereoisomerism
2.
Toxicol Appl Pharmacol ; 343: 29-39, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29458138

ABSTRACT

5-Methoxy-2-aminoindane (MEAI) is a novel psychoactive aminoindane derivative, exerting euphoric, alcohol-like tipsy experience and reduced desire to consume alcoholic beverages. Our previous toxicological evaluation of MEAI in rats, clearly indicated MEAI's potential to be further evaluated as a promising binge mitigating agent due to its favorable safety profile. In the light of these observations, we have determined MEAI's pharmacokinetic (PK) profile in rats and evaluated in-vitro its pharmacodynamics (PD) profile. Following oral and intravenous administration of MEAI, two metabolites were identified, namely, N-acetyl-MEAI and 5-hydroxy-N-acetyl-AI, arising from N-acetylation and oxidative demethylation. The PK-parameters of MEAI and N-acetyl-MEAI were derived from single i.v. bolus (10 mg/kg) and single oral doses (10 and 90 mg/kg) of MEAI to rats. MEAI displayed extensive total clearance (2.8 L/h/kg) and a very short plasma and brain half-life (0.5-0.7 h). At 10 mg/kg, MEAI displayed low oral bioavailability (25%) and a plasma to brain ratio in the range of 3-5.5, with brain MEAI peak levels attained rapidly. Non-linear pharmacokinetic behavior was observed in the 90 mg/kg oral group, in which the bioavailability increased by 500%. The non-linear behavior was also evident by the significant increase in plasma half-life of MEAI and its metabolite, N-acetyl-MEAI. N-acetyl-MEAI levels in plasma and brain were about ten times lower than the parent compound, indicative of its minor contribution to MEAI's pharmacological effect. MEAI displayed weak to moderate ligand binding inhibition at the 5-HT2B receptor, while the remaining neurochemical targets were unaffected. Further studies, in non-rodent species are required, in-order to assess MEAI's PK and PD profile adequately.


Subject(s)
Binge Drinking/metabolism , Brain/metabolism , Indans/chemistry , Indans/pharmacokinetics , Administration, Intravenous , Administration, Oral , Animals , Binge Drinking/drug therapy , Brain/drug effects , CHO Cells , Cricetinae , Cricetulus , Drug Evaluation, Preclinical/methods , Humans , Indans/administration & dosage , Male , Rats , Rats, Sprague-Dawley
3.
Pak J Pharm Sci ; 30(1): 103-97, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28603119

ABSTRACT

Chongmyungtang (CMT) is a famous Korean herbal medicine for improving learning and memory, which has been reported to have anti-cholinergic and neuroprotective effects. Therefore, drug-drug interactions were examined between CMT and donepezil as a first screening of combination therapy for cognitive deficits. Rats received oral co-administration of donepezil with distilled water as a control or donepezil with CMT as a combination. The distilled water or CMT was co-administered at intervals within 5min after donepezil or 1.5h intervals. The plasma samples were analyzed for donepezil concentration and its pharmacokinetic parameters of Tmax, Cmax, AUC, t1/2 and MRTinf. In the single co-administration at intervals within 5min, donepezil was detected lower in the combination than control at 0.5h and 2h post-treatment (P<0.05). In addition, the combination showed significant increases in MRTinf compared to the control (P<0.05). This suggests drug-drug interactions between donepezil and CMT in the co-administration within 5 min. However, no meaningful differences were found in the pharmacokinetic profiles of donepezil by single dosing with CMT at 1.5h intervals and even by the repeated dosing for a week at 1.5h intervals potential combination therapy of donepezil with CMT.


Subject(s)
Cholinesterase Inhibitors/pharmacokinetics , Herb-Drug Interactions , Indans/pharmacokinetics , Medicine, Korean Traditional , Piperidines/pharmacokinetics , Plant Extracts/administration & dosage , Administration, Oral , Animals , Area Under Curve , Cholinesterase Inhibitors/administration & dosage , Donepezil , Half-Life , Indans/administration & dosage , Indans/blood , Male , Metabolic Clearance Rate , Phytotherapy , Piperidines/administration & dosage , Piperidines/blood , Plants, Medicinal , Rats, Sprague-Dawley
4.
Drugs ; 74(4): 465-88, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24604791

ABSTRACT

QVA149 (indacaterol/glycopyrronium) [Xoterna(®) Breezhaler(®), Ultibro(®) Breezhaler(®)] is an inhaled fixed-dose combination of indacaterol (a long-acting selective ß2-adrenergic receptor agonist [LABA]) and glycopyrronium (a long-acting muscarinic receptor antagonist [LAMA]) that has been approved in the EU and Japan for the symptomatic control of chronic obstructive pulmonary disease (COPD) in adults. In phase III studies, QVA149 significantly improved bronchodilation versus indacaterol, glycopyrronium or tiotropium alone and the LABA/inhaled corticosteroid fixed-dose combination salmeterol/fluticasone. These improvements in lung function, which were rapid in onset and maintained during long-term treatment, were generally associated with significant improvements in dyspnoea, health status, COPD exacerbation risk, patient symptoms, and rescue medication use. The SHINE and ILLUMINATE studies in low (exacerbation) risk patients with moderate to severe disease suggest that QVA149 may offer more symptomatic relief than tiotropium and salmeterol/fluticasone. Similarly, the SPARK study in high (exacerbation) risk patients with severe or very severe disease showed that QVA149 was more effective than glycopyrronium in preventing moderate to severe exacerbations, and suggests that QVA149 may offer more symptomatic relief than LAMA monotherapy. Another phase III study comparing QVA149 with salmeterol/fluticasone in high-risk patients with moderate to very severe disease (FLAME) is ongoing. QVA149 is generally well tolerated, with no new safety signals identified compared with its monocomponents. Bronchodilators remain central to the symptomatic management of COPD. When dual bronchodilation is indicated, QVA149 offers the convenience of two bronchodilators in a single inhaler coupled with a simple, once-daily dosing regimen that may encourage better treatment adherence. Therefore, it is a valuable option in the treatment of COPD.


Subject(s)
Bronchodilator Agents/therapeutic use , Glycopyrrolate/analogs & derivatives , Indans/therapeutic use , Pulmonary Disease, Chronic Obstructive/drug therapy , Quinolones/therapeutic use , Administration, Inhalation , Bronchodilator Agents/administration & dosage , Bronchodilator Agents/adverse effects , Bronchodilator Agents/pharmacokinetics , Clinical Trials, Phase III as Topic , Drug Combinations , Glycopyrrolate/administration & dosage , Glycopyrrolate/adverse effects , Glycopyrrolate/pharmacokinetics , Glycopyrrolate/therapeutic use , Humans , Indans/administration & dosage , Indans/adverse effects , Indans/pharmacokinetics , Pulmonary Disease, Chronic Obstructive/diagnosis , Quinolones/administration & dosage , Quinolones/adverse effects , Quinolones/pharmacokinetics , Respiratory Function Tests , Severity of Illness Index , Treatment Outcome
5.
Expert Opin Drug Metab Toxicol ; 10(1): 129-37, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24295085

ABSTRACT

INTRODUCTION: Indacaterol is a ß2-agonist with a rapid onset of action and a bronchodilating effect that lasts for 24 h. AREAS COVERED: This review considers indacaterol in chronic obstructive pulmonary disease patients, in whom it is rapidly absorbed into the systemic circulation with serum levels measurable after 5 min and Cmax being reached approximately 15 min post-dose. Its disposition kinetics are characterized by at least two phases, a relatively fast decline of the concentrations within the first 12 h, followed by a terminal elimination phase. The increase in systemic exposure is dose-proportional, but systemic concentrations are low at the recommended doses. Indacaterol is relatively highly bound to plasma proteins regardless of concentration. Metabolic clearance and/or biliary clearance account for the majority of its systemic excretion. Weight, age, gender and ethnicity significantly influence its pharmacokinetic profile, but it is not necessary to adjust the dose based on these covariates. Substrates, inhibitors or inducers of UGT1A1 and CYP3A may also affect the pharmacokinetic profile of indacaterol. EXPERT OPINION: Blood concentrations of indacaterol are unable to predict its bronchodilator effects. Furthermore, at the recommended doses, systemic concentrations of indacaterol are low and this is the likely reason for its safe profile.


Subject(s)
Indans/pharmacokinetics , Indans/therapeutic use , Pulmonary Disease, Chronic Obstructive/drug therapy , Quinolones/pharmacokinetics , Quinolones/therapeutic use , Animals , Bronchodilator Agents/pharmacokinetics , Bronchodilator Agents/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Interactions , Humans , Indans/adverse effects , Indans/blood , Quinolones/adverse effects , Quinolones/blood
6.
J Chem Inf Model ; 53(8): 2033-46, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-23777291

ABSTRACT

Acetylcholinesterase (AChE) is a main drug target, and its inhibitors have demonstrated functionality in the symptomatic treatment of Alzheimer's disease (AD). In this study, a series of novel AChE inhibitors were designed and their inhibitory activity was evaluated with 2D quantitative structure-activity relationship (QSAR) studies using a training set of 20 known compounds for which IC50 values had previously been determined. The QSAR model was calculated based on seven unique descriptors. Model validation was determined by predicting IC50 values for a test set of 20 independent compounds with measured IC50 values. A correlation analysis was carried out comparing the statistics of the measured IC50 values with predicted ones. These selectivity-determining descriptors were interpreted graphically in terms of principal component analyses (PCA). A 3D pharmacophore model was also created based on the activity of the training set. In addition, absorption, distribution, metabolism, and excretion (ADME) descriptors were also determined to evaluate their pharmacokinetic properties. Finally, molecular docking of these novel molecules into the AChE binding domain indicated that three molecules (6c, 7c, and 7h) should have significantly higher affinities and solvation energies than the known standard drug donepezil. The docking studies of 2H-thiazolo[3,2-a]pyrimidines (6a-6j) and 5H-thiazolo[3,2-a] pyrimidines (7a-7j) with human AChE have demonstrated that these ligands bind to the dual sites of the enzyme. Simple and ecofriendly syntheses and diastereomeric crystallizations of 2H-thiazolo [3,2-a]pyrimidines and 5H-thiazolo[3,2-a] pyrimidines are described. The solid-state structures for the HBr salts of compounds 6a, 6e, 7a, and 7i have been determined using single-crystal X-ray diffraction techniques, and X-ray powder patterns were measured for the bulk solid remaining after solvent was removed from solutions containing 6a and 7a. These studies provide valuable insight for designing more potent and selective inhibitors for the treatment of AD.


Subject(s)
Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Drug Design , Molecular Docking Simulation , User-Computer Interface , Acetylcholinesterase/chemistry , Chemistry Techniques, Synthetic , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacokinetics , Donepezil , Drug Evaluation, Preclinical , Humans , Indans/chemical synthesis , Indans/metabolism , Indans/pharmacokinetics , Indans/pharmacology , Piperidines/chemical synthesis , Piperidines/metabolism , Piperidines/pharmacokinetics , Piperidines/pharmacology , Protein Conformation , Quantitative Structure-Activity Relationship
7.
Chemistry ; 19(21): 6641-9, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23536497

ABSTRACT

Acetylcholinesterase (AChE) inhibition is one of the most currently available therapies for the management of Alzheimer's disease (AD) symptoms. In this context, NMR spectroscopy binding studies were accomplished to explain the inhibition of AChE activity by Salvia sclareoides extracts. HPLC-MS analyses of the acetone, butanol and water extracts eluted with methanol and acidified water showed that rosmarinic acid is present in all the studied samples and is a major constituent of butanol and water extracts. Moreover, luteolin 4'-O-glucoside, luteolin 3',7-di-O-glucoside and luteolin 7-O-(6''-O-acetylglucoside) were identified by MS(2) and MS(3) data acquired during the LC-MS(n) runs. Quantification of rosmarinic acid by HPLC with diode-array detection (DAD) showed that the butanol extract is the richest one in this component (134 µg mg(-1) extract). Saturation transfer difference (STD) NMR spectroscopy binding experiments of S. sclareoides crude extracts in the presence of AChE in buffer solution determined rosmarinic acid as the only explicit binder for AChE. Furthermore, the binding epitope and the AChE-bound conformation of rosmarinic acid were further elucidated by STD and transferred NOE effect (trNOESY) experiments. As a control, NMR spectroscopy binding experiments were also carried out with pure rosmarinic acid, thus confirming the specific interaction and inhibition of this compound against AChE. The binding site of AChE for rosmarinic acid was also investigated by STD-based competition binding experiments using Donepezil, a drug currently used to treat AD, as a reference. These competition experiments demonstrated that rosmarinic acid does not compete with Donepezil for the same binding site. A 3D model of the molecular complex has been proposed. Therefore, the combination of the NMR spectroscopy based data with molecular modelling has permitted us to detect a new binding site in AChE, which could be used for future drug development.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/isolation & purification , Cholinesterase Inhibitors/pharmacology , Cinnamates/isolation & purification , Cinnamates/pharmacology , Depsides/isolation & purification , Depsides/pharmacology , Glucosides/isolation & purification , Glucosides/pharmacology , Luteolin/isolation & purification , Luteolin/pharmacology , Salvia/chemistry , Binding Sites , Cholinesterase Inhibitors/chemistry , Chromatography, High Pressure Liquid , Cinnamates/chemistry , Depsides/chemistry , Donepezil , Glucosides/chemistry , Indans/pharmacokinetics , Indans/therapeutic use , Luteolin/chemistry , Models, Molecular , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Piperidines/pharmacokinetics , Piperidines/therapeutic use , Plant Extracts/chemistry , Plant Extracts/pharmacology , Portugal , Rosmarinic Acid
9.
J Pharm Pharmacol ; 57(2): 191-5, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15720782

ABSTRACT

Fadolmidine, a novel selective alpha2-adrenoceptor agonist, was evaluated for its efficacy to lower intraocular pressure in normotensive rabbits (n=5-6). The dose-response profile between 0.004 microg and 12.5 microg of fadolmidine was determined. The effect of pH on the partition of fadolmidine was studied in order to select an optimal pH for topical fadolmidine administration. After topical administration, fadolmidine significantly lowered the intraocular pressure in normotensive rabbits. The onset of action was immediate, with no initial increase in intraocular pressure. A significant decrease in intraocular pressure was already observed at 1 h after dosing. The maximum decrease in intraocular pressure was observed after a 2.5 microg dose of fadolmidine in both eyes at 2 h after dosing. The mean maximum decrease in the treated and untreated eye was 6.4 mmHg and 3.9 mmHg, respectively. In conclusion, fadolmidine is a potent intraocular pressure lowering agent. In addition, fadolmidine does not cause a significant initial increase in intraocular pressure. Because of the strong dependence of the distribution coefficient on pH, the pH of the administered solution is important, with physiological pH being optimal in this respect.


Subject(s)
Imidazoles/pharmacokinetics , Indans/pharmacokinetics , Ocular Hypotension/chemically induced , Animals , Biological Availability , Chemistry, Pharmaceutical/methods , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Finland , Hydrogen-Ion Concentration , Imidazoles/administration & dosage , Imidazoles/chemistry , Indans/administration & dosage , Indans/chemistry , Intraocular Pressure/drug effects , Intraocular Pressure/physiology , Male , Rabbits
10.
J Physiol Pharmacol ; 55(1 Pt 1): 113-26, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15082872

ABSTRACT

In the present study we examined the effects of 1-aminoindan-1,5-dicarboxylic acid (AIDA), regarded as a selective and competitive mGluR1 antagonist, in animal models of anxiety. Diazepam (1-10 mg/kg) was used as a reference drug. After intraperitoneal administration, AIDA (0.5-2 mg/kg) produced anxiolytic-like effects in the conflict drinking test and the elevated plus-maze test in rats; however, in doses up to 8 mg/kg, it was inactive in the four-plate test in mice. AIDA tested at the effective doses in the conflict drinking test changed neither the treshold current nor water intake in rats compared to vehicle treatment. AIDA (in a dose of 4 mg/kg, but not lower) increased the exploratory locomotor activity of rats measured in the open-field test, but it did not disturb rat motor coordination in the rota-rod test. The above results indicate that selective mGluR1 antagonist AIDA induces antianxiety-like effects at a low risk of acute side effects characteristic of benzodiazepines. Further studies are required to identify the sites and the mechanism of action of AIDA.


Subject(s)
Anxiety/chemically induced , Indans/adverse effects , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/drug effects , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/pharmacokinetics , Anxiety/drug therapy , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Conflict, Psychological , Diazepam/administration & dosage , Diazepam/pharmacokinetics , Discrimination Learning/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Electric Stimulation/methods , Exploratory Behavior/drug effects , Indans/administration & dosage , Indans/pharmacokinetics , Injections, Intraperitoneal , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Motor Activity/drug effects , Physical Endurance/drug effects , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/administration & dosage , Water Deprivation/physiology
11.
CNS Drug Rev ; 9(4): 343-58, 2003.
Article in English | MEDLINE | ID: mdl-14647528

ABSTRACT

Traumatic brain injury (TBI) is the most common cause of mortality and morbidity in adults under 40 years of age in industrialized countries. Worldwide the incidence is increasing, about 9.5 million people are hospitalized per year due to TBI, and the death rate is estimated to be more than one million people per year. Recently BAY 38-7271 has been characterized as a structurally novel, selective and highly potent cannabinoid CB1/CB2 receptor agonist in vitro and in vivo with pronounced neuroprotective efficacy in a rat traumatic brain injury model, showing a therapeutic window of at least 5 h. Furthermore, neuroprotective efficacy was also found in models of transient and permanent occlusion of the middle cerebral artery and brain edema models as well. In this article we review the in vitro and in vivo pharmacology of BAY 38-7271, the results from acute and subacute toxicity studies, pharmacokinetics and drug metabolism in animals and healthy male volunteers. In phase I studies BAY 38-7271 was safe and well tolerated when administered by i.v. infusion for either 1 or 24 h. As the doses of BAY 38-7271 in animals needed for maximal neuroprotective efficacy were significantly lower than those inducing typical cannabinoid-like side effects, it is to be expected that the compound will offer a novel therapeutic approach with a favorable therapeutic window for the treatment of TBI or cerebral ischemia.


Subject(s)
Brain Injuries/drug therapy , Cannabinoid Receptor Agonists , Indans/therapeutic use , Neuroprotective Agents/therapeutic use , Sulfonic Acids/therapeutic use , Animals , Area Under Curve , Half-Life , Humans , Indans/metabolism , Indans/pharmacokinetics , Metabolic Clearance Rate , Sulfonic Acids/metabolism , Sulfonic Acids/pharmacokinetics
12.
Neuropharmacology ; 40(7): 866-78, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11378157

ABSTRACT

CHF3381 [n-(2-indanyl)-glycinamide hydrochloride] has been selected on the basis of a screening program as the compound displaying the highest anticonvulsant activity in the maximal electroshock seizure (MES) test and the best therapeutic index with reference to the rotarod test in mice and rats. In this study, the antiepileptic activity and the behavioural toxicity of CHF3381 were characterised in multiple model systems. CHF3381 effectively prevented MES-induced convulsions when administered i.p. (ED50, 24 mg/kg and 7.5 mg/kg) or p.o. (ED50, 21 mg/kg and 21 mg/kg) in both mice and rats, respectively. The time course of oral anti-MES activity in the rat was related to the brain concentration profile of unchanged CHF3381. Interestingly, the brain drug levels were about 4-5 times higher than in plasma. CHF3381 was very effective in mice against picrotoxin-, and i.c.v. N-methyl-D-aspartate (NMDA)-induced hind limb tonic extension (ED50 Approximately/=10 mg/kg), but was a weaker antagonist of 4-amynopyridine- and bicuculline-induced tonic seizures (ED50 approximately/=100 mg/kg), and ineffective against pentylentetrazole- and picrotoxin-induced clonic seizures. CHF3381 antagonised the behavioural effects and lethality of i.p. administered NMDA (ED50 = 57 mg/kg p.o.), indicating that the compound may act as a functional NMDA antagonist. In keeping with this idea, CHF3381 weakly displaced [(3)H]-TCP from binding to NMDA receptor channels (Ki, 8.8 microM). In the rat amygdala kindling model, CHF3381 was more efficient against kindling development than against kindled seizures (minimally active dose = 80 vs. 120 mg/kg i.p). Furthermore, it significantly increased the seizure threshold in kindled rats at relatively low doses (40 mg/kg i.p.). In contrast with MK-801-induced hyperactivity, CHF3381 moderately reduced the spontaneous locomotor activity in mice at anticonvulsant doses. Toxic effects on motor performance (rotarod test) were found at high doses only (TD50 approximately/= 300 mg/kg p.o., congruent with 100 mg/kg i.p. in both mice and rats). Furthermore, CHF3381 did not impair passive avoidance and Morris water maze responding in the therapeutic range of doses. Finally, the development of tolerance after repeated doses was negligible. These data indicate that CHF3381 exerts anticonvulsant and antiepileptogenic effects in various seizure models and possesses good therapeutic window, with scarce propensity to cause neurological side-effects.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Glycine/therapeutic use , Indans/therapeutic use , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacokinetics , Dizocilpine Maleate/pharmacology , Dizocilpine Maleate/therapeutic use , Drug Evaluation, Preclinical/methods , Epilepsy/chemically induced , Epilepsy/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Glycine/analogs & derivatives , Glycine/chemistry , Glycine/pharmacokinetics , Indans/chemistry , Indans/pharmacokinetics , Kindling, Neurologic/drug effects , Kindling, Neurologic/metabolism , Lamotrigine , Male , Mice , Motor Activity/drug effects , Motor Activity/physiology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Triazines/pharmacology , Triazines/therapeutic use
13.
J Med Chem ; 43(18): 3386-99, 2000 Sep 07.
Article in English | MEDLINE | ID: mdl-10978186

ABSTRACT

Recent results from human clinical trials have established the critical role of HIV protease inhibitors in the treatment of acquired immune-deficiency syndrome (AIDS). However, the emergence of viral resistance, demanding treatment protocols, and adverse side effects have exposed the urgent need for a second generation of HIV protease inhibitors. The continued exploration of our hydroxylaminepentanamide (HAPA) transition-state isostere series of HIV protease inhibitors, which initially resulted in the identification of Crixivan (indinavir sulfate, MK-639, L-735,524), has now yielded MK-944a (L-756,423). This compound is potent, is selective, and competitively inhibits HIV-1 PR with a K(i) value of 0.049 nM. It stops the spread of the HIV(IIIb)-infected MT4 lymphoid cells at 25.0-50.0 nM, even in the presence of alpha(1) acid glycoprotein, human serum albumin, normal human serum, or fetal bovine serum. MK-944a has a longer half-life in several animal models (rats, dogs, and monkeys) than indinavir sulfate and is currently in advanced human clinical trials.


Subject(s)
Antiviral Agents/chemical synthesis , HIV Protease Inhibitors/chemical synthesis , HIV-1/drug effects , Indans/chemical synthesis , Piperazines/chemical synthesis , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Cattle , Cell Culture Techniques , Dogs , Drug Evaluation, Preclinical , Drug Resistance, Microbial , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/pharmacology , Haplorhini , Humans , Indans/chemistry , Indans/pharmacokinetics , Indans/pharmacology , Male , Piperazines/chemistry , Piperazines/pharmacokinetics , Piperazines/pharmacology , Protein Binding , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Urinary Calculi/chemically induced , Urinary Calculi/urine
14.
Eur J Pharmacol ; 401(3): 419-28, 2000 Aug 11.
Article in English | MEDLINE | ID: mdl-10936502

ABSTRACT

For progression to clinical trials in stroke, putative neuroprotective compounds should show robust efficacy post-ischaemia in several experimental models of stroke. This paper describes the characterisation of (+)(1S, 2R)-cis-1-[4-(1-methyl-1-phenylethyl)phenoxy]-2-methylamino indane hydrochloride (SB-221420-A), a Ca(2+) and Na(+) channel antagonist. SB-221420-A inhibited (IC(50)=2.2 microM) N-type voltage-operated Ca(2+) channel currents in cultured superior cervical ganglion neurons, which were pretreated with 10 microM nimodipine to block L-type voltage-operated Ca(2+) channel currents. In dorsal root ganglion neurons pretreated with 1 microM omega-conotoxin GVIA to block N-type voltage-operated Ca(2+) channel currents, SB-221420-A inhibited the residual Ca(2+) current with an IC(50) of 7 microM. SB-221420-A also inhibited Na(+) currents in dorsal root ganglion neurons with an IC(50) of 8 microM. In rats, the pharmacokinetic profile of SB-221420-A shows that it has a half-life of 6.4 h, a high volume of distribution, is highly brain penetrating, and has no persistent metabolites. Following bilateral carotid artery occlusion in gerbils, SB-221420-A significantly reduced the level of ischaemia-induced hyperlocomotor activity and the extent of hippocampal CA1 cell loss compared to the ischaemic vehicle-treated group. SB-221420-A was also effective in focal models of ischaemia. In the mouse permanent middle cerebral artery occlusion model, SB-221420-A (10 mg/kg) administered intravenously, post-ischaemia significantly (P<0.05) reduced lesion volume compared to the ischaemic vehicle-treated group. In the normotensive rat permanent middle cerebral artery occlusion model, SB-221420-A (10 mg/kg) administered intravenously over 1 h, beginning 30 min postmiddle cerebral artery occlusion, significantly (P<0.05) reduced lesion volume from 291+/-16 to 153+/-30 mm(3), compared to ischaemic vehicle-treated controls when measured 24 h postmiddle cerebral artery occlusion. Efficacy was maintained when the same total dose of SB-221420-A was infused over a 6-h period, beginning 30 min postmiddle cerebral artery occlusion. SB-221420-A also significantly (P<0.05) reduced lesion volume following transient middle cerebral artery occlusion in normotensive rats and permanent middle cerebral artery occlusion in spontaneously hypertensive rats (SHR). Investigation of the side effect profile using the Irwin screen in mice revealed that, at neuroprotective doses, there were no overt behavioural or cardiovascular changes. These data demonstrate that robust neuroprotection can be seen post-ischaemia with SB-221420-A in both global and focal ischaemia with no adverse effects at neuroprotective doses, and indicate the potential utility of a mixed cation blocker to improve outcome in cerebral ischaemia.


Subject(s)
Calcium Channel Blockers/pharmacology , Indans/pharmacology , Neuroprotective Agents/pharmacology , Sodium Channel Blockers , Stroke/prevention & control , Anesthesia , Animals , Animals, Newborn , Brain/drug effects , Brain/pathology , Carotid Stenosis/physiopathology , Carotid Stenosis/prevention & control , Cells, Cultured , Consciousness , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Gerbillinae , Hemodynamics/drug effects , Hypertension/physiopathology , Indans/pharmacokinetics , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/prevention & control , Ischemic Attack, Transient/physiopathology , Ischemic Attack, Transient/prevention & control , Male , Membrane Potentials/drug effects , Metabolic Clearance Rate , Mice , Motor Activity/drug effects , Neurons, Afferent/cytology , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Stroke/physiopathology , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL