Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Pharmacol ; 45(5): 529-37, 2005 May.
Article in English | MEDLINE | ID: mdl-15831776

ABSTRACT

The pharmacokinetics and pharmacodynamics of adinazolam mesylate (10 mg), N-desmethyl adinazolam mesylate (NDMAD, 10 mg), and alprazolam (1 mg) were investigated in 9 healthy male subjects in a randomized, blinded, single-dose, 4-way crossover study. All drugs were intravenously infused over 30 minutes. Plasma adinazolam, NDMAD, and alprazolam concentrations, electroencephalographic (EEG) activity in the beta (12-30 Hz) range, performance on the Digit Symbol Substitution Test (DSST), and subjective measures of mood and sedation were monitored for 12 to 24 hours. Mean pharmacokinetic parameters for adinazolam, NDMAD, and alprazolam, respectively, were as follows: volume of distribution (L), 106, 100, and 77; elimination half-life (hours), 2.9, 2.8, and 14.6; and clearance (mL/min), 444, 321, and 84. More than 80% of the total infused adinazolam dose was converted to systemically appearing NDMAD. All 3 benzodiazepine agonists significantly increased beta EEG activity, with alprazolam showing the strongest agonist activity and adinazolam showing the weakest activity. Alprazolam and NDMAD significantly decreased DSST performance, whereas adinazolam had no effect relative to placebo. Adinazolam, NDMAD, and alprazolam all produced significant observer-rated sedation. Plots of EEG effect versus plasma alprazolam concentration demonstrated counterclockwise hysteresis, consistent with an effect site delay. This was incorporated into a kinetic-dynamic model in which hypothetical effect site concentration was related to pharmacodynamic EEG effect via the sigmoid E(max) model, yielding an effect site equilibration half-life of 4.8 minutes. The exponential effect model described NDMAD pharmacokinetics and EEG pharmacodynamics. The relation of both alprazolam and NDMAD plasma concentrations to DSST performance could be described by a modified exponential model. Pharmacokinetic-dynamic modeling was not possible for adinazolam, as the data did not conform to any known concentration-effect model. Collectively, these results indicate that the benzodiazepine-like effects occurring after adinazolam administration are mediated by mainly NDMAD.


Subject(s)
Alprazolam/pharmacology , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Benzodiazepines/pharmacology , Adult , Alprazolam/pharmacokinetics , Anti-Anxiety Agents/pharmacokinetics , Antidepressive Agents/pharmacokinetics , Benzodiazepines/pharmacokinetics , Cross-Over Studies , Electroencephalography , Emotions/drug effects , Half-Life , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Psychomotor Performance/drug effects
2.
Neuropsychopharmacology ; 29(10): 1823-30, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15138441

ABSTRACT

Sensorimotor gating, which is severely disrupted in schizophrenic patients, can be measured by assessing prepulse inhibition of the acoustic startle response (PPI). Acute administration of D2-like receptor agonists such as quinpirole reduces PPI, but tolerance occurs upon repeated administration. In the present study, PPI in rats was reduced by acute quinpirole (0.1 mg/kg, s.c.), but not following repeated quinpirole treatment once daily for 28 days. Repeated quinpirole treatment did not alter the levels of basal-, forskolin- (5 microM), or SKF 82958- (10 microM) stimulated adenylate cyclase activity in the nucleus accumbens (NAc), but significantly increased cAMP-dependent protein kinase (PKA) activity. Phosphorylation of cAMP response element-binding protein (CREB) was significantly greater in the NAc after repeated quinpirole treatment than after repeated saline treatment with or without acute quinpirole challenge. Activation of PKA by intra-accumbens infusion of the cAMP analog, Sp-cAMPS, prevented acute quinpirole-induced PPI disruption, similar to the behavioral effect observed following repeated quinpirole treatment. Thus, repeated quinpirole treatment increases NAc PKA activity and CREB phosphorylation, and this neuroadaptive response might facilitate the recovery of sensorimotor gating in schizophrenia.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine Antagonists/pharmacology , Nucleus Accumbens/metabolism , Quinpirole/antagonists & inhibitors , Quinpirole/pharmacology , Reflex, Startle/drug effects , Acoustic Stimulation , Adenylyl Cyclases/metabolism , Animals , Dose-Response Relationship, Drug , Enzyme Activators/pharmacology , Immunohistochemistry , Male , Nucleus Accumbens/drug effects , Phosphorylation , Rats , Rats, Sprague-Dawley
3.
J Clin Pharmacol ; 44(6): 605-11, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15145968

ABSTRACT

The objective of this study was to evaluate the kinetics and dynamics of midazolam when administered by three different infusion schemes, using electroencephalography to measure pharmacodynamic effects. In a three-way crossover study, 8 volunteers received midazolam (0.1 mg/kg) by constant-rate intravenous infusion. The durations of midazolam infusions for the three trials were 1 minute, 1 hour, and 3 hours. Plasma midazolam concentrations and electroencephalographic (EEG) activity in the 13- to 30-Hz range were monitored for 24 hours. Based on separate analysis of each subject-trial, mean values for volume of distribution and distribution or elimination half-life did not significantly vary. Central compartment volume and clearance differed among the three midazolam infusion trials; however, the magnitude of change was small. EEG activity in the 13- to 30-Hz range significantly increased for all three midazolam infusion trials. Plots of midazolam plasma concentration versus pharmacodynamic EEG effect for the 1-hour and 3-hour infusion trials did not reveal evidence of either counterclockwise or clockwise hysteresis. Plots from the 1-minute infusion trial demonstrated counterclockwise hysteresis, consistent with an equilibration effect-site delay. This was incorporated into a kinetic-dynamic model in which hypothetical effect-site concentration was related to pharmacodynamic EEG effect via the sigmoid E(max) model. Analysis of all three infusion trials together yielded the following mean estimates: maximum EEG effect, 16.3% over baseline; 50% maximum effective concentration, 31 ng/mL; and an apparent rate constant for drug disappearance from the effect compartment which approached infinity. Despite the delay in effect onset during the 1-minute midazolam infusion, midazolam infusions in duration of up to 3 hours produce CNS sedation without evidence of tolerance.


Subject(s)
Electroencephalography/drug effects , Hypnotics and Sedatives/pharmacokinetics , Midazolam/pharmacokinetics , Adult , Cross-Over Studies , Half-Life , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacology , Infusions, Intravenous , Male , Metabolic Clearance Rate , Midazolam/administration & dosage , Midazolam/pharmacology
4.
J Pharmacol Exp Ther ; 308(2): 487-94, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14593083

ABSTRACT

Sensorimotor gating, a neural process severely disrupted in patients with schizophrenia, can be measured by assessing prepulse inhibition (PPI) of acoustic startle responses. PPI is disrupted in experimental animals by stimulation of D(2)-like dopamine receptors in the nucleus accumbens (NAc). We examined the effect of repeated treatment with a selective dopamine D(2)-like receptor agonist, quinpirole, and characterized the molecular substrates of the resulting PPI adaptation. Animals were treated once daily for 10 or 28 consecutive days with quinpirole (0.0, 0.05, 0.1, or 0.3 mg/kg, s.c.), and the effect on PPI was assessed throughout the treatment period. PPI was reduced after acute quinpirole administration, but gradually increased with repeated treatment. Quinpirole-induced PPI disruption was attenuated after 10 days of treatment at lower doses, but complete recovery was not apparent until the treatment period was extended to 28 days. Since chronic drug exposure can alter the dopamine system, we sought to characterize the effects of repeated quinpirole treatment on G proteins coupled to D(2)-like receptors in the NAc. Guanosine 5'-O-(3-[(35)S]thiotriphosphate) ([(35)S]GTPgammaS) binding and Western blot analysis revealed that repeated quinpirole treatment had no effect on NAc D(2)-like receptor G protein function or G protein levels. These data indicate that repeated activation of D(2)-like receptors by quinpirole produces tolerance in the absence of receptor or G protein changes, suggesting that the locus of dopaminergic adaptation might be at the intracellular level.


Subject(s)
GTP-Binding Proteins/metabolism , Neural Inhibition/drug effects , Quinpirole/pharmacology , Receptors, Dopamine D2/metabolism , Animals , Dopamine Agonists/administration & dosage , Dopamine Agonists/pharmacology , GTP-Binding Proteins/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Motor Activity/drug effects , Psychomotor Performance/drug effects , Quinpirole/administration & dosage , Rats , Rats, Sprague-Dawley
5.
Brain Res ; 982(1): 12-8, 2003 Aug 22.
Article in English | MEDLINE | ID: mdl-12915235

ABSTRACT

Prepulse inhibition of the acoustic startle response (PPI) is a cross-species measure of sensorimotor gating, which is severely disrupted in patients with schizophrenia. PPI deficits can be produced in experimental animals by administration of selective D(2)-like dopamine receptor agonists in the nucleus accumbens (NAc). G proteins coupled to these receptors reportedly are altered in the NAc of patients with schizophrenia. Therefore, we sought to determine whether experimental inactivation of intracellular G proteins in the NAc alters PPI. In adult male Sprague-Dawley rats, baseline PPI was determined by presenting acoustic pulse stimuli (120 dB) alone or preceded 100 ms earlier by prepulse stimuli (3, 6 or 12 dB above 70 dB ambient noise). PPI disruption was assessed in the presence of quinpirole (0.0, 0.05, 0.1, 0.5 mg/kg, sc), and pertussis toxin (PTX; 0.05 microg/side) was then infused into the NAc bilaterally. Ten days later, quinpirole-mediated disruption of PPI was significantly reduced; neither PTX alone, nor heat-inactivated PTX had any effect on quinpirole-induced PPI reductions. PPI was significantly higher after PTX infusion upon moderate quinpirole challenge, suggesting that D(2)-like receptors were less effective. PTX treatment significantly reduced basal and dopamine-stimulated [35S]GTPgammaS binding in the NAc core and shell, and reduced G(i)(alpha) protein immunoreactivity in the NAc. The results suggest that PPI disruption mediated by D(2)-like receptor activation in the NAc depends on coupling to G(i) and G(o) proteins, alteration of which could cause sensorimotor gating deficits in schizophrenia.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Heterotrimeric GTP-Binding Proteins/physiology , Motor Activity/physiology , Nucleus Accumbens/metabolism , Proto-Oncogene Proteins/physiology , Sensation/physiology , Acoustic Stimulation , Animals , Behavior, Animal , Dopamine/pharmacology , GTP-Binding Protein alpha Subunit, Gi2 , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Male , Neural Inhibition , Pertussis Toxin/pharmacology , Quinpirole/pharmacology , Rats , Rats, Sprague-Dawley , Reflex, Startle/physiology , Sensation/drug effects
6.
J Clin Pharmacol ; 43(4): 414-22, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12723462

ABSTRACT

Antiretroviral agents may participate in drug interactions that influence the efficacy and toxicity of other antiretrovirals, as well as pharmacologic treatments of coincident or complicating diseases. The viral protease inhibitor, ritonavir, may cause drug interactions by inhibiting the activity of cytochrome P450-3A (CYP3A) isoforms. In a single-dose, blinded, four-way crossover study, 10 healthy volunteer subjects received 50 mg of trazodone hydrochloride or matching placebo concurrent with low-dose ritonavir (four doses of 200 mg each) or with placebo. Compared to the control condition, ritonavir significantly reduced apparent oral clearance of trazodone (155 +/- 23 vs. 75 +/- 12 ml/min, p < 0.001), prolonged elimination half-life (6.7 +/- 0.7 vs. 14.9 +/- 3.9 h, p < 0.05), and increased peak plasma concentrations (842 +/- 64 vs. 1125 +/- 111 ng/ml, p < 0.05) (mean +/- SE). Coadministration of trazodone with ritonavir increased sedation, fatigue, and performance impairment compared to trazodone plus placebo; differences reached significance only for the digitsymbol substitution test. Three subjects experienced nausea, dizziness, or hypotension when trazodone was given with ritonavir; 1 of these subjects also experienced syncope. Thus short-term low-dose administration of ritonavir impairs oral clearance of trazodone and increases the occurrence of adverse reactions. The findings are consistent with impairment of CYP3A-mediated trazodone metabolism by ritonavir.


Subject(s)
HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/pharmacology , Ritonavir/administration & dosage , Ritonavir/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacokinetics , Trazodone/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Chromatography, High Pressure Liquid , Cross-Over Studies , Drug Interactions , Drug Therapy, Combination , Female , Half-Life , Humans , Male , Middle Aged , Selective Serotonin Reuptake Inhibitors/adverse effects , Selective Serotonin Reuptake Inhibitors/blood , Time Factors , Trazodone/adverse effects , Trazodone/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...