Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Antimicrob Agents Chemother ; 59(9): 5503-10, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26124175

ABSTRACT

Hepatitis C virus (HCV) infection is common among people who inject drugs, including those managed with maintenance opioids. Pharmacokinetic interactions between opioids and emerging oral HCV antivirals merit evaluation. Daclatasvir is a potent pangenotypic inhibitor of the HCV NS5A replication complex recently approved for HCV treatment in Europe and Japan in combination with other antivirals. The effect of steady-state daclatasvir (60 mg daily) on stable plasma exposure to oral opioids was assessed in non-HCV-infected subjects receiving methadone (40 to 120 mg; n = 14) or buprenorphine plus naloxone (8 to 24 mg plus 2 to 6 mg; n = 11). No relevant interaction was inferred if the 90% confidence interval (CI) of the geometric mean ratio (GMR) of opioid area under the plasma concentration-time curve over the dosing interval (AUCτ) or maximum concentration in plasma (C max) with versus without daclatasvir was within literature-derived ranges of 0.7 to 1.43 (R- and S-methadone) or 0.5 to 2.0 (buprenorphine and norbuprenorphine). Dose-normalized AUCτ for R-methadone (GMR, 1.08; 90% CI, 0.94 to 1.24), S-methadone (1.13; 0.99 to 1.30), and buprenorphine (GMR, 1.37; 90% CI, 1.24 to 1.52) were within the no-effect range. The norbuprenorphine AUCτ was slightly elevated in the primary analysis (GMR, 1.62; 90% CI, 1.30 to 2.02) but within the no-effect range in a supplementary analysis of all evaluable subjects. Dose-normalized C max for both methadone enantiomers, buprenorphine and norbuprenorphine, were within the no-effect range. Standardized assessments of opioid pharmacodynamics were unchanged throughout daclatasvir administration with methadone or buprenorphine. Daclatasvir pharmacokinetics were similar to historical data. Coadministration of daclatasvir and opioids was generally well tolerated. In conclusion, these data suggest that daclatasvir can be administered with buprenorphine or methadone without dose adjustments.


Subject(s)
Buprenorphine, Naloxone Drug Combination/chemistry , Imidazoles/chemistry , Methadone/chemistry , Buprenorphine/analogs & derivatives , Buprenorphine/chemistry , Carbamates , Drug Interactions , Pyrrolidines , Valine/analogs & derivatives
2.
Clin Pharmacol Ther ; 97(2): 159-66, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25670521

ABSTRACT

Asunaprevir (ASV), an investigational, highly protein-bound inhibitor of hepatitis C virus NS3 protease, shows considerable hepatic compartmentalization in animal models. Preclinical data showed ASV inhibition of human OATP1B1 (IC50 = 0.3 µM), OATP2B1 (0.27 µM), and, to a lesser extent OATP1B3 (3.0 µM), confirmed by modest (<2-fold) clinical elevations in rosuvastatin exposure with concomitant ASV. Although no significant OATP transport of ASV was observed in vitro at standard micromolar assay concentrations, clinical coadministration of ASV with a single dose of the OATP inhibitor rifampin gave large, variable increases in ASV plasma Cmax (21-fold mean) and AUCinf (15-fold mean), consistent with reduced hepatic uptake. In vitro reevaluation at therapeutically relevant low-nanomolar concentrations of unbound ASV showed active, saturable human hepatocyte uptake (Km = 0.685 µM) and rifampin-reversible transport by OATP1B1 and OATP2B1, but not OATP1B3. At therapeutically relevant concentrations, ASV is therefore a sensitive substrate for, and weak inhibitor of, human OATP1B1, 1B3 and 2B1.


Subject(s)
Isoquinolines/metabolism , Isoquinolines/pharmacology , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters/antagonists & inhibitors , Sulfonamides/metabolism , Sulfonamides/pharmacology , Adolescent , Adult , Animals , Biological Transport/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Female , Fluorobenzenes/metabolism , Hepatocytes/metabolism , Humans , Liver-Specific Organic Anion Transporter 1 , Male , Middle Aged , Oocytes/drug effects , Pyrimidines/metabolism , Rifampin/metabolism , Rifampin/pharmacology , Rosuvastatin Calcium , Solute Carrier Organic Anion Transporter Family Member 1B3 , Xenopus laevis , Young Adult
3.
J Clin Pharmacol ; 54(6): 657-64, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24374821

ABSTRACT

The objectives of this study were to evaluate the safety, tolerability and pharmacokinetics (PK) of BMS-986001 as a single oral dose in healthy male subjects. Sixty-four healthy male subjects were randomized to receive a single dose of BMS-986001 or placebo in this single-blind, placebo-controlled, sequential ascending-dose study. There were eight treatment groups (10, 30, 100, 300, 600, and 900 mg fed; and 100 and 300 mg fasted) of eight subjects each (BMS-986001 n = 6/placebo n = 2). BMS-986001 was well tolerated, with no serious adverse events (AEs), deaths, or discontinuations due to AEs reported. AEs were experienced by 14.6% of subjects receiving BMS-986001; however, these did not appear to be dose related and were not considered to be related to study drug. BMS-986001 was rapidly absorbed and exhibited a linear dose-exposure relationship across the dose range studied. PK appeared similar whether administered with or without food. Administration of BMS-986001 as a single dose was generally safe and well tolerated. A linear dose-exposure relationship was seen across all doses studied, with no apparent food effect. Further clinical development is warranted.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Food-Drug Interactions , Reverse Transcriptase Inhibitors/pharmacokinetics , Thymidine/analogs & derivatives , Adult , Anti-HIV Agents/adverse effects , Anti-HIV Agents/blood , Anti-HIV Agents/urine , Dose-Response Relationship, Drug , Fasting/metabolism , Healthy Volunteers , Humans , Male , Middle Aged , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/blood , Reverse Transcriptase Inhibitors/urine , Single-Blind Method , Thymidine/adverse effects , Thymidine/blood , Thymidine/pharmacokinetics , Thymidine/urine , Young Adult
4.
HIV Med ; 12(9): 570-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21569187

ABSTRACT

OBJECTIVE: There are limited antiretroviral options for use in the treatment of HIV infection during pregnancy. The purpose of this study was to assess the safety, efficacy and appropriate dosing regimen for ritonavir (RTV)-boosted atazanavir in HIV-1-infected pregnant women. METHODS: In this nonrandomized, open-label study, HIV-infected pregnant women were dosed with either 300/100 mg (n=20) or 400/100 mg (n=21) atazanavir/RTV once-daily (qd) in combination with zidovudine (300 mg) and lamivudine (150 mg) twice daily in the third trimester. Pharmacokinetic parameters [maximum observed plasma concentration (C(max) ), trough observed plasma concentration 24 hour post dose (C(min) ) and area under concentration-time curve in one dosing interval (AUC(τ) )] were determined and compared with historical values (300/100 mg atazanavir/RTV) for HIV-infected nonpregnant adults (n=23). RESULTS: At or before delivery, all mothers achieved HIV RNA <50 HIV-1 RNA copies/mL and all infants were HIV DNA negative at 6 months of age. The third trimester AUC(τ) for atazanavir/RTV 300/100 mg was 21% lower than historical data, but the C(min) values were comparable. The C(min) value for atazanavir/RTV 400/100 mg was 39% higher than the C(min) for atazanavir/RTV 300/100 mg in historical controls, but the AUC(τ) values were comparable. Twice as many patients in the 400/100 mg group (62%) had an increase in total bilirubin (>2.5 times the upper limit of normal) as in the 300/100 mg group (30%). Atazanavir (ATV) was well tolerated with no unanticipated adverse events. CONCLUSIONS: In this study, use of atazanavir/RTV 300/100 mg qd produced C(min) comparable to historical data in nonpregnant HIV-infected adults. When used in combination with zidovudine/lamivudine, it suppressed HIV RNA in all mothers and prevented mother-to-child transmission of HIV-1 infection. During pregnancy, the pharmacokinetics, safety and efficacy demonstrated that a dose adjustment is not required for ATV.


Subject(s)
HIV Infections/prevention & control , HIV Protease Inhibitors/pharmacokinetics , HIV-1 , Infectious Disease Transmission, Vertical/prevention & control , Oligopeptides/pharmacokinetics , Pregnancy Complications, Infectious/drug therapy , Pyridines/pharmacokinetics , Ritonavir/pharmacokinetics , Adult , Atazanavir Sulfate , CD4 Lymphocyte Count , Drug Administration Schedule , Drug Therapy, Combination , Female , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/transmission , HIV Protease Inhibitors/administration & dosage , HIV-1/immunology , Humans , Oligopeptides/administration & dosage , Pregnancy , Pregnancy Complications, Infectious/virology , Puerto Rico/epidemiology , Pyridines/administration & dosage , Ritonavir/administration & dosage , South Africa/epidemiology , United States/epidemiology , Viral Load
5.
Clin Pharmacol Ther ; 84(6): 698-703, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18528434

ABSTRACT

Atazanavir (ATV) is known to inhibit UGT1A1-mediated glucuronidation. Here we report the effect of ATV and ATV/ritonavir (RTV) on another UGT1A isoenzyme, UGT1A4. Twenty-one healthy volunteers received a single dose of 100 mg of oral lamotrigine on days 1, 13, and 27; on each occasion blood was sampled before the dose was administered and through 120 h after ingestion of the drug. On days 8-17 the subjects received oral ATV 400 mg q.d. On days 18-30 the subjects received oral ATV 300 mg plus oral RTV 100 mg q.d. Seventeen subjects were evaluable for pharmacokinetic analysis. Geometric mean ratios (+90% confidence intervals (CIs)) of lamotrigine area under the plasma concentration-time curve (AUC)(0-inf) and peak plasma concentration (C(max)) for ATV + lamotrigine and for lamotrigine alone were 0.88 (0.86-0.91) and 0.99 (0.95-1.02), respectively; the corresponding ratios for ATV/RTV and for lamotrigine were 0.68 (0.65-0.70) and 0.94 (0.90-0.97), respectively. The mean ratio of lamotrigine-2N-glucuronide to lamotrigine AUC(0-inf) increased from 0.45 for lamotrigine to 0.71 for ATV/RTV + lamotrigine. ATV alone does not significantly influence glucuronidation of lamotrigine. In contrast, ATV/RTV results in moderately decreased exposure to lamotrigine.


Subject(s)
Anti-HIV Agents/pharmacology , Glucuronosyltransferase/drug effects , Glucuronosyltransferase/metabolism , Oligopeptides/pharmacology , Pyridines/pharmacology , Ritonavir/pharmacokinetics , Triazines/pharmacokinetics , Administration, Oral , Adult , Atazanavir Sulfate , Confidence Intervals , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Humans , Lamotrigine , Male , Middle Aged , Molecular Probe Techniques , Phenotype , Prospective Studies , Reference Values , Ritonavir/administration & dosage , Sensitivity and Specificity , Triazines/administration & dosage , Young Adult
6.
Antimicrob Agents Chemother ; 50(10): 3336-42, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17005814

ABSTRACT

Mycobacterium tuberculosis is a concern in patients with human immunodeficiency virus (HIV) infection. Rifampin (RIF), an agent used against M. tuberculosis, is contraindicated with most HIV protease inhibitors. Atazanavir (ATV) has clinical efficacy comparable to a standard of care regimen in naive patients and, when dosed with low-dose ritonavir (RTV), also in treatment-experienced patients. We evaluated here the safety and pharmacokinetics of ATV, resulting from three regimens of ATV, RTV, and RIF in 71 healthy subjects. The pharmacokinetics for ATV and RTV were assessed after 6 and 10 days of dosing with ATV 400 mg (n = 53) and with ATV-RTV at 300 and 100 mg (ATV/RTV 300/100; n = 52), respectively. Steady-state pharmacokinetics for ATV, RTV, RIF, and desacetyl-rifampin (des-RIF) were measured after 10 days of dosing of ATV/RTV/RIF 300/100/600 (n = 17), ATV/RTV/RIF 300/200/600 (n = 17), or ATV/RTV/RIF 400/200/600 (n = 14). An RIF 600-alone arm was enrolled as a control group (n = 18). With ATV/RTV/RIF 400/200/600, ATV area under the concentration-time curve values were comparable, but the C(min) values were lower relative to ATV 400 alone. ATV exposures were substantially reduced for the other RIF-containing regimens relative to ATV 400 alone and for all regimens relative to ATV/RTV 300/100 alone. RIF and des-RIF exposures were 1.6- to 2.5-fold higher than with RIF 600 alone. The incidence of grade 3/4 alanine aminotransferase/aspartate aminotransferase values was limited to 1 subject each in both the ATV/RTV/RIF 300/200/600 and the ATV/RTV/RIF 400/200/600 treatments. Coadministration of ATV with RIF was safe and generally well tolerated. Since ATV exposures were reduced in all regimens, ATV and RIF should not be coadministered at the dosing regimens studied.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Oligopeptides/pharmacokinetics , Pyridines/pharmacokinetics , Rifampin/pharmacokinetics , Ritonavir/pharmacokinetics , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Area Under Curve , Atazanavir Sulfate , Drug Interactions , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Female , Humans , Male , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Pyridines/administration & dosage , Pyridines/adverse effects , Rifampin/administration & dosage , Rifampin/adverse effects , Ritonavir/administration & dosage , Ritonavir/adverse effects
7.
Antimicrob Agents Chemother ; 48(5): 1553-60, 2004 May.
Article in English | MEDLINE | ID: mdl-15105105

ABSTRACT

Coadministration of lopinavir-ritonavir, an antiretroviral protease inhibitor, at the standard dose (400/100 mg twice a day [BID]) with the antituberculous agent rifampin is contraindicated because of a significant pharmacokinetic interaction due to induction of cytochrome P450 3A by rifampin. In the present study, two adjusted-dose regimens of lopinavir-ritonavir were tested in combination with rifampin. Thirty-two healthy subjects participated in a randomized, two-arm, open-label, multiple-dose, within-subject controlled study. All subjects were treated with lopinavir-ritonavir at 400/100 mg BID from days 1 to 15. From days 16 to 24, the subjects in arm 1 received lopinavir-ritonavir at 800/200 mg BID in a dose titration, and the subjects in arm 2 received lopinavir-ritonavir at 400/400 mg BID in a dose titration. Rifampin was given at 600 mg once daily to all subjects from days 11 to 24. The multiple-dose pharmacokinetics of lopinavir, ritonavir, and rifampin were assessed. Twelve of 32 subjects withdrew from the study. For nine subjects lopinavir-ritonavir combined with rifampin resulted in liver enzyme level elevations. Pharmacokinetic data for 19 subjects were evaluable. Geometric mean ratios for the lopinavir minimum concentration in serum and the maximum concentration in serum (C(max)) on day 24 versus that on day 10 were 0.43 (90% confidence interval [CI], 0.19 to 0.96) and 1.02 (90% CI, 0.85 to 1.23), respectively, for arm 1 (n = 10) and 1.03 (90% CI, 0.68 to 1.56) and 0.93 (90% CI, 0.81 to 1.07), respectively, for arm 2 (n = 9). Ritonavir exposure increased from days 10 to 24 in both arms. The geometric mean C(max) of rifampin was 13.5 mg/liter (day 24) and was similar between the two arms. Adjusted-dose regimens of lopinavir-ritonavir in combination with therapeutic drug monitoring and monitoring of liver function may allow concomitant use of rifampin.


Subject(s)
Antibiotics, Antitubercular/pharmacokinetics , HIV Protease Inhibitors/pharmacokinetics , Pyrimidinones/pharmacokinetics , Rifampin/pharmacokinetics , Ritonavir/pharmacokinetics , Adult , Aged , Antibiotics, Antitubercular/adverse effects , Area Under Curve , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Combinations , Drug Interactions , Female , HIV Protease Inhibitors/adverse effects , Half-Life , Humans , Lopinavir , Male , Middle Aged , Pyrimidinones/adverse effects , Rifampin/adverse effects , Ritonavir/adverse effects , Spectrophotometry, Ultraviolet
8.
AIDS ; 15(1): F1-9, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11192874

ABSTRACT

OBJECTIVE: To evaluate the safety and antiviral activity of different dose levels of the HIV protease inhibitor ABT-378 combined with low-dose ritonavir, plus stavudine and lamivudine in antiretroviral-naive individuals. DESIGN: Prospective, randomized, double-blind, multicenter. METHODS: Eligible patients with plasma HIV-1 RNA > 5000 copies/ml received ABT-378 200 or 400 mg with ritonavir 100 mg every 12 h; after 3 weeks stavudine 40 mg and lamivudine 150 mg every 12 h were added (group I, n = 32). A second group initiated treatment with ABT-378 400 mg and ritonavir 100 or 200 mg plus stavudine and lamivudine every 12 h (group II, n = 68). RESULTS: Mean baseline HIV-1 RNA was 4.9 log10 copies/ml in both groups and CD4 cell count was 398 x 10(6)/l and 310 x 10(6)/l in Groups I and II respectively. In the intent-to-treat (ITT; missing value = failure) analysis at 48 weeks, HIV-1 RNA was < 400 copies/ml for 91% (< 50 copies/ml, 75%) and 82% (< 50 copies/ml, 79%) of patients in groups I and II respectively. Mean steady-state ABT-378 trough concentrations exceeded the wild-type HIV-1 EC50 (effective concentration to inhibit 50%) by 50-100-fold. The most common adverse events were abnormal stools, diarrhea and nausea. No patient discontinued before 48 weeks because of treatment-related toxicity or virologic rebound. CONCLUSIONS: ABT-378 is a potent, well-tolerated protease inhibitor. The activity and durable suppression of HIV-1 observed in this study is probably attributable to the observed tolerability profile and the achievement of high ABT-378 plasma concentrations.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , HIV-1/drug effects , Lamivudine/therapeutic use , Pyrimidinones/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Ritonavir/therapeutic use , Stavudine/therapeutic use , Adult , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , CD4 Lymphocyte Count , Drug Therapy, Combination , Female , HIV Infections/immunology , HIV Infections/physiopathology , HIV Infections/virology , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/pharmacokinetics , Humans , Lamivudine/adverse effects , Lamivudine/pharmacokinetics , Lopinavir , Male , Pyrimidinones/adverse effects , Pyrimidinones/pharmacokinetics , RNA, Viral/blood , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/pharmacokinetics , Ritonavir/adverse effects , Ritonavir/pharmacokinetics , Stavudine/adverse effects , Stavudine/pharmacokinetics , Viral Load
9.
Pharmacotherapy ; 20(5): 549-53, 2000 May.
Article in English | MEDLINE | ID: mdl-10809341

ABSTRACT

STUDY OBJECTIVE: To determine the effects of ritonavir on the pharmacokinetics of meperidine and normeperidine. DESIGN: Open-label, crossover, pharmacokinetic study. SETTING: United States government research hospital. SUBJECTS: Eight healthy volunteers who tested negative for the human immunodeficiency virus. INTERVENTION: Subjects received oral meperidine 50 mg and had serial blood samples collected for 48 hours. They then received ritonavir 500 mg twice/day for 10 days, followed by administration of a second 50-mg meperidine dose and collection of serial samples. MEASUREMENTS AND MAIN RESULTS: Plasma samples were assayed for meperidine, normeperidine, and ritonavir. Meperidine's area under the curve (AUC) decreased in all subjects by a mean of 67+/-4% in the presence of ritonavir (p<0.005). Mean +/- SD maximum concentration was decreased from 126+/-47 to 51+/-21 ng/ml. Normeperidine's mean AUC was increased 47%, suggesting induction of hepatic metabolism. CONCLUSION: Meperidine's AUC is significantly reduced, not increased, by concomitant ritonavir. Based on these findings, the risk of narcotic-related adverse effects from this combination appears to be minimal. However, increased concentrations of normeperidine suggest a potential for toxicity with increased dosages or long-term therapy.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Cholinesterase Inhibitors/pharmacokinetics , HIV Protease Inhibitors/pharmacokinetics , Meperidine/analogs & derivatives , Meperidine/pharmacokinetics , Ritonavir/pharmacokinetics , Adult , Analgesics, Opioid/blood , Area Under Curve , Cholinesterase Inhibitors/blood , Confidence Intervals , Cross-Over Studies , Drug Interactions , Female , HIV Protease Inhibitors/blood , Humans , Male , Meperidine/blood , Ritonavir/blood
10.
Int J Pharm ; 191(2): 131-40, 1999 Nov 30.
Article in English | MEDLINE | ID: mdl-10564839

ABSTRACT

The purpose of this study was to evaluate the feasibility of intrapulmonary delivery of ABT-431, a selective D1 receptor agonist. Following intratracheal instillation of the drug solution, the lung bioavailability was found to be approximately 75% in dogs. An aerosol suspension formulation was then developed by dispersing the drug in tetrafluoroethane, HFC-134a, with the aid of poloxamer 124 and vitamin E. This ABT-431 MDI aerosol formulation showed about 40% of the particles emitted from the valve and actuator system to be under 5 microm in diameter. Also, the primary package (15 mL aluminum container, DF10/ACT-150 valve, and Micron-4-actuator with the orifice 0.4 mm) was satisfactory for accurate and reproducible dosimetry. Using tracheostomized beagle dogs, the C(max) following tracheal administration of 5 mg aerosolized ABT-431 was found to be 13.3+/-0.9 ng ml(-1) and the AUC(0-24) was estimated at 33.2+/-10.6 h ng ml(-1). The lung bioavailability of the aerosolized drug was 34% compared to intravenous injection in dogs. In humans, results from a single rising dose study demonstrated that rapid absorption of ABT-431 following oral inhalation administration resulted in a dose-dependent increase in the area under the plasma-time curve at dosage levels between 3.3 and 13.2 mg. There is a possibility of up to 25% absorption of the drug from human lung. Thus, pulmonary bioavailability of ABT-431 is significantly greater than that of oral administration. Also, these findings suggest that small and lipophilic compounds, especially with hepatic first pass effect, may be effectively delivered systemically using oral inhalation aerosols.


Subject(s)
Dopamine Agonists/administration & dosage , Lung/metabolism , Prodrugs/administration & dosage , Pyridines/administration & dosage , Quinolones/administration & dosage , Receptors, Dopamine D1/agonists , Tetrahydronaphthalenes/administration & dosage , Thiophenes/administration & dosage , Absorption , Adult , Aerosol Propellants , Aerosols , Animals , Area Under Curve , Biological Availability , Dogs , Dopamine Agonists/pharmacokinetics , Double-Blind Method , Humans , Injections, Intravenous , Male , Nebulizers and Vaporizers , Particle Size , Prodrugs/pharmacokinetics , Pyridines/pharmacokinetics , Quinolones/pharmacokinetics , Tetrahydronaphthalenes/pharmacokinetics , Thiophenes/pharmacokinetics
11.
J Clin Psychopharmacol ; 19(2): 114-24, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10211912

ABSTRACT

The gamma-aminobutyric acid (GABA) agonist alprazolam is known to decrease adrenocorticotropic hormone and cortisol concentrations. Dehydroepiandrosterone (DHEA) is secreted synchronously with cortisol by the adrenal glands and demonstrates diurnal variation. The major objective of this study was to determine whether alprazolam affects concentrations of DHEA and DHEA-S, the sulfated metabolite. In vitro studies have demonstrated that DHEA-S, and perhaps DHEA, have GABA antagonistic activity. Another objective was to determine whether DHEA-S and/or DHEA concentrations are related to psychomotor impairment after alprazolam. Thirty-eight healthy volunteers (25 young men, aged 22-35, and 13 elderly men, aged 65-75) received a single intravenous dose of alprazolam 2 mg/2 min (part 1). Fifteen young and 13 elderly men responded to alprazolam and agreed to participate in part 2 of the study, which was a crossover of placebo and alprazolam infusion to plateau for 9 hours. Plasma samples at 0, 1, 4, and 7 hours were assayed for steroid concentrations. Alprazolam produced (1) significant increases in DHEA concentrations at 7 hours in both young and elderly men; (2) significant decreases in cortisol concentrations; and (3) no change in DHEA-S concentrations. The relationship between psychomotor decrement and DHEA concentrations at 7 hours after alprazolam 2 mg/2 min was described by a u-shaped curve (p < 0.0047). Both the linear and quadratic components of the equations for the tests were significant (p < 0.002). These results suggest that alprazolam modulates peripheral concentrations of DHEA and that DHEA and/or DHEA-S may have an in vivo role in modulating GABA receptor-mediated responses.


Subject(s)
Alprazolam/pharmacology , Dehydroepiandrosterone/blood , Adult , Age Factors , Aged , Alprazolam/administration & dosage , Humans , Hydrocortisone/blood , Male , Psychomotor Performance/drug effects , Time Factors
12.
Clin Pharmacokinet ; 35(4): 275-91, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9812178

ABSTRACT

Ritonavir is 1 of the 4 potent synthetic HIV protease inhibitors, approved by the US Food and Drug Administration (FDA) between 1995 and 1997, that have revolutionised HIV therapy. The extent of oral absorption is high and is not affected by food. Within the clinical concentration range, ritonavir is approximately 98 to 99% bound to plasma proteins, including albumin and alpha 1-acid glycoprotein. Cerebrospinal fluid (CSF) drug concentrations are low in relation to total plasma concentration. However, parallel decreases in the viral burden have been observed in the plasma, CSF and other tissues. Ritonavir is primarily metabolised by cytochrome P450 (CYP) 3A isozymes and, to a lesser extent, by CYP2D6. Four major oxidative metabolites have been identified in humans, but are unlikely to contribute to the antiviral effect. About 34% and 3.5% of a 600 mg dose is excreted as unchanged drug in the faeces and urine, respectively. The clinically relevant t1/2 beta is about 3 to 5 hours. Because of autoinduction, plasma concentrations generally reach steady state 2 weeks after the start of administration. The pharmacokinetics of ritonavir are relatively linear after multiple doses, with apparent oral clearance averaging 7 to 9 L/h. In vitro, ritonavir is a potent inhibitor of CYP3A. In vivo, ritonavir significantly increases the AUC of drugs primarily eliminated by CYP3A metabolism (e.g. clarithromycin, ketoconazole, rifabutin, and other HIV protease inhibitors, including indinavir, saquinavir and nelfinavir) with effects ranging from an increase of 77% to 20-fold in humans. It also inhibits CYP2D6-mediated metabolism, but to a significantly lesser extent (145% increase in desipramine AUC). Since ritonavir is also an inducer of several metabolising enzymes [CYP1A4, glucuronosyl transferase (GT), and possibly CYP2C9 and CYP2C19], the magnitude of drug interactions is difficult to predict, particularly for drugs that are metabolised by multiple enzymes or have low intrinsic clearance by CYP3A. For example, the AUC of CYP3A substrate methadone was slightly decreased and alprazolam was unaffected. Ritonavir is minimally affected by other CYP3A inhibitors, including ketoconazole. Rifampicin (rifampin), a potent CYP3A inducer, decreased the AUC of ritonavir by only 35%. The degree and duration of suppression of HIV replication is significantly correlated with the plasma concentrations. Thus, the large increase in the plasma concentrations of other protease inhibitors when coadministered with ritonavir forms the basis of rational dual protease inhibitor regimens, providing patients with 2 potent drugs at significantly reduced doses and less frequent dosage intervals. Combination treatment of ritonavir with saquinavir and indinavir results in potent and sustained clinical activity. Other important factors with combination regimens include reduced interpatient variability for high clearance agents, and elimination of the food effect on the bioavailibility of indinavir.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV Protease Inhibitors/pharmacokinetics , Ritonavir/pharmacokinetics , Animals , Anti-HIV Agents/pharmacology , Drug Interactions , HIV Protease Inhibitors/pharmacology , Humans , Ritonavir/pharmacology
13.
J Pharmacol Exp Ther ; 281(3): 1317-29, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9190868

ABSTRACT

This study was designed to determine whether age influences sensitivity to alprazolam and/or rate of acute tolerance development to the effects of alprazolam. Three treatments were each separated by 4 weeks. Twenty-five young (ages 22-35) and 13 elderly (ages 65-75) men received 2 mg of alprazolam/2 min i.v. Blood samples were obtained over 48 hr, and sedative, psychomotor and memory effects were assessed serially for 12 hr. Clearance was lower (P = .05) and elimination t[1/2] was longer (P = .005) in the elderly, but area under the concentration curve to 12 hr and maximum concentration did not differ by age group. Maximum impairment was greater in the elderly for all assessments. Mean EC50 values differed between the elderly (25.3 and 25.0 ng/ml) and the young (39.8 and 36.5 ng/ml) on card sorting and digit symbol substitution, respectively (P < .001). Bolus treatment data were used to individualize doses for the crossover of placebo and alprazolam; infusions were designed to maintain a plateau alprazolam concentration between 1 and 9 hr. Alprazolam concentrations through 12 hr did not differ between the young and elderly. Median t[1/2] for offset of effect for digit symbol substitution was 2.8 hr in the young and 4.9 hr in the elderly (P = .05). Therefore, aging decreases alprazolam clearance and increases sensitivity to effects of alprazolam through a mechanism other than pharmacokinetics; aging also decreases the rate of offset of effect of alprazolam. In addition, the data provide insight into the intensity of initial effect as a determinant of rate of tolerance development.


Subject(s)
Aging/drug effects , Alprazolam/pharmacology , Drug Tolerance , Memory/drug effects , Psychomotor Performance/drug effects , Adult , Aged , Alprazolam/pharmacokinetics , Double-Blind Method , Humans , Male , Sensitivity and Specificity
14.
Clin Pharmacokinet ; 32(3): 210-58, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9084960

ABSTRACT

This article reviews the information available to assist pharmacokineticists in the prediction of metabolic drug interactions. Significant advances in this area have been made in the last decade, permitting the identification in early drug development of dominant cytochrome P450 (CYP) isoform(s) metabolising a particular drug as well as the ability of a drug to inhibit a specific CYP isoform. The major isoforms involved in human drug metabolism are CYP3A, CYP2D6, CYP2C, CYP1A2 and CYP2E1. Often patients are taking multiple concurrent medications, and thus an assessment of potential drug-drug interactions is imperative. A database containing information about the clearance routes for over 300 drugs from multiple therapeutic classes, including analgesics, anti-infectives, psychotropics, anticonvulsants, cancer chemotherapeutics, gastrointestinal agents, cardiovascular agents and others, was constructed to assist in the semiquantitative prediction of the magnitude of potential interactions with drugs under development. With knowledge of the in vitro inhibition constant of a drug (Ki) for a particular CYP isoform, it is theoretically possible to assess the likelihood of interactions for a drug cleared through CYP-mediated metabolism. For many agents, the CYP isoform involved in metabolism has not been identified and there is substantial uncertainty given the current knowledge base. The mathematical concepts for prediction based on competitive enzyme inhibition are reviewed in this article. These relationships become more complex if the inhibition is of a mixed competitive/noncompetitive nature. Sources of uncertainty and inaccuracy in predicting the magnitude of in vivo inhibition includes the nature and design of in vitro experiments to determine Ki, inhibitor concentration in the hepatic cytosol compared with that in plasma, prehepatic metabolism, presence of active metabolites and enzyme induction. The accurate prospective prediction of drug interactions requires rigorous attention to the details of the in vitro results, and detailed information about the pharmacokinetics and metabolism of the inhibitor and inhibited drug. With the discussion of principles and accompanying tabulation of literature data concerning the clearance of various drugs, a framework for reasonable semiquantitative predictions is offered in this article.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Pharmacokinetics , Humans , Isoenzymes/metabolism
15.
Neuropharmacology ; 34(9): 1169-75, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8532187

ABSTRACT

Modulation of benzodiazepine receptor ligand binding to the GABAA receptor complex by the neuroactive steroids 3 alpha-hydroxy-dihydroprogesterone (3 alpha-OH-DHP) and 3 alpha-hydroxycorticosterone (3 alpha- THDOC) was assessed in an in vitro binding assay with the benzodiazepine antagonist [3H]flumazenil using rat cortical membranes. Neuroactive steroids, pentobarbital, GABA and bicuculline did not significantly affect flumazenil binding. However, the addition of neuroactive steroids significantly decreased the Ki of benzodiazepine agonists, including alprazolam, diazepam and clonazepam, indicating an increase in agonist affinity. Only the addition of 3 beta-OH-DHP, an inactive stereoisomer had no effect on the Ki of these agonists. The binding of the benzodiazepine inverse agonist FG 7142 was not significantly affected by these steroids, but the addition of GABA significantly increased the Ki of FG 7142 indicating a decrease in inverse agonist affinity. High concentrations of GABA or bicuculline were able to occlude the 3 alpha-THDOC mediated decrease in alprasolam Ki, indicating a GABA dependent mechanism of binding enhancement. An advantage of using [3H]flumazenil is that neither the Ki nor the Bmax change in the presence of allosteric site modulators, permitting the simple and direct assessment of alterations in benzodiazepine ligand affinity for the GABAA receptor complex by neuroactive steroids.


Subject(s)
Binding, Competitive , Brain/drug effects , Flumazenil/pharmacology , Receptors, GABA-A/drug effects , Alprazolam/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Desoxycorticosterone/analogs & derivatives , Desoxycorticosterone/pharmacology , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Radioligand Assay , Rats , Rats, Sprague-Dawley
16.
J Clin Psychopharmacol ; 15(4): 259-62, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7593708

ABSTRACT

This study was designed to evaluate the relative and absolute bioavailability of triazolam, 0.25 mg, after the administration of the marketed oral tablet and a sublingual prototype wafer; an intravenous dose was used as a reference. Twelve men were evaluated in a three-way crossover study; study days were separated by 1 week. A single dose was administered to each subject at approximately 8 a.m.; serial blood samples were obtained for the determination of triazolam concentration. The fraction absorbed relative to intravenous was 20% higher in the sublingual than in the oral treatment (p = 0.0128); the difference between treatments was greatest in the first 2 hours as indicated by the area under the curve from 0 to 2 hours (p < 0.05). The extraction ratio ranged from 0.05 to 0.25, and the predicted availability after oral administration was 86% with a range of 75 to 95%. In contrast, the observed mean absolute availability was 44% (oral) and 53% (sublingual). A potential explanation for this discrepancy between predicted and observed bioavailability is that after oral administration, a fraction of triazolam may be metabolized by cytochrome P450IIIA4 in the gut wall, with a separate fraction subject to first-pass metabolism in the liver. Although this study was not designed to identify sites of triazolam metabolism, the proposed explanation is consistent with the occurrence of P450IIIA4 in the stomach, small intestine, and liver. Doses administered sublingually avoid first-pass metabolism, producing earlier and higher peak concentrations than do doses administered orally.


Subject(s)
Hypnotics and Sedatives/pharmacokinetics , Triazolam/pharmacokinetics , Administration, Oral , Administration, Sublingual , Adult , Cross-Over Studies , Double-Blind Method , Female , Humans , Hypnotics and Sedatives/administration & dosage , Injections, Intravenous , Male , Triazolam/administration & dosage
17.
J Pharmacol Exp Ther ; 264(3): 1047-55, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8450449

ABSTRACT

Although acute tolerance to selected effects of many benzodiazepines is known to occur, acute tolerance to triazolam has not been documented even in studies that have included pharmacodynamic modeling. The purpose of this investigation was to determine whether acute tolerance to triazolam occurs in humans. Intravenous bolus doses of triazolam were used to individualize two subsequent intravenous infusions: one to achieve and maintain a constant triazolam concentration and one to achieve a series of incremental steady-state concentrations; a placebo treatment was also included. Ten healthy men completed the four single-dose treatments. Serial blood sampling and psychomotor and memory testing were done. In the constant infusion treatment, mean performance impairment was greatest at 1 h and then decreased rapidly despite maintenance of a mean triazolam concentration of 2.48 ng/ml for 9 h. Neither learning nor changes in free concentration account for the observations. Additionally, data from the step-infusion treatment indicate that the triazolam effect-concentration relationship after a single dose can be altered by rate of administration. Because tolerance develops, the administration of drug in small increments results in an increased effect at a lower concentration, with a blunted maximal response. Furthermore, our data suggest intersubject variability in the rate of development of acute tolerance. Patients who develop tolerance more slowly would experience a longer duration of effect. Further study regarding the rate of development of tolerance to specific effects and in different patient populations is warranted.


Subject(s)
Triazolam/pharmacology , Adult , Dose-Response Relationship, Drug , Drug Tolerance , Humans , Infusions, Intravenous , Injections, Intravenous , Male , Psychomotor Performance/drug effects , Triazolam/administration & dosage , Triazolam/blood
18.
Ann Pharmacother ; 27(3): 298-301, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8453164

ABSTRACT

OBJECTIVE: To report a case of status epilepticus in a patient with anticonvulsant hypersensitivity syndrome (AHS) that was controlled successfully using continuous intravenous infusion diazepam. AHS and alternatives for treatment of status epilepticus are also reviewed. DESIGN: Single case report. SETTING: 217-bed children's university hospital. PATIENT: Four-year-old, 20-kg girl, diagnosed with idiopathic tonic-clonic epilepsy, who developed AHS to phenobarbital and phenytoin and status epilepticus unresponsive to lorazepam. RESULTS: Diazepam intravenous infusion at dosages titrated to 8 mg/h was used successfully to control seizures for eight days until signs and symptoms of AHS had resolved and maintenance therapy with valproic acid was started. CONCLUSIONS: Continuous intravenous infusion diazepam is a reasonable therapeutic choice for the management of status epilepticus in a patient with AHS when traditional therapy such as phenytoin and phenobarbital cannot be used.


Subject(s)
Anticonvulsants/adverse effects , Diazepam/administration & dosage , Drug Hypersensitivity/etiology , Status Epilepticus/drug therapy , Child, Preschool , Diazepam/therapeutic use , Female , Hospitals, Pediatric , Hospitals, University , Humans , Infusions, Intravenous , Phenobarbital/adverse effects , Phenytoin/adverse effects , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...