Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Clin Infect Dis ; 67(2): 193-201, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29415190

ABSTRACT

Background: Once-weekly isoniazid and rifapentine for 3 months is a treatment option in persons with human immunodeficiency virus and latent tuberculosis infection. This study aimed to examine pharmacokinetic drug-drug interactions between this regimen and dolutegravir, a first-line antiretroviral medication. Methods: This was a single-center, open-label, fixed-sequence, drug-drug interaction study in healthy volunteers. Subjects received oral dolutegravir 50 mg once daily alone (days 1-4) and concomitantly with once-weekly isoniazid 900 mg, rifapentine 900 mg, and pyridoxine 50 mg (days 5-19). Dolutegravir concentrations were measured on days 4, 14, and 19, and rifapentine, 25-desacetyl-rifapentine, and isoniazid concentrations were measured on day 19. Cytokines and antidrug antibodies to isoniazid and rifapentine were examined at select time points. Results: The study was terminated following the development of flu-like syndrome and elevated aminotransferase levels in 2 of 4 subjects after the third isoniazid-rifapentine dose. Markedly elevated levels of interferon-γ, CXCL10, C-reactive protein, and other cytokines were temporally associated with symptoms. Antidrug antibodies were infrequently detected. Dolutegravir area under the curve (AUC) was decreased by 46% (90% confidence interval, 27-110%; P = .13) on day 14. Rifapentine and 25-desacetyl rifapentine levels on day 19 were comparable to reference data, whereas isoniazid AUCs were approximately 67%-92% higher in the subjects who developed toxicities. Conclusions: The combined use of dolutegravir with once-weekly isoniazid-rifapentine resulted in unexpected and serious toxicities that were mediated by endogenous cytokine release. Additional investigations are necessary to examine the safety and efficacy of coadministering these medications. Clinical Trials Registration: NCT02771249.


Subject(s)
Antibiotics, Antitubercular/adverse effects , Cytokines/immunology , Drug Administration Schedule , Heterocyclic Compounds, 3-Ring/adverse effects , Isoniazid/adverse effects , Rifampin/analogs & derivatives , Adolescent , Adult , Aged , Antibiotics, Antitubercular/pharmacokinetics , Cytokines/blood , Drug Interactions , Female , HIV Infections/microbiology , Healthy Volunteers , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Humans , Isoniazid/pharmacokinetics , Latent Tuberculosis/drug therapy , Male , Middle Aged , Oxazines , Piperazines , Pyridones , Rifampin/adverse effects , Rifampin/pharmacokinetics , Young Adult
2.
Article in English | MEDLINE | ID: mdl-28848011

ABSTRACT

Dabigatran etexilate (DE) is a P-glycoprotein (P-gp) probe substrate, and its active anticoagulant moiety, dabigatran, is a substrate of the multidrug and toxin extrusion protein-1 (MATE-1) transporter. The antiretroviral pharmacokinetic enhancers, ritonavir and cobicistat, inhibit both these transporters. Healthy volunteers received single doses of DE at 150 mg alone, followed by ritonavir at 100 mg or cobicistat at 150 mg daily for 2 weeks. DE was then given 2 h before ritonavir or cobicistat. One week later, DE was given simultaneously with ritonavir or cobicistat. No significant increases in dabigatran pharmacokinetic (PK) exposure or thrombin time (TT) measures were observed with the simultaneous administration of ritonavir. Separated administration of ritonavir resulted in a mean decrease in dabigatran PK exposure of 29% (90% confidence interval [CI], 18 to 40%) but did not significantly change TT measures. However, cobicistat increased dabigatran PK exposure (area under the concentration-versus-time curve from time zero to infinity and maximum plasma concentration) by 127% each (90% CI, 81 to 173% and 59 to 196%, respectively) and increased TT measures (33% for the area-under-the-effect curve from time zero to 24 h [90% CI, 22 to 44%] and 51% for TT at 24 h [90% CI, 22 to 78%]) when given simultaneously with dabigatran. Similar increases were observed when cobicistat was administered separately by 2 h from the administration of dabigatran. In all comparisons, no significant increase in the dabigatran elimination half-life was observed. Therefore, it is likely safe to coadminister ritonavir with DE, while there is a potential need for reduced dosing and prudent clinical monitoring with the coadministration of cobicistat due to the greater net inhibition of intestinal P-gp transport and increased bioavailability. (This study has been registered at ClinicalTrials.gov under identifier NCT01896622.).


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cobicistat/pharmacokinetics , Dabigatran/pharmacokinetics , Intestinal Mucosa/metabolism , Ritonavir/pharmacokinetics , Adult , Antithrombins/administration & dosage , Antithrombins/pharmacokinetics , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Area Under Curve , Cobicistat/administration & dosage , Dabigatran/administration & dosage , Drug Interactions , Female , Healthy Volunteers , Humans , Intestines/drug effects , Male , Middle Aged , Ritonavir/administration & dosage , Thrombin Time
3.
J Clin Pharmacol ; 57(2): 219-229, 2017 02.
Article in English | MEDLINE | ID: mdl-27407018

ABSTRACT

Steady-state population pharmacokinetics of a noncommercial immediate-release metformin (hydrochloride) drug product were characterized in 28 severely obese children with insulin resistance. The concentration-time profiles with double peaks were well described by a 1-compartment model with 2 absorption sites. Mean population apparent clearance (CL/F) was 68.1 L/h, and mean apparent volume of distribution (V/F) was 28.8 L. Body weight was a covariate of CL/F and V/F. Estimated glomerular filtration rate was a significant covariate of CL/F (P < .001). SLC22A1 genotype did not significantly affect metformin pharmacokinetics. The response to 6 months of metformin treatment (HbA1c , homeostasis model assessment for insulin resistance, fasting insulin, and glucose changes) did not differ between SLC22A1 wild-type subjects and carriers of presumably low-activity SLC22A1 alleles. However, SLC22A1 variant carriers had smaller reductions in percentage of total trunk fat after metformin therapy, although the percentage reduction in trunk fat was small. The median % change in trunk fat was -2.20% (-9.00% to 0.900%) and -1.20% (-2.40% to 7.30%) for the SLC22A1 wild-type subjects and variant carriers, respectively. Future study is needed to evaluate the effects of SLC22A1 polymorphisms on metformin-mediated weight reduction in obese children.


Subject(s)
Adiposity/drug effects , Adiposity/genetics , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/pharmacokinetics , Insulin Resistance/genetics , Metformin/pharmacology , Metformin/pharmacokinetics , Obesity/drug therapy , Obesity/genetics , Octamer Transcription Factor-1/genetics , Octamer Transcription Factor-1/metabolism , Body Weight , Child , Double-Blind Method , Female , Genotype , Glomerular Filtration Rate , Humans , Male , Polymorphism, Genetic/genetics , Weight Loss/drug effects
5.
Pharmacotherapy ; 36(1): 49-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26799348

ABSTRACT

STUDY OBJECTIVE: Because we previously observed a significant 41% reduction in gemfibrozil exposure after 2 weeks of lopinavir-ritonavir administration, we sought to determine the influence of lopinavir-ritonavir and ritonavir alone on the pharmacokinetics of fenofibric acid, an alternative to gemfibrozil for the treatment of elevated triglyceride levels. DESIGN: Open-label, single-sequence pharmacokinetic study. SETTING: Clinical Research Center at the National Institutes of Health. SUBJECTS: Thirteen healthy adult volunteers. INTERVENTION: Subjects received a single oral dose of fenofibrate 145 mg during three study phases: before ritonavir administration, after 2 weeks of administration of ritonavir 100 mg twice/day, and after 2 weeks of administration of lopinavir 400 mg-ritonavir 100 mg twice/day. MEASUREMENTS AND MAIN RESULTS: Serial blood samples were collected over 120 hours for determination of fenofibric acid concentrations. Fenofibric acid pharmacokinetic parameter values were compared before and after concomitant ritonavir or lopinavir-ritonavir administration. The geometric mean ratios (90% confidence intervals) for fenofibric acid area under the plasma concentration-time curve were 0.89 (0.77-1.01) after 14 days of ritonavir alone compared with baseline (p>0.05) and 0.87 (0.69-1.05) after 14 days of lopinavir-ritonavir compared with baseline (p>0.05). Study drugs were generally well tolerated; all adverse events were mild or moderate, transient, and resolved without intervention. CONCLUSION: In contrast to a significant interaction between gemfibrozil and lopinavir-ritonavir, neither lopinavir-ritonavir nor ritonavir alone altered the pharmacokinetics of fenofibric acid in healthy volunteers. These data suggest that fenofibrate remains an important option in human immunodeficiency virus-infected patients receiving common ritonavir-boosted therapy.


Subject(s)
Fenofibrate/analogs & derivatives , Hypolipidemic Agents/pharmacokinetics , Lopinavir/pharmacology , Ritonavir/pharmacology , Adult , Drug Administration Schedule , Drug Interactions , Drug Therapy, Combination , Female , Fenofibrate/blood , Fenofibrate/pharmacokinetics , Humans , Hypolipidemic Agents/blood , Lopinavir/administration & dosage , Male , Middle Aged , Ritonavir/administration & dosage , Young Adult
6.
Clin Infect Dis ; 62(8): 1036-1042, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26797214

ABSTRACT

BACKGROUND: The current study was conducted to determine if efavirenz (EFV) or atazanavir/ritonavir (ATV/r)-based combination antiretroviral therapy (cART) impacted steady-state atovaquone plasma concentrations in human immunodeficiency virus (HIV)-infected patients receiving treatment doses of atovaquone. METHODS: Thirty HIV-infected volunteers were recruited, 10 taking no cART and 10 each taking cART that included EFV or ATV/r. Subjects were randomly assigned to atovaquone 750 mg twice daily (BID) for 14 days followed by atovaquone 1500 mg BID for 14 days, or vice-versa, with a washout period in between. On day 14 of each phase, blood was sampled for pharmacokinetic studies, and the area under the concentration-time curve (AUCτ) and average concentration (C avg) were calculated and compared using an unpaired t test. RESULTS: Twenty-nine subjects completed both dosing cohorts. Subjects receiving EFV-based cART had 47% and 44% lower atovaquone AUCτ than subjects not receiving cART at atovaquone doses of 750 mg BID and 1500 mg BID, respectively (P≤ .01). Only 5 of 10 subjects receiving EFV-based cART plus atovaquone 750 mg BID had an atovaquone C avg>15 µg/mL, which has previously been associated with successful treatment of Pneumocystis jirovecipneumonia. AUCτ and Cavg did not significantly differ for concurrent ATV/r for 750 mg BID or 1500 mg BID when compared to the group not receiving cART. Nine of 10 subjects not receiving cART, 8 of 10 subjects receiving ATV/r, and 2 of 10 subjects receiving EFV in combination with atovaquone 750 mg BID achieved an atovaquone C avg>18.5 µg/mL, a concentration that has previously been associated with successful treatment of Toxoplasmaencephalitis (TE). CONCLUSIONS: These data suggest that the currently recommended dose of atovaquone 750 mg BID for treatment of mild to moderate PCP may not be adequate in patients receiving concurrent EFV. Furthermore, doses lower than the currently recommended dose of 1500 mg BID may achieve plasma concentrations adequate to treat TE in HIV-infected patients not receiving EFV. CLINICAL TRIALS REGISTRATION: NCT01479361.


Subject(s)
Anti-HIV Agents/therapeutic use , Anti-Infective Agents/pharmacokinetics , Anti-Infective Agents/therapeutic use , Atovaquone/pharmacokinetics , Atovaquone/therapeutic use , Benzoxazines/therapeutic use , Ritonavir/therapeutic use , AIDS-Related Opportunistic Infections/drug therapy , Administration, Oral , Adolescent , Adult , Aged , Alkynes , Anti-Infective Agents/blood , Atazanavir Sulfate/adverse effects , Atazanavir Sulfate/therapeutic use , Atovaquone/blood , Benzoxazines/adverse effects , Cyclopropanes , Drug Interactions , Drug Therapy, Combination/adverse effects , Encephalitis/drug therapy , Encephalitis/prevention & control , Female , HIV Infections/drug therapy , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/therapeutic use , Humans , Male , Middle Aged , Pneumonia, Pneumocystis/drug therapy , Pneumonia, Pneumocystis/prevention & control , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/therapeutic use , Ritonavir/adverse effects , Toxoplasmosis, Cerebral/drug therapy , Toxoplasmosis, Cerebral/prevention & control , Young Adult
7.
Pharmacotherapy ; 34(11): 1151-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25142999

ABSTRACT

STUDY OBJECTIVE: Panax ginseng has been shown in preclinical studies to modulate cytochrome P450 enzymes involved in the metabolism of HIV protease inhibitors. Therefore, the purpose of this study was to determine the influence of P. ginseng on the pharmacokinetics of the HIV protease inhibitor combination lopinavir-ritonavir (LPV-r) in healthy volunteers. DESIGN: Single-sequence, open-label, single-center pharmacokinetic investigation. SETTING: Government health care facility. SUBJECTS: Twelve healthy human volunteers. MEASUREMENTS AND MAIN RESULTS: Twelve healthy volunteers received LPV-r (400-100 mg) twice/day for 29.5 days. On day 15 of LPV-r administration, serial blood samples were collected over 12 hours for determination of lopinavir and ritonavir concentrations. On study day 16, subjects began taking P. ginseng 500 mg twice/day, which they continued for 2 weeks in combination with LPV-r. On day 30 of LPV-r administration, serial blood samples were again collected over 12 hours for determination of lopinavir and ritonavir concentrations. Lopinavir and ritonavir pharmacokinetic parameter values were determined using noncompartmental methods, and preadministration and postadministration ginseng values were compared using a Student t test, where p<0.05 was accepted as statistically significant. CONCLUSION: Neither lopinavir nor ritonavir steady-state pharmacokinetics were altered by 2 weeks of P. ginseng administration to healthy human volunteers. Thus, a clinically significant interaction between P. ginseng and LPV-r is unlikely to occur in HIV-infected patients who choose to take these agents concurrently. It is also unlikely that P. ginseng will interact with other ritonavir-boosted protease inhibitor combinations, although confirmatory data are necessary.


Subject(s)
Cytochrome P-450 CYP3A Inducers/adverse effects , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Dietary Supplements/adverse effects , Food-Drug Interactions , HIV Protease Inhibitors/pharmacokinetics , Lopinavir/pharmacokinetics , Panax/adverse effects , Ritonavir/pharmacokinetics , Adult , Cytochrome P-450 CYP3A Inhibitors/blood , Drug Combinations , Female , HIV Protease Inhibitors/blood , Half-Life , Humans , Immunologic Factors/adverse effects , Lopinavir/blood , Male , Maryland , Metabolic Clearance Rate , National Institute of Allergy and Infectious Diseases (U.S.) , Nootropic Agents/adverse effects , Plant Roots/adverse effects , Ritonavir/blood , United States , Young Adult
8.
J Acquir Immune Defic Syndr ; 63(3): 355-61, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23535292

ABSTRACT

OBJECTIVE: To identify an alternative inhaled corticosteroid to fluticasone propionate that can be safely coadministered with HIV protease inhibitors, the safety and pharmacokinetics of beclomethasone dipropionate (BDP) and its active metabolite, beclomethasone 17-monopropionate (17-BMP), in combination with ritonavir (RTV) and darunavir/ritonavir (DRV/r) were assessed. DESIGN: Open-label, prospective, randomized pharmacokinetic and pharmacodynamic study in healthy volunteers. METHODS: Thirty healthy volunteers received inhaled 160 µg bid BDP for 14 days and were then randomized (1:1:1) into 3 groups: group 1 (control) remained on BDP alone for 28 days, group 2 received 100 mg bid BDP + RTV for 28 days, and group 3 received 600/100 mg bid BDP + DRV/r for 28 days. Pharmacokinetic sampling for 17-BMP was performed on days 14 and 28, and pharmacokinetic parameter values were compared within patients and between groups. Cortisol stimulation testing was also performed on days 1, 14, 28, and 42 and compared within and between groups. RESULTS: Geometric mean ratios (day 28:day 14) (90% confidence interval) for 17-BMP area under the concentration-time curve in groups 1, 2, and 3, respectively, were 0.93 (0.81 to 1.06, P = 0.27), 2.08 (1.52 to 2.65, P = 0.006), and 0.89 (0.68 to 1.09, P = 0.61). There were no significant reductions in serum cortisol levels within or between groups (P > 0.05). CONCLUSIONS: DRV/r did not increase 17-BMP exposure, whereas RTV alone produced a statistically significant but clinically inconsequential 2-fold increase in 17-BMP exposure. Adrenal suppression was not observed in any of the study groups. These data suggest that BDP can be safely coadministered with DRV/r and likely other RTV-boosted protease inhibitors.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Beclomethasone/pharmacokinetics , Ritonavir/administration & dosage , Sulfonamides/administration & dosage , Adolescent , Adult , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Beclomethasone/administration & dosage , Beclomethasone/analogs & derivatives , Beclomethasone/blood , Beclomethasone/therapeutic use , Darunavir , Drug Interactions , Female , HIV Infections/drug therapy , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/therapeutic use , Humans , Male , Middle Aged , Ritonavir/adverse effects , Ritonavir/therapeutic use , Sulfonamides/adverse effects , Sulfonamides/therapeutic use , Young Adult
9.
J Clin Pharmacol ; 52(6): 932-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21646440

ABSTRACT

A number of herbal preparations have been shown to interact with prescription medications secondary to modulation of cytochrome P450 (CYP) and/or P-glycoprotein (P-gp). The purpose of this study was to determine the influence of Panax ginseng on CYP3A and P-gp function using the probe substrates midazolam and fexofenadine, respectively. Twelve healthy participants (8 men) completed this open-label, single-sequence pharmacokinetic study. Healthy volunteers received single oral doses of midazolam 8 mg and fexofenadine 120 mg, before and after 28 days of P ginseng 500 mg twice daily. Midazolam and fexofenadine pharmacokinetic parameter values were calculated and compared before and after P ginseng administration. Geometric mean ratios (postginseng/preginseng) for midazolam area under the concentration-time curve from zero to infinity (AUC(0-∞)), half-life (t(1/2)), and maximum concentration (C(max)) were significantly reduced at 0.66 (0.55-0.78), 0.71 (0.53-0.90), and 0.74 (0.56-0.93), respectively. Conversely, fexofenadine pharmacokinetics were unaltered by P ginseng administration. Based on these results, P ginseng appeared to induce CYP3A activity in the liver and possibly the gastrointestinal tract. Patients taking P ginseng in combination with CYP3A substrates with narrow therapeutic ranges should be monitored closely for adequate therapeutic response to the substrate medication.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cytochrome P-450 CYP3A/metabolism , Drugs, Chinese Herbal/pharmacology , Herb-Drug Interactions , Midazolam/pharmacokinetics , Panax/chemistry , Terfenadine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Adult , Cross-Over Studies , Cytochrome P-450 CYP3A Inhibitors , Drug Monitoring , Drugs, Chinese Herbal/adverse effects , Female , Half-Life , Humans , Intestinal Absorption/drug effects , Male , Metabolic Clearance Rate/drug effects , Midazolam/blood , Plant Roots/chemistry , Terfenadine/blood , Terfenadine/pharmacokinetics , Young Adult
10.
Pharmacotherapy ; 30(8): 797-805, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20653355

ABSTRACT

STUDY OBJECTIVE: . To determine the influence of Echinacea purpurea on the pharmacokinetics of lopinavir-ritonavir and on cytochrome P450 (CYP) 3A and P-glycoprotein activity by using the probe substrates midazolam and fexofenadine, respectively. DESIGN: Open-label, single-sequence pharmacokinetic study. SETTING: Outpatient clinic in a federal government research center. SUBJECTS: Thirteen healthy volunteers (eight men, five women). INTERVENTION: Subjects received lopinavir 400 mg-ritonavir 100 mg twice/day with meals for 29.5 days. On day 16, subjects received E. purpurea 500 mg 3 times/day for 28 days: 14 days in combination with lopinavir-ritonavir and 14 days of E. purpurea alone. In order to assess CYP3A and P-glycoprotein activity, subjects received single oral doses of midazolam 8 mg and fexofenadine 120 mg, respectively, before and after the 28 days of E. purpurea. MEASUREMENTS AND MAIN RESULTS: On days 15 and 30 of lopinavir-ritonavir administration (before and after E. purpurea administration, respectively), serial blood samples were collected over 12 hours to determine lopinavir and ritonavir concentrations and subsequent pharmacokinetic parameters by using noncompartmental methods. Neither lopinavir nor ritonavir pharmacokinetics were significantly altered by 14 days of E. purpurea coadministration. The post-echinacea: pre-echinacea geometric mean ratios (GMRs) for lopinavir area under the concentration-time curve (AUC) from 0-12 hours and for maximum concentration were 0.96 (90% confidence interval [CI] 0.83-1.10, p=0.82) and 1.00 (90% CI 0.88-1.12, p=0.72), respectively. Conversely, GMRs for midazolam AUC from time zero extrapolated to infinity and oral clearance were 0.73 (90% CI 0.61-0.85, p=0.008) and 1.37 (90% CI 1.10-1.63, p=0.02), respectively. Fexofenadine pharmacokinetics did not significantly differ before and after E. purpurea administration (p>0.05). CONCLUSION: Echinacea purpurea induced CYP3A activity but did not alter lopinavir concentrations, most likely due to the presence of the potent CYP3A inhibitor, ritonavir. Echinacea purpurea is unlikely to alter the pharmacokinetics of ritonavir-boosted protease inhibitors but may cause modest decreases in plasma concentrations of other CYP3A substrates.


Subject(s)
Cytochrome P-450 CYP3A/biosynthesis , Echinacea , Herb-Drug Interactions , Pyrimidinones/administration & dosage , Ritonavir/administration & dosage , Adult , Cytochrome P-450 CYP3A/metabolism , Drug Combinations , Enzyme Induction/drug effects , Enzyme Induction/physiology , Female , Herb-Drug Interactions/physiology , Humans , Lopinavir , Male , Middle Aged , Plant Extracts/administration & dosage , Young Adult
11.
J Acquir Immune Defic Syndr ; 52(2): 235-9, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19648824

ABSTRACT

OBJECTIVE: The objective of this study was to determine the influence of a 2-week course of lopinavir-ritonavir on the pharmacokinetics of the triglyceride-lowering agent, gemfibrozil. METHODS: The study was conducted as an open label, single-sequence pharmacokinetic study in healthy human volunteers. Gemfibrozil pharmacokinetic parameter values were compared using a Student t test after a single 600-mg dose was administered to healthy volunteers before and after 2 weeks of lopinavir-ritonavir (400/100 mg) twice daily. RESULTS: Fifteen healthy volunteers (eight males) completed the study. All study drugs were generally well tolerated and no subjects withdrew participation. The geometric mean ratio (90% confidence interval) for gemfibrozil area under the plasma concentration-time curve after 14 days of lopinavir-ritonavir compared with baseline was 0.59 (0.52, 0.67) (P < 0.001). All 15 study subjects experienced a reduction in gemfibrozil area under the plasma concentration-time curve after lopinavir-ritonavir (range, -6% to -74%). The geometric mean ratios for gemfibrozil apparent oral clearance and maximum concentration were 1.69 (1.41, 1.97) and 0.67 (0.49, 0.86) after 14 days of lopinavir-ritonavir versus baseline, respectively (P < 0.0001 and 0.01, respectively). Gemfibrozil elimination half-life did not change after lopinavir-ritonavir administration (P = 0.60). CONCLUSION: Lopinavir-ritonavir significantly reduced the systemic exposure of gemfibrozil by reducing gemfibrozil absorption. Clinicians treating HIV-infected patients with hypertriglyceridemia should be aware of this drug interaction.


Subject(s)
Anti-HIV Agents/administration & dosage , Gemfibrozil/administration & dosage , Gemfibrozil/pharmacokinetics , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/pharmacokinetics , Pyrimidinones/administration & dosage , Ritonavir/administration & dosage , Adult , Drug Antagonism , Female , Healthy Volunteers , Humans , Lopinavir , Male , Middle Aged , Plasma/chemistry
12.
J Acquir Immune Defic Syndr ; 49(5): 513-9, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18989234

ABSTRACT

OBJECTIVE: To characterize the effect of efavirenz on bupropion hydroxylation as a marker of cytochrome P450 (CYP) 2B6 activity in healthy subjects. METHODS: Thirteen subjects received a single oral dose of bupropion SR 150 mg before and after 2 weeks of efavirenz administration for comparison of bupropion and hydroxybupropion pharmacokinetics. Efavirenz plasma concentrations were also assessed. Subjects were genotyped for CYP2B6 (G516T, C1459T, and A785G), CYP3A4 (A-392G), CYP3A5 (A6986G), and multidrug resistance protein 1 (C3435T). RESULTS: The area under the concentration vs. time curve ratio of hydroxybupropion:bupropion increased 2.3-fold after efavirenz administration (P=0.0001). Bupropion area under the concentration vs. time curve and Cmax decreased by 55% and 34%, respectively (P<0.002). None of the CYP2B6 or CYP3A genotypes evaluated were associated with a difference in bupropion or efavirenz clearance. The 2 individuals homozygous for multidrug resistance protein 1 3435-T/T had 2.5- and 1.8-fold greater bupropion and efavirenz clearance, respectively, relative to C/C and C/T individuals (P<0.05). CONCLUSIONS: Our results confirm that efavirenz induces CYP2B6 enzyme activity in vivo, as demonstrated by an increase in bupropion hydroxylation after 2 weeks of efavirenz administration.


Subject(s)
Anti-HIV Agents/pharmacology , Antidepressive Agents, Second-Generation/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/metabolism , Benzoxazines/pharmacology , Bupropion/pharmacokinetics , Oxidoreductases, N-Demethylating/metabolism , Adult , Alkynes , Anti-HIV Agents/blood , Antidepressive Agents, Second-Generation/blood , Antidepressive Agents, Second-Generation/pharmacology , Aryl Hydrocarbon Hydroxylases/drug effects , Aryl Hydrocarbon Hydroxylases/genetics , Benzoxazines/administration & dosage , Benzoxazines/blood , Benzoxazines/pharmacokinetics , Bupropion/administration & dosage , Bupropion/analogs & derivatives , Bupropion/blood , Bupropion/metabolism , Cyclopropanes , Cytochrome P-450 CYP2B6 , Delayed-Action Preparations , Drug Interactions , Enzyme Induction , Female , Genetic Variation , Humans , Hydroxylation/drug effects , Male , Middle Aged , Oxidoreductases, N-Demethylating/drug effects , Oxidoreductases, N-Demethylating/genetics , Pharmacogenetics , Young Adult
13.
J Acquir Immune Defic Syndr ; 48(5): 561-6, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18645517

ABSTRACT

BACKGROUND: Corticosteroids are cytochrome P450 3A4 substrates, which have been associated with toxicities in patients receiving cytochrome P450 3A4 inhibitors such as human immunodeficiency virus protease inhibitors. In a study in healthy volunteers, ritonavir significantly increased prednisolone exposure. METHODS: We investigated the influence of antiretroviral (ARV) medications on prednisolone pharmacokinetics in 3 groups of 10 human immunodeficiency virus-infected subjects. One group received lopinavir/ritonavir, and another efavirenz, as part of their ARV regimen; a third group did not receive ARV medications. Each subject received a single 20-mg prednisone dose followed by serial blood sampling for prednisolone. Prednisolone pharmacokinetics were compared among the groups. RESULTS: Area under the concentration-time curve was significantly lower in efavirenz recipients versus subjects receiving lopinavir/ritonavir (geometric mean ratio = 0.60, P = 0.01). Average prednisolone area under the concentration-time curve was higher in subjects taking lopinavir/ritonavir versus subjects not on ARVs; however, this difference was not significant (P > 0.05). CONCLUSIONS: These data indicate that prednisolone concentrations may fluctuate widely when human immunodeficiency virus-positive individuals established on efavirenz therapy change to lopinavir/ritonavir or vice versa.


Subject(s)
Anti-HIV Agents/therapeutic use , Benzoxazines/therapeutic use , HIV Infections/metabolism , Prednisolone/pharmacokinetics , Pyrimidinones/therapeutic use , Ritonavir/therapeutic use , Adult , Alkynes , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacology , Benzoxazines/administration & dosage , Benzoxazines/pharmacology , Cyclopropanes , Drug Interactions , Female , HIV Infections/drug therapy , HIV Infections/virology , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , Humans , Lopinavir , Male , Middle Aged , Prednisolone/administration & dosage , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacology , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcriptase Inhibitors/therapeutic use , Ritonavir/administration & dosage , Ritonavir/pharmacology
14.
J Clin Pharmacol ; 48(6): 671-80, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18420532

ABSTRACT

Midazolam is a common probe used to predict CYP3A activity, but multiple blood samples are necessary to determine midazolam's area under the concentration-time curve (AUC). As such, single sampling strategies have been examined. The purpose of this study was to assess the ability of single midazolam concentrations to predict midazolam AUC in the presence and absence of CYP3A modulation by Ginkgo biloba extract (GBE). Subjects received oral midazolam 8 mg before and after 28 days of GBE administration. Postdose blood samples were collected during both study periods and midazolam AUC determined. Linear regression was used to generate measures of predictive performance for each midazolam concentration. The geometric mean ratio (90% confidence intervals) of midazolam AUC(0-infinity) post-GBE/AUC(0-infinity) pre-GBE was 0.66 (0.49-0.84) (P = .03). Before and after GBE administration, optimal midazolam sampling times were identified at 3.5 to 5 hours and 2 to 3 hours, respectively. Single midazolam concentrations between 2 and 5 hours correctly predicted the reduction in midazolam AUC following GBE exposure, but confidence intervals were generally wide. Intersubject variability in CYP3A activity (either inherent or from drug administration) alters the prediction of optimal midazolam sampling times; therefore, midazolam AUC is preferred for assessing CYP3A activity in drug-drug interaction studies.


Subject(s)
Cytochrome P-450 CYP3A/drug effects , Herb-Drug Interactions , Midazolam/pharmacokinetics , Plant Extracts/pharmacology , Administration, Oral , Adult , Area Under Curve , Cytochrome P-450 CYP3A/metabolism , Female , Forecasting/methods , Ginkgo biloba/chemistry , Humans , Linear Models , Longitudinal Studies , Male , Middle Aged , Time Factors
15.
Curr Med Res Opin ; 24(2): 591-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18205997

ABSTRACT

OBJECTIVE: Animal and in vitro data suggest that Ginkgo biloba extract (GBE) may modulate CYP3A4 activity. As such, GBE may alter the exposure of HIV protease inhibitors metabolized by CYP3A4. It is also possible that GBE could alter protease inhibitor pharmacokinetics (PK) secondary to modulation of P-glycoprotein (P-gp). The primary objective of the study was to evaluate the effect of GBE on the exposure of lopinavir in healthy volunteers administered lopinavir/ritonavir. Secondary objectives were to compare ritonavir exposure pre- and post-GBE, and assess the effect of GBE on single doses of probe drugs midazolam and fexofenadine. METHODS: This open-label study evaluated the effect of 2 weeks of standardized GBE administration on the steady-state exposure of lopinavir and ritonavir in 14 healthy volunteers administered lopinavir/ritonavir to steady-state. In addition, single oral doses of probe drugs midazolam and fexofenadine were administered prior to and after 4 weeks of GBE (following washout of lopinavir/ritonavir) to assess the influence of GBE on CYP3A and P-gp activity, respectively. RESULTS: Lopinavir, ritonavir and fexofenadine exposures were not significantly affected by GBE administration. However, GBE decreased midazolam AUC(0-infinity) and C(max) by 34% (p = 0.03) and 31% (p = 0.03), respectively, relative to baseline. In general, lopinavir/ritonavir and GBE were well tolerated. Abnormal laboratory results included mild elevations in hepatic enzymes, cholesterol and triglycerides, and mild-to-moderate increases in total bilirubin. CONCLUSIONS: Our results suggest that GBE induces CYP3A metabolism, as assessed by a decrease in midazolam concentrations. However, there was no change in the exposure of lopinavir, likely due to ritonavir's potent inhibition of CYP3A4. Thus, GBE appears unlikely to reduce the exposure of ritonavir-boosted protease inhibitors, while concentrations of unboosted protease inhibitors may be affected. Limitations to our study include the single sequence design and the evaluation of a ritonavir-boosted protease inhibitor exclusively.


Subject(s)
Ginkgo biloba/metabolism , HIV Protease Inhibitors/pharmacology , Midazolam/pharmacokinetics , Phytotherapy , Plant Extracts/metabolism , Protease Inhibitors/pharmacokinetics , Pyrimidinones/pharmacokinetics , Ritonavir/pharmacokinetics , Terfenadine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Female , Ginkgo biloba/adverse effects , Humans , Lopinavir , Male , Plant Extracts/adverse effects , Terfenadine/pharmacokinetics
16.
Antimicrob Agents Chemother ; 50(10): 3418-23, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17005824

ABSTRACT

We investigated the compartmentalized intrapulmonary pharmacokinetics of amphotericin B and its lipid formulations in healthy rabbits. Cohorts of three to seven noninfected, catheterized rabbits received 1 mg of amphotericin B deoxycholate (DAMB) per kg of body weight or 5 mg of either amphotericin B colloidal dispersion (ABCD), amphotericin B lipid complex (ABLC), or liposomal amphotericin B (LAMB) per kg once daily for a total of 8 days. Following sparse serial plasma sampling, rabbits were sacrificed 24 h after the last dose, and epithelial lining fluid (ELF), pulmonary alveolar macrophages (PAM), and lung tissue were obtained. Pharmacokinetic parameters in plasma were derived by model-independent techniques, and concentrations in ELF and PAM were calculated based on the urea dilution method and macrophage cell volume, respectively. Mean amphotericin B concentrations +/- standard deviations (SD) in lung tissue and PAM were highest in ABLC-treated animals, exceeding concurrent plasma levels by 70- and 375-fold, respectively (in lung tissue, 16.24 +/- 1.62 versus 2.71 +/- 1.22, 6.29 +/- 1.17, and 6.32 +/- 0.57 microg/g for DAMB-, ABCD-, and LAMB-treated animals, respectively [P = 0.0029]; in PAM, 89.1 +/- 37.0 versus 8.92 +/- 2.89, 5.43 +/- 1.75, and 7.52 +/- 2.50 mug/ml for DAMB-, ABCD-, and LAMB-treated animals, respectively [P = 0.0246]). By comparison, drug concentrations in ELF were much lower than those achieved in lung tissue and PAM. Among the different cohorts, the highest ELF concentrations were found in LAMB-treated animals (2.28 +/- 1.43 versus 0.44 +/- 0.13, 0.68 +/- 0.27, and 0.90 +/- 0.28 microg/ml in DAMB-, ABCD-, and ABLC-treated animals, respectively [P = 0.0070]). In conclusion, amphotericin B and its lipid formulations displayed strikingly different patterns of disposition in lungs 24 h after dosing. Whereas the disposition of ABCD was overall not fundamentally different from that of DAMB, ABLC showed prominent accumulation in lung tissue and PAM, while LAMB achieved the highest concentrations in ELF.


Subject(s)
Amphotericin B/chemistry , Amphotericin B/pharmacokinetics , Antifungal Agents/chemistry , Antifungal Agents/pharmacokinetics , Lung/metabolism , Amphotericin B/administration & dosage , Animals , Antifungal Agents/administration & dosage , Deoxycholic Acid/administration & dosage , Deoxycholic Acid/pharmacokinetics , Drug Combinations , Female , Leukocytes, Mononuclear/metabolism , Liposomes/administration & dosage , Liposomes/pharmacokinetics , Lung/cytology , Macrophages, Alveolar/metabolism , Phosphatidylcholines/administration & dosage , Phosphatidylcholines/pharmacokinetics , Phosphatidylglycerols/administration & dosage , Phosphatidylglycerols/pharmacokinetics , Rabbits , Tissue Distribution
17.
Pharmacotherapy ; 26(8): 1051-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16863481

ABSTRACT

STUDY OBJECTIVE: To investigate a potential correlation between exposure to oral midazolam, a commonly used cytochrome P450 (CYP) 3A probe, and saquinavir and indinavir exposure. DESIGN: Open-label, prospective, pharmacokinetic study. SETTING: Outpatient research center. SUBJECTS: Thirty-six healthy volunteers aged 22-50 years. INTERVENTION: Subjects received a single oral dose of midazolam 8 mg; 4 hours later, blood was drawn to determine their serum midazolam concentrations. Midazolam phenotyping was followed by successive administration of the protease inhibitors indinavir and saquinavir, with blood sampling and pharmacokinetic analyses performed at steady state. MEASUREMENTS AND MAIN RESULTS: Pharmacokinetic parameters of each protease inhibitor were evaluated to assess for a potential relationship with 4-hour concentrations of midazolam. No correlations between phenotype results for midazolam and any pharmacokinetic parameter for indinavir or saquinavir were identified (r(2)=0.00002-0.073). When the results were analyzed based on race, significant correlations were identified in five African-American subjects, including correlations between 4-hour midazolam levels and apparent oral clearance of saquinavir (r(2)=0.734, p=0.064), area under the plasma concentration-time curve from 0-8 hours (r(2)=0.914, p=0.011), minimum concentration (r(2)=0.857, p=0.024), and maximum concentration (r(2)=0.969, p=0.002). These findings for African-American subjects were not seen with indinavir. No correlation was found between indinavir and saquinavir pharmacokinetic parameters (r(2)=0.017-0.261). CONCLUSION: Oral midazolam was not a useful probe for predicting saquinavir or indinavir exposure at steady state. Reasons for the lack of correlation likely included differences between midazolam and protease inhibitor P-glycoprotein specificity, differences in the relative contribution of CYP3A5-mediated metabolism, and/or variation in intestinal and hepatic CYP3A specificity. The strong correlation between midazolam phenotype and pharmacokinetic parameters for saquinavir in African-American subjects indicated a racial difference in one or more of these confounding variables.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , HIV Protease Inhibitors/pharmacokinetics , Indinavir/pharmacokinetics , Midazolam/pharmacokinetics , Saquinavir/pharmacokinetics , Administration, Oral , Adult , Black or African American , Area Under Curve , Cytochrome P-450 CYP3A , Female , HIV Protease Inhibitors/blood , Humans , Indinavir/blood , Male , Metabolic Clearance Rate , Midazolam/administration & dosage , Midazolam/blood , Middle Aged , Phenotype , Prospective Studies , Reference Values , Saquinavir/blood
18.
Br J Clin Pharmacol ; 61(4): 379-88, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16542198

ABSTRACT

AIMS: To examine the influence of sex on steady-state saquinavir pharmacokinetics in HIV-seronegative volunteers administered saquinavir without a concomitant protease inhibitor. METHODS: Thirty-eight healthy volunteers (14 female) received saquinavir soft-gel capsules 1200 mg three times daily for 3 days to achieve steady-state conditions. Following administration of the 10th dose, blood was collected serially over 8 h for measurement of saquinavir plasma concentrations. Saquinavir pharmacokinetic parameter values were determined using noncompartmental methods and compared between males and females. CYP3A phenotype (using oral midazolam) and MDR-1 genotypes at positions 3435 and 2677 were determined for all subjects in order to characterize possible mechanisms for any observed sex-related differences. RESULTS: There was no significant difference in saquinavir AUC(0-8) or any other pharmacokinetic parameter value between the sexes. These findings persisted after mathematically correcting for total body weight. The mean weight-normalized AUC(0-8) was 29.9 (95% confidence interval 15.5, 44.3) and 29.8 (18.6, 40.9) ng h(-1) ml(-1) kg(-1) for males and females, respectively. No significant difference in CYP3A phenotype was observed between the groups; likewise, the distribution of MDR-1 genotypes was similar for males and females. CONCLUSION: In contrast to previous study findings, results from this investigation showed no difference in saquinavir pharmacokinetics between males and females. The discrepancy between our findings and those previously reported may be explained by the fact that we evaluated HIV-seronegative volunteers and administered saquinavir in the absence of concomitant protease inhibitors such as ritonavir. Caution must be exercised when extrapolating pharmacokinetic data from healthy volunteer studies (including sex-based pharmacokinetic differences) to HIV-infected populations or to patients receiving additional concurrent medications.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , HIV Seronegativity/physiology , Saquinavir/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Cohort Studies , Cytochrome P-450 CYP3A/genetics , Female , Genes, MDR/genetics , Genotype , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/blood , HIV Seronegativity/genetics , Humans , Male , Midazolam/administration & dosage , Midazolam/blood , Middle Aged , Phenotype , Saquinavir/adverse effects , Saquinavir/blood , Sex Factors
19.
J Acquir Immune Defic Syndr ; 40(5): 573-80, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16284534

ABSTRACT

Corticosteroid therapy has been associated with bone toxicities (eg, osteonecrosis) and Cushing syndrome in HIV-infected patients; this may be partially attributable to a pharmacokinetic drug interaction between HIV protease inhibitors and corticosteroids. The purpose of this study was to characterize the influence of low-dose ritonavir on prednisolone pharmacokinetics in healthy subjects. Ten HIV-seronegative volunteers were given single oral doses of prednisone, 20 mg, before (baseline) and after receiving ritonavir, 200 mg, twice daily for 4 and 14 days. After each prednisone dose, serial blood samples were collected and prednisolone concentrations were determined; pharmacokinetic parameter values were compared between the groups. Geometric mean ratios (GMRs, 90% confidence interval [CI]) of the prednisolone area under the plasma concentration versus time curve (AUC0-infinity) after 4 and 14 days of ritonavir versus baseline were 1.41 (90% CI: 1.08 to 1.74) and 1.30 (90% CI: 1.09 to 1.49), respectively (P = 0.002 and P = 0.004, respectively). GMRs of prednisolone apparent oral clearance (Cl/F) were 0.71 (09% CI: 0.57 to 0.93) and 0.77 (90% CI: 0.67 to 0.92) after 4 and 14 days of ritonavir versus baseline, respectively (P = 0.0004 and P = 0.0003, respectively). Ritonavir significantly increased the systemic exposure of prednisolone in healthy subjects. Results from this investigation suggest that corticosteroid exposure is likely elevated in HIV-infected patients receiving protease inhibitors.


Subject(s)
Glucocorticoids/pharmacokinetics , HIV Protease Inhibitors/pharmacokinetics , Prednisolone/pharmacokinetics , Ritonavir/pharmacokinetics , Adult , Area Under Curve , Drug Interactions , Female , Glucocorticoids/administration & dosage , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/pharmacology , HIV Seronegativity , Humans , Male , Prednisolone/administration & dosage , Ritonavir/administration & dosage , Ritonavir/pharmacology
20.
Am J Health Syst Pharm ; 62(5): 475-84, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15745910

ABSTRACT

PURPOSE: The results of three studies that describe the external contamination of chemotherapy drug vials are presented. New techniques for the improved decontamination of vials containing cisplatin are also described. SUMMARY: Study 1 evaluated the external contamination of drug vials with cyclophosphamide and ifosfamide in a pharmacy setting. Widespread contamination of the outside of drug vials was found with each drug. Study 2 evaluated the surface contamination of drug vials with cyclophosphamide and fluorouracil in three pharmacies. Sporadic contamination with fluorouracil was detected, while cyclophosphamide was found on most vials. In study 3, investigators compared the decontamination abilities of a standard decontamination procedure at the manufacturer level with an improved decontamination procedure and the use of sleeves to further decrease contamination. Though the methods of each study reported herein differed, the outcomes were similar. All chemotherapy drug vials studied demonstrated levels of contamination with the drug well above the limit of detection. Improved decontamination procedures, combined with the use of protective sleeves, reduced the level of platinum contamination by 90%, suggesting that standard decontamination procedures should be reconsidered. CONCLUSION: The results of these studies are consistent with several others that have reported contamination of the outside surface of drug vials for a number of chemotherapy drugs. Contamination can be reduced by using decontamination equipment and protective sleeves during the manufacturing process.


Subject(s)
Antineoplastic Agents/isolation & purification , Decontamination/methods , Antineoplastic Agents/analysis , Drug Packaging , Equipment Contamination/prevention & control , Pharmacy Service, Hospital
SELECTION OF CITATIONS
SEARCH DETAIL