Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Chirality ; 23(7): 495-503, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21671287

ABSTRACT

Itraconazole is an antifungal drug widely used in a variety of fungal infections, which have become a significant public-health problem in recent decades. Itraconazole is a chiral drug consisting of two diastereoisomeric racemates, i.e., four stereoisomers. Data in the literature suggests that stereochemistry may play a significant role in the action and disposition of the drug and therefore stereoselective analytical methods for the determination of the drug in biological fluids are needed for the elucidation of that role. We report a stereoselective HPLC method that incorporates solvent extraction, the use of an internal standard, two chiral stationary phases in series, and fluorescence detection. The procedure is enantioselective and partially diastereoselective and provides the concentrations in blood plasma of the two epimer mixtures 2R,4S,2'R/2R,4S2'S and 2S,4R,2'R/2S,4R,2'S, respectively, each of which is a combination of the two epimers that differ in the configuration at the sec-butyl group. The analytical method has suitable sensitivity, recovery, precision, and accuracy. Analysis of the plasma of a human subject six hours after the oral administration of a single 200-mg dose of itraconazole showed a 3.4-fold difference between the concentrations of the epimer mixtures. The method has certain advantages over the published alternative procedure that uses LC-MS.


Subject(s)
Blood Chemical Analysis/methods , Itraconazole/blood , Itraconazole/chemistry , Blood Chemical Analysis/standards , Chromatography, High Pressure Liquid , Humans , Itraconazole/isolation & purification , Limit of Detection , Linear Models , Reference Standards , Spectrometry, Fluorescence , Stereoisomerism
2.
HIV Clin Trials ; 11(6): 312-24, 2010.
Article in English | MEDLINE | ID: mdl-21239359

ABSTRACT

BACKGROUND AND OBJECTIVE: Maximizing the durability of viral suppression is a key goal of antiretroviral therapy. The objective of AIDS Clinical Trials Group Study 372A was to determine whether the intensification strategy of adding abacavir to an effective indinavir-dual nucleoside regimen would delay the time to virologic failure. METHODS: Zidovudine-experienced subjects (n=229) on therapy with indinavir + zidovudine + lamivudine with plasma HIV-1 RNA levels<500 copies/mL were randomized to abacavir 300 mg twice daily or placebo. The primary endpoint was the time to treatment failure, defined as a composite of confirmed virologic failure (2 consecutive HIV-1 RNAs>200 copies/mL) and treatment discontinuation. RESULTS: At baseline, the study population was 88% male with a median age of 41 years and median CD4 cell count of 250/mm3. Median follow-up was 4.4 years. The primary endpoint was reached in 61/116 of abacavir versus 62/113 of placebo recipients (P=.77); virologic failure occurred in 34/116 and 42/113 patients, respectively (P=.22). There were no differences in the proportions of subjects with plasma HIV-1 RNA levels below 50 copies/mL, in CD4 cell count increases, nor adverse events between the arms. In the study, 17% of subjects developed nephrolithiasis, 2% experienced abacavir hypersensitivity, and 4.8% experienced at least 1 serious cardiovascular event (7 [6%] in the abacavir arm, 4 [3.5%] in the placebo arm). In additional secondary and post hoc analyses, rates of intermittent viremia, suppression below a plasma HIV-1 RNA level of 6 copies/mL, and HIV-1 proviral DNA levels in peripheral blood mononuclear cells were not significantly different in the 2 arms. CONCLUSIONS: The strategy of intensification with abacavir in patients who are virologically suppressed on a stable antiretroviral regimen does not confer a clinical or virologic benefit. As antiretroviral regimens have become more potent since this trial was completed, it will be even more difficult to prove that late intensification of already virologically suppressed patients will add benefit. However, studies are warranted with drugs with new mechanisms of action to determine whether the level of persistent viremia below 50 copies/ mL can be further reduced and what influence this may have on latent HIV reservoirs.


Subject(s)
Anti-HIV Agents/pharmacology , Dideoxynucleosides/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Adult , Aged , Anti-HIV Agents/standards , CD4 Lymphocyte Count , Dideoxynucleosides/standards , Double-Blind Method , Drug Therapy, Combination , Female , Genotype , HIV Protease Inhibitors/administration & dosage , HIV-1/genetics , Humans , Indinavir/administration & dosage , Male , Middle Aged , Placebos , Treatment Failure , Young Adult , Zidovudine/administration & dosage
3.
J Acquir Immune Defic Syndr ; 50(3): 290-3, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19194314

ABSTRACT

BACKGROUND: Rifampin is the cornerstone of antituberculosis therapy, but induction of hepatic cytochrome P4503A by rifampin markedly lowers HIV protease inhibitor plasma concentrations. METHODS: This phase 1, open-label, one-arm study was designed to assess pharmacokinetic interactions and safety of atazanavir, ritonavir, and rifampin among 14 evaluable HIV-seronegative volunteers. The study included 3 sequential periods of study drug dosing, with plasma sampling for pharmacokinetic analyses to occur on the last day of each period. During period 1, participants received rifampin 600 mg every 24 hours for 8 days. During period 2, participants continued rifampin 600 mg every 24 hours, and added atazanavir 300 mg and ritonavir 100 mg every 12 hours, to continue for at least 11 days. During period 3, atazanavir was to be increased to 400 mg every 12 hours. RESULTS: Upon adding atazanavir and ritonavir, the first 3 subjects developed vomiting and transaminase elevations resulting in study drug discontinuation. The study was therefore terminated. CONCLUSIONS: Coadministration of rifampin with HIV protease inhibitors may not be a viable treatment option if rifampin administration precedes protease inhibitor initiation. Future studies, which explore concomitant HIV protease inhibitors with rifampin must carefully consider the sequence in which drugs are initiated.


Subject(s)
Antitubercular Agents/adverse effects , Gastrointestinal Tract/drug effects , HIV Protease Inhibitors/adverse effects , Liver/drug effects , Oligopeptides/adverse effects , Pyridines/adverse effects , Rifampin/adverse effects , Ritonavir/adverse effects , Adult , Antitubercular Agents/administration & dosage , Atazanavir Sulfate , Female , HIV Protease Inhibitors/administration & dosage , Humans , Male , Oligopeptides/administration & dosage , Pyridines/administration & dosage , Rifampin/administration & dosage , Ritonavir/administration & dosage
4.
J Acquir Immune Defic Syndr ; 47(5): 570-8, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18176327

ABSTRACT

OBJECTIVES: This open-label, single-arm, pharmacokinetic (PK) study in HIV-seronegative volunteers evaluated the bioequivalence of rosuvastatin and lopinavir/ritonavir when administered alone and in combination. Tolerability and lipid changes were also assessed. METHODS: Subjects took 20 mg of rosuvastatin alone for 7 days, then lopinavir/ritonavir alone for 10 days, and then the combination for 7 days. Intensive PK sampling was performed on days 7, 17, and 24. RESULTS: Twenty subjects enrolled, and PK data were available for 15 subjects. Geometric mean (+/-SD) rosuvastatin area under the concentration time curve (AUC)[0,tau] and maximum concentration (Cmax) were 47.6 ng.h/mL (+/-15.3) and 4.34 ng/mL (+/-1.8), respectively, when given alone versus 98.8 ng.h/mL (+/-65.5) and 20.2 ng/mL (+/-16.9) when combined with lopinavir/ritonavir (P < 0.0001). The geometric mean ratio was 2.1 (90% confidence interval [CI]: 1.7 to 2.6) for rosuvastatin AUC[0,tau] and 4.7 (90% CI: 3.4 to 6.4) for rosuvastatin Cmax with lopinavir/ritonavir versus rosuvastatin alone (P < 0.0001). There was 1 asymptomatic creatine phosphokinase elevation 17 times the upper limit of normal (ULN) and 1 liver function test elevation between 1.1 and 2.5 times the ULN with the combination. CONCLUSIONS: Rosuvastatin low-density lipoprotein reduction was attenuated with lopinavir/ritonavir. Rosuvastatin AUC and Cmax were unexpectedly increased 2.1- and 4.7-fold in combination with lopinavir/ritonavir. Rosuvastatin and lopinavir/ritonavir should be used with caution until the safety, efficacy, and appropriate dosing of this combination have been demonstrated in larger populations.


Subject(s)
Fluorobenzenes/pharmacokinetics , Pyrimidines/pharmacokinetics , Pyrimidinones/pharmacokinetics , Ritonavir/pharmacokinetics , Sulfonamides/pharmacokinetics , Adolescent , Adult , Drug Interactions , Female , Fluorobenzenes/administration & dosage , Fluorobenzenes/adverse effects , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/pharmacokinetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Lipids/blood , Lopinavir , Male , Middle Aged , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidinones/administration & dosage , Pyrimidinones/adverse effects , Ritonavir/administration & dosage , Ritonavir/adverse effects , Rosuvastatin Calcium , Sulfonamides/administration & dosage , Sulfonamides/adverse effects
5.
J Acquir Immune Defic Syndr ; 47(4): 459-66, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-17971707

ABSTRACT

INTRODUCTION: Fish oil has been shown to reduce serum triglyceride (TG) concentrations. In HIV-infected patients on antiretroviral therapy, high TG concentrations likely contribute to increased risk of cardiovascular disease. AIDS Clinical Trials Group A5186 examined the safety and efficacy of fish oil plus fenofibrate in subjects not achieving serum TG levels < or =200 mg/dL with either agent alone. METHODS: One hundred subjects on highly active antiretroviral therapy with serum TG concentrations > or =400 mg/dL and low-density lipoprotein cholesterol < or =160 mg/dL were randomized to 3 g of fish oil twice daily or 160 mg of fenofibrate daily for 8 weeks. Subjects with a fasting TG level >200 mg/dL at week 8 received a combination of fish oil and fenofibrate in the same doses from week 10 to week 18. RESULTS: Median baseline TG was 662 mg/dL in the fish oil group and 694 mg/dL in the fenofibrate group (P = not significant). Fish oil reduced TG levels by a median of 283 mg/dL (46%), fenofibrate reduced them by 367 mg/dL (58%), and combination therapy reduced them by 65.5%. Combination therapy achieved TG levels of < or =200 mg/dL in 22.7% subjects. Fish oil had no measurable effect on immunologic parameters or the pharmacokinetics of lopinavir. CONCLUSIONS: Fish oil was safe when administered alone or combined with fenofibrate and significantly reduced TG levels in HIV-infected subjects with hypertriglyceridemia.


Subject(s)
Fenofibrate/therapeutic use , Fish Oils/therapeutic use , HIV Infections/drug therapy , Hypertriglyceridemia/drug therapy , Adult , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Drug Therapy, Combination , Female , HIV Infections/blood , HIV Infections/complications , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/complications , Hypolipidemic Agents/therapeutic use , Male , Middle Aged , Time Factors , Treatment Outcome , Triglycerides/blood
6.
Antimicrob Agents Chemother ; 51(9): 3104-10, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17576825

ABSTRACT

The potent induction of hepatic cytochrome P450 3A isoforms by rifampin complicates therapy for coinfection with human immunodeficiency virus (HIV) and Mycobacterium tuberculosis. We performed an open-label, single-arm study to assess the safety and pharmacokinetic interactions of the HIV protease inhibitor atazanavir coadministered with rifampin. Ten healthy HIV-negative subjects completed pharmacokinetic sampling at steady state while receiving 300 mg atazanavir every 12 h without rifampin (period 1), 300 mg atazanavir every 12 h with 600 mg rifampin every 24 h (period 2), and 400 mg atazanavir every 12 h with 600 mg rifampin every 24 h (period 3). During period 1, the mean concentration of drug in serum at 12 h (C(12 h)) was 811 ng/ml (range, 363 to 2,484 ng/ml) for atazanavir, similar to historic seronegative data for once-daily treatment with 300 mg atazanavir boosted with 100 mg ritonavir. During periods 2 and 3, the mean C(12 h) values for atazanavir were 44 ng/ml (range, <25 to 187 ng/ml) and 113 ng/ml (range, 39 to 260 ng/ml), respectively, well below historic seronegative data for once-daily treatment with 400 mg atazanavir without ritonavir. Although safe and generally well tolerated, 300 mg or 400 mg atazanavir administered every 12 h did not maintain adequate plasma exposure when coadministered with rifampin.


Subject(s)
Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacokinetics , Antitubercular Agents/administration & dosage , Antitubercular Agents/pharmacokinetics , HIV Infections/drug therapy , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Rifampin/administration & dosage , Rifampin/pharmacokinetics , Tuberculosis/drug therapy , Adolescent , Adult , Anti-HIV Agents/adverse effects , Antitubercular Agents/adverse effects , Area Under Curve , Atazanavir Sulfate , Biotransformation , Drug Interactions , Female , HIV Infections/complications , Half-Life , Humans , Male , Middle Aged , Oligopeptides/adverse effects , Pyridines/adverse effects , Rifampin/adverse effects , Tuberculosis/complications
7.
Antivir Ther ; 11(5): 619-23, 2006.
Article in English | MEDLINE | ID: mdl-16964830

ABSTRACT

BACKGROUND: Amdoxovir (2,6-diaminopurine dioxolane; DAPD) is a nucleoside reverse transcriptase inhibitor (NRTI) of human immunodeficiency virus-1 (HIV-1) with activity against wild-type and NRTI-resistant viruses. METHODS: ACTG A5118 assessed the antiretroviral activity and safety of DAPD (300 mg orally, twice daily) versus placebo in combination with enfuvirtide (ENF) plus an optimized background (OB) regimen in subjects with failure of two or more antiretroviral (ARV) regimens. The primary endpoints for comparison were time-averaged area under the curve minus baseline (AAUCMB) of plasma HIV-1 RNA concentration at 24 weeks and time to first serious (DAIDS toxicity table Grade > or = 3) adverse event (AE). An unplanned interim review recommended closing enrollment because the study was unlikely to demonstrate a difference between arms. The 18 subjects on study, nine in each arm, were unblinded and allowed to continue study treatment through 48 weeks. RESULTS: Intention-to-treat analysis showed the median AAUCMB was -0.9 log10 copies/mL (95% CI = -2.2, -.0.1) in the DAPD arm and -0.9 log10 copies/ml (95% CI = -1.1, -0.1) in the placebo arm (P = 0.69). Median CD4+ T-cell increase was 79 cells/mm3 (95% CI =1, 115) in the DAPD arm and 60 (95% CI =1, 101) in the placebo arm (P = 0.45). Time to first serious AE did not differ between arms (P = 0.91). Mild decreases of creatinine clearance were observed with similar frequency between arms; no subject developed lens opacities. CONCLUSIONS: Addition of DAPD to ENF plus OB in advanced subjects with highly resistant virus appeared safe, but did not add statistically significant antiretroviral activity at 24 weeks in this small study.


Subject(s)
Dioxolanes/therapeutic use , HIV Envelope Protein gp41/therapeutic use , HIV Fusion Inhibitors/therapeutic use , HIV Infections/drug therapy , HIV-1 , Peptide Fragments/therapeutic use , Purine Nucleosides/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Adult , CD4 Lymphocyte Count , Dioxolanes/adverse effects , Drug Resistance, Viral , Drug Therapy, Combination , Enfuvirtide , Female , HIV Infections/immunology , HIV Infections/virology , Humans , Male , Middle Aged , Pilot Projects , Purine Nucleosides/adverse effects , Reverse Transcriptase Inhibitors/adverse effects , Treatment Failure , Viral Load
8.
J Pharmacokinet Pharmacodyn ; 33(4): 399-419, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16583266

ABSTRACT

We developed a novel HIV-1 dynamic model with consideration of pharmacokinetics, drug adherence and drug susceptibility to link plasma drug concentration to the long-term changes in HIV-1 RNA observation after initiation of therapy. A Bayesian approach is proposed to fit this model to clinical data from ACTG A5055, a study of two dosage regimens of indinavir (IDV) with ritonavir (RTV) in subjects failing their first protease inhibitor treatment. The HIV RNA testing was completed at days 0, 7, 14, 28, 56, 84, 112, 140, and 168. An intensive pharmacokinetic (PK) evaluation was performed on day 14 and multiple trough concentrations were subsequently collected. Pill counts were used to monitor adherence. IC(50) for IDV and RTV were determined at baseline and at virologic failure. Viral dynamic model fitting residuals were used to assess the significance of covariate effects on long-term virologic response. As univariate predictors, none of the four PK parameters C(trough), C(12 hour), C(max), and AUC was significantly related to virologic response (p > 0.05). By including drug susceptibility (IC(50)), or IC(50) and adherence measured by pill counts together, C(trough), C(12 hour), C(max) and AUC were each significantly correlated to long-term virologic response (p = 0.0055,0.0002,0.0136,0.0002 with IC(50) and adherence measured by pill counts considered). The IC(50) and adherence measured by pill counts alone were not related to the virologic response. In predicting virologic response adherence measured by pill counts did not provide any additional information to PK parameters (p = 0.064), to drug susceptibility IC(50) (p = 0.086), and to their combination (p = 0.22). Simple regression approaches did not detect any significant pharmacodynamic (PD) relationships. Any single factor of PK, adherence measured by pill counts and drug susceptibility did not contribute to long-term virologic response. But their combinations in viral dynamic modeling significantly predicted virologic response. The HIV dynamic modeling can appropriately capture complicated nonlinear relationships and interactions among multiple covariates.


Subject(s)
Anti-Retroviral Agents/pharmacokinetics , HIV Infections/drug therapy , HIV-1/drug effects , Models, Biological , Algorithms , Anti-Retroviral Agents/administration & dosage , Antiretroviral Therapy, Highly Active , Bayes Theorem , Drug Resistance, Viral , HIV Infections/metabolism , HIV-1/isolation & purification , Humans , Indinavir/administration & dosage , Indinavir/pharmacokinetics , Regression Analysis , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/pharmacokinetics , Ritonavir/administration & dosage , Ritonavir/pharmacokinetics , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Viral Load
9.
AIDS ; 20(5): 725-9, 2006 Mar 21.
Article in English | MEDLINE | ID: mdl-16514303

ABSTRACT

BACKGROUND: Nelfinavir, an HIV protease inhibitor with numerous drug-drug interactions, is associated with dyslipidemia. Pravastatin is the preferred statin prescribed for HIV-associated dyslipidemia. OBJECTIVE: To examine the effect of nelfinavir on pravastatin pharmacokinetics. DESIGN: Open-label study in healthy HIV-seronegative adults conducted at the AIDS Clinical Trials Group sites in the United States. METHODS: Subjects received pravastatin 40 mg daily and underwent intensive sampling for pharmacokinetics on day 3. Subjects took only nelfinavir 1250 mg twice daily on days 4-12. On days 13-15, subjects continued nelfinavir and reinitiated pravastatin. Plasma samples were collected over 24 h for the calculation of pravastatin area under the concentration-time curve for 0-24 h on days 3 and 16. RESULTS: Data from 14 subjects with complete pharmacokinetic samples were available for analysis. The median within-subject percentage change in pravastatin AUC was a decrease of 46.5%. Pravastatin maximum plasma concentrations were also lower when pravastatin was administered with nelfinavir. Median values for the maximum plasma concentrations were 27.9 and 12.4 ng/ml for days 3 and 16, respectively, and the median within-subject decrease was 40.1%. CONCLUSIONS: Coadministration of pravastatin and nelfinavir led to a substantial reduction in pravastatin plasma concentrations. Higher doses of pravastatin may need to be prescribed in order to achieve optimal lipid-lowering activity.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Nelfinavir/pharmacokinetics , Pravastatin/pharmacokinetics , Adult , Area Under Curve , Drug Administration Schedule , Drug Interactions , Female , HIV Protease Inhibitors/blood , HIV Seronegativity , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Male , Nelfinavir/blood , Pravastatin/blood , Time Factors , Volunteers
10.
Curr HIV/AIDS Rep ; 2(2): 61-7, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16091250

ABSTRACT

Therapeutic drug monitoring (TDM) is the process by which a patient's dosing regimen is guided by repeated measures of plasma drug concentrations. An enormous challenge with regard to TDM of antiretroviral drugs (ARV) is that the concentration goals can be moving targets. Well-designed prospective studies demonstrating that prospectively altering ARV doses based on TDM leads to virologic success and increased tolerability are needed. Nevertheless, there are specific clinical instances where this experimental intervention should be considered to potentially reduce toxicity and improve therapeutic outcomes.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Drug Monitoring/methods , HIV Infections/drug therapy , Adolescent , Adult , Anti-Retroviral Agents/administration & dosage , Anti-Retroviral Agents/adverse effects , Child , Drug Administration Schedule , Drug Interactions , Drug Monitoring/economics , Female , HIV Infections/blood , Humans , Patient Compliance , Prospective Studies
11.
Clin Pharmacol Ther ; 78(2): 143-53, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16084849

ABSTRACT

BACKGROUND: Hypertension is an important modifiable cardiac risk factor in human immunodeficiency virus (HIV)-infected patients. Calcium channel blockers are substrates of cytochrome P450 3A and are commonly prescribed for hypertension. We evaluated potential bidirectional pharmacokinetic interactions between calcium channel blockers and coadministered indinavir and ritonavir. METHODS: Healthy HIV- seronegative subjects received 120 mg diltiazem daily or 5 mg amlodipine daily for days 1 to 7 and 20 to 26. All subjects received 100 mg ritonavir and 800 mg indinavir every 12 hours on days 8 to 26. Twenty-four-hour pharmacokinetic collection was performed on days 7 and 26, with 12-hour collection on day 19. RESULTS: Indinavir plus ritonavir increased the median amlodipine area under the curve from 0 to 24 hours (AUC) by 89.8%, from 122 to 230 ng.h/mL (n = 18, P < .0001), and increased the median diltiazem AUC by 26.5%, from 800 to 1060 ng.h/mL (n = 13, P = .06). Of 13 subjects, 2 (15%) had greater than 4-fold increases in diltiazem AUC. Desacetyldiltiazem AUC increased by 102.2% (P = .001), and desmethyldiltiazem AUC decreased by 27.4% (P = .01). Neither amlodipine nor diltiazem affected steady-state AUCs of the protease inhibitors. No serious cardiovascular adverse effects were observed. CONCLUSIONS: Indinavir plus ritonavir increases the AUCs of both amlodipine and diltiazem, which may result in an increased response. If coadministration is indicated, amlodipine or diltiazem should be initiated at low doses with careful titration to response and side effects.


Subject(s)
Amlodipine/pharmacology , Calcium Channel Blockers/pharmacology , Diltiazem/pharmacology , HIV Infections , HIV Protease Inhibitors/pharmacokinetics , Indinavir/pharmacokinetics , Ritonavir/pharmacokinetics , Adolescent , Adult , Drug Interactions , Drug Therapy, Combination , Female , HIV Protease Inhibitors/administration & dosage , Humans , Hypertension/drug therapy , Indinavir/administration & dosage , Male , Middle Aged , Ritonavir/administration & dosage
12.
J Acquir Immune Defic Syndr ; 39 Suppl 1: S1-23, quiz S24-25, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15990598

ABSTRACT

The trend toward once-daily dosing in HIV antiretroviral therapy is based on the association between adherence, treatment outcome, and patient preferences. Patients prefer simpler treatments, fewer pills, less frequent dosing, and no food restrictions. When a regimen meets a patient's preferences, the patient is more likely to be adherent, and with good adherence, the regimen is more likely to be effective. Nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) have been a prime focus for developing once-daily therapies primarily because they form the backbone of most current regimens. Within the NRTI class, however, drugs differ in their pharmacokinetic properties, such as plasma and intracellular half-lives, and thus in their suitability for once-daily dosing. For example, newer NRTIs, such as tenofovir and emtricitabine, combine longer plasma half-lives with longer intracellular half-lives, prolonging exposure and the period of pharmacologic activity. Of equal importance, the clinical impact of systemic and intracellular interactions between concomitant drugs defines which once-daily drugs may be combined in once-daily regimens. To construct simplified and effective therapies for individual patients, clinicians require an understanding of the plasma and intracellular pharmacokinetic properties of NRTIs and how these properties determine a drug's appropriateness for once-daily dosing and placement within a once-daily regimen.


Subject(s)
HIV Infections/drug therapy , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/pharmacokinetics , Drug Administration Schedule , Drug Resistance, Multiple, Viral , Drug Therapy, Combination , HIV-1/drug effects , HIV-1/genetics , Humans , Mutation , Patient Compliance , Reverse Transcriptase Inhibitors/chemistry
13.
AIDS ; 19(10): 1059-63, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15958837

ABSTRACT

OBJECTIVE: To describe the in vivo protein-binding characteristics of indinavir (IDV) in the presence of ritonavir (RTV) relative to total IDV plasma concentrations. DESIGN: The ACTG protocol 5055 was a multicenter study comparing the safety and pharmacokinetics of IDV/RTV at doses of 800/200 and 400/400 mg twice daily in HIV-infected adults. METHODS: Forty-four patients underwent a 12-h intensive pharmacokinetic assessment after 2 weeks of therapy. Three plasma samples from 35 patients at Cmax, 6 and 12 h post dose were used to determine the unbound IDV concentrations. Unbound IDV was separated in plasma samples using ultra-filtration and measured using high-performance liquid chromatography with UV detection. RESULTS: Mean IDV protein-bound fraction across all time points in the 800/200 and 400/400 arm were 53.4 and 51.8%, respectively. In the 800/200 arm, percentage binding at Cmax was 50% compared with 56% at 12 h (P = 0.008). In the 400/400 arm, percentage binding at Cmax was 49% compared with 54% at 12 h (P = 0.008). CONCLUSIONS: The extent of plasma protein binding of IDV in this study was less than in previously published data with IDV alone. Although IDV concentrations differed across the arms, the percentage of IDV protein binding at all time points was not different between the 800/200 and 400/400 arms. However, the percentage of IDV protein binding at Cmax was significantly lower compared with 12 h in each arm, possibly suggesting that IDV protein binding is concentration-dependent. These data suggest that RTV affects IDV protein-binding characteristics and IDV also exhibits concentration dependent binding when administered with RTV.


Subject(s)
Anti-HIV Agents/metabolism , HIV Infections/drug therapy , HIV Protease Inhibitors/metabolism , Indinavir/metabolism , Ritonavir/metabolism , Adult , Anti-HIV Agents/administration & dosage , Dose-Response Relationship, Drug , Drug Combinations , Female , HIV Protease Inhibitors/administration & dosage , Humans , Indinavir/administration & dosage , Male , Middle Aged , Protein Binding , Ritonavir/administration & dosage
14.
J Acquir Immune Defic Syndr ; 39(3): 272-83, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15980686

ABSTRACT

We propose a long-term HIV-1 dynamic model by considering drug potency, drug exposure, and drug susceptibility. Using a Bayesian approach, HIV-1 dynamic parameters were estimated by fitting the model to viral load data from a phase 1/2 randomized clinical study of 2 indinavir (IDV)/ritonavir (RTV)-containing highly active antiretroviral (ARV) therapy regimens in HIV-infected subjects who had previously failed protease inhibitor-containing ARV therapies. A large between-subject variation in estimated viral dynamic parameters was observed, even after accounting for variations in drug exposure and drug susceptibility, suggesting that characteristics of HIV-1 dynamics are host dependent. Significant correlations of baseline factors such as HIV-1 RNA levels and CD4 cell counts with viral dynamic parameters were found. These correlations coincide with biologic interaction mechanisms between HIV and the host immune system and also provide an explanation for the correlations between the baseline viral load and phase 1 viral decay rate, for which inconsistent results have been reported in the literature. The relations between viral dynamic parameters and virologic response were established, and these results suggest that viral dynamic parameters may play an important role in determining treatment success or failure. In particular, we estimated a drug efficacy threshold for each patient that can be used to assess whether an ARV regimen is potent enough to suppress HIV viruses in the individual patient. Our findings indicate that it is necessary to individualize the ARV regimen to treat HIV-1-infected patients. The proposed mathematic models and statistical techniques may provide a framework to simulate and predict antiviral response for individual patients.


Subject(s)
Anti-HIV Agents/pharmacology , Anti-HIV Agents/pharmacokinetics , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , Models, Biological , Bayes Theorem , CD4 Lymphocyte Count , Drug Resistance, Viral , HIV Infections/immunology , HIV Infections/metabolism , Humans , Mathematics , Patient Compliance , RNA, Viral/blood
15.
J Acquir Immune Defic Syndr ; 39(3): 307-12, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15980690

ABSTRACT

Efavirenz (EFV) is associated with hyperlipidemia when used in combination with other antiretroviral drugs. EFV is a mixed inducer/inhibitor of cytochrome P450 (CYP) 3A4 isozyme and may interact with hydroxymethylglutaryl coenzyme A (HMG-CoA) reductase inhibitors that are primarily metabolized via CYP3A4. To assess the drug-drug interaction of EFV used in combination with simvastatin (SIM), atorvastatin (ATR), or pravastatin (PRA), an open-label trial was conducted in 52 healthy adult HIV-seronegative subjects across AIDS Clinical Trials Group sites in the United States. Subjects received 40 mg of SIM, 10 mg of ATR, or 40 mg of PRA daily on days 0 through 3 and days 15 through 18. EFV was administered daily at a dose of 600 mg on days 4 through 18. SIM, ATR, and PRA concentrations were determined before and after EFV, and EFV concentrations were determined before and after statins. EFV reduced SIM acid exposure (area under the curve at 0 to 24 hours [AUC0-24 h]) by 58% (Wilcoxon signed rank test, P=0.003) and active HMG-CoA reductase inhibitory activity by 60% (P<0.001). EFV reduced ATR exposure by 43% (P<0.001) and the total active ATR exposure by 34% (P=0.005). EFV administration resulted in a 40% decrease in PRA exposure (P=0.005). SIM, ATR, and PRA had no effect on non-steady-state EFV concentrations. In conclusion, EFV, when administered with SIM, ATR, or PRA, can result in significant induction of statin metabolism. The reduced inhibition of HMG-CoA reductase activity during coadministration of EFV may result in diminished antilipid efficacy at usual doses of SIM, ATR, and PRA.


Subject(s)
Anti-HIV Agents/adverse effects , Heptanoic Acids/administration & dosage , Heptanoic Acids/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hyperlipidemias/etiology , Oxazines/administration & dosage , Oxazines/adverse effects , Pravastatin/administration & dosage , Pravastatin/pharmacokinetics , Pyrroles/administration & dosage , Pyrroles/pharmacokinetics , Simvastatin/administration & dosage , Simvastatin/pharmacokinetics , Adult , Alkynes , Atorvastatin , Benzoxazines , Cholesterol, LDL/blood , Cyclopropanes , Drug Interactions , Female , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Seronegativity , Humans , Hyperlipidemias/drug therapy , Hyperlipidemias/metabolism , Male
17.
Chirality ; 17 Suppl: S101-8, 2005.
Article in English | MEDLINE | ID: mdl-15772975

ABSTRACT

The absolute configuration assignments of three antifungal agents, (+)-(2R,4S)-ketoconazole, (+)-(2R,4S)-itraconazole (with (S)-configuration at the sec-butyl group) and (+)-(S)-miconazole nitrate have been confirmed by using vibrational circular dichroism (VCD). For these three antifungal drugs, this study also provides evidence for the most abundant conformations of miconazole and for the relative conformations of the azole, dichlorophenyl, and methoxyphenyl groups in ketoconazole and itraconazole, in chloroform solution.


Subject(s)
Antifungal Agents/chemistry , Itraconazole/chemistry , Ketoconazole/chemistry , Miconazole/chemistry , Circular Dichroism , Models, Molecular , Molecular Conformation , Solutions , Spectroscopy, Fourier Transform Infrared , Stereoisomerism
18.
Clin Infect Dis ; 40(1): 174-81, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15614709

ABSTRACT

Chronic viral hepatitis is common among persons with HIV-1 infection, because of shared modes of transmission, and coinfection results in accelerated liver damage, compared with persons with chronic viral hepatitis alone. The use of highly active antiretroviral therapy (HAART) has led to a significant decrease in the morbidity and mortality associated with HIV-1 infection. A number of the medications that are commonly used in HAART regimens are metabolized by the hepatic CYP enzymes, which raises the possibility of significant interactions between antiretroviral medications and hepatic impairment induced by chronic viral hepatitis. Although the data are still very scant, the pharmacokinetics of several antiretroviral medications have been shown to be significantly altered in the presence of liver disease. In the present report, we review the available data and consider potential options, such as dose adjustment and therapeutic drug monitoring, for the administration of antiretroviral therapy to patients with significant hepatic impairment.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Antiretroviral Therapy, Highly Active , HIV Infections/metabolism , Hepatitis/metabolism , Alkynes , Benzoxazines , Cyclopropanes , Dideoxynucleosides/pharmacokinetics , Drug Therapy, Combination , HIV Infections/complications , Hepatitis/complications , Humans , Indinavir/pharmacokinetics , Liver Function Tests , Lopinavir , Nelfinavir/pharmacokinetics , Nevirapine/pharmacokinetics , Oxazines/pharmacokinetics , Protease Inhibitors/pharmacokinetics , Pyrimidinones/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Ritonavir/pharmacokinetics , Saquinavir/pharmacokinetics , Zidovudine/pharmacokinetics
19.
J Acquir Immune Defic Syndr ; 37(3): 1358-66, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15483465

ABSTRACT

BACKGROUND: Pharmacokinetic enhancement of protease inhibitors (PIs) with low-dose ritonavir (RTV) for salvage therapy is increasingly common. The purpose of this study was to compare the pharmacokinetics, safety, and tolerability of indinavir (IDV)/RTV at 800/200 mg (arm A) and 400/400 mg (arm B) administered twice daily in HIV-infected subjects failing their first PI-based regimen. METHODS: A phase I/II, randomized, open-label, 24-week study was conducted. Formal 12-hour pharmacokinetic evaluations were performed, and study visits occurred at baseline; at weeks 1, 2, and 4; and every 4 week thereafter for 24 weeks. Clinical symptoms and laboratory assessments were collected. Subjects were allowed to switch arms because of toxicity. RESULTS: Forty-four subjects were enrolled (22 per arm). IDV predose concentration, maximum plasma concentration and area under the curve were significantly higher in arm A. Fifty-five percent and 45% of subjects in arms A and B responded (<200 copies/mL at week 24; P = 0.76), respectively. CD4 cell responses were similar. All subjects had IDV-sensitive virus at baseline and at virologic failure. Tolerability was comparable, but all grade 3 or higher triglyceride increases occurred in arm B and more subjects in arm B switched because of toxicity (5 vs. 1 triglyceride increases). CONCLUSIONS: This is the largest formal pharmacokinetic evaluation of 2 dosage combinations of IDV/RTV in HIV-infected individuals. Pharmacokinetic parameters were consistent with previous results in patients but lower than in seronegative controls. Both regimens exhibited similar tolerability and response rates. High toxicity with a low response suggests that the optimum IDV/RTV combination would include an RTV dose <400 mg and an IDV dose <800 mg in this population.


Subject(s)
HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , Indinavir/therapeutic use , Ritonavir/therapeutic use , Adolescent , Adult , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/immunology , Drug Administration Schedule , Drug Therapy, Combination , Ethnicity , Female , HIV Infections/blood , HIV Infections/immunology , HIV-1/genetics , HIV-1/isolation & purification , Humans , Indinavir/administration & dosage , Indinavir/pharmacokinetics , Male , Middle Aged , RNA, Viral/blood , RNA, Viral/genetics , Ritonavir/administration & dosage , Ritonavir/pharmacokinetics , Substance Abuse, Intravenous/complications , Substance Abuse, Intravenous/epidemiology , Viral Load
20.
Chirality ; 16(2): 79-85, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14712470

ABSTRACT

Racemic ketoconazole (KTZ) was the first orally active azole antifungal agent used in clinical practice and has become widely used in the treatment of mucosal fungal infections associated with AIDS immunosuppression and cancer chemotherapy. However, the use of KTZ has been limited because of adverse drug-drug interactions. KTZ blocks ergosterol biosynthesis by inhibiting the fungal cytochrome P450 (CYP51). KTZ is also a potent inhibitor of human cytochrome P450 3A4 (CYP3A4) enzyme, the major drug-metabolizing CYP isozyme in the human liver. We examined the enantioselective differences of KTZ in the inhibition of human CYP3A4 and in antifungal action. Dextro- and levo-KTZ exhibited modest enantioselective differences with respect to CYP3A4 inhibition of testosterone and methadone metabolism. For both substrates levo-KTZ was approximately a 2-fold more potent inhibitor. We examined the enantioselective differences in the in vitro activity of KTZ against medically relevant species of Candida and Aspergillus, as well as Cryptococcus neoformans. Overall, levo-KTZ was 2-4-fold more active than dextro-KTZ. Therefore, levo-KTZ is a more potent inhibitor of CYP3A4 and has stronger in vitro antifungal activity. Chirality 16:79-85, 2004.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Ketoconazole/chemistry , Ketoconazole/pharmacology , Methadone/metabolism , Testosterone/metabolism , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Candida/drug effects , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Humans , Inhibitory Concentration 50 , Molecular Structure , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...