Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 261
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Vet Pharmacol Ther ; 44(1): 116-125, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32744755

ABSTRACT

Orosomucoid polymorphisms influence plasma drug binding in humans; however, canine variants and their effect on drug plasma protein binding have not yet been reported. In this study, the orosomucoid gene (ORM1) was sequenced in 100 dogs to identify the most common variant and its allele frequency determined in 1,464 dogs (from 64 breeds and mixed-breed dogs). Plasma protein binding extent of amitriptyline, indinavir, verapamil, and lidocaine were evaluated by equilibrium dialysis using plasma from ORM1 genotyped dogs (n = 12). Free and total drug plasma concentrations were quantified by liquid chromatography-mass spectrometry. From the five polymorphisms identified in canine ORM1, two were nonsynonymous. The most common was c.70G>A (p.Ala24Thr) with an allele frequency of 11.2% (n = 1464). Variant allele frequencies varied by breed, reaching 74% in Shetland Sheepdogs (n = 21). Free drug fractions did not differ significantly (p > .05; Mann-Whitney U) between plasma collected from dogs with c.70AA (n = 4) and those with c.70GG (n = 8) genotypes. While c.70G>A did not affect the extent of plasma protein binding in our study, the potential biological and pharmacological implication of this newly discovered ORM1 variant in dogs should be further investigated.


Subject(s)
Blood Proteins/metabolism , Dogs/genetics , Genotype , Orosomucoid/metabolism , Polymorphism, Genetic , Amitriptyline/pharmacokinetics , Anesthetics, Local/pharmacokinetics , Animals , Anti-Arrhythmia Agents/pharmacokinetics , Antidepressive Agents, Tricyclic/pharmacokinetics , Dogs/blood , Dogs/metabolism , Gene Expression Regulation/physiology , HIV Protease Inhibitors/pharmacokinetics , Indinavir/pharmacokinetics , Lidocaine/pharmacokinetics , Orosomucoid/genetics , Protein Binding , Verapamil/pharmacokinetics
2.
Drug Dev Ind Pharm ; 45(5): 736-744, 2019 May.
Article in English | MEDLINE | ID: mdl-30640551

ABSTRACT

OBJECTIVE: Indinavir (IDV), an antiretroviral protease inhibitor used in treatment of HIV infection, has limited entry into brain due to efflux by the P-glycoprotein presented in blood-brain barrier. The aim of present study was to develop lactoferrin-treated nanoemulsion containing indinavir (Lf-IDV-NEs) for delivery to brain. METHODS: Indinavir-loaded nanoemulsions (IDV-NEs) were prepared by high-speed homogenization method, and then lactoferrin was coupled to IDV-NEs by water soluble EDC method. RESULTS: The hydrodynamic diameters, polydispersity index, and zeta potential of IDV-NEs were 112 ± 3.5 nm, 0.20 ± 0.02, and -33.2 ± 2.6 mV, respectively. From in vivo studies in animal model of rats, the AUC0-4 h of brain concentration-time profile of IDV-NEs and Lf-IDV-NEs were 1.6 and 4.1 times higher than free drug, respectively. The brain uptake clearance of IDV-NEs and Lf-IDV-NEs were, interestingly, 393- and 420-times higher than the free drug. CONCLUSIONS: It can be concluded that applying both lactoferrin-treated and non-treated nanoemulsions clearly leads to significant brain penetration enhancement of indinavir, an effect which is more pronounced in the case of Lf-IDV-NEs with the higher drug residence time in brain.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Carriers/chemistry , HIV Protease Inhibitors/pharmacokinetics , Indinavir/pharmacokinetics , Lactoferrin/chemistry , Animals , Area Under Curve , Drug Liberation , Emulsions , HIV Infections/drug therapy , HIV Protease Inhibitors/administration & dosage , Indinavir/administration & dosage , Injections, Intravenous , Male , Nanoparticles/chemistry , Permeability , Polysorbates/chemistry , Rats , Rats, Sprague-Dawley
3.
Planta Med ; 84(12-13): 895-901, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29672818

ABSTRACT

Hypoxis hemerocallidea (African potato) is a popular medicinal plant that has been used traditionally for the treatment of various disorders. Some HIV/AIDS patients use this traditional medicine together with their antiretroviral therapy. This study aimed to determine the impact of selected H. hemerocallidea materials (i.e., a commercial product, an aqueous extract, and biomass reference plant material) on the bidirectional permeability of indinavir across Caco-2 cell monolayers as well as the bioavailability of indinavir during an acute, single administration study in Sprague-Dawley rats. All of the selected H. hemerocallidea test materials demonstrated inhibition effects on indinavir efflux across Caco-2 cell monolayers, albeit to different extents. An increase in the bioavailability of indinavir was obtained in vivo when administered concomitantly with the H. hemerocallidea materials, albeit not statistically significantly. The change in bioavailability directly correlated with the in vitro permeability results. It can therefore be concluded that the change in permeability and bioavailability of indinavir was caused by efflux inhibition and this effect was dependent on the type of H. hemerocallidea material investigated, which was found to be in the following order: commercial product > aqueous extract > reference plant material. The clinical significance of the combined effect of efflux and metabolism inhibition by H. hemerocallidea should be determined in another in vivo model that expresses the cytochrome P450 3A4 enzyme.


Subject(s)
HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Herb-Drug Interactions , Hypoxis/chemistry , Indinavir/pharmacokinetics , Plant Extracts/pharmacology , Animals , Biological Availability , Caco-2 Cells , Chromatography, Liquid , Humans , Male , Mass Spectrometry , Medicine, Traditional , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Rats, Sprague-Dawley
4.
Am J Ther ; 24(4): e405-e418, 2017.
Article in English | MEDLINE | ID: mdl-26291590

ABSTRACT

In the present day antiretroviral therapy, Ctrough is a key tool for efficacy assessment. The present work explored the feasibility of using Ctrough or Cmax in the area under the concentration-time curve (AUC) prediction of indinavir. A simple unweighted linear regression model was developed to describe the relationship between Cmax versus AUC (r = 0.8101, P < 0.001) and Ctrough versus AUC (r = 0.8127, P < 0.001) for indinavir. The regression lines were used to predict the AUC values from literature Cmax or Ctrough data of indinavir in HIV and healthy subjects. The fold difference, defined as the quotient of the observed and predicted AUC values, was evaluated along with statistical comparison, including root mean square error (RMSE) prediction for the 2 models. The correlation between Cmax versus AUC and Ctrough versus AUC was established. Majority of the predicted values for Cmax versus AUC were within 0.75- to 1.5-fold differences. However, the Ctrough versus AUC model showed larger variability with approximately one-third of the predictions within 0.75- to 1.5-fold differences. The r value and %RMSE for observed versus predicted AUC for Ctrough (r = 0.5925, n = 65, P < 0.001, and RMSE: 67%) were inferior to the Cmax (r = 0.8773, n = 86, P < 0.001, and RMSE: 46%). In conclusion, Cmax versus AUC and Ctrough versus AUC relationships were established for indinavir showing the utility of a single concentration time point for therapeutic drug monitoring purpose. The Cmax model for indinavir may be more relevant for AUC prediction as determined by the statistical criteria.


Subject(s)
Area Under Curve , Drug Monitoring/methods , HIV Protease Inhibitors/pharmacokinetics , Indinavir/pharmacokinetics , Retroviridae Infections/drug therapy , Feasibility Studies , HIV Protease Inhibitors/therapeutic use , Humans , Indinavir/therapeutic use , Linear Models
5.
Br J Clin Pharmacol ; 76(3): 475-83, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23305215

ABSTRACT

AIM: To describe the pharmacokinetics and safety of indinavir boosted with ritonavir (IDV/r) during the second and third trimesters of pregnancy and in the post-partum period. METHODS: IMPAACT P1026s is an on-going, prospective, non-blinded study of antiretroviral pharmacokinetics (PK) in HIV-infected pregnant women with a Thai cohort receiving IDV/r 400/100 mg twice daily during pregnancy through to 6-12 weeks post-partum as part of clinical care. Steady-state PK profiles were performed during the second (optional) and third trimesters and at 6-12 weeks post-partum. PK targets were the estimated 10(th) percentile IDV AUC (12.9 µg ml(-1)h) in non-pregnant historical Thai adults and a trough concentration of 0.1 µg ml(-1), the suggested minimum target. RESULTS: Twenty-six pregnant women were enrolled; thirteen entered during the second trimester. Median (range) age was 29.8 (18.9-40.8) years and weight 60.5 (50.0-85.0) kg at the third trimester PK visit. The 90% confidence limits for the geometric mean ratio of the indinavir AUC(0,12 h) and Cmax during the second trimester and post-partum (ante : post ratios) were 0.58 (0.49, 0.68) and 0.73 (0.59, 0.91), respectively; third trimester/post-partum AUC(0,12 h) and Cmax ratios were 0.60 (0.53, 0.68) and 0.63 (0.55, 0.72), respectively. IDV/r was well tolerated and 21/26 women had a HIV-1 viral load < 40 copies ml(-1) at delivery. All 26 infants were confirmed HIV negative. CONCLUSION: Indinavir exposure during the second and third trimesters was significantly reduced compared with post-partum and ∼30% of women failed to achieve a target trough concentration. Increasing the dose of IDV/r during pregnancy to 600/100 mg twice daily may be preferable to ensure adequate drug concentrations.


Subject(s)
HIV Infections/metabolism , HIV Protease Inhibitors/pharmacokinetics , Indinavir/pharmacokinetics , Pregnancy Complications, Infectious/metabolism , Ritonavir/pharmacokinetics , Adolescent , Adult , Antiretroviral Therapy, Highly Active , Dose-Response Relationship, Drug , Female , HIV Infections/drug therapy , HIV Infections/transmission , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/therapeutic use , Humans , Indinavir/administration & dosage , Indinavir/blood , Indinavir/therapeutic use , Infant , Infant, Newborn , Infectious Disease Transmission, Vertical/prevention & control , Postpartum Period , Pregnancy , Pregnancy Complications, Infectious/drug therapy , Pregnancy Complications, Infectious/virology , Pregnancy Trimester, Second , Pregnancy Trimester, Third , Prospective Studies , Ritonavir/administration & dosage , Ritonavir/blood , Ritonavir/therapeutic use , Young Adult
6.
Eur J Clin Pharmacol ; 68(5): 715-21, 2012 May.
Article in English | MEDLINE | ID: mdl-22173281

ABSTRACT

PURPOSE: To assess the effects of venlafaxine extended-release (XR) capsules and desvenlafaxine extended-release (XR) tablets upon indinavir pharmacokinetic properties when co-administrated to healthy volunteers. METHODS: This was an open-label, two-period, fixed-dose study conducted at the clinical research unit located on a university campus. Twenty-four healthy volunteers enrolled in the study (mean age 28.3 ± 8.0 years). Each subject received a single dose of indinavir 800 mg on day 1. Subsequently, subjects were then randomly assigned to either the venlafaxine XR group (N = 12) or the desvenlafaxine XR group (N = 12). Starting on day 2, venlafaxine XR was dosed at 37.5 mg/day for 4 days and increased to 75 mg/day for 6 days. Desvenlafaxine XR was dosed at 50 mg/day for 10 days. On day 12, indivanvir 800 mg was co-administered to both the venlafaxine XR and the desvenlafaxine XR groups. The pharmacokinetics of indinavir were determined both before and at the end of antidepressant dosing. Plasma indinavir, venlafaxine, and desvenlafaxine concentrations were assayed by high-performance liquid chromatography with ultra-violet (UV) detection. Indinavir pharmacokinetic parameters were calculated by noncompartmental analysis using validated computer software. RESULTS: Venlafaxine XR and desvenlafaxine XR did not produce any significant changes in indinavir disposition. Both antidepressants were well tolerated by the subjects with only minor adverse side effects. CONCLUSIONS: No pharmacokinetic drug-drug interaction was demonstrated between venlafaxine XR and indinavir or between desvenlafaxine XR and indinvair. The lack of interaction could be due to the venlafaxine and desvenlafaxine extended-release formulation.


Subject(s)
Antidepressive Agents/pharmacology , Cyclohexanols/pharmacology , HIV Protease Inhibitors/pharmacokinetics , Indinavir/pharmacokinetics , Adult , Antidepressive Agents/administration & dosage , Antidepressive Agents/blood , Antidepressive Agents/pharmacokinetics , Capsules , Cyclohexanols/administration & dosage , Cyclohexanols/blood , Cyclohexanols/pharmacokinetics , Delayed-Action Preparations , Desvenlafaxine Succinate , Dose-Response Relationship, Drug , Drug Interactions , Female , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/blood , Half-Life , Humans , Indinavir/administration & dosage , Indinavir/blood , Male , Metabolic Clearance Rate/drug effects , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/blood , Selective Serotonin Reuptake Inhibitors/pharmacokinetics , Tablets , Venlafaxine Hydrochloride , Young Adult
7.
Biostatistics ; 11(3): 537-45, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20203258

ABSTRACT

We introduce a dose-finding algorithm to be used to identify a level of dose that corresponds to some given targeted response. Our motivation arises from problems where the response is a continuously measured quantity, typically some pharmacokinetic parameter. We consider the case where an agreed level of response has been determined from earlier studies on some population and the purpose of the current trial is to obtain the same, or a comparable, level of response in a new population. This relates to bridging studies. The example driving our interest comes from studies on drugs for HIV that have already been evaluated in adults and where the new studies are to be carried out in children. These drugs have the ability to produce some given mean pharmacokinetic response in the adult population, and the goal is to calibrate the dose in order to obtain a comparable response in the childhood population. In practice, it may turn out that the dose producing some desired mean response is also associated with an unacceptable rate of toxicity. In this case, we may need to reevaluate the target response and this is readily achieved. In simulations, the algorithm can be seen to work very well. In the most challenging situations for the method, those where the targeted response corresponds to a region of the dose-response curve that is relatively flat, the algorithm can still perform satisfactorily.


Subject(s)
Algorithms , Clinical Trials, Phase I as Topic/methods , Pharmacokinetics , Adult , Child , Computer Simulation , HIV Infections/drug therapy , Humans , Indinavir/pharmacokinetics
8.
Ther Drug Monit ; 33(1): 25-31, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21233689

ABSTRACT

Indinavir boosted with ritonavir (IDV/r) dosing with 400/100 mg, twice daily, is preferred in Thai adults, but this dose can lead to concentrations close to the boundaries of its therapeutic window. The objectives of this analysis were to validate a population pharmacokinetic model to describe IDV/r concentrations in HIV-infected Thai patients and to investigate the impact of patient characteristics on achieving adequate IDV concentrations. IDV/r concentration data from 513 plasma samples were available. Population means and variances of pharmacokinetic parameters were estimated using a nonlinear mixed effects regression model (NONMEM Version VI). Monte Carlo simulations were performed to estimate the probability of achieving IDV concentrations within its therapeutic window. IDV/r pharmacokinetics were best described by a one-compartment model coupled with a single transit compartment absorption model. Body weight influenced indinavir apparent oral clearance and volume of distribution and allometric scaling significantly reduced the interindividual variability. Final population estimates (interindividual variability in percentage) of indinavir apparent oral clearance and volume of distribution were 21.3 L/h/70 kg (30%) and 90.7 L/70 kg (22%), respectively. Based on model simulations, the probability of achieving an IDV trough concentration greater than 0.1 mg/L was greater than 99% for 600/100 mg and greater than 98% for 400/100 mg, twice daily, in patients weighing 40 to 80 kg. However, the probability of achieving IDV concentrations associated with an increased risk of drug toxicity (greater than 10.0 mg/L) increased from 1% to 10% with 600/100 mg compared with less than 1% with 400/100 mg when body weight decreased from 80 to 40 kg. The validated model developed predicts that 400/100 mg of IDV/r, twice daily, provides indinavir concentrations within the recommended therapeutic window for the majority of patients. The risk of toxic drug concentrations increases rapidly with IDV/r dose of 600/100 mg for patients less than 50 kg and therapeutic drug monitoring of IDV concentrations would help to reduce the risk of IDV-induced nephrotoxicity.


Subject(s)
Body Weight , Drug Monitoring , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Indinavir/pharmacokinetics , Ritonavir/pharmacokinetics , Adult , Drug Therapy, Combination , Female , HIV Infections/blood , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/therapeutic use , Humans , Indinavir/adverse effects , Indinavir/blood , Indinavir/therapeutic use , Male , Middle Aged , Monte Carlo Method , Ritonavir/adverse effects , Ritonavir/blood , Ritonavir/therapeutic use , Thailand , Young Adult
9.
J Immunol ; 183(1): 661-9, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19535632

ABSTRACT

Antiretroviral therapy (ART) shows variable blood-brain barrier penetration. This may affect the development of neurological complications of HIV infection. In attempts to attenuate viral growth for the nervous system, cell-based nanoformulations were developed with the focus on improving drug pharmacokinetics. We reasoned that ART carriage could be facilitated within blood-borne macrophages traveling across the blood-brain barrier. To test this idea, an HIV-1 encephalitis (HIVE) rodent model was used where HIV-1-infected human monocyte-derived macrophages were stereotactically injected into the subcortex of severe combined immunodeficient mice. ART was prepared using indinavir (IDV) nanoparticles (NP, nanoART) loaded into murine bone marrow macrophages (BMM, IDV-NP-BMM) after ex vivo cultivation. IDV-NP-BMM was administered i.v. to mice resulting in continuous IDV release for 14 days. Rhodamine-labeled IDV-NP was readily observed in areas of HIVE and specifically in brain subregions with active astrogliosis, microgliosis, and neuronal loss. IDV-NP-BMM treatment led to robust IDV levels and reduced HIV-1 replication in HIVE brain regions. We conclude that nanoART targeting to diseased brain through macrophage carriage is possible and can be considered in developmental therapeutics for HIV-associated neurological disease.


Subject(s)
Brain/virology , Encephalitis, Viral/drug therapy , HIV-1/drug effects , Indinavir/administration & dosage , Macrophages/transplantation , Macrophages/virology , Nanocapsules/administration & dosage , Severe Combined Immunodeficiency/drug therapy , Animals , Biological Availability , Bone Marrow Cells/pathology , Bone Marrow Cells/virology , Brain/pathology , Cell Movement , Cells, Cultured , Disease Models, Animal , Drug Administration Schedule , Encephalitis, Viral/metabolism , HIV-1/growth & development , Humans , Indinavir/pharmacokinetics , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Severe Combined Immunodeficiency/virology , Virus Replication/drug effects
10.
Parasitology ; 138(11): 1384-91, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21819637

ABSTRACT

There is much evidence to indicate the ability of Indinavir (IND) to reduce Cryptosporidium parvum infection in both in vitro and in vivo models. However, there are limitations to the administration of IND as such, due to its renal toxicity and the high rate of metabolism and degradation. We aimed to encapsulate IND in biodegradable poly (D,L-lactide-co-glycolide) nanoparticles (Np) and to engineer their surface by conjugation with an anti-Cryptosporidium IgG polyclonal antibody (Ab). Tetramethylrhodamine-labelled Np were loaded with IND and modified by conjugation with an Ab. The IND-loaded modified Np (Ab-TMR-IND-Np) did not show any change, as demonstrated by chemical analysis studies. Simultaneous addition of 50µM Ab-TMR-IND-Np and excysted oocysts to the cell culture resulted in complete inhibition of the infection. In C. parvum-infected cells, the extent to which the infection decreased depended on the duration of treatment with the Ab-TMR-IND-Np. The antibody-engineered Np loaded with IND were able to target C. parvum in infected cells and therefore might represent a novel therapeutic strategy against Cryptosporidium sp. infection. Moreover, the use of Np as an IND delivery device, allows the development of a more appropriate dose formulation thereby reducing the IND side effects.


Subject(s)
Chemistry, Pharmaceutical/methods , Cryptosporidiosis/drug therapy , Cryptosporidium parvum/drug effects , Drug Carriers/chemistry , HIV Protease Inhibitors/pharmacokinetics , Immunoconjugates/pharmacokinetics , Indinavir/pharmacokinetics , Molecular Targeted Therapy , Nanoparticles/chemistry , Animals , Antibodies, Protozoan/chemistry , Antibodies, Protozoan/immunology , Biocompatible Materials/chemistry , Cattle , Cell Line, Tumor , Cryptosporidiosis/immunology , Cryptosporidiosis/parasitology , Drug Compounding , HIV Protease Inhibitors/therapeutic use , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Indinavir/therapeutic use , Lactic Acid/chemistry , Microscopy, Fluorescence , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rhodamines/analysis , Spectrum Analysis
11.
AAPS PharmSciTech ; 12(1): 422-30, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21360313

ABSTRACT

The aim of our present work was to develop indinavir O/W submicron lipid emulsions (SLEs) loaded with lipoamino acids for specific delivery to brain. Tetradecyl aspartic acid (A) and decyl glutamic acid (G) loaded stable SLEs of indinavir having a mean size range of 210-220 nm and average zeta potential of -23.54±1.2 mV were developed using homogenization and ultrasonication. The cumulative % drug release from different SLEs varied in between 26% and 85%. The formulations, SLE, SLE-A3, and SLE-G3 were stable to the centrifugal stress, dilution stress, and storage at RT. The total drug content and entrapment efficiency were determined by HPLC method. During pharmacokinetic studies in male Wistar rats there was no significant difference in the serum levels of indinavir for SLE, SLE-A3 and SLE-G3 formulations at all time points. In tissue distribution studies, the therapeutic availability (TA) of indinavir in brain and kidneys for SLE-A3 were 4.27- and 2.66-fold whereas for SLE-G3 were 2.94 and 2.12 times, respectively, higher than that of indinavir solution. But when compared with that of SLE, in brain tissue the levels of indinavir from SLE-G3 and SLE-A3 varied in between 2.5- and 3.38-fold. While in case of the kidney, it was between 1.23- and 1.54-fold only. However, the TA is not significantly different in tissues like the heart, liver, and spleen. Thus, brain-specific delivery of indinavir was improved by including tetradecyl aspartic acid and decyl glutamic acid in submicron lipid emulsions.


Subject(s)
Brain/drug effects , Drug Delivery Systems , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/pharmacokinetics , Indinavir/administration & dosage , Indinavir/pharmacokinetics , Amino Acids/chemistry , Animals , Drug Carriers , Drug Stability , Emulsions/chemistry , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/chemistry , Indinavir/blood , Indinavir/chemistry , Lipids/chemistry , Male , Rats , Rats, Wistar , Solubility , Tissue Distribution
12.
Therapie ; 66(3): 239-46, 2011.
Article in French | MEDLINE | ID: mdl-21819808

ABSTRACT

The HIV protease inhibitor indinavir presents a wide inter-individual variability related to an intense hepatic metabolism. Published studies were analyzed to establish whether there is evidence that therapeutic drug monitoring of indinavir could improve patient care. It was reported that indinavir virological efficacy in HIV-infected patients with wild-type virus was significantly associated with trough concentrations > 100-150 ng/mL. Concerning the exposure-toxicity relationship, the risk of occurrence of nephrotoxicity was more frequently associated with trough concentrations > 500-1 000 ng/mL. Studies with concentration-controlled indinavir therapy suggest that therapeutic drug monitoring allows to achieve safe and effective concentrations, therefore, the level of evidence of the interest of indinavir therapeutic drug monitoring is highly recommended when indinavir is not associated with ritonavir and recommended when ritonavir is combined with ritonavir.


Subject(s)
HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , Indinavir/therapeutic use , Dose-Response Relationship, Drug , Drug Interactions , Drug Monitoring , Evidence-Based Medicine , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/economics , HIV Protease Inhibitors/pharmacokinetics , Humans , Indinavir/adverse effects , Indinavir/economics , Indinavir/pharmacokinetics , Kidney Diseases/chemically induced
13.
Antimicrob Agents Chemother ; 54(11): 4903-6, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20805393

ABSTRACT

Patterns of HIV-1 protease inhibitor (PI) resistance-associated mutations (RAMs) and effects on PI susceptibility associated with the L76V mutation were studied in a large database. Of 20,501 sequences with ≥1 PI RAM, 3.2% contained L76V; L76V was alone in 0.04%. Common partner mutations included M46I, I54V, V82A, I84V, and L90M. L76V was associated with a 2- to 6-fold decrease in susceptibility to lopinavir, darunavir, amprenavir, and indinavir and a 7- to 8-fold increase in susceptibility to atazanavir and saquinavir.


Subject(s)
HIV Protease Inhibitors/pharmacology , HIV Protease/genetics , Antiviral Agents/pharmacology , Carbamates/pharmacology , Darunavir , Furans , HIV Protease Inhibitors/antagonists & inhibitors , Humans , Indinavir/pharmacokinetics , Lopinavir , Mutation , Pyrimidinones/pharmacology , Sulfonamides/pharmacology
14.
Drug Metab Dispos ; 38(5): 841-50, 2010 May.
Article in English | MEDLINE | ID: mdl-20100816

ABSTRACT

The 2-methyl substituted indole, 2MI [2-(4-(4-(2,4-dichlorophenylsulfonamido)-2-methyl-1H-indol-5-yloxy)-3-methoxyphenyl)acetic acid] is a potent dual inhibitor of 1) chemoattractant receptor-homologous molecule expressed on T-helper type-2 cells and 2) d-prostanoid receptor. During evaluation as a potential treatment for asthma and allergic rhinitis, 2MI was identified as a mechanism-based inactivator of CYP3A4 in vitro. The inactivation was shown to be irreversible by dialysis and accompanied by an NADPH-dependent increase in 2MI covalent binding to a 55- to 60-kDa microsomal protein, consistent with irreversible binding to CYP3A4. Two glutathione (GSH) adducts, G1 and G2, were identified in vitro, and the more abundant adduct (G1) was unambiguously determined via NMR to be GSH adducted to the 3-position of the 2-methylindole moiety. The potential for a clinical drug-drug interaction arising from mechanism-based inactivation of CYP3A4 by 2MI was predicted using a steady-state model, and a 4.3- to 7.5-fold increase in the exposure of midazolam was predicted at anticipated therapeutic concentrations. To better assess the potential for in vivo drug-drug interactions, the Sprague-Dawley rat was used as an in vivo model. An excellent in vitro-in vivo correlation was observed for the reduction in enzyme steady-state concentration (E'(ss/Ess)) as well as the change in the exposure of a prototypical CYP3A substrate, indinavir (area under the curve (AUC) for indinavir/AUC). In summary, 2MI was identified as a potent mechanism-based inactivator of CYP3A and was predicted to elicit a clinically relevant drug-drug interaction in humans at an anticipated therapeutic concentration.


Subject(s)
Cytochrome P-450 CYP3A/drug effects , Cytochrome P-450 CYP3A/metabolism , Glutathione/metabolism , Indoles/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Animals , Area Under Curve , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/drug effects , Aryl Hydrocarbon Hydroxylases/metabolism , Biocatalysis/drug effects , Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Horseradish Peroxidase/metabolism , Humans , Hydrogen Peroxide/metabolism , Indinavir/metabolism , Indinavir/pharmacokinetics , Indoles/pharmacology , Kinetics , Magnetic Resonance Spectroscopy , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Midazolam/metabolism , Midazolam/pharmacokinetics , Models, Biological , Molecular Structure , NADP/metabolism , Pharmacokinetics , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
15.
Intervirology ; 53(3): 176-82, 2010.
Article in English | MEDLINE | ID: mdl-20197684

ABSTRACT

OBJECTIVE: To investigate the potential for a pharmacokinetic interaction between darunavir (DRV, TMC114, Prezista), indinavir (IDV, Crixivan) and low-dose ritonavir (RTV, Norvir). METHODS: In three 7-day sessions, 17 HIV-negative healthy volunteers received treatment A (DRV/r 400/100 mg b.i.d.), treatment B (IDV/r 800/100 mg b.i.d.) and treatment C (DRV/r 400/100 mg b.i.d. + IDV 800 mg b.i.d.). On day 7, full pharmacokinetic profiles of DRV, IDV and RTV were determined. Safety and tolerability were also assessed. RESULTS: Based on the least-squares means ratios, the steady-state exposure (area under the curve, AUC(12h)) and plasma concentrations (C(min) and C(max)) of IDV were increased by 23, 125 and 8%, respectively, when DRV was co-administered. The co-administration of IDV with DRV/r resulted in increases of 24, 44 and 11% for, respectively, DRV AUC(12h), C(min) and C(max), compared with administration of DRV/r alone. Eight volunteers discontinued due to an adverse event. Overall, adverse events and laboratory abnormalities were more commonly reported during treatments including IDV. CONCLUSIONS: When used in combination with DRV/r, dose adjustment of IDV from 800 mg b.i.d. to 600 mg b.i.d. may be warranted in cases of intolerance.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Indinavir/pharmacokinetics , Ritonavir/pharmacokinetics , Sulfonamides/pharmacokinetics , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Darunavir , Drug Interactions , Female , Healthy Volunteers , Humans , Indinavir/administration & dosage , Indinavir/adverse effects , Male , Plasma/chemistry , Ritonavir/administration & dosage , Ritonavir/adverse effects , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Young Adult
16.
Biopharm Drug Dispos ; 31(2-3): 178-88, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20238377

ABSTRACT

Hepatotoxicity has been reported as a side-effect in some patients on HIV protease inhibitors (PI). Since transporter interaction has been implicated as a mechanism underlying drug-mediated hepatotoxicity and drug-drug interactions, the interaction of PI with the hepatic canalicular efflux transporter ABCC2 (MRP2; multidrug resistance associated protein-2) was studied. Interaction with ABCC2/Abcc2 was evaluated in human and rat sandwich-cultured hepatocytes using 5(6)-carboxy-2',7'-dichlorofluorescein (CDF) as substrate. In rat hepatocytes, interaction with estradiol-17-beta-D-glucuronide (E17G) efflux was also studied. In human hepatocytes, saquinavir, ritonavir and atazanavir were the most efficient inhibitors of ABCC2-mediated biliary excretion of CDF, whereas in rat hepatocytes indinavir, lopinavir and nelfinavir were the most efficient. No species-similarity was found for ABCC2/Abcc2 inhibition. In rat hepatocytes, the effects on Abcc2 were substrate-dependent as inhibition of biliary excretion of E17G was most pronounced for saquinavir (completely blocked), amprenavir (82% inhibition) and indinavir (68% inhibition). In conclusion, several HIV PI showed substantial ABCC2 inhibition, which, combined with the effects of PI on other hepatobiliary disposition mechanisms, will determine the clinical relevance of these in vitro interaction data.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV Protease Inhibitors/pharmacokinetics , Hepatocytes/metabolism , Liver/pathology , Multidrug Resistance-Associated Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Biological Transport , Carbamates/pharmacokinetics , Cells, Cultured , Fluoresceins/pharmacokinetics , Furans , Humans , Indinavir/pharmacokinetics , Multidrug Resistance-Associated Protein 2 , Nelfinavir/pharmacokinetics , Rats , Rats, Wistar , Ritonavir/pharmacokinetics , Saquinavir/pharmacokinetics , Sulfonamides/pharmacokinetics
18.
Drug Dev Ind Pharm ; 36(3): 355-61, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19722914

ABSTRACT

BACKGROUND: Compartmental as well as noncompartmental analyses are used routinely in pharmacokinetic analysis. MATERIALS AND METHODS: Pharmacokinetic parameters of the anti-HIV agent, indinavir, have been determined in six male rats applying both the compartmental and the noncompartmental analysis. RESULTS AND DISCUSSION: A very slow declining phase was found in the indinavir plasma concentration profile using an extended sampling time period and applying a sensitive high-performance liquid chromatography assay method. This apparent terminal elimination phase can cause some significant errors when applying noncompartmental kinetic analysis to the data, with mean residence time (MRT) (544.2 +/- 123.2 minutes), total systemic clearance (12.0 +/- 2.1 mL/min/kg), and steady-state volume of distribution (V(d) (ss)) (6.4 +/- 1.0 L/kg) being highly different from the results of compartmental kinetic analysis (MRT, Cl(total), and V(d) (ss) values of 23.7 +/- 5.9 minutes, 35.18 +/- 5.1 mL/min/kg, and 0.84 +/- 0.28 L/kg, respectively). The parameters estimated by our noncompartmental approach were also significantly different from the results of the same type of data analysis reported in the literature. CONCLUSION: The differences in parameter estimations, while being a result of the extended plasma sampling period, which is recommended in noncompartmental analysis, support the priority of applying the compartmental analysis approach in the similar cases with some pre-assumptions, mainly ignoring the final apparent terminal elimination phase(s), very deep tissue, which involves very low drug concentrations.


Subject(s)
Body Fluid Compartments/physiology , HIV Protease Inhibitors/pharmacokinetics , Indinavir/pharmacokinetics , Models, Biological , Animals , HIV Protease Inhibitors/blood , Half-Life , Indinavir/blood , Male , Metabolic Clearance Rate , Rats , Rats, Sprague-Dawley
19.
Eur J Clin Pharmacol ; 65(7): 667-78, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19440701

ABSTRACT

AIMS: To assess the relationship between genetic polymorphisms and indinavir pharmacokinetic variability and to study the link between concentrations and short-term response or metabolic safety. METHODS: Forty protease inhibitor-naive patients initiating highly active antiretroviral therapy (HAART) including indinavir/ritonavir and enrolled in the COPHAR 2-ANRS 111 trial were studied. At week 2, four blood samples were taken before and up to 6 h following drug intake. A population pharmacokinetic analysis was performed using the stochastic approximation expectation maximization (SAEM) algorithm implemented in MONOLIX software. The area under the concentration-time curve (AUC) and maximum (C(max)) and trough concentrations (C(trough)) of indinavir were derived from the population model and tested for their correlation with short-term viral response and safety measurements, while for ritonavir, these same three parameters were tested for their correlation with short-term biochemical safety RESULTS: A one-compartment model with first-order absorption and elimination best described both indinavir and ritonavir concentrations. For indinavir, the estimated clearance and volume of distribution were 22.2 L/h and 97.3 L, respectively. The eight patients with the *1B/*1B genotype for the CYP3A4 gene showed a 70% decrease in absorption compared to those with the *1A/*1B or *1A/*1A genotypes (0.5 vs. 2.1, P = 0.04, likelihood ratio test by permutation). The indinavir AUC and C(trough) were positively correlated with the decrease in human immunodeficiency virus RNA between week 0 and week 2 (r = 0.4, P = 0.03 and r = -0.4, P = 0.03, respectively). Patients with the *1B/*1B genotype also had a significantly lower indinavir C(max) (median 3.6, range 2.1-5.2 ng/mL) than those with the *1A/*1B or *1A/*1A genotypes (median 4.4, range 2.2-8.3 ng/mL) (P = 0.04) and a lower increase in triglycerides during the first 4 weeks of treatment (median 0.1, range -0.7 to 1.4 vs. median 0.6, range -0.5 to 1.7 mmol/L, respectively; P = 0.02). For ritonavir, the estimated clearance and volume of distribution were 8.3 L/h and 60.7 L, respectively, and concentrations were not found to be correlated to biochemical safety. Indinavir and ritonavir absorption rate constants were found to be correlated, as well as their apparent volumes of distribution and clearances, indicating correlated bioavailability of the two drugs. CONCLUSION: The CYP3A4*1B polymorphism was found to influence the pharmacokinetics of indinavir and, to some extent, the biochemical safety of indinavir.


Subject(s)
Antiretroviral Therapy, Highly Active , HIV Protease Inhibitors/pharmacokinetics , HIV-1 , Indinavir/pharmacokinetics , Pharmacogenetics , Adult , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , Area Under Curve , Clinical Trials as Topic , Cohort Studies , Cytochrome P-450 CYP3A/drug effects , Female , Genotype , HIV Infections/drug therapy , HIV Protease Inhibitors/adverse effects , Humans , Indinavir/adverse effects , Male , Metabolic Clearance Rate , Middle Aged , Models, Statistical , Multicenter Studies as Topic , Pilot Projects , Polymorphism, Genetic , Prospective Studies , Treatment Outcome
20.
Pharmacology ; 83(1): 59-66, 2009.
Article in English | MEDLINE | ID: mdl-19052483

ABSTRACT

The aim of this work is to: (1) assess therapeutic drug monitoring of indinavir (IDV) during clinical routine practice in HIV-infected children, whose antiretroviral treatment includes IDV boosted with ritonavir (RTV), and (2) describe a possible relationship between IDV pharmacokinetics and MDR1 genotypes. In 21 ambulatory pediatric patients receiving IDV plus RTV, IDV plasma levels and MDR1 genotypes on exon 26 (C3435T) were determined. Nine of the 21 patients initially receiving 250 mg/m(2) IDV yielded trough levels below 0.10 microg/ml (median: 0.21, range: 0.04-1.31 microg/ml). When the dosage was increased to 400 mg/m(2) IDV plus 100 mg/m(2) RTV b.i.d., all, except 1 patient, achieved levels above 0.10 microg/ml. Pharmacokinetic analysis showed higher volume of distribution median values related to the C/C genotype in comparison with C/T or T/T genotypes for exon 26 (4.57 vs. 1.20 and 1.50 l/kg, respectively; p = 0.002). Although a higher median value of clearance was observed with the C/C genotype, the difference was not statistically significant (1.43 vs. 0.27 and 0.42 l/h, respectively; p = 0.052). These results may be explained by a reduced absorption of the drug, related with lower plasma IDV levels in patients carrying the C/C genotype in exon 26.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Indinavir/pharmacokinetics , Adolescent , Child , Dose-Response Relationship, Drug , Drug Monitoring/methods , Drug Therapy, Combination , Exons , Female , Genotype , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/therapeutic use , Humans , Indinavir/administration & dosage , Indinavir/therapeutic use , Male , Polymorphism, Genetic , Ritonavir/therapeutic use , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL