Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Antimicrob Chemother ; 74(9): 2690-2697, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31119275

ABSTRACT

OBJECTIVES: Dolutegravir is widely prescribed owing to its potent antiviral activity, high genetic barrier and good tolerability. The aim of this study was to characterize dolutegravir's pharmacokinetic profile and variability in a real-life setting and to identify individual factors and co-medications affecting dolutegravir disposition. METHODS: A population pharmacokinetic model was developed using NONMEM®. Relevant demographic factors, clinical factors and co-medications were tested as potential covariates. Simulations based on the final model served to compare expected dolutegravir concentrations under standard and alternative dosage regimens in the case of drug-drug interactions. RESULTS: A total of 620 dolutegravir plasma concentrations were collected from 521 HIV-infected individuals under steady-state conditions. A one-compartment model with first-order absorption and elimination best characterized dolutegravir pharmacokinetics. Typical dolutegravir apparent clearance (CL/F) was 0.93 L/h with 32% between-subject variability, the apparent volume of distribution was 20.2 L and the absorption rate constant was fixed to 2.24 h-1. Older age, higher body weight and current smoking were associated with higher CL/F. Atazanavir co-administration decreased dolutegravir CL/F by 38%, while darunavir modestly increased CL/F by 14%. Rifampicin co-administration showed the largest impact on CL/F. Simulations suggest that average dolutegravir trough concentrations are 63% lower after 50 mg/12h with rifampicin compared with a standard dosage of 50 mg/24h without rifampicin. Average trough concentrations after 100 mg/24h and 100 mg/12h with rifampicin are 92% and 25% lower than the standard dosage without rifampicin, respectively. CONCLUSIONS: Patients co-treated with dolutegravir and rifampicin might benefit from therapeutic drug monitoring and individualized dosage increase, up to 100 mg/12 h in some cases.


Subject(s)
Drug Interactions , HIV Integrase Inhibitors/pharmacokinetics , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Models, Theoretical , Adolescent , Adult , Aged , Antibiotics, Antitubercular/pharmacology , Antiretroviral Therapy, Highly Active/adverse effects , Antiretroviral Therapy, Highly Active/methods , Cohort Studies , Drug Monitoring , Female , HIV Infections/drug therapy , Humans , Male , Middle Aged , Oxazines , Piperazines , Pyridones , Young Adult
2.
Malar J ; 18(1): 139, 2019 Apr 18.
Article in English | MEDLINE | ID: mdl-30999915

ABSTRACT

BACKGROUND: The World Health Organization (WHO) recommends combinations of an artemisinin derivative plus an anti-malarial drug of longer half-life as treatment options for uncomplicated Plasmodium falciparum infections. In Africa, artesunate-mefloquine (ASMQ) is an infrequently used artemisinin-based combination therapy (ACT) because of perceived poor tolerance to mefloquine. However, the WHO has recommended reconsideration of the use of ASMQ in Africa. In this large clinical study, the pharmacokinetics (PK) of a fixed dose combination of ASMQ was investigated in an African paediatric population to support dosing recommendations used in Southeast Asia and South America. METHODS: Among the 472 paediatric patients aged 6-59 months from six African centres included in the large clinical trial, a subset of 50 Kenyan children underwent intensive sampling to develop AS, its metabolite dihydroartemisinin (DHA) and MQ PK models. The final MQ PK model was validated using sparse data collected in the remaining participants (NONMEM®). The doses were one or two tablets containing 25/55 mg AS/MQ administered once a day for 3 days according to patients' age. A sensitive LC-MS/MS method was used to quantify AS, DHA and MQ concentrations in plasma. An attempt was made to investigate the relationship between the absence/presence of malaria recrudescence and MQ area under the curve (AUC) using logistic regression. RESULTS: AS/DHA concentration-time profiles were best described using a one-compartment model for both compounds with irreversible AS conversion into DHA. AS/DHA PK were characterized by a significant degree of variability. Body weight affected DHA PK parameters. MQ PK was characterized by a two-compartment model and a large degree of variability. Allometric scaling of MQ clearances and volumes of distribution was used to depict the relationship between MQ PK and body weight. No association was found between the model predicted AUC and appearance of recrudescence. CONCLUSIONS: The population pharmacokinetic models developed for both AS/DHA and MQ showed a large variability in drug exposure in the investigated African paediatric population. The largest contributor to this variability was body weight, which is accommodated for by the ASMQ fixed dose combination (FDC) dosing recommendation. Besides body weight considerations, there is no indication that the dosage should be modified in children with malaria compared to adults. Trial registration Pan African Clinical Trials Registry PACTR201202000278282 registration date 2011/02/16.


Subject(s)
Antimalarials/pharmacology , Artesunate/pharmacology , Malaria, Falciparum/drug therapy , Mefloquine/pharmacology , Antimalarials/pharmacokinetics , Artesunate/pharmacokinetics , Child, Preschool , Dose-Response Relationship, Drug , Drug Combinations , Female , Humans , Infant , Kenya , Male , Mefloquine/pharmacokinetics , Prospective Studies , Recurrence
3.
Article in English | MEDLINE | ID: mdl-27799217

ABSTRACT

Rilpivirine (RPV), the latest nonnucleoside reverse transcriptase inhibitor active against HIV-1, is prescribed in a standard dosage of 25 mg once a day in combination with emtricitabine (FTC) and tenofovir disoproxil fumarate (TDF). The aim of this observational study was to characterize the RPV pharmacokinetic profile, to quantify interpatient variability, and to identify potential factors that could influence drug exposure. RPV concentration data were collected from HIV-infected patients as part of routine therapeutic drug monitoring performed in our center (Laboratory of Clinical Pharmacology). A population pharmacokinetic analysis was performed with NONMEM by comparing various structural models. The influence of demographic and clinical covariates, as well as frequent genetic polymorphisms in 5 genes (CYP3A4*22, CYP3A5*3, CYP2C19*2, CYP2C19*17, UGT1A1*28, and UGT1A4*2), on RPV elimination was explored. A total of 325 plasma concentration measurements were obtained from 249 HIV-positive patients. Plasma concentrations ranged from 12 to 255 ng/ml. A one-compartment model with zero-order absorption best characterized RPV pharmacokinetics. The average RPV clearance (CL) was 11.7 liters/h, the average volume of distribution was 401 liters, and the mean absorption time was 4 h. The interinterindividual variability (IIV) for CL was estimated to be 33%. None of the available demographic or genetic covariates showed any influence on RPV pharmacokinetics, but 29% of the patients were predicted to present minimal concentrations below the recently identified target cutoff value of 50 ng/ml. The variability in RPV pharmacokinetics appears to be lower than that for most other antiretroviral drugs. However, under the standard regimen of 25 mg daily, a significant number of patients might be underdosed. It remains to be investigated whether the underexposure has an impact on the development of resistance while patients are on maintenance therapy.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV-1/pathogenicity , Rilpivirine/pharmacokinetics , Adult , Aged , Aged, 80 and over , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP3A/genetics , Female , Genotype , Glucuronosyltransferase/genetics , HIV Infections/genetics , HIV Infections/metabolism , HIV-1/drug effects , Humans , Male , Middle Aged , Polymorphism, Genetic/genetics , Rilpivirine/therapeutic use , Young Adult
4.
J Antimicrob Chemother ; 71(7): 1933-42, 2016 07.
Article in English | MEDLINE | ID: mdl-27029846

ABSTRACT

OBJECTIVES: Co-formulated elvitegravir, cobicistat, tenofovir disoproxil fumarate and emtricitabine is among the preferred regimens for first-line ART. A population approach was used to characterize the pharmacokinetics of elvitegravir and cobicistat and identify individual factors and co-medications influencing their disposition, taking into consideration the interaction between the two compounds. METHODS: The study population included 144 HIV-infected individuals who provided 186 and 167 elvitegravir and cobicistat plasma concentrations, respectively. First, distinct NONMEM(®) analyses were conducted for elvitegravir and cobicistat, including individual demographic, clinical and genetic factors as potential covariates. Elvitegravir and cobicistat interaction was then assessed through different inhibitory models. Simulations based on the final model served to compare expected drug concentrations under standard and alternative dosage regimens. RESULTS: Clearance with between-subject variability was 7.6 L/h [coefficient of variation (CV) 16.6%] and volume of distribution 61 L for elvitegravir and 16.0 L/h (CV 41.9%) and 88.3 L, respectively, for cobicistat. Concomitant administration of non-ritonavir-boosted atazanavir decreased elvitegravir clearance by 35%, likely due to UDP-glucuronosyl transferase (UGT) 1A1 inhibition. Concomitant administration of non-ritonavir-boosted atazanavir and ritonavir-boosted darunavir decreased cobicistat clearance by 47% and 27%, respectively. The final interaction model included cobicistat exposure (AUC0-24) on elvitegravir clearance. Simulations confirmed that a reduced elvitegravir dose of 85 mg co-administered with cobicistat and atazanavir produces a concentration-time course comparable to the standard regimen without atazanavir. CONCLUSIONS: Elvitegravir and cobicistat pharmacokinetic variability appears to be mainly explained by drug-drug interactions that may be encountered in routine clinical practice. In these cases, therapeutic drug monitoring and surveillance for potential toxicities would be justified.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Cobicistat/pharmacokinetics , HIV Infections/drug therapy , HIV-1/isolation & purification , Quinolones/pharmacokinetics , Adolescent , Adult , Aged , Anti-HIV Agents/administration & dosage , Area Under Curve , Cobicistat/administration & dosage , Cohort Studies , Computer Simulation , Drug Interactions , Female , HIV Infections/virology , Humans , Male , Middle Aged , Models, Statistical , Quinolones/administration & dosage , Young Adult
5.
Genet Med ; 18(8): 814-22, 2016 08.
Article in English | MEDLINE | ID: mdl-26765343

ABSTRACT

PURPOSE: The implementation of genomic-based medicine is hindered by unresolved questions regarding data privacy and delivery of interpreted results to health-care practitioners. We used DNA-based prediction of HIV-related outcomes as a model to explore critical issues in clinical genomics. METHODS: We genotyped 4,149 markers in HIV-positive individuals. Variants allowed for prediction of 17 traits relevant to HIV medical care, inference of patient ancestry, and imputation of human leukocyte antigen (HLA) types. Genetic data were processed under a privacy-preserving framework using homomorphic encryption, and clinical reports describing potentially actionable results were delivered to health-care providers. RESULTS: A total of 230 patients were included in the study. We demonstrated the feasibility of encrypting a large number of genetic markers, inferring patient ancestry, computing monogenic and polygenic trait risks, and reporting results under privacy-preserving conditions. The average execution time of a multimarker test on encrypted data was 865 ms on a standard computer. The proportion of tests returning potentially actionable genetic results ranged from 0 to 54%. CONCLUSIONS: The model of implementation presented herein informs on strategies to deliver genomic test results for clinical care. Data encryption to ensure privacy helps to build patient trust, a key requirement on the road to genomic-based medicine.Genet Med 18 8, 814-822.


Subject(s)
Computer Security , Genetic Privacy , HIV Infections/genetics , Genetic Variation , Genomics/ethics , Humans , Models, Theoretical
6.
Drug Metab Dispos ; 44(1): 151-61, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26553012

ABSTRACT

Efavirenz (EFV) is principally metabolized by CYP2B6 to 8-hydroxy-efavirenz (8OH-EFV) and to a lesser extent by CYP2A6 to 7-hydroxy-efavirenz (7OH-EFV). So far, most metabolite profile analyses have been restricted to 8OH-EFV, 7OH-EFV, and EFV-N-glucuronide, even though these metabolites represent a minor percentage of EFV metabolites present in vivo. We have performed a quantitative phase I and II metabolite profile analysis by tandem mass spectrometry of plasma, cerebrospinal fluid (CSF), and urine samples in 71 human immunodeficiency virus patients taking efavirenz, prior to and after enzymatic (glucuronidase and sulfatase) hydrolysis. We have shown that phase II metabolites constitute the major part of the known circulating efavirenz species in humans. The 8OH-EFV-glucuronide (gln) and 8OH-EFV-sulfate (identified for the first time) in humans were found to be 64- and 7-fold higher than the parent 8OH-EFV, respectively. In individuals (n = 67) genotyped for CYP2B6, 2A6, and CYP3A metabolic pathways, 8OH-EFV/EFV ratios in plasma were an index of CYP2B6 phenotypic activity (P < 0.0001), which was also reflected by phase II metabolites 8OH-EFV-glucuronide/EFV and 8OH-EFV-sulfate/EFV ratios. Neither EFV nor 8OH-EFV, nor any other considered metabolites in plasma were associated with an increased risk of central nervous system (CNS) toxicity. In CSF, 8OH-EFV levels were not influenced by CYP2B6 genotypes and did not predict CNS toxicity. The phase II metabolites 8OH-EFV-gln, 8OH-EFV-sulfate, and 7OH-EFV-gln were present in CSF at 2- to 9-fold higher concentrations than 8OH-EFV. The potential contribution of known and previously unreported EFV metabolites in CSF to the neuropsychological effects of efavirenz needs to be further examined in larger cohort studies.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Benzoxazines/adverse effects , Benzoxazines/pharmacokinetics , HIV Infections/drug therapy , Metabolomics/methods , Reverse Transcriptase Inhibitors/pharmacokinetics , Tandem Mass Spectrometry , Alkynes , Anti-HIV Agents/adverse effects , Anti-HIV Agents/blood , Anti-HIV Agents/cerebrospinal fluid , Anti-HIV Agents/urine , Benzoxazines/blood , Benzoxazines/cerebrospinal fluid , Benzoxazines/urine , Cyclopropanes , Cytochrome P-450 CYP2A6/genetics , Cytochrome P-450 CYP2A6/metabolism , Cytochrome P-450 CYP2B6/genetics , Cytochrome P-450 CYP2B6/metabolism , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Genotype , Glucuronides/blood , Glucuronides/cerebrospinal fluid , Glucuronides/urine , HIV Infections/diagnosis , HIV Infections/metabolism , Humans , Hydroxylation , Metabolic Detoxication, Phase I , Metabolic Detoxication, Phase II , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/metabolism , Phenotype , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/blood , Reverse Transcriptase Inhibitors/cerebrospinal fluid , Reverse Transcriptase Inhibitors/urine , Risk Assessment , Sulfates/blood , Sulfates/cerebrospinal fluid , Sulfates/urine
7.
PLoS One ; 10(7): e0133028, 2015.
Article in English | MEDLINE | ID: mdl-26176696

ABSTRACT

BACKGROUND: The efficacy of first-generation protease inhibitor based triple-therapy against hepatitis C virus (HCV) infection is limited in HIV/HCV-coinfected patients with advanced liver fibrosis and non-response to previous peginterferon-ribavirin. These patients have a low chance of achieving a sustained virologic response (SVR) using first generation triple-therapy, with a success rate of only 20%. We investigated the efficacy and safety of lead-in therapy with intravenous silibinin followed by triple-therapy in this difficult-to-treat patient group. METHODOLOGY: Inclusion criteria were HIV/HCV coinfection with advanced liver fibrosis and documented previous treatment failure on peginterferon-ribavirin. The intervention was a lead-in therapy with intravenous silibinin 20 mg/kg/day for 14 days, followed by triple-therapy (peginterferon-ribavirin and telaprevir) for 12 weeks, and peginterferon-ribavirin alone for 36 weeks. Outcome measurements were HCV-RNA after silibinin lead-in and during triple-therapy, SVR data at week 12, and safety and tolerability of silibinin. RESULTS: We examined sixteen HIV/HCV-coinfected patients with previous peginterferon-ribavirin failure, of whom 14 had a fibrosis grade METAVIR ≥F3. All were on successful antiretroviral therapy. Median (IQR) HCV-RNA decline after silibinin therapy was 2.65 (2.1-2.8) log10 copies/mL. Fifteen of sixteen patients (94%) had undetectable HCV RNA at weeks 4 and 12, eleven patients (69%) showed end-of-treatment response (i.e., undetectable HCV-RNA at week 48), and ten patients (63%) reached SVR at week 12 (SVR 12). Six of the sixteen patients (37%) did not reach SVR 12: One patient had rapid virologic response (RVR) (i.e., undetectable HCV-RNA at week 4) but stopped treatment at week 8 due to major depression. Five patients had RVR, but experienced viral breakthroughs at week 21, 22, 25, or 32, or a relapse at week 52. The HIV RNA remained below the limit of detection in all patients during the complete treatment period. No serious adverse events and no significant drug-drug interactions were associated with silibinin. CONCLUSION: A lead-in with silibinin before triple-therapy was safe and highly effective in difficult-to-treat HIV/HCV coinfected patients, with a pronounced HCV-RNA decline during the lead-in phase, which translates into 63% SVR. An add-on of intravenous silibinin to standard of care HCV treatment is worth further exploration in selected difficult-to-treat patients. TRIAL REGISTRATION: ClinicalTrials.gov NCT01816490.


Subject(s)
Antiretroviral Therapy, Highly Active , Antiviral Agents/therapeutic use , HIV Infections/drug therapy , Hepatitis C, Chronic/drug therapy , Liver Cirrhosis/drug therapy , RNA, Viral/antagonists & inhibitors , Silymarin/therapeutic use , Adult , Coinfection , Drug Administration Schedule , Female , HIV/drug effects , HIV/growth & development , HIV Infections/blood , HIV Infections/pathology , HIV Infections/virology , Hepacivirus/drug effects , Hepacivirus/growth & development , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/pathology , Hepatitis C, Chronic/virology , Humans , Injections, Intravenous , Interferon-alpha/therapeutic use , Liver Cirrhosis/blood , Liver Cirrhosis/pathology , Liver Cirrhosis/virology , Male , Middle Aged , Oligopeptides/therapeutic use , Patient Safety , Polyethylene Glycols/therapeutic use , Prospective Studies , Protease Inhibitors/therapeutic use , RNA, Viral/blood , Recombinant Proteins/therapeutic use , Ribavirin/therapeutic use , Silybin , Treatment Outcome , Viral Load/drug effects
8.
Antivir Ther ; 20(2): 165-75, 2015.
Article in English | MEDLINE | ID: mdl-24964403

ABSTRACT

BACKGROUND: Management of persistent low-level viraemia (pLLV) in patients on combined antiretroviral therapy (cART) with previously undetectable HIV viral loads (VLs) is challenging. We examined virological outcome and management among patients enrolled in the Swiss HIV Cohort Study (SHCS). METHODS: In this retrospective study (2000-2011), pLLV was defined as a VL of 21-400 copies/ml on ≥ three consecutive plasma samples with ≥8 weeks between first and last analyses, in patients undetectable for ≥24 weeks on cART. Control patients had ≥ three consecutive undetectable VLs over ≥32 weeks. Virological failure (VF), analysed in the pLLV patient group, was defined as a VL>400 copies/ml. RESULTS: Among 9,972 patients, 179 had pLLV and 5,389 were controls. Compared to controls, pLLV patients were more often on unboosted protease inhibitor (PI)-based (adjusted odds ratio [aOR; 95% CI] 3.2 [1.8, 5.9]) and nucleoside/nucleotide reverse transcriptase inhibitor (NRTI)-only combinations (aOR 2.1 [1.1, 4.2]) than on non-nucleoside reverse transcriptase inhibitor and boosted PI-based regimens. At 48 weeks, 102/155 pLLV patients (66%) still had pLLV, 19/155 (12%) developed VF and 34/155 (22%) had undetectable VLs. Predictors of VF were previous VF (aOR 35 [3.8, 315]), unboosted PI-based (aOR 12.8 [1.7, 96]) or NRTI-only combinations (aOR 115 [6.8, 1,952]), and VLs>200 during pLLV (aOR 3.7 [1.1, 12]). No VF occurred in patients with persistent very LLV (21-49 copies/ml; n=26). At 48 weeks, 29/39 patients (74%) who changed cART had undetectable VLs, compared with 19/74 (26%) without change (P<0.001). CONCLUSIONS: Among patients with pLLV, VF was predicted by previous VF, cART regimen and VL≥200. Most patients who changed cART had undetectable VLs 48 weeks later. These findings support cART modification for pLLV>200 copies/ml.


Subject(s)
Antiretroviral Therapy, Highly Active , HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Viremia/drug therapy , Adult , CD4 Lymphocyte Count , Disease Management , Female , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , HIV-1/drug effects , HIV-1/physiology , Humans , Male , Middle Aged , Retrospective Studies , Switzerland , Treatment Outcome , Viral Load/drug effects , Viremia/immunology , Viremia/virology , Virus Replication/drug effects
9.
J Mass Spectrom ; 48(5): 616-25, 2013 May.
Article in English | MEDLINE | ID: mdl-23674286

ABSTRACT

Because of the large variability in the pharmacokinetics of anti-HIV drugs, therapeutic drug monitoring in patients may contribute to optimize the overall efficacy and safety of antiretroviral therapy. An LC-MS/MS method for the simultaneous assay in plasma of the novel antiretroviral agents rilpivirine (RPV) and elvitegravir (EVG) has been developed to that endeavor. Plasma samples (100 µL) extraction is performed by protein precipitation with acetonitrile, and the supernatant is subsequently diluted 1:1 with 20-mM ammonium acetate/MeOH 50:50. After reverse-phase chromatography, quantification of RPV and EVG, using matrix-matched calibration samples, is performed by electrospray ionization-triple quadrupole mass spectrometry by selected reaction monitoring detection using the positive mode. The stable isotopic-labeled compounds RPV-(13) C6 and EVG-D6 were used as internal standards. The method was validated according to FDA recommendations, including assessment of extraction yield, matrix effects variability (<6.4%), as well as EVG and RPV short and long-term stability in plasma. Calibration curves were validated over the clinically relevant concentrations ranging from 5 to 2500 ng/ml for RPV and from 50 to 5000 ng/ml for EVG. The method is precise (inter-day CV%: 3-6.3%) and accurate (3.8-7.2%). Plasma samples were found to be stable (<15%) in all considered conditions (RT/48 h, +4°C/48 h, -20°C/3 months and 60°C/1 h). Selected metabolite profiles analysis in patients' samples revealed the presence of EVG glucuronide, that was well separated from parent EVG, allowing to exclude potential interferences through the in-source dissociation of glucuronide to parent drug. This new, rapid and robust LCMS/MS assay for the simultaneous quantification of plasma concentrations of these two major new anti-HIV drugs EVG and RPV offers an efficient analytical tool for clinical pharmacokinetics studies and routine therapeutic drug monitoring service.


Subject(s)
Anti-Retroviral Agents/blood , Chromatography, Liquid/methods , HIV Infections/blood , HIV Infections/drug therapy , Nitriles/blood , Pyrimidines/blood , Quinolones/blood , Tandem Mass Spectrometry/methods , Anti-Retroviral Agents/chemistry , Anti-Retroviral Agents/therapeutic use , Drug Stability , Humans , Nitriles/chemistry , Nitriles/therapeutic use , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Quinolones/chemistry , Quinolones/therapeutic use , Reproducibility of Results , Rilpivirine , Sensitivity and Specificity
10.
Antimicrob Agents Chemother ; 57(7): 3147-58, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23629707

ABSTRACT

New directly acting antivirals (DAAs) that inhibit hepatitis C virus (HCV) replication are increasingly used for the treatment of chronic hepatitis C. A marked pharmacokinetic variability and a high potential for drug-drug interactions between DAAs and numerous drug classes have been identified. In addition, ribavirin (RBV), commonly associated with hemolytic anemia, often requires dose adjustment, advocating for therapeutic drug monitoring (TDM) in patients under combined antiviral therapy. However, an assay for the simultaneous analysis of RBV and DAAs constitutes an analytical challenge because of the large differences in polarity among these drugs, ranging from hydrophilic (RBV) to highly lipophilic (telaprevir [TVR]). Moreover, TVR is characterized by erratic behavior on standard octadecyl-based reversed-phase column chromatography and must be separated from VRT-127394, its inactive C-21 epimer metabolite. We have developed a convenient assay employing simple plasma protein precipitation, followed by high-performance liquid chromatography coupled to tandem mass spectrometry (HPLC-MS/MS) for the simultaneous determination of levels of RBV, boceprevir, and TVR, as well as its metabolite VRT-127394, in plasma. This new, simple, rapid, and robust HPLC-MS/MS assay offers an efficient method of real-time TDM aimed at maximizing efficacy while minimizing the toxicity of antiviral therapy.


Subject(s)
Antiviral Agents/blood , Drug Monitoring/methods , Oligopeptides/blood , Proline/analogs & derivatives , Ribavirin/blood , Chromatography, Liquid , Hepacivirus/drug effects , Hepatitis/drug therapy , Hepatitis/prevention & control , Hepatitis/virology , Humans , Liver Cirrhosis/drug therapy , Liver Cirrhosis/prevention & control , Liver Cirrhosis/virology , Oligopeptides/therapeutic use , Proline/blood , Proline/therapeutic use , Ribavirin/therapeutic use , Tandem Mass Spectrometry
11.
Antivir Ther ; 18(5): 739-43, 2013.
Article in English | MEDLINE | ID: mdl-23640958

ABSTRACT

Here, we describe severe neuropsychiatric symptoms in an HIV-positive Asian man with extremely high efavirenz plasma levels while receiving standard treatment with efavirenz/tenofovir/emtricitabine fixed-dose regimen. Genetic examination revealed compound homozygosity for loss-of-function alleles of CYP2B6, including coding for a rare truncated protein. Neuropsychiatric symptoms resolved completely after efavirenz discontinuation.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/metabolism , Benzoxazines/metabolism , Reverse Transcriptase Inhibitors/pharmacokinetics , Adult , Alkynes , Alleles , Anti-HIV Agents/therapeutic use , Aryl Hydrocarbon Hydroxylases/genetics , Cyclopropanes , Cytochrome P-450 CYP2B6 , Genotype , HIV Infections/drug therapy , HIV Infections/enzymology , HIV Infections/genetics , HIV-1 , Humans , Male , Pharmacogenetics , Polymorphism, Single Nucleotide , Reverse Transcriptase Inhibitors/therapeutic use
12.
Chimia (Aarau) ; 66(5): 291-5, 2012.
Article in English | MEDLINE | ID: mdl-22867538

ABSTRACT

Pharmacokinetic variability in drug levels represent for some drugs a major determinant of treatment success, since sub-therapeutic concentrations might lead to toxic reactions, treatment discontinuation or inefficacy. This is true for most antiretroviral drugs, which exhibit high inter-patient variability in their pharmacokinetics that has been partially explained by some genetic and non-genetic factors. The population pharmacokinetic approach represents a very useful tool for the description of the dose-concentration relationship, the quantification of variability in the target population of patients and the identification of influencing factors. It can thus be used to make predictions and dosage adjustment optimization based on Bayesian therapeutic drug monitoring (TDM). This approach has been used to characterize the pharmacokinetics of nevirapine (NVP) in 137 HIV-positive patients followed within the frame of a TDM program. Among tested covariates, body weight, co-administration of a cytochrome (CYP) 3A4 inducer or boosted atazanavir as well as elevated aspartate transaminases showed an effect on NVP elimination. In addition, genetic polymorphism in the CYP2B6 was associated with reduced NVP clearance. Altogether, these factors could explain 26% in NVP variability. Model-based simulations were used to compare the adequacy of different dosage regimens in relation to the therapeutic target associated with treatment efficacy. In conclusion, the population approach is very useful to characterize the pharmacokinetic profile of drugs in a population of interest. The quantification and the identification of the sources of variability is a rational approach to making optimal dosage decision for certain drugs administered chronically.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV Infections/metabolism , HIV Seropositivity/metabolism , Nevirapine/pharmacokinetics , Adult , Aged , Aged, 80 and over , Algorithms , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Aryl Hydrocarbon Hydroxylases/genetics , Cohort Studies , Computer Simulation , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A/genetics , Drug Monitoring , Female , Genotype , HIV Infections/drug therapy , HIV Seropositivity/drug therapy , Humans , Male , Middle Aged , Models, Biological , Models, Statistical , Nevirapine/administration & dosage , Nevirapine/therapeutic use , Oxidoreductases, N-Demethylating/genetics , Polymorphism, Genetic , Population , Treatment Outcome , Young Adult
14.
Antimicrob Agents Chemother ; 56(6): 2959-66, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22371894

ABSTRACT

The objectives of this study were to characterize raltegravir (RAL) population pharmacokinetics in HIV-positive (HIV(+)) and healthy individuals, identify influential factors, and search for new candidate genes involved in UDP glucuronosyltransferase (UGT)-mediated glucuronidation. The pharmacokinetic analysis was performed with NONMEM. Genetic association analysis was performed with PLINK using the relative bioavailability as the phenotype. Simulations were performed to compare once- and twice-daily regimens. A 2-compartment model with first-order absorption adequately described the data. Atazanavir, gender, and bilirubin levels influenced RAL relative bioavailability, which was 30% lower in HIV(+) than in healthy individuals. UGT1A9*3 was the only genetic variant possibly influencing RAL pharmacokinetics. The majority of RAL pharmacokinetic variability remains unexplained by genetic and nongenetic factors. Owing to the very large variability, trough drug levels might be very low under the standard dosing regimen, raising the question of a potential relevance of therapeutic drug monitoring of RAL in some situations.


Subject(s)
HIV Infections/drug therapy , Pharmacogenetics/methods , Ritonavir/pharmacology , Adolescent , Adult , Aged , Anti-HIV Agents , Female , Genotype , HIV Infections/genetics , Humans , Male , Middle Aged , Models, Statistical , Models, Theoretical , Ritonavir/therapeutic use , Young Adult
15.
Clin Pharmacokinet ; 49(11): 741-65, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20923248

ABSTRACT

Oseltamivir is the ester-type prodrug of the neuraminidase inhibitor oseltamivir carboxylate. It has been shown to be an effective treatment for both seasonal influenza and the recent pandemic 2009 A/H1N1 influenza, reducing both the duration and severity of the illness. It is also effective when used preventively. This review aims to describe the current knowledge of the pharmacokinetic and pharmacodynamic characteristics of this agent, and to address the issue of possible therapeutic drug monitoring. According to the currently available literature, the pharmacokinetics of oseltamivir carboxylate after oral administration of oseltamivir are characterized by mean ± SD bioavailability of 79 ± 12%, apparent clearance of 25.3 ± 7.0 L/h, an elimination half-life of 7.4 ± 2.5 hours and an apparent terminal volume of distribution of 267 ± 122 L. A maximum plasma concentration of 342 ± 83 µg/L, a time to reach the maximum plasma concentration of 4.2 ± 1.1 hours, a trough plasma concentration of 168 ± 32 µg/L and an area under the plasma concentration-time curve from 0 to 24 hours of 6110 ± 1330 µg · h/L for a 75 mg twice-daily regimen were derived from literature data. The apparent clearance is highly correlated with renal function, hence the dosage needs to be adjusted in proportion to the glomerular filtration rate. Interpatient variability is moderate (28% in apparent clearance and 46% in the apparent central volume of distribution); there is no indication of significant erratic or limited absorption in given patient subgroups. The in vitro pharmacodynamics of oseltamivir carboxylate reveal wide variation in the concentration producing 50% inhibition of influenza A and B strains (range 0.17-44 µg/L). A formal correlation between systemic exposure to oseltamivir carboxylate and clinical antiviral activity or tolerance in influenza patients has not yet been demonstrated; thus no formal therapeutic or toxic range can be proposed. The pharmacokinetic parameters of oseltamivir carboxylate after oseltamivir administration (bioavailability, apparent clearance and the volume of distribution) are fairly predictable in healthy subjects, with little interpatient variability outside the effect of renal function in all patients and bodyweight in children. Thus oseltamivir carboxylate exposure can probably be controlled with sufficient accuracy by thorough dosage adjustment according to patient characteristics. However, there is a lack of clinical study data on naturally infected patients. In addition, the therapeutic margin of oseltamivir carboxylate is poorly defined. The usefulness of systematic therapeutic drug monitoring in patients therefore appears to be questionable; however, studies are still needed to extend the knowledge to particular subgroups of patients or dosage regimens.


Subject(s)
Antiviral Agents/pharmacokinetics , Disease Outbreaks , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Influenza, Human/drug therapy , Oseltamivir/analogs & derivatives , Pandemics , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Area Under Curve , Dose-Response Relationship, Drug , Drug Monitoring , Humans , Influenza Vaccines , Influenza, Human/epidemiology , Oseltamivir/pharmacokinetics , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/therapeutic use , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...