Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Eric Singlas is active.

Publication


Featured researches published by Eric Singlas.


Clinical Pharmacology & Therapeutics | 1989

Zidovudine disposition in patients with severe renal impairment: Influence of hemodialysis

Eric Singlas; Jean‐Claude Pioger; Anne‐Marie Taburet; Jean‐Noel Colin; Jean‐Paul Fillastre

Pharmacokinetics of zidovudine (azidothymidine, AZT) was investigated after oral administration (200 mg) in 25 HIV seronegative subjects: 14 patients with severe renal impairment (creatinine clearance 6 to 31 ml/min), five hemodialyzed anuric patients, and six healthy subjects. Plasma and urine concentrations of zidovudine and its glucuronidated metabolite (GAZT) were measured simultaneously by HPLC assay. In healthy subjects, GAZT concentrations were higher than those of AZT; AUC values were 23.7 ± 1.9 and 5.2 ± 0.6 µmol · hr/L, respectively. Formation of GAZT rate‐limits its elimination: GAZT half‐life (t½) parallels that of AZT, which is around 1 hour. In uremic patients, AZT concentrations were moderately increased (AUC = 11.7 ± 1.1 µmol · hr/L), whereas t½ and mean residence time (MRT) remain unchanged despite the decreased renal clearance (16 ± 2 versus 220 ± 58 ml/min) and decreased urinary excretion (1.6 ± 0.3 versus 8.1 ± 1.0% of the dose). In contrast, GAZT concentrations are markedly increased (AUC = 402.9 ± 88.6 µmol · hr/L). As a consequence of the decreased renal clearance (27 ± 3 versus 331 ± 42 ml/min), elimination is the rate‐limiting step and t½ is increased (8 ± 2 versus 0.9 ± 0.1 hr). Contribution of a 4‐hour hemodialysis session to AZT elimination appears to be negligible, whereas elimination of GAZT is enhanced. On the sole basis of AZT pharmacokinetic data, no particular dose adjustment appears to be necessary in patients who have severe renal impairment (creatinine clearance between 10 and 30 ml/min). However, high levels of GAZT should be anticipated with the usual dosage regimen.


Clinical Pharmacology & Therapeutics | 1990

Pharmacokinetics of zidovudine in patients with liver cirrhosis

Anne‐Marie Taburet; Sylvie Naveau; Grégoire Zorza; Jean‐Noel Colin; Jean‐François Delfraissy; Chaput Jc; Eric Singlas

The pharmacokinetics of zidovudine (azidothymidine, AZT) was investigated after oral administration (200 mg) in 14 human immunodeficiency virus seronegative patients with liver cirrhosis. They were divided in three groups according to the severity of the liver disease quantitated by the Child‐Pugh score. Plasma and urine concentrations of zidovudine and its glucuronidated metabolite (GAZT) were measured simultaneously by HPLC assay. Findings were compared with those previously measured in six healthy volunteers. As a consequence of a marked drop in oral clearance (10 ± 4 versus 38 ± 15 ml/min/kg), zidovudine concentrations, half‐life, and mean residence time were increased in patients with cirrhosis. No difference could be established between the three groups. The reason for such a decrease in oral clearance of zidovudine was the reduction in the GAZT formation clearance (236 ± 73 versus 1540 ± 540 ml/min); this led to a decrease in the AUC ratio of GAZT and zidovudine (1.3 ± 0.6 versus 4.6 ± 0.7), which was directly related to the severity of the cirrhosis. In patients, as in volunteers, formation of GAZT rate limits its elimination. To avoid important cumulation of zidovudine after repeated dosing in patients with acquired immunodeficiency syndrome who have hepatic impairment, a dosage adjustment could be proposed.


British Journal of Pharmacology | 2004

Cerebral uptake of mefloquine enantiomers with and without the P-gp inhibitor elacridar (GF1210918) in mice

Sylvie Barraud de Lagerie; Emmanuelle Comets; Céline Gautrand; Christine Fernandez; Daniel Auchere; Eric Singlas; François Gimenez

Mefloquine is a chiral neurotoxic antimalarial agent showing stereoselective brain uptake in humans and rats. It is a substrate and an inhibitor of the efflux protein P‐glycoprotein. We investigated the stereoselective uptake and efflux of mefloquine in mice, and the consequences of the combination with an efflux protein inhibitor, elacridar (GF120918) on its brain transport. Racemic mefloquine (25 mg kg−1) was administered intraperitoneally with or without elacridar (10 mg kg−1). Six to seven mice were killed at each of 11 time‐points between 30 min and 168 h after administration. Blood and brain concentrations of mefloquine enantiomers were determined using liquid chromatography. A three‐compartment model with zero‐order absorption from the injection site was found to best represent the pharmacokinetics of both enantiomers in blood and brain. (−)Mefloquine had a lower blood and brain apparent volume of distribution and a lower efflux clearance from the brain, resulting in a larger brain/blood ratio compared to (+)mefloquine. Elacridar did not modify blood concentrations or the elimination rate from blood for either enantiomers. However, cerebral AUCinf of both enantiomers were increased, with a stronger effect on (+)mefloquine. The efflux clearance from the brain decreased for both enantiomers, with a larger decrease for (+)mefloquine. After administration of racemic mefloquine in mice, blood and brain pharmacokinetics are stereoselective, (+)mefloquine being excreted from brain more rapidly than its antipode, showing that mefloquine is a substrate of efflux proteins and that mefloquine enantiomers undergo efflux in a stereoselective manner. Moreover, pretreatment with elacridar reduced the brain efflux clearances with a more pronounced effect on (+)mefloquine.


Clinical Pharmacokinectics | 1996

Drug interactions with antiviral drugs.

Anne-Marie Taburet; Eric Singlas

SummaryAntiviral drug interactions are a particular problem among immuno-compromised patients because these patients are often receiving multiple different drugs, i.e. antiretroviral drugs and drugs effective against herpesvirus. The combination of zidovudine and other antiretroviral drugs with different adverse event profiles, such as didanosine, zalcitabine and lamivudine, appears to be well tolerated and no relevant pharmacokinetic interactions have been detected. The adverse effects of didanosine and zalcitabine (i.e. peripheral neuropathy and pancreatitis) should be taken into account when administering these drugs with other drugs with the same tolerability profile.Coadministration of zidovudine and ganciclovir should be avoided because of the high rate of haematological intolerance. In contrast, zidovudine and foscarnet have synergistic effect and no pharmacokinetic interaction has been detected. No major change in zidovudine pharmacokinetics was seen when the drug was combined with aciclovir, famciclovir or interferons. However, concomitant use of zidovudine and ribavirin is not advised.Although no pharmacokinetic interaction was documented when didanosine was first administered with intravenous ganciclovir, recent studies have shown that concentrations of didanosine are increased by 50% or more when coadministered with intravenous or oral ganciclovir. The mechanism of this interaction has not been elucidated. Lack of pharmacokinetic interaction was demonstrated between foscarnet and didanosine or ganciclovir.Clinical trials have shown that zidovudine can be administered safely with paracetamol (acetaminophen), nonsteroidal anti-inflammatory drugs, oxazepam or codeine. Inhibition of zidovudine glucuronidation has been demonstrated with fluconazole, atovaquone, valproic acid (valproate sodium), methadone, probenecid and inosine pranobex; however, the clinical consequences of this have not been fully investigated.No interaction has been demonstrated with didanosine per se but care should be taken of interaction with the high pH buffer included in the tablet formulation. Drugs that need an acidic pH for absorption (ketoconazole, itraconazole but not fluconazole, dapsone, pyrimethamine) or those that can be chelated by the ions of the buffer (quinolones and tetracyclines) should be administered 2 hours before or 6 hours after didanosine.Very few interaction studies have been undertaken with other antiviral drugs. Coadministration of zalcitabine with the antacid ‘Maalox’ results in a reduction of its absorption. Dapsone does not influence the disposition of zalcitabine. Cotrimoxazole (trimethoprim-sulfamethoxazole) causes an increase in lamivudine concentrations by 43%. Saquinavir, delavirdine and atevirdine appeared to be metabolised by cytochrome P450 and interactions with enzyme inducers or inhibitors could be anticipated.Some studies showed that interferons can reduce drug metabolism but only a few studies have evaluated the pathways involved.Further studies are required to better understand the clinical consequences of drug interactions with antiviral drugs. Drug-drug interactions should be considered in addition to individual drug clinical benefits and safety profiles.


Journal of Chromatography B: Biomedical Sciences and Applications | 2000

Simultaneous high-performance liquid chromatographic determination of the antiretroviral agents amprenavir, nelfinavir, ritonavir, saquinavir, delavirdine and efavirenz in human plasma

Virginie Proust; Katalin Toth; Anne Hulin; Anne-Marie Taburet; François Gimenez; Eric Singlas

This article describes a method for the simultaneous determination of four licensed HIV protease inhibitors (amprenavir, nelfinavir, saquinavir and ritonavir) and two novel non-nucleoside reverse transcriptase inhibitors (efavirenz and delavirdine) in human plasma in a single run. Plasma samples (500 microl) were treated by liquid-liquid extraction with methyl tert.-butyl ether. The compounds were separated by reversed-phase liquid chromatography on a C(18) column with spectrophotometric detection at 260 nm. The method is linear over the specific ranges investigated, accurate (inaccuracy <11.7%) and showed intra-day and inter-day precision within the ranges of 0.9-7.0 and 1.9-8.8%, respectively. The six compounds were stable in plasma after 6 months of storage at -20 degrees C and after five freeze-thaw cycles.


Clinical Pharmacokinectics | 2004

Evaluation of pharmacokinetic interactions after oral administration of mycophenolate mofetil and valaciclovir or aciclovir to healthy subjects.

François Gimenez; Estelle Foeillet; Olivier Bourdon; Steve Weller; Christophe Garret; Roselyne Bidault; Eric Singlas

ObjectiveTo investigate a potential pharmacokinetic interaction between mycophenolate mofetil (MMF) and aciclovir or valaciclovir.Study design and participantsFifteen healthy subjects were enrolled in a prospective, randomised, open-label, single-dose, cross-over study conducted at a single centre. Subjects received each of the following five oral treatments: (i) aciclovir 800mg alone; (ii) valaciclovir 2g alone; (iii) MMF 1g alone; (iv) valaciclovir 2g + MMF 1g; and (v) aciclovir 800mg + MMF 1g. The following pharmacokinetic parameters were estimated for aciclovir, mycophenolic acid (MPA) and its inactive glucuronide metabolite (MPAG) from the plasma concentration-time data using noncompartmental methods: area under the concentration-time curve from zero to infinity (AUC∞), terminal elimination half-life (t½z), peak concentration (Cmax) and time to Cmax (tmax). The renal clearance (CLR) of aciclovir was also calculated. These parameters were compared when aciclovir or valaciclovir were coadministered with MMF relative to aciclovir, valaciclovir or MMF given alone.Results and discussionAciclovir Cmax, tmax and AUC∞ were significantly increased by 40%, 0.38 hour and 31%, respectively, following coadministration of aciclovir and MMF, whereas aciclovir t½z was significantly decreased by 11%. Following coadministration of valaciclovir and MMF, aciclovir pharmacokinetic parameters were not significantly modified except for tmax (about 0.5 hour shorter with MMF). MPA and MPAG pharmacokinetic parameters were not significantly modified following coadministration of MMF with valaciclovir or aciclovir except for MPAG AUC∞, which was decreased by 12% with valaciclovir. Our results are similar to those reported in the literature, except for MPAG AUC. In urine, following coadministration of aciclovir and MMF, aciclovir CLR was significantly decreased by 19%. Competition between MPAG and aciclovir for renal tubular secretion could partly explain this phenomenon. Following coadmin-istration of valaciclovir and MMF, aciclovir CLR was not significantly modified.ConclusionIn healthy subjects, interactions are observed after coadministration of MMF and aciclovir, but the extent of the interactions is unlikely to be of clinical significance. These interactions should be investigated in patients with abnormal renal function.


Clinical Pharmacokinectics | 1991

Pharmacokinetics of Newer Drugs in Patients with Renal Impairment (Part I)

Fillastre Jp; Eric Singlas

SummaryMany drugs are eliminated via the renal route and the usual dose must be modified in patients with severe renal impairment. This review is an attempt to supply physicians with the more recent data on pharmacokinetic studies of new drugs administered in uraemic patients. The review is in 2 parts: the first indicates the results of studies on the pharmacokinetics of antibiotic agents, antifungal, antiviral and antiulcer drugs, and nonsteroidal anti-inflammatory drugs. Special mention is made of epoetin (recombinant human erythropoietin). It was not possible to give all the information collected from the recent literature: since mild renal failure has little effect on the fate of a drug, pharmacokinetic data obtained in patients with a creatinine clearance (CLCR) of more than 50 ml/min has been omitted. Both the text and tables give recommendations for treating patients with moderate renal insufficiency (CLCR of about 50 ml/min), more severe renal impairment (CLCR between 10 and 50 ml/min) and end-stage renal failure with a very low creatinine clearance (below 10 ml/min). It was not possible to give uniform recommendations (i.e. reducing the dose while maintaining the same interval, or giving the same dose and prolonging the interval). This article follows the recommendations of the authors, which may vary for drugs in similar classes.


Clinical Pharmacokinectics | 1990

Disposition of fleroxacin, a new trifluoroquinolone, and its metabolites. Pharmacokinetics in renal failure and influence of haemodialysis.

Eric Singlas; A. Leroy; Eric Sultan; Michel Godin; Bruno Moulin; Anne Marie Taburet; Malek Dhib; Fillastre Jp

SummaryThe pharmacokinetics of fleroxacin and its metabolites following a single oral dose of fleroxacin 400mg were examined in 6 healthy subjects and 24 patients with various degrees of renal insufficiency. Plasma and urine samples, collected at various times after administration, were assayed by high performance liquid chromatography (HPLC).In healthy subjects. Cmax was 6.8 ± 0.7 mg/L; Xmax = about 1h. t½ = 14 ± 2h, total clearance = 4.86 ± 0.72 L/h and the percentage of unchanged fleroxacin excreted in urine in 48 hours was 48 ± 4% (HPLC). Plasma concentrations of metabolites were very low and accounted for no more than 5% of the levels of unchanged fleroxacin.In uraemic patients Cmax did not change, whatever the degree of renal failure; tmax was increased in patients with a glomerular filtration rate below 0.6 L/h, and Vd/f was independent of the seventy of renal failure. These data suggest that bioavailability of the drug is unchanged. In uraemic patients t½, was prolonged and AUC multiplied by a factor of 2 to 3. A linear relationship was found between total and renal clearances of fleroxacin and crcatinine clearance. Accumulation of N-demethyl-fleroxacin and N-oxide-fleroxacin was very high in uraemic patients, due to slow formation of these metabolites and decreased urinary elimination.Dialysance of fleroxacin and of its metabolites was approximately 3.6 to 4.8 L/h. These findings suggest that fleroxacin dosage may need to be reduced in patients with severe renal disease; in haemodialysed patients, treated every 2 days, a single dose of fleroxacin 400mg is recommended at the end of each dialysis session.


Clinical Pharmacokinectics | 1990

Disposition of fleroxacin, a new trifluoroquinolone, and its metabolites. Pharmacokinetics in elderly patients.

Anne Marie Taburet; Alain Devillers; Patrick Thomare; Jean‐Paul Fillastre; Pierre Veyssier; Eric Singlas

SummaryThe pharmacokinetics of fleroxacin and its main metabolites, N-demethyl-fleroxacin and N-oxide-fleroxacin, were studied in 12 elderly patients aged 63 to 88 years. Plasma and urine samples collected at different times after drug administration were analysed by a specific reverse phase high performance liquid chromatography (HPLC) method.The peak plasma concentration (Cmax) of fleroxacin was 15.6 ± 1.6 mg/L, time to Cmax (tmax) was about 3h, elimination half-life (t½) was 16 ± 1h and the percentage of unchanged fleroxacin excreted in urine was 39 ± 3% of the dose. The plasma concentrations of metabolites were very low and accounted for no more than 4% of the concentration of unchanged fleroxacin.Plasma parameters were mainly correlated with age and weight; urinary parameters were correlated with creatinine clearance. Compared with results in younger normal patients, no significant change in the t½, of fleroxacin or metabolites was observed. Assuming that the bioavailability (f) is complete, the apparent volume of distribution (Vd/f) was lower in elderly (0.9 ± 0.1 L/kg) than in younger patients (1.3 ± 0.1 L/kg) and a 2-fold decrease in apparent total clearance (CL/f) was noted (2.58 ± 0.42 vs 4.86 ±0.72 L/h); plasma concentrations were consequently higher in elderly patients. Compared with patients with renal failure, the pharmacokinetics of fleroxacin and metabolites in the elderly were similar to those of patients with mild to moderate renal insufficiency.On the basis of the findings of this single dose study, no major dosage adjustments are needed for patients of this age range except for those with creatinine clearance < 30 ml/min.


Drug Metabolism and Disposition | 2003

Effect of Efflux Inhibition on Brain Uptake of Itraconazole in Mice Infected with Cryptococcus neoformans

Frédéric Imbert; Méryam Jardin; Christine Fernandez; Jean Charles Gantier; Françoise Dromer; Gabriel Baron; Ludy van Beijsterveldt; Eric Singlas; François Gimenez

Collaboration


Dive into the Eric Singlas's collaboration.

Top Co-Authors

Avatar

François Gimenez

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Virginie Proust

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

François Gimenez

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrick Niaudet

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Agnès Ferroni

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

Agnès Mogenet

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

Anne Hulin

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

Bruno Moulin

University of Strasbourg

View shared research outputs
Researchain Logo
Decentralizing Knowledge