Network


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

Hotspot


Dive into the research topics where Gilbert Lefèvre is active.

Publication


Featured researches published by Gilbert Lefèvre.


BMC Medicine | 2015

Artemether-lumefantrine treatment of uncomplicated Plasmodium falciparum malaria: a systematic review and meta-analysis of day 7 lumefantrine concentrations and therapeutic response using individual patient data

Elizabeth A. Ashley; Francesca T. Aweeka; Karen I. Barnes; Quique Bassat; Steffen Borrmann; Prabin Dahal; Tme Davis; Philippe Deloron; Mey Bouth Denis; Abdoulaye Djimde; Jean-François Faucher; Blaise Genton; Philippe J Guerin; Kamal Hamed; Eva Maria Hodel; Liusheng Huang; Jullien; Harin Karunajeewa; Kiechel; Poul-Erik Kofoed; Gilbert Lefèvre; Niklas Lindegardh; Kevin Marsh; Andreas Mårtensson; Mayfong Mayxay; Rose McGready; C Moreira; Paul N. Newton; Billy Ngasala; François Nosten

Achieving adequate antimalarial drug exposure is essential for curing malaria. Day 7 blood or plasma lumefantrine concentrations provide a simple measure of drug exposure that correlates well with artemether-lumefantrine efficacy. However, the ‘therapeutic’ day 7 lumefantrine concentration threshold needs to be defined better, particularly for important patient and parasite sub-populations. The WorldWide Antimalarial Resistance Network (WWARN) conducted a large pooled analysis of individual pharmacokinetic-pharmacodynamic data from patients treated with artemether-lumefantrine for uncomplicated Plasmodium falciparum malaria, to define therapeutic day 7 lumefantrine concentrations and identify patient factors that substantially alter these concentrations. A systematic review of PubMed, Embase, Google Scholar, ClinicalTrials.gov and conference proceedings identified all relevant studies. Risk of bias in individual studies was evaluated based on study design, methodology and missing data. Of 31 studies identified through a systematic review, 26 studies were shared with WWARN and 21 studies with 2,787 patients were included. Recrudescence was associated with low day 7 lumefantrine concentrations (HR 1.59 (95 % CI 1.36 to 1.85) per halving of day 7 concentrations) and high baseline parasitemia (HR 1.87 (95 % CI 1.22 to 2.87) per 10-fold increase). Adjusted for mg/kg dose, day 7 concentrations were lowest in very young children (<3 years), among whom underweight-for-age children had 23 % (95 % CI −1 to 41 %) lower concentrations than adequately nourished children of the same age and 53 % (95 % CI 37 to 65 %) lower concentrations than adults. Day 7 lumefantrine concentrations were 44 % (95 % CI 38 to 49 %) lower following unsupervised treatment. The highest risk of recrudescence was observed in areas of emerging artemisinin resistance and very low transmission intensity. For all other populations studied, day 7 concentrations ≥200 ng/ml were associated with >98 % cure rates (if parasitemia <135,000/μL). Current artemether-lumefantrine dosing recommendations achieve day 7 lumefantrine concentrations ≥200 ng/ml and high cure rates in most uncomplicated malaria patients. Three groups are at increased risk of treatment failure: very young children (particularly those underweight-for-age); patients with high parasitemias; and patients in very low transmission intensity areas with emerging parasite resistance. In these groups, adherence and treatment response should be monitored closely. Higher, more frequent, or prolonged dosage regimens should now be evaluated in very young children, particularly if malnourished, and in patients with hyperparasitemia.


The Lancet | 2008

Efficacy and safety of artemether-lumefantrine dispersible tablets compared with crushed commercial tablets in African infants and children with uncomplicated malaria: a randomised, single-blind, multicentre trial

Salim Abdulla; Issaka Sagara; Steffen Borrmann; Umberto D'Alessandro; Raquel González; Mary J. Hamel; Bernhards Ogutu; Andreas Mårtensson; John Lyimo; Hamma Maiga; Philip Sasi; Alain Nahum; Quique Bassat; Elizabeth Juma; Lucas Otieno; Anders Björkman; Hans-Peter Beck; Kim Andriano; Marc Cousin; Gilbert Lefèvre; David Ubben; Zulfikarali Premji

BACKGROUND Combination treatments, preferably containing an artemisinin derivative, are recommended to improve efficacy and prevent Plasmodium falciparum drug resistance. Our aim was to show non-inferiority of a new dispersible formulation of artemether-lumefantrine to the conventional crushed tablet in the treatment of young children with uncomplicated malaria. METHODS We did a randomised non-inferiority study on children weighing 5-35 kg with uncomplicated P falciparum malaria in Benin, Kenya, Mali, Mozambique, and Tanzania. The primary outcome measure was PCR-corrected 28-day parasitological cure rate. We aimed to show non-inferiority (with a margin of -5%) of dispersible versus crushed tablet. We constructed an asymptotic one-sided 97.5% CI on the difference in cure rates. A computer-generated randomisation list was kept centrally and investigators were unaware of the study medication administered. We used a modified intention-to-treat analysis. This trial is registered with ClinicalTrials.gov, number NCT00386763. FINDINGS 899 children aged 12 years or younger were randomly assigned to either dispersible (n=447) or crushed tablets (n=452). More than 85% of patients in each treatment group completed the study. 812 children qualified for the modified intention-to-treat analysis (n=403 vs n=409). The PCR-corrected day-28 cure rate was 97.8% (95% CI 96.3-99.2) in the group on dispersible formulation and 98.5% (97.4-99.7) in the group on crushed formulation. The lower bound of the one-sided 97.5% CI was -2.7%. The most common drug-related adverse event was vomiting (n=33 [7%] and n=42 [9%], respectively). No signs of ototoxicity or relevant cardiotoxicity were seen. INTERPRETATION A six-dose regimen of artemether-lumefantrine with the new dispersible formulation is as efficacious as the currently used crushed tablet in infants and children, and has a similar safety profile.


The New England Journal of Medicine | 2014

Spiroindolone KAE609 for Falciparum and Vivax Malaria

Nicholas J. White; Sasithon Pukrittayakamee; Aung Pyae Phyo; Ronnatrai Rueangweerayut; François Nosten; Podjanee Jittamala; Atthanee Jeeyapant; Jay Prakash Jain; Gilbert Lefèvre; Ruobing Li; Baldur Magnusson; Thierry T. Diagana; Leong Fj

BACKGROUND KAE609 (cipargamin; formerly NITD609, Novartis Institute for Tropical Diseases) is a new synthetic antimalarial spiroindolone analogue with potent, dose-dependent antimalarial activity against asexual and sexual stages of Plasmodium falciparum. METHODS We conducted a phase 2, open-label study at three centers in Thailand to assess the antimalarial efficacy, safety, and adverse-event profile of KAE609, at a dose of 30 mg per day for 3 days, in two sequential cohorts of adults with uncomplicated P. vivax malaria (10 patients) or P. falciparum malaria (11). The primary end point was the parasite clearance time. RESULTS The median parasite clearance time was 12 hours in each cohort (interquartile range, 8 to 16 hours in patients with P. vivax malaria and 10 to 16 hours in those with P. falciparum malaria). The median half-lives for parasite clearance were 0.95 hours (range, 0.68 to 2.01; interquartile range, 0.85 to 1.14) in the patients with P. vivax malaria and 0.90 hours (range, 0.68 to 1.64; interquartile range, 0.78 to 1.07) in those with P. falciparum malaria. By comparison, only 19 of 5076 patients with P. falciparum malaria (<1%) who were treated with oral artesunate in Southeast Asia had a parasite clearance half-life of less than 1 hour. Adverse events were reported in 14 patients (67%), with nausea being the most common. The adverse events were generally mild and did not lead to any discontinuations of the drug. The mean terminal half-life for the elimination of KAE609 was 20.8 hours (range, 11.3 to 37.6), supporting a once-daily oral dosing regimen. CONCLUSIONS KAE609, at dose of 30 mg daily for 3 days, cleared parasitemia rapidly in adults with uncomplicated P. vivax or P. falciparum malaria. (Funded by Novartis and others; ClinicalTrials.gov number, NCT01524341.).


Clinical Pharmacology & Therapeutics | 2008

Pharmacokinetics and Pharmacodynamics of the Novel Daily Rivastigmine Transdermal Patch Compared With Twice‐daily Capsules in Alzheimer's Disease Patients

Gilbert Lefèvre; Greg Sedek; S. S. Jhee; M. T. Leibowitz; Hsun‐Lun Aaron Huang; A. Enz; Steve Maton; L. Ereshefsky; Françoise Pommier; H. Schmidli; Silke Appel-Dingemanse

A transdermal patch has been developed for the cholinesterase inhibitor rivastigmine. This study investigated the pharmacokinetics and pharmacodynamics of rivastigmine and NAP226–90, and compared drug exposure between patch and capsule administrations. This was an open‐label, parallel‐group study in Alzheimers disease patients randomized to receive either capsule (1.5–6 mg Q12H, i.e., 3–12 mg/day) or patch (5–20 cm2) in ascending doses through four 14‐day periods. The patch showed lower Cmax (ca. 30% lower at 20 cm2, 19.5 versus 29.3 ng/ml), longer tmax (8.0 versus 1.0 h), and greater AUC (ca. 1.8‐fold at 20 cm2, 345 versus 191 ng·h/ml) compared with the 6 mg Q12H capsule dose, with markedly less fluctuation between peak and trough plasma levels (80% at 20 cm2 versus 620% at 1.5 mg Q12H). Plasma butyrylcholinesterase inhibition rose slowly after patch administration, whereas two distinct peaks were seen after capsule administration. Average exposure with the 10 cm2 patch was comparable to the highest capsule dose (6 mg Q12H, i.e., 12 mg/day).


Malaria Journal | 2009

Understanding the pharmacokinetics of Coartem

Abdoulaye Djimde; Gilbert Lefèvre

Artemether and lumefantrine (AL), the active constituents of Coartem® exhibit complementary pharmacokinetic profiles. Artemether is absorbed quickly; peak concentrations of artemether and its main active metabolite, dihydroartemisinin (DHA) occur at approximately two hours post-dose, leading to a rapid reduction in asexual parasite mass and a prompt resolution of symptoms. Lumefantrine is absorbed and cleared more slowly (terminal elimination half-life 3-4 days in malaria patients), and accumulates with successive doses, acting to prevent recrudescence by destroying any residual parasites that remain after artemether and DHA have been cleared from the body. Food intake significantly enhances the bioavailability of both artemether and lumefantrine, an effect which is more apparent for the highly lipophilic lumefantrine. However, a meal with only a small amount of fat (1.6 g) is considered sufficient to achieve adequate exposure to lumefantrine. The pharmacokinetics of artemether or lumefantrine are similar in children, when dosed according to their body weight, compared with adults. No randomized study has compared the pharmacokinetics of either agent in pregnant versus non-pregnant women. Studies in healthy volunteers and in children with malaria have confirmed that the pharmacokinetic characteristics of crushed standard AL tablets and the newly-developed Coartem® Dispersible tablet formulation are similar. Studies to date in healthy volunteers have not identified any clinically relevant drug-drug interactions; data relating to concomitant administration of HIV therapies are limited. While dose-response analyses are difficult to undertake because of the low rate of treatment failures under AL, it appears that artemether and DHA exposure impact on parasite clearance time while lumefantrine exposure is associated with cure rate, consistent with their respective modes of action. In conclusion, knowledge of the pharmacokinetic profiles of artemether and lumefantrine is increasing within a range of settings, including infants and children. However, additional data would be warranted to better characterize artemether and lumefantrine pharmacokinetics in patients with hepatic impairment, in pregnant women, and in patients undergoing HIV/AIDS chemotherapy.


European Journal of Pharmaceutical Sciences | 2000

Pharmacokinetic interaction trial between co-artemether and mefloquine

Gilbert Lefèvre; Margaretha Bindschedler; Farkad Ezzet; Nicole Schaeffer; Ingo Meyer; Mikael S. Thomsen

Forty-two healthy subjects were randomized in a parallel three-group design trial to investigate potential electrocardiographic and pharmacokinetic interactions between the new antimalarial co-artemether, a combination of artemether and lumefantrine (both of which are predominantly metabolized through CYP3A4), and mefloquine, another antimalarial described as a substrate (and possible inhibitor) of CYP3A4. Subjects were assigned to one of the three possible treatment groups (i.e., co-artemether alone or mefloquine alone or the combination of both). The dosage was 1000 mg mefloquine (divided into three doses over 12 h) followed 12 h later by six applications of co-artemether (40 mg artemether+480 mg lumefantrine each) over 60 h. The study medications were generally well tolerated after all treatments. Concomitant administration with mefloquine caused statistically significant lower (around 30-40%) plasma concentrations of lumefantrine than when co-artemether was administered alone. Even if important, this decrease in lumefantrine exposure was considered unlikely to impact clinical efficacy given the wide therapeutic index of co-artemether and the usual high variability in lumefantrine plasma levels, mostly and more importantly influenced by food intake. However, patients should be encouraged to eat at dosing times to compensate for this decreased bioavailability. The pharmacokinetics of artemether, DHA or mefloquine were not affected. Artemether concentrations significantly decreased over doses, independently of mefloquine co-administration, while DHA concentrations slightly (not significantly) increased. Therefore, no clinically relevant risks due to pharmacokinetic drug-drug interaction are expected at the enzymatic level following co-administration of co-artemether with CYP3A4 substrates with similar affinity to that of mefloquine.


Current Medical Research and Opinion | 2007

Rivastigmine exposure provided by a transdermal patch versus capsules.

Francois Mercier; Gilbert Lefèvre; Hsun‐Lun Aaron Huang; Heinz Schmidli; Billy Amzal; Silke Appel-Dingemanse

ABSTRACT Objectives: The rivastigmine transdermal patch is the first transdermal treatment for Alzheimers disease (AD) and dementia associated with Parkinsons disease. The objective of this study was to evaluate the pharmacokinetics of rivastigmine following transdermal delivery by a patch versus oral delivery with conventional capsules in a population of AD patients. Methods: Both non-compartmental and compartmental analyses were performed on the same database showing relatively large inter-patient variations in pharmacokinetic parameters (up to 73% for the capsule group). The compartmental analysis provided model-based predictions of pharmacokinetic parameters, with the aim of comparing the two modes of administration when adjusting for confounding factors such as patient body weight and gender. Results: According to both non-compartmental and compartmental analyses, the patch provided significantly lower peak rivastigmine plasma concentrations (Cmax) and slower times to Cmax (tmax), compared with capsules. However, drug exposure (area under the curve; AUC) was not significantly different between the 4.6 mg/24 hour (5 cm2) patch and 3 mg BID (6 mg/day) capsule doses, or between the 9.5 mg/24 hour (10 cm2) patch and 6 mg BID (12 mg/day) capsule doses, according to both analyses. This suggests comparable exposure from these two rivastigmine delivery systems. Conclusion: The analyses were consistent with previous reports of a markedly less fluctuating, more continuous drug delivery with the rivastigmine patch. This characteristic delivery profile is associated with similar efficacy yet improved tolerability, compared with capsules.


European Journal of Pharmaceutical Sciences | 1999

Binding of artemether and lumefantrine to plasma proteins and erythrocytes.

Danielle Colussi; Carole Parisot; Francois Legay; Gilbert Lefèvre

The serum/plasma protein binding and blood distribution of artemether and lumefantrine was studied in vitro. The techniques used were the erythrocyte partitioning and ultrafiltration methods with 1499%, respectively. Under physiological protein concentrations, the distribution in blood showed that 33% of artemether was bound to alpha(1)-acid glycoprotein, 17% to albumin, 12% to high density lipoproteins (HDL), 9.3% to low density lipoproteins (LDL) and 12% to very low density lipoproteins (VLDL), with binding capacities (nKa) of 3.2 x 10(5), 6.2 x 10(3), 2.1 x 10(5), 1.7 x 10(6) and 2.0 x 10(7) lmol(-1), respectively. 77% of lumefantrine was bound to HDL, 7.3% to LDL and 6.6% to VLDL, with binding capacities of 2.7 x 10(7), 2. 6 x 10(7) and 2.4 x 10(8) lmol(-1), respectively. A negligible fraction of lumefantrine was bound to albumin and alpha(1)-acid glycoprotein. The fraction in erythrocytes was around 10% for both artemether and lumefantrine.


Clinical Drug Investigation | 1999

Clinical Pharmacokinetics of Artemether and Lumefantrine (Riamet

Gilbert Lefèvre; Mikael S. Thomsen

This paper reports the clinical pharmacokinetics of artemether and lumefantrine in healthy volunteers and in malaria patients. These two drugs are the active components of the fixed-dose oral combination tablet co-artemether (Riamet®), used for the treatment of Plasmodium falciparum malaria.Artemether has a fast absorption rate followed by rapid clearance from plasma [terminal elimination half-life (t1/2β) 2 to 3 hours]. Its major metabolite dihydroartemisinin (DHA) is formed rapidly and has a similar clearance pattern to artemether. Lumefantrine is slowly absorbed (2 hours lag time) followed by a slow clearance from plasma (t1/2β up to 10 days). Food intake significantly increases the bioavailability of both artemether (>2-fold) and lumefantrine (approximately 16-fold).Artemether and lumefantrine are predominantly metabolised by the cytochrome P450 3A4 (CYP3A4) isoenzyme. The intersubject variability for artemether, DHA and lumefantrine is high in both healthy volunteers and patients.CYP3A4 substrates/inhibitors such as the antimalarial mefloquine can be coadministered with co-artemether without clinically relevant risk of drug-drug interaction. Similarly, potential for a significant medical hazard in the management of P. falciparum malaria with co-artemether following co-administration in close temporal relationship with quinine (a CYP3A4 substrate) is unlikely. The wide therapeutic index of artemether and lumefantrine and the short duration of administration of co-artemether suggest that the risk for any tolerability problems from drug accumulation is minimal.


Neurology | 2007

Pharmacokinetic rationale for the rivastigmine patch

Jeffrey L. Cummings; Gilbert Lefèvre; Gary W. Small; Silke Appel-Dingemanse

The dual cholinesterase inhibitor rivastigmine is approved in capsule form in many countries for the symptomatic treatment of dementia associated with Alzheimer disease (AD) and Parkinson disease (PD). All orally administered cholinesterase inhibitors are associated with central cholinergic gastrointestinal side effects, particularly during the titration phase, which are believed to be caused by a rapid increase in brain acetylcholine levels after effective inhibition of the target enzymes. A recently developed rivastigmine transdermal patch may have the potential to reduce such side effects. Pharmacokinetic studies have shown that transdermal administration of rivastigmine prolongs tmax, lowers Cmax, and reduces fluctuations in plasma concentration. The 10-cm2 rivastigmine patch provides comparable exposure (area under the curve, AUC) to the highest capsule dose (6-mg BID) and may be the target maintenance dose for most patients, delivering optimal rivastigmine exposure to produce a therapeutic effect. The potential of a patch to improve the tolerability of rivastigmine (e.g., nausea and vomiting) while permitting similar exposure to the highest doses of capsules may, in turn, lead to improved efficacy and compliance.

Collaboration


Dive into the Gilbert Lefèvre's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andreas Mårtensson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge