Network


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

Hotspot


Dive into the research topics where Yannick Le Meur is active.

Publication


Featured researches published by Yannick Le Meur.


Therapeutic Drug Monitoring | 2009

Opportunities to optimize tacrolimus therapy in solid organ transplantation: report of the European consensus conference.

Pierre Wallemacq; Victor W. Armstrong; Mercè Brunet; Vincent Haufroid; David W. Holt; Atholl Johnston; Dirk Kuypers; Yannick Le Meur; Pierre Marquet; Michael Oellerich; Eric Thervet; Burkhand Toenshoff; Nas Undre; Lutz T. Weber; Ian S. Westley; Michel Mourad

In 2007, a consortium of European experts on tacrolimus (TAC) met to discuss the most recent advances in the drug/dose optimization of TAC taking into account specific clinical situations and the analytical methods currently available and drew some recommendations and guidelines to help clinicians with the practical use of the drug. Pharmacokinetic, pharmacodynamic, and more recently pharmacogenetic approaches aid physicians to individualize long-term therapies as TAC demonstrates a high degree of both between- and within-individual variability, which may result in an increased risk of therapeutic failure if all patients are administered a uniform dose. TAC has undoubtedly benefited from therapeutic drug monitoring, but interpretation of the blood concentration is confounded by the relative differences between the assays. Single time points, limited sampling strategies, and area under concentration-time curve have all been considered to determine the most appropriate sampling procedure that correlates with efficacy. Therapeutic trough TAC concentration ranges have changed since the initial introduction of the drug, while still maintaining adequate immunosuppression and avoiding drug-related adverse effects. Pharmacodynamic markers have also been considered advantageous to the clinician, which may better reflect efficacy and safety, taking into account the between-individual variability rather than whole blood concentrations. The choice of method, differences between methods, and potential pitfalls of the method should all be considered when determining TAC concentrations. The recommendations of this consensus meeting regarding the analytical methods include the following: encourage the development and promote the use of analytical methods displaying a lower limit of quantification (1 ng/mL), perform careful validation when implementing a new analytical assay, participate in external proficiency testing programs, promote the use of certified material as calibrators in high-performance liquid chromatography with mass spectrometric detection methods, and take account of the assay and intermethod bias when comparing clinical trial outcomes. It is also important to consider that TAC concentrations may also be influenced by other factors such as specific pharmacokinetic characteristics associated with the population, drug interactions, pharmacogenetics, adverse events that may alter TAC concentrations, and any change in the oral formulation that may result in pharmacokinetic changes. This meeting emphasized the importance of obtaining multicenter prospective trials to assess the efficacy of alternative strategies to TAC trough concentrations whether it is other single time points or area under the concentration-time curve Bayesian estimation using limited sampling strategies and to select, standardize, and validate routine biomarkers of TAC pharmacodynamics.


American Journal of Transplantation | 2002

Immunoprophylaxis with basiliximab compared with antithymocyte globulin in renal transplant patients receiving MMF-containing triple therapy.

Yvon Lebranchu; Frank Bridoux; M. Buchler; Yannick Le Meur; Isabelle Etienne; Olivier Toupance; Bruno Hurault de Ligny; Guy Touchard; Bruno Moulin; Patrick Le Pogamp; Olivier Reigneau; Michel Guignard; Gérard Rifle

Acute graft rejection remains a major problem in renal transplant recipients, and there is no consensus on the optimal immunosuppressive strategy. Immunoprophylaxis with Thymoglobulin® or basiliximab has significantly reduced the incidence of acute rejection episodes and graft loss following kidney transplantation. This open, randomized, multicenter study investigated the efficacy and tolerability of basiliximab (20u2003mg day 0– day 4) plus early cyclosporine from day 0 (nu2003=u200350) compared with Thymoglobulin® plus delayed cyclosporine (nu2003=u200350) in adult kidney transplant patients. In addition, all patients received steroids and mycophenolate mofetil (MMF) at standard doses from day 0. Patient and graft survival rates at 12u2003months were 98 and 94% in the basiliximab group, respectively, compared with 100 and 96% in the Thymoglobulin® group. The incidences of biopsy‐confirmed acute rejection (8.0% in each group) and treatment failure (14% in the basiliximab group vs. 8% in the Thymoglobulin group) were comparable in the two groups. There was a nonsignificant tendency to more dialysis (14 vs. 6%), and fewer cytomegalovirus (CMV) infections (p =u200a0.005) in the basiliximab group, but the percentage of clinical CMV was not different between the two groups (6 vs. 12%). Both strategies give excellent results, despite the differences in patterns, in nonhyperimmunized patients receiving their first cadaveric renal allograft.


Clinical Pharmacology & Therapeutics | 2004

CYP3A5 and MDR1 Genetic Polymorphisms and Cyclosporine Pharmacokinetics After Renal Transplantation

Dany Anglicheau; Eric Thervet; Isabelle Etienne; Bruno Hurault de Ligny; Yannick Le Meur; Guy Touchard; M. Buchler; Pierre Laurent-Puig; David Tregouet; Philippe Beaune; Ann K. Daly; Christophe Legendre; Pierre Marquet

The immunosuppressive drug cyclosporine (INN, ciclosporin), whose pharmacokinetic characteristics vary greatly among individuals, is a substrate for cytochrome P450 (CYP) 3A and P‐glycoprotein, the product of the multidrug resistance 1 (MDR1) gene. Some of the single nucleotide polymorphisms (SNPs) in these genes are associated with deficient protein expression and reduced in vivo activity. We postulated that, in renal transplant recipients, these SNPs could be associated with interindividual variations in cyclosporine pharmacokinetics.


Clinical Journal of The American Society of Nephrology | 2010

Consensus Report on Therapeutic Drug Monitoring of Mycophenolic Acid in Solid Organ Transplantation

Dirk Kuypers; Yannick Le Meur; Marcelo Cantarovich; Michael Tredger; Susan E. Tett; Dario Cattaneo; Burkhard Tönshoff; David W. Holt; Jeremy R. Chapman; Teun van Gelder

With the increasing use of mycophenolic acid (MPA) in solid organ transplantation, the need for more accurate drug dosing has become evident. Personalized immunosuppressive therapy requires better strategies for avoidance of drug-related toxicity while maintaining efficacy. Few studies have assessed the clinical usefulness of therapeutic drug monitoring (TDM) of MPA in solid organ transplantation in a prospective way, and they have produced opposing results. To provide clinicians with an objective and balanced clinical interpretation of the current scientific evidence on TDM of MPA, a consensus meeting involving 47 experts from around the world was commissioned by The Transplantation Society and held in Rome on November 20 to 21, 2008. The goal of this consensus meeting was to offer information to transplant practitioners on clinically relevant pharmacokinetic characteristics of MPA, to rationalize the basis for currently advised target exposure ranges for MPA in various types of organ transplantation, and to summarize available methods for application of MPA TDM in clinical practice. Although this consensus report does not evaluate the final role of MPA TDM in transplantation, it seeks to examine the current scientific evidence for concentration-controlled dosing of MPA.


Clinical Pharmacokinectics | 2009

Tacrolimus population pharmacokinetic-pharmacogenetic analysis and Bayesian estimation in renal transplant recipients.

Khaled Benkali; Aurélie Prémaud; Nicolas Picard; Jean-Philippe Rerolle; Olivier Toupance; Guillaume Hoizey; Alain Turcant; Florence Villemain; Yannick Le Meur; Pierre Marquet; Annick Rousseau

AbstractObjectives: The aims of this study were (i) to investigate the population pharmacokinetics of tacrolimus in renal transplant recipients, including the influence of biological and pharmacogenetic covariates; and (ii) to develop a Bayesian estimator able to reliably estimate the individual pharmacokinetic parameters and inter-dose area under the blood concentration-time curve (AUC) from 0 to 12 hours (AUC12) in renal transplant patients.n Methods: Full pharmacokinetic profiles were obtained from 32 renal transplant patients at weeks 1 and 2, and at months 1, 3 and 6 post-transplantation. The population pharmacokinetic analysis was performed using the nonlinear mixed-effect modelling software NONMEM® version VI. Patients’ genotypes were characterized by allelic discrimination for PXR −25385C>T genes.n Results: Tacrolimus pharmacokinetics were well described by a two-compartment model combined with an Erlang distribution to describe the absorption phase, with low additive and proportional residual errors of 1.6 ng/mL and 9%, respectively. Both the haematocrit and PXR −25385C>T single nucleotide polymorphism (SNP) were identified as significant covariates for apparent oral clearance (CL/F) of tacrolimus, which allowed improvement of prediction accuracy. Specifically, CL/F decreased gradually with the number of mutated alleles for the PXR −25385C>T SNP and was inversely proportional to the haematocrit value. However, clinical criteria of relevance, mainly the decrease in interindividual variability and residual error, led us to retain only the haematocrit in the final model. Maximum a posteriori Bayesian forecasting allowed accurate prediction of the tacrolimus AUC12 using only three sampling times (at 0 hour [predose] and at 1 and 3 hours postdose) in addition to the haematocrit value, with a nonsignificant mean AUC bias of 2% and good precision (relative mean square error = 11%).n Conclusion: Population pharmacokinetic analysis of tacrolimus in renal transplant recipients showed a significant influence of the haematocrit on its CL/F and led to the development of a Bayesian estimator compatible with clinical practice and able to accurately predict tacrolimus individual pharmacokinetic parameters and the AUC12.


Clinical Pharmacology & Therapeutics | 2006

CYP3A5*3 influences sirolimus oral clearance in de novo and stable renal transplant recipients.

Yannick Le Meur; Nassim Djebli; Jean-Christophe Szelag; Guillaume Hoizey; Olivier Toupance; Jean Philippe Rérolle; Pierre Marquet

The low and highly variable oral bioavailability of the immunosuppressant sirolimus is thought to result partly from genetic polymorphism of the CYP3A5 gene.


American Journal of Human Genetics | 2016

Mutations in GANAB, Encoding the Glucosidase IIα Subunit, Cause Autosomal-Dominant Polycystic Kidney and Liver Disease.

Binu Porath; Vladimir G. Gainullin; Emilie Cornec-Le Gall; Elizabeth K. Dillinger; Christina M. Heyer; Katharina Hopp; Marie E. Edwards; Charles D. Madsen; Sarah R. Mauritz; Carly J. Banks; Saurabh Baheti; Bharathi Reddy; José Ignacio Herrero; Jesus M. Banales; Marie C. Hogan; Velibor Tasic; Terry Watnick; Arlene B. Chapman; Cécile Vigneau; Frédéric Lavainne; Marie Pierre Audrezet; Claude Férec; Yannick Le Meur; Vicente E. Torres; Peter C. Harris

Autosomal-dominant polycystic kidney disease (ADPKD) is a common, progressive, adult-onset disease that is an important cause of end-stage renal disease (ESRD), which requires transplantation or dialysis. Mutations in PKD1 or PKD2 (∼85% and ∼15% of resolved cases, respectively) are the known causes of ADPKD. Extrarenal manifestations include an increased level of intracranial aneurysms and polycystic liver disease (PLD), which can be severe and associated with significant morbidity. Autosomal-dominant PLD (ADPLD) with no or very few renal cysts is a separate disorder caused by PRKCSH, SEC63, or LRP5 mutations. After screening, 7%-10% of ADPKD-affected and ∼50% of ADPLD-affected families were genetically unresolved (GUR), suggesting further genetic heterogeneity of both disorders. Whole-exome sequencing of six GUR ADPKD-affected families identified one with a missense mutation in GANAB, encoding glucosidase II subunit α (GIIα). Because PRKCSH encodes GIIβ, GANAB is a strong ADPKD and ADPLD candidate gene. Sanger screening of 321 additional GUR families identified eight further likely mutations (six truncating), and a total of 20 affected individuals were identified in seven ADPKD- and two ADPLD-affected families. The phenotype was mild PKD and variable, including severe, PLD. Analysis of GANAB-null cells showed an absolute requirement of GIIα for maturation and surface and ciliary localization of the ADPKD proteins (PC1 and PC2), and reduced mature PC1 was seen in GANAB(+/-) cells. PC1 surface localization in GANAB(-/-) cells was rescued by wild-type, but not mutant, GIIα. Overall, we show that GANAB mutations cause ADPKD and ADPLD and that the cystogenesis is most likely driven by defects in PC1 maturation.


Journal of Leukocyte Biology | 2002

Macrophage accumulation at a site of renal inflammation is dependent on the M-CSF/c-fms pathway

Yannick Le Meur; Gregory H Tesch; Prudence A. Hill; Wei Mu; Rita Foti; David J. Nikolic-Paterson; Robert C. Atkins

Production of macrophage‐colony stimulating factor (M‐CSF), the major macrophage growth factor, is increased in tissues during inflammation. Therefore, w determined whether M‐CSF, acting through its receptor c‐fms, contributes to macrophage accumulation at a site of tissue injury. Daily treatment with anti‐c‐fms or control antibody was given to mice with renal inflammation resulting from unilateral ureteric obstruction (UUO). Following UUO, kidney M‐CSF mRNA increased in association with macrophage accumulation (days 1, 5, and 10) and local macrophage proliferation (days 5 and 10). Anti‐c‐fms treatment caused a minor inhibition of monocyte recruitment at day 1, reduced macrophage accumulation by 75% at day 10, but did not affect blood monocyte counts or the CD4 and CD8 lymphocytic infiltrate. Prevention of macrophage accumulation by anti‐c‐fms treatment was associated with a 90% reduction in local macrophage proliferation at days 5 and 10 without evidence of increased macrophage apoptosis. Therefore, M‐CSF/c‐fms signaling plays a key role in macrophage accumulation during tissue injury.


Therapeutic Drug Monitoring | 2005

Maximum a posteriori bayesian estimation of mycophenolic acid pharmacokinetics in renal transplant recipients at different postgrafting periods.

Aurélie Prémaud; Yannick Le Meur; Jean Debord; Jean-Christophe Szelag; Annick Rousseau; Guillaume Hoizey; Olivier Toupance; Pierre Marquet

The aim of this study was to develop maximum a posteriori probability (MAP) Bayesian estimators of mycophenolic acid (MPA) pharmacokinetics (PK) capable of accurately estimating the MPA interdose AUC in renal transplant patients using a limited number of blood samples. The individual MPA plasma concentration-time profiles of 44 adult kidney transplant recipients were retrospectively studied: in 24 de novo transplant patients, 2 profiles were obtained on day 7 and day 30 after transplantation, and in 20 stable transplant patients, 1 profile was obtained in the stable period (>3 months). MPA was assayed by liquid chromatography-mass spectrometry. Concentration data were fitted using previously designed PK models, including 1 or 2Γ-distribution to describe the absorption rate. MAP-Bayesian estimations were performed using an in-house program. For each posttransplantation period, the limited sampling strategies (LSS) providing either the best determination coefficient or the lowest bias for AUC estimates with respect to trapezoidal AUCs were selected and compared with respect to the percentage of “clinically acceptable” AUC estimates (ie, within −20% to +20% of the true value) they yielded. A common LSS (blood samples collected at T20min, T1h, and T3h postdosing), convenient for all 3 periods, was also selected and validated: bias (RMSE%) values were −5.7% (20.5%), −8.2% (14.4%), and +0.4% (12.0%) on D7, D30, and for >M3 with respect to the reference values obtained using the trapezoidal rule, respectively. For the first time, MAP-Bayesian estimators of MPA systemic exposure at different posttransplantation periods (early as well as later periods) could be designed. They have since been used for MPA dose adaptation in concentration-controlled studies as well as for MPA therapeutic drug monitoring in clinical practice.


American Journal of Transplantation | 2003

Blockade of Macrophage Colony-Stimulating Factor Reduces Macrophage Proliferation and Accumulation in Renal Allograft Rejection

Matthew D. Jose; Yannick Le Meur; Robert C. Atkins; Steven J. Chadban

Macrophage accumulation within an acutely rejecting allograft occurs by recruitment and local proliferation. To determine the importance of M‐CSF in driving macrophage proliferation during acute rejection, we blocked the M‐CSF receptor, c‐fms, in a mouse model of acute renal allograft rejection. C57BL/6 mouse kidneys (allografts, nu2003=u200320) or BALB/c kidneys (isografts, nu2003=u20035) were transplanted into BALB/c mice. Anti‐c‐fms antibody (AFS98) or control Ig (50u2003mg/kg/day, i.p.) was given daily to allografts from days 0–5. All mice were killed day 6 postoperatively. Expression of the M‐CSF receptor, c‐fms, was restricted to infiltrating CD68+ macrophages. Blockade of c‐fms reduced proliferating (CD68+/BrdU+) macrophages by 82% (1.1 v 6.2%, pu2003<u20030.001), interstitial CD68+ macrophage accumulation by 53% (595 v 1270/mm2, pu2003<u20030.001), and glomerular CD68+ macrophage accumulation by 71% (0.73u2003V 2.48u2003CD68+ cells per glomerulus, pu2003<u20030.001). Parameters of T‐cell involvement (intragraft CD4+, CD8+ and CD25+ lymphocyte numbers) were not affected. The severity of tubulointerstitial rejection was reduced in the treatment group as shown by decreased tubulitis and tubular cell proliferation. Macrophage proliferation during acute allograft rejection is dependent on the interaction of M‐CSF with its receptor c‐fms. This pathway plays a significant and specific role in the accumulation of macrophages within a rejecting renal allograft.

Collaboration


Dive into the Yannick Le Meur's collaboration.

Top Co-Authors

Avatar

Olivier Toupance

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

Yvon Lebranchu

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

M. Buchler

François Rabelais University

View shared research outputs
Researchain Logo
Decentralizing Knowledge