Network


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

Hotspot


Dive into the research topics where Valérie Gouilleux-Gruart is active.

Publication


Featured researches published by Valérie Gouilleux-Gruart.


Clinical Cancer Research | 2011

Cetuximab Pharmacokinetics Influences Progression-Free Survival of Metastatic Colorectal Cancer Patients

Nicolas Azzopardi; Thierry Lecomte; David Ternant; Michelle Boisdron-Celle; Friedrich Piller; Alain Morel; Valérie Gouilleux-Gruart; Céline Vignault-Desvignes; Hervé Watier; Erick Gamelin; Gilles Paintaud

Purpose: An ancillary phase II study was conducted to study interindividual variability in cetuximab pharmacokinetics and its influence on progression-free survival (PFS) in metastatic colorectal cancer patients cotreated with irinotecan and 5-fluorouracil. Experimental Design: Ninety-six patients received cetuximab as an infusion loading dose of 400 mg/m2 followed by weekly infusions of 250 mg/m2. Doses of irinotecan and 5-fluorouracil were adjusted individually. Cetuximab concentrations were measured by ELISA. Compartmental pharmacokinetic parameters were estimated by a population approach, and PFS was analyzed using a Cox model. Results: Cetuximab pharmacokinetics was best described using a two-compartment model with both first-order and saturable (zero-order) elimination. Estimated pharmacokinetic parameters (% standard error) were as follows: central volume of distribution V1 = 2.96 L (4%), peripheral volume of distribution V2 = 4.65 L (6%), elimination clearance CL = 0.497 L/d (4%), distribution clearance Q = 0.836 L/d (8%), and zero-order elimination rate k0 = 8.71 mg/d (10%). Body surface area influenced V1, V2, and k0. Pretreatment serum albumin influenced CL. Risk of disease progression decreased with cetuximab global clearance (cumulative dose/cumulative area under the concentration versus time curve; P = 0.00016). Median PFS of patients with a cetuximab residual concentration on day 14 below median value was 3.3 months as compared with 7.8 months for the other patients (P = 0.004). Conclusions: Cetuximab pharmacokinetics in colorectal cancer patients can be described using a model combining linear and nonlinear elimination rates. PFS is influenced by global clearance of cetuximab, a parameter that can be estimated using cetuximab residual concentration on day 14. Clin Cancer Res; 17(19); 6329–37. ©2011 AACR.


Journal of Controlled Release | 2014

Fate of inhaled monoclonal antibodies after the deposition of aerosolized particles in the respiratory system.

Laurent Guilleminault; Nicolas Azzopardi; Christophe Arnoult; Julien Sobilo; V. Hervé; Jérôme Montharu; A. Guillon; C. Andres; O. Herault; A. Le Pape; P. Diot; Etienne Lemarié; Gilles Paintaud; Valérie Gouilleux-Gruart; Nathalie Heuzé-Vourc'h

Monoclonal antibodies (mAbs) are usually delivered systemically, but only a small proportion of the drug reaches the lung after intravenous injection. The inhalation route is an attractive alternative for the local delivery of mAbs to treat lung diseases, potentially improving tissue concentration and exposure to the drug while limiting passage into the bloodstream and adverse effects. Several studies have shown that the delivery of mAbs or mAb-derived biopharmaceuticals via the airways is feasible and efficient, but little is known about the fate of inhaled mAbs after the deposition of aerosolized particles in the respiratory system. We used cetuximab, an anti-EGFR antibody, as our study model and showed that, after its delivery via the airways, this mAb accumulated rapidly in normal and cancerous tissues in the lung, at concentrations twice those achieved after intravenous delivery, for early time points. The spatial distribution of cetuximab within the tumor was heterogeneous, as reported after i.v. injection. Pharmacokinetic (PK) analyses were carried out in both mice and macaques and showed aerosolized cetuximab bioavailability to be lower and elimination times shorter in macaques than in mice. Using transgenic mice, we showed that FcRn, a key receptor involved in mAb distribution and PK, was likely to make a greater contribution to cetuximab recycling than to the transcytosis of this mAb in the airways. Our results indicate that the inhalation route is potentially useful for the treatment of both acute and chronic lung diseases, to boost and ensure the sustained accumulation of mAbs within the lungs, while limiting their passage into the bloodstream.


Blood | 2012

Polymorphisms of protein tyrosine phosphatase CD148 influence FcγRIIA-dependent platelet activation and the risk of heparin-induced thrombocytopenia

Jérôme Rollin; Claire Pouplard; Marie-Pierre Gratacap; Dorothee Leroux; Marc-Antoine May; Michel Aupart; Valérie Gouilleux-Gruart; Bernard Payrastre; Yves Gruel

Heparin-induced thrombocytopenia (HIT) is due primarily to IgG antibodies specific to platelet factor 4/heparin complexes (PF4/Hs) that activate platelets via FcγRIIA. CD148 is a protein tyrosine phosphatase that regulates Src kinases and collagen-induced platelet activation. Three polymorphisms affecting CD148 (Q276P, R326Q, and D872E) were studied in HIT patients and 2 control groups, with or without antibodies to PF4/Hs. Heterozygote status for CD148 276P or 326Q alleles was less frequent in HIT patients, suggesting a protective effect of these polymorphisms. Aggregation tests performed with collagen, HIT plasma, and monoclonal antibodies cross-linking FcγRIIA showed consistent hyporesponsiveness of platelets expressing the 276P/326Q alleles. In addition, platelets expressing the 276P/326Q alleles exhibited a greater sensitivity to the Src family kinases inhibitor dasatinib in response to collagen or ALB6 cross-linking FcγRIIA receptors. Moreover, the activatory phosphorylation of Src family kinases was considerably delayed as well as the phosphorylation of Linker for activation of T cells and phospholipase Cγ2, 2 major signaling proteins downstream from FcγRIIA. In conclusion, this study shows that CD148 polymorphisms affect platelet activation and probably exert a protective effect on the risk of HIT in patients with antibodies to PF4/Hs.


Clinical Pharmacokinectics | 2015

Assessment of the Influence of Inflammation and FCGR3A Genotype on Infliximab Pharmacokinetics and Time to Relapse in Patients with Crohn's Disease.

David Ternant; Zahir Berkane; Laurence Picon; Valérie Gouilleux-Gruart; Jean-Frederic Colombel; Matthieu Allez; Edouard Louis; Gilles Paintaud

AbstractBackground and ObjectivesInfliximab is a monoclonal anti-tumor necrosis factor-α (anti-TNFα) antibody that profoundly modified the treatment of Crohn’s disease (CD). The polymorphism of Fc fragment of IgG, low affinity IIIa, receptor (CD16a) [FCGR3A]n influences the biological response to infliximab in patients with CD. Our aim was to study its influence on infliximab pharmacokinetics and risk of relapse after infliximab discontinuation.MethodsIn 111 CD patients in remission, infliximab was discontinued and its concentrations were measured for 30xa0months or until relapse. Infliximab pharmacokinetics were described using monocompartmental population modeling.ResultsThe elimination rate of infliximab increased with C-reactive protein (CRP) [pxa0=xa00.00018] and was 16xa0% higher in FCGR3A-158V/V patients than in F carriers (pxa0=xa00.0028). Risk of relapse was higher in patients with baseline CRPxa0≥5xa0mg/L than in those with a lower value (pxa0=xa00.0000029). In addition, there was a first-order interaction between CRP and the FCGR3A genotype; in patients with high CRP, risk of relapse was higher for V/V patients than for F carriers (hazard ratio 4.80 and 2.84 for V/V and F carriers, respectively; pxa0=xa00.013).ConclusionBoth increased inflammation and FCGR3A-158V/V genotype are associated with increased infliximab elimination and risk of relapse after infliximab discontinuation in patients with CD.


mAbs | 2015

Therapeutic drug monitoring of eculizumab: Rationale for an individualized dosing schedule.

Philippe Gatault; Guillaume Brachet; David Ternant; Danielle Degenne; Guillaume Récipon; Christelle Barbet; Emmanuel Gyan; Valérie Gouilleux-Gruart; Cécile Bordes; Alexandra Farrell; Jean Michel Halimi; Hervé Watier

The annual cost of eculizumab maintenance therapy in paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic–uremic syndrome (aHUS) exceeds


Journal of Immunology | 2014

ADAM17-Mediated Shedding of FcγRIIIA on Human NK Cells: Identification of the Cleavage Site and Relationship with Activation

Laurie Lajoie; Nicolas Congy-Jolivet; Armelle Bolzec; Valérie Gouilleux-Gruart; Elodie Sicard; Hsueh Cheng Sung; Frank Peiretti; Thierry Moreau; Henri Vié; Béatrice Clémenceau; Gilles Thibault

300,000 per patient. A better understanding of eculizumab pharmacokinetics and subsequent individual dose adjustment could reduce this cost. We measured the trough eculizumab concentration in 9 patients with maintenance therapy (aHUS, n = 7; PNH, n = 2) and determined: 1) the intra- and inter-individual variability; 2) the influence of weight on eculizumab pharmacokinetics; and 3) the rate of elimination of eculizumab following discontinuation. A one-compartment model was developed to describe the pharmacokinetics of eculizumab and predicted complement activity by body weight. Trough eculizumab concentrations were >50 µg/mL in 9/9, >100 µg/mL in 8/9, and >300 µg/mL in 5/9 of patients. Intra-individual variability was low but eculizumab concentrations, closely correlated with patient weight (R2 = 0.66, p = 0.034), varied broadly (55 ± 12 to 733 ± 164 µg/mL). Pharmacokinetic modeling showed that the elimination half-life varied greatly, with an increase from 7.8 d in a patient weighing 100 kg to 19.5 d in a 40 kg patient. We predicted that infusions of 1200 mg could be spaced every 4 or 6 weeks in patients weighing <90 and <70 kg, respectively. In this pilot study, the current recommended use of a fixed eculizumab dose for maintenance therapy is associated with excessively high trough concentrations in many patients. Further prospective larger studies are now required to support an individualized schedule adjusted for patient weight and based on the observed trough serum eculizumab concentration.


JAK-STAT | 2013

Oxidative metabolism in cancer: A STAT affair?

Jérome Bourgeais; Valérie Gouilleux-Gruart; Fabrice Gouilleux

FcγRIIIA/CD16A, the low-affinity receptor for the IgG Fc portion expressed on human CD56dim NK cells and involved in Ab-dependent cell cytotoxicity, is shed upon NK cell activation. We found that recombinant a disintegrin and metalloprotease (ADAM) 17 cleaved the ectodomain of FcγRIIIA/CD16A and a peptide for which the sequence encompasses aa 191–201 of the FcγRIIIA/CD16A stalk region but not ADAM10. MALDI-TOF analysis revealed that the peptide was cleaved between Ala195 and Val196 (i.e., 1 aa upstream of the expected position). This location of the cleavage site was confirmed by the finding that ADAM17 failed to cleave a peptide in which Ala and Val were reversed. ADAM17 was found to be expressed on NK cells, and stimulation with PMA or N-ethyl-maleimide resulted in the shedding of FcγRIIIA/CD16A and CD62L, a specific substrate of ADAM17. Selective inhibition of ADAM17 prevented the shedding of both molecules. Moreover, the shedding of FcγRIIIA/CD16A was strongly correlated with degranulation when a wide range of CD56dim NK cell activating receptors were stimulated, whereas both ADAM17-dependent shedding and internalization were involved in FcγRIIIA/CD16A downmodulation when the latter was engaged. Finally, the shedding of FcγRIIIA/CD16A was restricted to activated cells, suggesting that ADAM17 acts mainly, if not exclusively, in cis. Taken together, our results demonstrated for the first time, to our knowledge, at the molecular level that ADAM17 cleaves the stalk region of FcγRIIIA/CD16A and identified its cleavage site. The shedding of FcγRIIIA/CD16A was at least partially ADAM17 dependent, and it may be considered as a marker of FcγRIIIA/CD16A-independent NK cell activation highly correlated with degranulation.


Biochimica et Biophysica Acta | 2011

Regulation of IGF-1-dependent cyclin D1 and E expression by hEag1 channels in MCF-7 cells: the critical role of hEag1 channels in G1 phase progression.

Anne-Sophie Borowiec; Frédéric Hague; Valérie Gouilleux-Gruart; Kaiss Lassoued; Halima Ouadid-Ahidouch

STAT3 and STAT5 (STAT3/5) proteins are crucial mediators of cytokine- or growth factor-induced cell survival and proliferation. These transcription factors are frequently overactivated in a variety of solid tumors and hematopoietic neoplasms and are targets of various oncogenes with tyrosine kinase activity. STAT3/5 proteins regulate expression of genes involved in survival and proliferation in the nucleus and interact with signaling pathways in the cytoplasm. Evidences for a cross-talk between STAT3/5 and oxidative metabolism have recently emerged. This review summarizes the current knowledge on the cross-regulation between STAT3/5 and oxidative metabolism in normal and cancer cells.


Science Translational Medicine | 2015

Regulation of immune responses to protein therapeutics by transplacental induction of T cell tolerance

Nimesh Gupta; Slobodan Culina; Yann Meslier; Jordan D. Dimitrov; Christophe Arnoult; Sandrine Delignat; Bagirath Gangadharan; Maxime Lecerf; Sune Justesen; Valérie Gouilleux-Gruart; Benoît L. Salomon; David W. Scott; Srinivas V. Kaveri; Roberto Mallone; Sébastien Lacroix-Desmazes

Insulin-like Growth Factor-1 (IGF-1) plays a key role in breast cancer development and cell cycle regulation. It has been demonstrated that IGF-1 stimulates cyclin expression, thus regulating the G1 to S phase transition of the cell cycle. Potassium (K(+)) channels are involved in the G1 phase progression of the cell cycle induced by growth factors. However, mechanisms that allow growth factors to cooperate with K(+) channels in order to modulate the G1 phase progression and cyclin expression remain unknown. Here, we focused on hEag1 K(+) channels which are over-expressed in breast cancer and are involved in the G1 phase progression of breast cancer cells (MCF-7). As expected, IGF-1 increased cyclin D1 and E expression of MCF-7 cells in a cyclic manner, whereas the increase of CDK4 and 2 levels was sustained. IGF-1 stimulated p21(WAF1/Cip1) expression with a kinetic similar to that of cyclin D1, however p27(Kip1) expression was insensitive to IGF-1. Interestingly, astemizole, a blocker of hEag1 channels, but not E4031, a blocker of HERG channels, inhibited the expression of both cyclins after 6-8h of co-stimulation with IGF-1. However, astemizole failed to modulate CDK4, CDK2, p21(WAF1/Cip1) and p27(Kip1) expression. The down-regulation of hEag1 by siRNA provoked a decrease in cyclin expression. This study is the first to demonstrate that K(+) channels such as hEag1 are directly involved in the IGF-1-induced up-regulation of cyclin D1 and E expression in MCF-7 cells. By identifying more specifically the temporal position of the arrest site induced by the inhibition of hEag1 channels, we confirmed that hEag1 activity is predominantly upstream of the arrest site induced by serum-deprivation, prior to the up-regulation of both cyclins D1 and E. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.


Journal of Immunology | 2016

IgG1 Allotypes Influence the Pharmacokinetics of Therapeutic Monoclonal Antibodies through FcRn Binding

David Ternant; Christophe Arnoult; Martine Pugnière; Christine Dhommée; Daniel Drocourt; Eric Perouzel; Christophe Passot; Nadine Baroukh; Denis Mulleman; Gérard Tiraby; Hervé Watier; Gilles Paintaud; Valérie Gouilleux-Gruart

Transplacental modulation of T cell repertoire during immune ontogeny leads to antigen-specific immune tolerance to an immunogenic protein therapeutic. Can’t we all get along? The past century brought great advances for the treatment of some genetic disorders caused by single protein deficiencies. In particular, hemophilia A, which is caused by a deficiency of coagulation factor VIII, can now be treated by replacing the missing factor. Unfortunately, the most severely affected patients, who completely lack endogenous factor VIII, often develop antibodies against the replacement protein because their bodies recognize it as a foreign substance. To overcome this problem, Gupta et al. have now developed a method for transplacental exposure of fetuses to the missing antigen, thus ensuring immune tolerance for protein therapy that will be delivered after birth. Central tolerance plays a key role in modulating immune responses to self and exogenous antigens. The absence of self-antigen expression, as in patients with genetic deficiencies, prevents the development of antigen-specific immune tolerance. Hence, a substantial number of patients develop neutralizing antibodies to the corresponding protein therapeutics after replacement treatment. In this context, the administration of missing antigens during fetal development, a key period for self-tolerance establishment, should confer early and long-lasting antigen-specific tolerance. To this end, we exploited the physiological pathway of the neonatal Fc receptor (FcRn) through which maternal immunoglobulins are transplacentally transferred to fetuses. We demonstrate that Fc-fused antigens administered to pregnant mice reach fetal lymphoid organs in an FcRn-dependent manner, accumulate in antigen-presenting cells of myeloid origin, and promote the generation of both thymic and peripheral antigen-specific regulatory T cells. This strategy was successfully pursued in a mouse model of hemophilia A, where maternofetal transfer of the Fc-fused immunodominant domains of coagulation factor VIII conferred antigen-specific tolerance. Transplacental tolerance induction with Fc-fused proteins may thus prove valuable to prevent alloimmunization after replacement protein therapy for congenital deficiencies.

Collaboration


Dive into the Valérie Gouilleux-Gruart's collaboration.

Top Co-Authors

Avatar

Hervé Watier

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

Gilles Paintaud

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

David Ternant

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

Christophe Arnoult

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar

Nicolas Azzopardi

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

Christine Dhommée

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

Christophe Passot

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

Guillaume Brachet

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

Thierry Lecomte

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

Jérôme Rollin

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge