Network


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

Hotspot


Dive into the research topics where Nicolas Tournier is active.

Publication


Featured researches published by Nicolas Tournier.


The International Journal of Neuropsychopharmacology | 2010

Interaction of drugs of abuse and maintenance treatments with human P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2)

Nicolas Tournier; Lucie Chevillard; Bruno Mégarbane; Stéphane Pirnay; Jean-Michel Scherrmann; Xavier Declèves

Drug interaction with P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) may influence its tissue disposition including blood-brain barrier transport and result in potent drug-drug interactions. The limited data obtained using in-vitro models indicate that methadone, buprenorphine, and cannabinoids may interact with human P-gp; but almost nothing is known about drugs of abuse and BCRP. We used in vitro P-gp and BCRP inhibition flow cytometric assays with hMDR1- and hBCRP-transfected HEK293 cells to test 14 compounds or metabolites frequently involved in addiction, including buprenorphine, norbuprenorphine, methadone, ibogaine, cocaine, cocaethylene, amphetamine, N-methyl-3,4-methylenedioxyamphetamine, 3,4-methylenedioxyamphetamine, nicotine, ketamine, Delta9-tetrahydrocannabinol (THC), naloxone, and morphine. Drugs that in vitro inhibited P-gp or BCRP were tested in hMDR1- and hBCRP-MDCKII bidirectional transport studies. Human P-gp was significantly inhibited in a concentration-dependent manner by norbuprenorphine>buprenorphine>methadone>ibogaine and THC. Similarly, BCRP was inhibited by buprenorphine>norbuprenorphine>ibogaine and THC. None of the other tested compounds inhibited either transporter, even at high concentration (100 microm). Norbuprenorphine (transport efflux ratio approoximately 11) and methadone (transport efflux ratio approoximately 1.9) transport was P-gp-mediated; however, with no significant stereo-selectivity regarding methadone enantiomers. BCRP did not transport any of the tested compounds. However, the clinical significance of the interaction of norbuprenorphine with P-gp remains to be evaluated.


Current Pharmaceutical Design | 2011

Opioid Transport by ATP-Binding Cassette Transporters at the Blood-Brain Barrier: Implications for Neuropsychopharmacology

Nicolas Tournier; Xavier Declèves; Bruno Saubaméa; Jean-Michel Scherrmann; Salvatore Cisternino

Some of the ATP-binding cassette (ABC) transporters like P-glycoprotein (P-gp; ABCB1, MDR1), BCRP (ABCG2) and MRPs (ABCCs) that are present at the blood-brain barrier (BBB) influence the brain pharmacokinetics (PK) of their substrates by restricting their uptake or enhancing their clearance from the brain into the blood, which has consequences for their CNS pharmacodynamics (PD). Opioid drugs have been invaluable tools for understanding the PK-PD relationships of these ABC-transporters. The effects of morphine, methadone and loperamide on the CNS are modulated by P-gp. This review examines the ways in which other opioid drugs and some of their active metabolites interact with ABC transporters and suggests new mechanisms that may be involved in the variability of the response of the CNS to these drugs like carrier-mediated system belonging to the solute carrier (SLC) superfamily. Exposure to opioids may also alter the expression of ABC transporters. P-gp can be overproduced during morphine treatment, suggesting that the drug has a direct or, more likely, an indirect action. Variations in cerebral neurotransmitters during exposure to opioids and the release of cytokines during pain could be new endogenous stimuli affecting transporter synthesis. This review concludes with an analysis of the pharmacotherapeutic and clinical impacts of the interactions between ABC transporters and opioids.


Antimicrobial Agents and Chemotherapy | 2009

Preparation and Stability of Voriconazole Eye Drop Solution

Antoine Dupuis; Nicolas Tournier; Gwenael Le Moal; Nicolas Venisse

ABSTRACT The combined systemic and topical administration of voriconazole has successfully been used to treat keratomycosis. Because no voriconazole eye drop product is commercially available, we prepared a sterile eye drop solution (10 mg/ml). Voriconazole remains stable over 30 days, providing an eye drop solution suitable for use for the topical treatment of fungal keratitis.


The Journal of Nuclear Medicine | 2011

Transport of Selected PET Radiotracers by Human P-Glycoprotein (ABCB1) and Breast Cancer Resistance Protein (ABCG2): An In Vitro Screening

Nicolas Tournier; Héric Valette; Marie-Anne Peyronneau; Wadad Saba; Sébastien Goutal; Bertrand Kuhnast; Frédéric Dollé; Jean-Michel Scherrmann; Salvatore Cisternino; Michel Bottlaender

Radiolabeled compounds used for brain imaging with PET must readily cross the blood–brain barrier (BBB) to reach their target. Efflux transporters at the BBB—P-glycoprotein (P-gp) and the breast cancer resistance protein (BCRP)—could limit their uptake by the brain. Methods: We developed and validated an in vitro model using MDCKII cells transfected with human multidrug resistance (MDR1) or BCRP genes and assessed the transport of selected PET ligands by the concentration equilibrium technique. The tested compounds included befloxatone, (R,S)-CGP-12177, clorgyline, R-(−)-deprenyl, diprenorphine, DPA-714, fallypride, flumazenil, 2-fluoro-A-85380, LBT-999, loperamide, p-MPPF, PE2I, Pittsburgh compound B (PIB), (R,S)-PK11195, raclopride, R-(+)-verapamil, and WAY-100635. The assays were performed using the nonradioactive form of each compound (ultraviolet high-performance liquid chromatography analysis) and, when available, the 18F-labeled analogs (γ-counting). Results: Befloxatone appeared to be transported solely by BCRP. Loperamide, verapamil, and diprenorphine were the only P-gp substrates. Other ligands were transported by neither P-gp nor BCRP. Conclusion: The present method can readily be used to screen new-compound transport by P-gp or BCRP, even before any radiolabeling. Compounds that were previously thought to be transported by P-gp in rodents, such as p-MPPF, WAY-100635, and flumazenil, cannot be considered substrates of human P-gp. The impact of BCRP and P-gp at the BBB on the transport of befloxatone and diprenorphine in vivo remains to be evaluated with PET.


Critical Care Medicine | 2012

Respiratory toxicity of buprenorphine results from the blockage of P-glycoprotein-mediated efflux of norbuprenorphine at the blood-brain barrier in mice.

Hisham Alhaddad; Salvatore Cisternino; Xavier Declèves; Nicolas Tournier; Joël Schlatter; Fouad Chiadmi; Patricia Risède; Maria Smirnova; Capucine Besengez; Jean-Michel Scherrmann; Frédéric J. Baud; Bruno Mégarbane

Objectives:Deaths due to asphyxia as well as following acute poisoning with severe respiratory depression have been attributed to buprenorphine in opioid abusers. However, in human and animal studies, buprenorphine exhibited ceiling respiratory effects, whereas its metabolite, norbuprenorphine, was assessed as being a potent respiratory depressor in rodents. Recently, norbuprenorphine, in contrast to buprenorphine, was shown in vitro to be a substrate of human P-glycoprotein, a drug-transporter involved in all steps of pharmacokinetics including transport at the blood–brain barrier. Our objectives were to assess P-glycoprotein involvement in norbuprenorphine transport in vivo and study its role in the modulation of buprenorphine-related respiratory effects in mice. Setting:University-affiliated research laboratory, INSERM U705, Paris, France. Subjects:Wild-type and P-glycoprotein knockout female Friend virus B-type mice. Interventions:Respiratory effects were studied using plethysmography and the P-glycoprotein role at the blood–brain barrier using in situ brain perfusion. Measurements and Main Results:Norbuprenorphine(≥1 mg/kg) and to a lesser extent buprenorphine (≥10 mg/kg) were responsible for dose-dependent respiratory depression combining increased inspiratory (TI) and expiratory times (TE). PSC833, a powerful P-glycoprotein inhibitor, significantly enhanced buprenorphine-related effects on TI (p < .01) and TE (p < .05) and norbuprenorphine-related effects on minute volume (VE, p < .05), TI, and TE (p < .001). In P-glycoprotein-knockout mice, buprenorphine-related effects on VE (p < .01), TE (p < .001), and TI (p < .05) and norbuprenorphine-related effects on VE (p < .05) and TI (p < .001) were significantly enhanced. Plasma norbuprenorphine concentrations were significantly increased in PSC833-treated mice (p < .001), supporting a P-glycoprotein role in norbuprenorphine pharmacokinetics. Brain norbuprenorphine efflux was significantly reduced in PSC833-treated and P-glycoprotein-knockout mice (p < .001), supporting P-glycoprotein-mediated norbuprenorphine transport at the blood–brain barrier. Conclusions:P-glycoprotein plays a key-protective role in buprenorphine-related respiratory effects, by allowing norbuprenorphine efflux at the blood–brain barrier. Our findings suggest a major role for drug–drug interactions that lead to P-glycoprotein inhibition in buprenorphine-associated fatalities and respiratory depression.


Aaps Journal | 2013

Effects of Selected OATP and/or ABC Transporter Inhibitors on the Brain and Whole-Body Distribution of Glyburide

Nicolas Tournier; Wadad Saba; Salvatore Cisternino; Marie-Anne Peyronneau; Annelaure Damont; Sébastien Goutal; Albertine Dubois; Frédéric Dollé; Jean-Michel Scherrmann; Héric Valette; Bertrand Kuhnast; Michel Bottlaender

Glyburide (glibenclamide, GLB) is a widely prescribed antidiabetic with potential beneficial effects in central nervous system injury and diseases. In vitro studies show that GLB is a substrate of organic anion transporting polypeptide (OATP) and ATP-binding cassette (ABC) transporter families, which may influence GLB distribution and pharmacokinetics in vivo. In the present study, we used [11C]GLB positron emission tomography (PET) imaging to non-invasively observe the distribution of GLB at a non-saturating tracer dose in baboons. The role of OATP and P-glycoprotein (P-gp) in [11C]GLB whole-body distribution, plasma kinetics, and metabolism was assessed using the OATP inhibitor rifampicin and the dual OATP/P-gp inhibitor cyclosporine. Finally, we used in situ brain perfusion in mice to pinpoint the effect of ABC transporters on GLB transport at the blood–brain barrier (BBB). PET revealed the critical role of OATP on liver [11C]GLB uptake and its subsequent impact on [11C]GLB metabolism and plasma clearance. OATP-mediated uptake also occurred in the myocardium and kidney parenchyma but not the brain. The inhibition of P-gp in addition to OATP did not further influence [11C]GLB tissue and plasma kinetics. At the BBB, the inhibition of both P-gp and breast cancer resistance protein (BCRP) was necessary to demonstrate the role of ABC transporters in limiting GLB brain uptake. This study demonstrates that GLB distribution, metabolism, and elimination are greatly dependent on OATP activity, the first step in GLB hepatic clearance. Conversely, P-gp, BCRP, and probably multidrug resistance protein 4 work in synergy to limit GLB brain uptake.


British Journal of Pharmacology | 2016

Blood-brain and retinal barriers show dissimilar ABC transporter impacts and concealed effect of P-glycoprotein on a novel verapamil influx carrier.

Hélène Chapy; Bruno Saubaméa; Nicolas Tournier; Fanchon Bourasset; Francine Behar-Cohen; Xavier Declèves; Jean-Michel Scherrmann; Salvatore Cisternino

The respective impact and interplay between ABC (P‐glycoprotein/P‐gp/Abcb1a, BCRP/ABCG2, MRP/ABCC) and SLC transporter functions at the blood–brain barrier (BBB) and blood–retinal barriers (BRB) are incompletely understood.


Journal of Neurochemistry | 2009

Changes in dipole membrane potential at the mouse blood–brain barrier enhance the transport of 99mTechnetium Sestamibi more than inhibiting Abcb1, Abcc1, or Abcg2

Julie Cattelotte; Nicolas Tournier; Nathalie Rizzo-Padoin; Alfred H. Schinkel; Jean-Michel Scherrmann; Salvatore Cisternino

Cationic 99mTc‐agents like 99mTc‐hexakis‐2‐methoxyisobutyl isonitrile (99mTc‐MIBI) cannot be used for brain imaging because they do not enter the brain as readily as some uncharged 99mTc‐compounds. The mechanism by which cationic 99mTc‐agents are transported across the blood–brain barrier (BBB) remains unclear. We explored 99mTc‐MIBI transport by in situ mouse brain perfusion to determine the influence of BBB features like the ATP‐binding cassette transporters (Abcb1/P‐glycoprotein (P‐gp), Abcc1/Mrp1, and Abcg2/Bcrp), organic cation transporters (Slc22a1‐3/Oct1‐3), the transmembrane potential and the dipole membrane potential. P‐gp reduced 99mTc‐MIBI transport across the BBB of P‐gp‐deficient mice 2.2‐fold, as confirmed by PSC833 and GF120918 inhibition. Paradoxically verapamil decreased its transport ‘0.6‐fold’. Reducing the BBB dipole membrane potential with tetraphenylborate or phloretin increased 99mTc‐MIBI transport about 12‐ and 20‐fold, respectively. Guanidine, diphenhydramine, and carnitine significantly decreased 99mTc‐MIBI transport, but tetraethylammonium did not. 99mTc‐MIBI transport at the BBB is restricted by P‐gp but not by Mrp1 or Bcrp. Some organic cations reduced the influx of 99mTc‐MIBI into the brain independently of Oct1, 2 and 3, but this could be due to their effect on another cation transporter. The membrane dipole potential of the luminal BBB membrane appeared to be the main factor restricting 99mTc‐MIBI permeability.


Pharmaceutical Research | 2012

Discrepancies in the P-glycoprotein-Mediated Transport of 18F-MPPF: A Pharmacokinetic Study in Mice and Non-human Primates

Nicolas Tournier; Salvatore Cisternino; Marie-Anne Peyronneau; Sébastien Goutal; Frédéric Dollé; Jean-Michel Scherrmann; Michel Bottlaender; Wadad Saba; Héric Valette

PurposeSeveral in vivo studies have found that the 5-HT1A PET radioligand 18F-MPPF is a substrate of rodent P-glycoprotein (P-gp). However, in vitro assays suggest that MPPF is not a substrate of human P-gp. We have now tested the influence of inhibiting P-gp on the brain kinetics of 18F-MPPF in mice and non-human primates.MethodsWe measured the peripheral kinetics (arterial input function, metabolism, free fraction in plasma (fP)) during 18F-MPPF brain PET scanning in baboons with or without cyclosporine A (CsA) infusion. We measured 3H-MPPF transport at the mouse BBB using in situ brain perfusion in P-gp/Bcrp deficient mice and after inhibiting P-gp with PSC833.ResultsThere was an unexpected 1.9-fold increase in brain area under the curve in CsA-treated baboons (n = 4), with no change in radiometabolite-corrected arterial input. However, total volume of distribution corrected for fP (VT/fP) remained unchanged. In situ brain perfusion showed that P-gp restricted the permeability of the mouse BBB to 3H-MPPF while Bcrp did not.ConclusionThese and previous in vitro results suggest that P-gp may not influence the permeability of human BBB to 18F-MPPF. However, CsA treatment increased 18F-MPPF free fraction, which is responsible for a misleading, P-gp unrelated enhanced brain uptake.


The Journal of Nuclear Medicine | 2017

Strategies to Inhibit ABCB1- and ABCG2-Mediated Efflux Transport of Erlotinib at the Blood–Brain Barrier: A PET Study on Nonhuman Primates

Nicolas Tournier; Sébastien Goutal; Sylvain Auvity; Alexander Traxl; Severin Mairinger; Thomas Wanek; Ourkia-Badia Helal; Irène Buvat; Michaël Soussan; Fabien Caillé; Oliver Langer

The tyrosine kinase inhibitor erlotinib poorly penetrates the blood–brain barrier (BBB) because of efflux transport by P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2), thereby limiting its utility in the treatment of non–small cell lung cancer metastases in the brain. Pharmacologic strategies to inhibit ABCB1/ABCG2-mediated efflux transport at the BBB have been successfully developed in rodents, but it remains unclear whether these can be translated to humans given the pronounced species differences in ABCG2/ABCB1 expression ratios at the BBB. We assessed the efficacy of two different ABCB1/ABCG2 inhibitors to enhance brain distribution of 11C-erlotinib in nonhuman primates as a model of the human BBB. Methods: Papio anubis baboons underwent PET scans of the brain after intravenous injection of 11C-erlotinib under baseline conditions (n = 4) and during intravenous infusion of high-dose erlotinib (10 mg/kg/h, n = 4) or elacridar (12 mg/kg/h, n = 3). Results: Under baseline conditions, 11C-erlotinib distribution to the brain (total volume of distribution [VT], 0.22 ± 0.015 mL/cm3) was markedly lower than its distribution to muscle tissue surrounding the skull (VT, 0.86 ± 0.10 mL/cm3). Elacridar infusion resulted in a 3.5 ± 0.9-fold increase in 11C-erlotinib distribution to the brain (VT, 0.81 ± 0.21 mL/cm3, P < 0.01), reaching levels comparable to those in muscle tissue, without changing 11C-erlotinib plasma pharmacokinetics. During high-dose erlotinib infusion, 11C-erlotinib brain distribution was also significantly (1.7 ± 0.2-fold) increased (VT, 0.38 ± 0.033 mL/cm3, P < 0.05), with a concomitant increase in 11C-erlotinib plasma exposure. Conclusion: We successfully implemented ABCB1/ABCG2 inhibition protocols in nonhuman primates resulting in pronounced increases in brain distribution of 11C-erlotinib. For patients with brain tumors, such inhibition protocols may ultimately be applied to create more effective treatments using drugs that undergo efflux transport at the BBB.

Collaboration


Dive into the Nicolas Tournier's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sylvain Auvity

Université Paris-Saclay

View shared research outputs
Top Co-Authors

Avatar

Fabien Caillé

Université Paris-Saclay

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xavier Declèves

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Oliver Langer

Austrian Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge