JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 JPET ThisFast article Forward. has not been Published copyedited andon formatted.June 13, The 2013 final versionas DOI:10.1124/jpet.113.204990 may differ from this version. JPET#_204990R

Pharmacological characterization of JNJ-40068782, a new potent, selective

and systemically active positive of the mGlu2 receptor

and its radioligand [3H]JNJ-40068782

Hilde Lavreysen, Xavier Langlois, Abdel Ahnaou, Wilhelmus Drinkenburg, Paula te Riele,

Ilse Biesmans, Ilse Van der Linden, Luc Peeters, Anton Megens, Cindy Wintmolders, José

Cid, Andrés A. Trabanco, Ignacio Andrés, Frank M. Dautzenberg, Robert Lütjens, Gregor Downloaded from

Macdonald, John Atack

jpet.aspetjournals.org Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium (HL, XL, AA,

WD, PtR, IB, IVdL, LP, AM, CW, FD, GM, JA)

Janssen Research and Development, Jarama 75, 45007 Toledo, Spain (JC, AT, IA) at ASPET Journals on September 30, 2021 Addex Therapeutics, Chemin des Aulx 12, 1228 Plan-les-Ouates, Geneva, Switzerland (RL)

1

Copyright 2013 by the American Society for Pharmacology and Experimental Therapeutics. JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Running title: Pharmacological characterization of JNJ-40068782, a mGlu2 receptor PAM

Address correspondence, proofs and reprint requests to Dr. Hilde Lavreysen at: Neuroscience,

Janssen, Turnhoutseweg 30, B-2340 Beerse, Belgium

Tel: +32 (0)14 607680

Fax: +32 (0)14 606121

E-mail: [email protected] Downloaded from

Number of text pages: 43

Number of tables: 2 jpet.aspetjournals.org

Number of figures: 10

Number of references: 53

Words in abstract: 260 at ASPET Journals on September 30, 2021

Words in Introduction: 783

Words in Discussion: 1771

Abbreviations:

JNJ-40068782, 3-cyano-1-cyclopropylmethyl-4-(4-phenyl-piperidin-1-yl)-pyridine-2(1H)- one; JNJ-40264796, 1-Butyl-4-[4-(2-methylpyridin-4-yloxy)phenyl]-2-oxo-1,2- dihydropyridine-3-carbonitrile; DCG-IV, (2S,2′R,3′R)-2-(2′,3′- dicarboxylcyclopropyl)glycine; BINA, biphenyl-indanone A; LY487379, 2,2,2-Trifluoro-N-

[4-(2-methoxyphenoxy)phenyl]-N-(3-pyridinylmethyl)-ethanesulfonamide; LY341495, 2S-2- amino-2-(1S,2S-2-carboxycyclopropyl-1-yl)-3-(xanth-9-yl) propanoic acid; LY404039, (-)-

(1R,4S,5S,6S)-4-amino-2-sulfonylbicyclo[3.1.0]hexane-4,6-dicarboxylic acid;

THIIC/LY2607540, N-(4-((2-(trifluoromethyl)-3-hydroxy-4-

2

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

(isobutyryl)phenxoy)methylbenzyl)-1-methyl-1H-imidazo-4-carboxamide; GDP, guanosin 5’- diphosphate sodium salt; EGTA, ethylene glycol-bis(2-amino-ethylether)-N,N,N’,N’-tetra- acetic acid; BSA, albumin from bovine serum; Tris, Tris(hydroxymethyl)-aminomethane;

PMSF, phenylmethanesulfonyl fluoride; HBSS, Hank’s Buffered Salt Solution; REM, rapid eye movement

Recommended Section Assignment: Neuropharmacology Downloaded from jpet.aspetjournals.org at ASPET Journals on September 30, 2021

3

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Abstract

Modulation of the metabotropic glutamate type 2 (mGlu2) receptor is considered a promising target for the treatment of CNS diseases like schizophrenia. Here, we describe the pharmacological properties of the novel mGlu2 receptor positive allosteric modulator (PAM)

3-cyano-1-cyclopropylmethyl-4-(4-phenyl-piperidin-1-yl)-pyridine-2(1H)-one (JNJ-

3 35 40068782) and its radioligand [ H]JNJ-40068782. In [ S]GTPγS binding, JNJ-40068782

produced a left- and up-ward shift in the glutamate concentration-effect curve at human Downloaded from recombinant mGlu2 receptors. The EC50 of JNJ-40068782 for potentiation of an EC20- equivalent concentration of glutamate was 143 nM. While JNJ-40068782 did not affect jpet.aspetjournals.org binding of the orthosteric antagonist [3H] 2S-2-amino-2-(1S,2S-2-carboxycyclopropyl-1-yl)-

3-(xanth-9-yl) propanoic acid (LY-341495), it did potentiate the binding of the agonist [3H]

(2S,2′R,3′R)-2-(2′,3′-dicarboxylcyclopropyl)glycine (DCG-IV), demonstrating that it can at ASPET Journals on September 30, 2021 allosterically affect binding at the agonist recognition site. The binding of [3H]JNJ-40068782 to human recombinant mGlu2 receptors in CHO cells and rat brain receptors was saturable

3 with a KD of ∼10 nM. In rat brain, the anatomical distribution of [ H]JNJ-40068782 was consistent with mGlu2 expression previously described and was most abundant in cortex and hippocampus. The ability of structurally unrelated PAMs to displace [3H]JNJ-40068782 suggests that PAMs may bind to common determinants within the same site. Interestingly, agonists also increased the binding affinity of [3H]JNJ-40068782. JNJ-40068782 influenced rat sleep-wake organization by decreasing REM sleep with a lowest active dose of 3 mg/kg p.o. In mice, JNJ-40068782 reversed PCP-induced hyperlocomotion with an ED50 of 5.7 mg/kg s.c. Collectively, the present data demonstrate that JNJ-40068782 has utility in investigating the potential of mGlu2 modulation for the treatment of diseases characterized by disturbed signalling, and highlight the value of [3H]JNJ-40068782 in exploring allosteric binding.

4

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Introduction

Glutamate is the major excitatory neurotransmitter in the central nervous system of vertebrates and elicits and modulates synaptic responses by activating ionotropic or metabotropic glutamate (mGlu) receptors (Kew and Kemp, 2005). To date, eight mGlu receptor subtypes have been cloned and classified into three groups based upon sequence homology, pharmacological profile and preferential signal transduction pathway (Niswender and Conn, 2010). Downloaded from

The drug discovery efforts related to group III mGlu receptors have lagged behind

(Lavreysen and Dautzenberg, 2008), and various research groups have focussed on the development of ligands acting at group I and II receptors. For example, mGlu1 receptor jpet.aspetjournals.org blockade is thought to have anxiolytic-like properties and to relieve pain (Tatarczynska et al.,

2001; Steckler et al., 2005; Varney and Gereau, 2002), whereas inhibitors of the mGlu5 receptor are believed to be alternative treatments for anxiety, depression and Fragile X, while at ASPET Journals on September 30, 2021 mGlu5 activators may have beneficial effects in schizophrenia (Spooren et al., 2003,

Auerbach et al., 2011). Since group II mGlu receptors reduce transmission at glutamatergic synapses in brain regions where excessive glutamatergic transmission may be implicated in the pathophysiology of anxiety and schizophrenia, it is hypothesized that activation of group

II mGlu receptors may provide anxiolytic and/or effects (Swanson et al., 2005).

Accordingly, mGlu2/3 receptor agonists, such as (-)-(1R,4S,5S,6S)-4-amino-2- sulfonylbicyclo[3.1.0]hexane-4,6-dicarboxylic acid (LY404039) are effective in a number of animal models of schizophrenia (Rorick-Kehn et al., 2007) and anxiety (Helton et al, 1998;

Tizzano et al., 2002; Swanson et al., 2005; Witkin et al., 2007). These observations have been extended into the clinic where LY2140023, a methionine amide prodrug of LY404039, has been shown to demonstrate efficacy in a Phase II study in schizophrenia patients (Patil et al.,

2007; Mezler et al., 2010), though these observations could not be confirmed in a follow-up

5

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R trial (Kinon et al., 2011). The structurally-related analogue LY354740 was able to reduce anxiety in healthy volunteers in a fear-potentiated startle model (Grillon et al., 2003) and attenuated CO2-induced panic in subjects with panic disorder (Schoepp et al., 2003). These anxiolytic-like properties were reproduced in patients with generalised anxiety disorder using

LY354740 (Michelson et al., 2005) as well as its prodrug LY544344 (Dunayevich et al.,

2008).

The use of knockout mice lacking either mGlu2 or mGlu3 receptors has further refined Downloaded from our understanding of the mechanism of action of mGlu2/3 receptor agonists. For example, the anxiolytic actions of LY354740 in the elevated plus maze test require the expression of one or

both of these receptors (Linden et al., 2005). On the other hand, mGlu2 rather than mGlu3 jpet.aspetjournals.org receptors appear to be responsible for the antipsychotic-like properties of LY404039 (Fell et al, 2008), LY379268 (Wooley et al., 2008) and LY314882, which is the racemate of

LY354740 (Spooren et al., 2000). Based upon the collective data using mGlu2/3 selective at ASPET Journals on September 30, 2021 agonists as well as mice lacking either the mGlu2 or mGlu3 receptor, the mGlu2 receptor would appear to be an attractive potential therapeutic target (Niswender and Conn, 2010).

mGlu2 positive allosteric modulators (PAMs) that bind at a receptor site that is different

(i.e. allosteric) from glutamate offer an alternative approach for enhancing mGlu2 activity. mGlu2 PAMs have minimal effects on their own but increase the response of glutamate.

Consequently, mGlu2 receptor PAMs offer an opportunity to enhance mGlu2 receptor signalling by maintaining activity based on transient release of glutamate, but have a lower risk of tolerance development (Conn et al, 2008 and 2009). 2,2,2-Trifluoro-N-[4-(2- methoxyphenoxy)phenyl]-N-(3-pyridinylmethyl)-ethanesulfonamide (LY487379) was identified as the first mGlu2 receptor-specific PAM (Johnson et al., 2003) and was active in animal models predictive of anxiolytic or antipsychotic activity (Johnson et al., 2003, 2005;

Galici et al., 2005), providing preclinical proof-of-concept that PAMs can mimic aspects of

6

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R the pharmacology of agonists. Minimal efficacy at sufficiently low doses as well as short duration of action however limited its use for further development. Pinkerton et al. (2005) reported an alternative series of mGlu2 PAMs, of which the prototypic example biphenyl- indanone A (BINA) demonstrates long-lasting activity in some animal models used to predict potential antipsychotic and anxiolytic activity (Galici et al., 2006). Recently, the structurally novel allosteric potentiator N-(4-((2-(trifluoromethyl)-3-hydroxy-4-

(isobutyryl)phenxoy)methylbenzyl)-1-methyl-1H-imidazo-4-carboxamide (THIIC, also Downloaded from known as LY2607540), which seems to have activity in models predicting either anxiolytic or antidepressant effects, was reported (Fell et al., 2010). While preclinical data hold promise

for mGlu2 PAMs as novel therapeutics, clinical proof-of-concept is awaited with more drug- jpet.aspetjournals.org like compounds compared to the first generation PAMs.

Here, we describe the in vitro and in vivo properties of the novel PAM 3-cyano-1-

cyclopropylmethyl-4-(4-phenyl-piperidin-1-yl)-pyridine-2(1H)-one (JNJ-40068782) (Cid et at ASPET Journals on September 30, 2021 al., Patent WO 2008/107481 A1 2008 and Imogai et al. Patent WO 2007/104783 A2 2007].

We furthermore describe [3H]JNJ-40068782 as the first high affinity, selective mGlu2 PAM radioligand and show that this is an excellent tool to study allosteric mGlu2 interactions.

7

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Methods

Materials.

BINA, LY487379, THIIC (LY2607540), 1-Butyl-4-[4-(2-methylpyridin-4-yloxy)phenyl]-2- oxo-1,2-dihydropyridine-3-carbonitrile (JNJ-40264796) and JNJ-40068782 and [3H]JNJ-

40068782 (Figure 1) were synthesized at Janssen Research and Development. Both [3H]-

DCG-IV and [3H]-LY341495 were obtained from American Radiolabeled Chemicals. [35S]-

γ γ GTP S (specific activity 1250 Ci/mmol; guanosine 5’-( -thio)triphosphate was obtained from Downloaded from

PerkinElmer (Boston MA). GDP, HEPES, EGTA, BSA, probenecid and glutamate were purchased from Sigma (St-Louis, MO); MgCl2 was purchased from VWR Prolabo (Belgium), jpet.aspetjournals.org saponin from Calbiochem (US & Canada), HBSS and fluo-4-AM from Invitrogen (Oregon,

USA), and PMSF was obtained from Fluka (Germany). All other reagents were obtained from Merck (Germany). For in vitro experiments, concentration-response curves of JNJ- at ASPET Journals on September 30, 2021 40068782 were prepared in 100% DMSO and further diluted in assay buffer with final assay concentrations reaching 1% DMSO. For in vivo studies, JNJ-40068782 solutions were prepared in 20% hydroxypropyl-β-cyclodextrine.

Tritiation of JNJ-40068782

The brominated precursor 4-(4-(4-bromophenyl)piperidin-1-yl)-1-(cyclopropylmethyl)-2-oxo-

1,2-dihydropyridine-3-carbonitrile was dissolved in tetrahydrofuran. Next N,N-

Diisopropylethylamine and Pd/C were added. The reaction mixture was attached to a tritium manifold (RC Tritec) and 183 mbar of tritium gas was placed on top of the reaction mixture and was stirred for 1 h at room temperature. The remaining tritium gas was trapped on a

Uranium bed and all volatile compounds were lyophilized to a waste ampoule. The crude reaction mixture was rinsed with methanol, dissolved in ethanol (and filtered over an

Acrodisk filter, yielding 16.4 GBq. A small portion of this material was further purified and

8

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R analyzed by HPLC to obtain the tritiated compound 4-(4-(4-tritiumphenyl)piperidin-1-yl)-

1(cyclopropylmethyl)-2-oxo-1,2-dihydropyridine-3-carbonitrile (radiochemical purity of

>98%, 865 GBq/mmol).

Functional mGlu2 assays.

Ca2+ assay. Intracellular Ca2+ levels were measured using the Functional Drug

Screening System (FDSS 6000, Hamamatsu). Human mGlu2 receptor expressing Gα16-HEK Downloaded from

293 cells were seeded at 10,000 cells/well in 384-well black/clear bottom poly-D-lysine coated plates 24 h before the experiment. On the day of the experiment, medium was jpet.aspetjournals.org removed by brief centrifugation and 20 μl of a fluo-4-AM solution (HBSS buffer containing

20 mM HEPES, 2.5 mM probenecid and ∼2 μM fluo-4-AM, pH 7.4) was added. After an incubation period of 1 h at room temperature in the dark, 40 μl buffer was added and cells at ASPET Journals on September 30, 2021 were kept at room temperature for another 10 min before measurement. Fluorescence was recorded upon the addition of compound (20 μl), and the ratio of the peak over basal fluorescence was exported and used for further calculation. For the measurement of intrinsic agonism, compound was added alone (i.e. in the absence of glutamate); for measurement of

PAM activity, compound was added together with an EC20 equivalent concentration of glutamate (0.3 μM).

[35S]GTPγS binding. CHO cells expressing the rat or human mGlu2 receptor were grown until 80% confluence, washed in ice-cold phosphate-buffered saline and stored at -

20°C until membrane preparation. After thawing, cells were suspended in 50 mM Tris-HCl, pH 7.4 and collected through centrifugation for 10 min at 23,500 g at 4°C. Cells were lysed in 5 mM hypotonic Tris-HCl, pH 7.4 and, after recentrifugation for 20 min at 30,000 g at 4°C, the pellet was homogenized with an Ultra Turrax homogenizer in 50 mM Tris-HCl, pH 7.4.

9

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Protein concentrations were measured by the Bio-Rad protein assay using bovine serum albumin as standard.

For [35S]GTPγS measurements, compound and glutamate were diluted in buffer containing 10 mM HEPES acid, 10 mM HEPES salt, pH 7.4, containing 100 mM NaCl, 3 mM MgCl2 and 10 μM GDP. Membranes were thawed on ice and diluted in the same buffer, supplemented with 14 μg/ml saponin (final assay concentration of 2 μg/ml saponin). Final assay mixtures contained 7 (human mGlu2) or 10 (rat mGlu2) μg of membrane protein and Downloaded from were pre-incubated with compound alone (determination of agonist effects) or together with an EC20 concentration (4 μM) of glutamate (determination of PAM effects) for 30 min at

30°C. [35S]GTPγS was added at a final concentration of 0.1 nM and incubated for another 30 jpet.aspetjournals.org min at 30°C. Reactions were terminated by rapid filtration through Unifilter-96 GF/B filter plates (PerkinElmer, Meriden, CT) using a Unifilter-96 harvester (PerkinElmer). Filters were at ASPET Journals on September 30, 2021 washed 3 times with ice-cold 10 mM NaH2PO4/10 mM Na2HPO4, pH 7.4 and filter-bound radioactivity was counted in a Topcount NXT Microplate Scintillation and Luminescence

Counter from PerkinElmer. Typically, [35S]GTPγS binding in the absence of agonist was in the region of 400 cpm whereas a maximal glutamate response corresponded to ∼2500 cpm.

[35S]GTPγS binding to rat brain sections. Autoradiography of agonist-stimulated

[35S]GTPγS binding in brain sections was performed as previously described (Happe et al.,

2001). Hence, 20 μm thick sagital sections were incubated twice in assay buffer pH 7.5 (50 mM Tris-HCl, 10 mM MgCl2, 0,2 mM EGTA, 100 mM NaCl, 0,1 mM PMSF and 1% BSA) for 10 min at 25°C and once in assay buffer with 2 mM GDP for 15 min at 25°C. Slides were then incubated in assay buffer containing 0.04 nM [35S]GTPγS, in presence or absence of 10

µM glutamate and increasing concentrations of JNJ-40068782 for 2 h at 25°C. The incubation was stopped by washing twice for 3 min in ice-cold 50 mM Tris-HCl (pH 7.4) and a quick dip in ice-cold distilled water.

10

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Sections were then dried under a cold stream of air and placed in a light-tight cassette together with commercially available radioactive polymer standards and exposed to Kodak

Biomax MR film (Kodak, PerkinElmer, Cambridge, UK) for 4 days. Films were developed by standard techniques, digitized and analyzed using the MCID basic 7.0 system (Imaging

Research, Canada).

Agonist-stimulated activity in brain sections was calculated by subtracting the optical density in basal sections (incubated with GDP alone) from that of agonist-stimulated sections; Downloaded from results were expressed as percentage basal activity.

Radioligand binding to human mGlu2-CHO membranes or rat cortical membranes. jpet.aspetjournals.org

Membranes from human mGlu2 receptor stably transfected CHO cells were prepared as above (see [35S]GTPγS binding). For cortical membranes, male Wistar rats (150 g) were

sacrificed and decapitated. Brains were removed and cortex tissue was dissected and kept on at ASPET Journals on September 30, 2021 ice. Tissue was homogenized with an Ultra-Turrax homogenizer in 50 mM Tris-HCl, pH 7.4 and the homogenate was centrifuged for 10 min at 16000 RPM. The pellet was resuspended and homogenized again with a glass-teflon homogenizer (Dual). After 2 washing/centrifugation steps, the final pellet was resuspended in 50 mM Tris-HCl, pH 7.4.

Aliquots were made and frozen at -80°C until further use. Protein concentration was determined with the Bio-Rad Protein Assay method using bovine serum albumin as standard.

After thawing, membranes were homogenized using an Ultra Turrax homogenizer and suspended in ice-cold binding buffer containing 50 mM Tris-HCl (pH 7.4), 10 mM MgCl2, and 2 mM CaCl2.

[3H]LY341495 binding. For competition binding studies, assay mixtures were incubated for 60 min at room temperature in a volume of 0.5 ml containing 10 μg membrane protein (hmGlu2 CHO), appropriate concentrations of test compound and 2 nM 11

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

[3H]LY341495. Non-specific binding was determined in the presence of 1000 μM glutamate and was about 10% of total binding. The incubation was stopped by rapid filtration using

Unifilter-96 GF/B filter plates and a Unifilter-96 harvester (PerkinElmer).

[3H]DCG-IV binding. For saturation experiments, 50-75 μg membrane protein

(hmGlu2 CHO), test compound and increasing concentrations ranging from 2 to 800 nM of

[3H]DCG-IV were incubated for 60 min at room temperature (in total, 11 or 12 concentrations

were tested). Non-specific binding (about 30% of total binding) was determined in the Downloaded from presence of 10 μM LY341495. Displacement studies were done with 50 nM of [3H]DCG-IV.

Filtration was performed using Unifilter-96 GF/C filters pre-soaked in 0.1% PEI and a jpet.aspetjournals.org Unifilter-96 harvester (PerkinElmer).

[3H]JNJ-40068782 binding. For saturation experiments, 75 μg (hmGlu2 CHO) or 75-

μ

125 g (cortex) membrane protein, test compound and increasing concentrations at ASPET Journals on September 30, 2021

(concentrations in assay ranged from 0.5 to 200 nM and a total of 12 concentrations were tested) of [3H]JNJ-40068782 were incubated for 60 min at room temperature, in a total volume of 0.5 ml. Non-specific binding (about 30% of total binding) was determined in the presence of 10 μM 1-butyl-4-[4-(2-methylpyridin-4-yloxy)phenyl]-2-oxo-1,2- dihydropyridine-3-carbonitrile JNJ-40264796, a structurally-related molecule. Displacement studies were performed using 10 nM of radioligand. Filtration was performed using Unifilter-

96 GF/C filters pre-soaked in 0.1% PEI and a 40-well manifold or 96-well Brandell harvester

96.

For all radioligands, [3H]LY341495, [3H]DCG-IV, and [3H]JNJ-40068782, assay conditions including radioligand concentrations were chosen such that <10% of the free radioligand concentration was receptor-bound. After the addition of scintillation liquid,

12

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R radioactivity on the filters was counted in a Microplate Scintillation and Luminescence

Counter or Liquid Scintillation Analyzer from Perkin Elmer.

[3H]JNJ-40068782 rat brain autoradiography.

Tissue Preparation. Male Wistar rats (200 g) were sacrificed by decapitation. Brains were immediately removed from the skull and then rapidly frozen in dry-ice-cooled 2- methylbutane (-40°C). Brains were then stored at -20°C until sectioning. Sagittal sections (20

μm) were cut using a Leica C3050 cryostat microtome (Leica Microsystems, Wetzlar, Downloaded from

Germany) and thaw-mounted on SuperFrost Plus microscope slides (Menzel-Glaser GmbH,

Braunschweig, Germany). The sections were then kept at -20°C until use. Male wild type jpet.aspetjournals.org C57BL/6 and mGlu2 knock-out mice (Deltagen, Inc.) weighing 25-30 g were sacrificed by decapitation and their brain prepared as stated above.

Receptor Autoradiography. Sections were thawed and dried under a stream of cold air, at ASPET Journals on September 30, 2021 and preincubated (2 times 10 min) at room temperature in incubation buffer (50 mM Tris-

HCl, 2 mM MgCl2, 2 mM CaCl2, pH 7.4). Sections were then incubated for 60 min at room temperature in buffer containing 10 nM [3H]JNJ-40068782 with nonspecific binding determined by addition of 10 μM JNJ-40068782. After incubation, the excess of radioligand was washed off (2 times 10 min) in ice-cold buffer, followed by a rapid dip in cold distilled water. The sections were dried under a stream of cold air. Rat brain sections were exposed to

[3H]Hyperfilm (Amersham Biosciences, Buckinghamshire, UK) for 6 weeks at room temperature. The films were developed manually in Kodak D19 and fixed with Kodak

Readymatic (Eastman Kodak, Rochester, NY). Mouse brain sections were exposed for 6 days to a Phosphorimager screen. Digital autoradiograms were obtained using a PhosphorImager

FLA7000 (Fuji).

13

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Selectivity panel.

Ca2+ assays with human mGlu1, 3, 5, 7 or 8 receptor-expressing HEK 293 cells.

Measurement of intracellular Ca2+ mobilization in HEK293 cells stably transfected with the human mGlu1a receptor proceeded as follows. Cells were seeded at 10,000 cells/well in a

384-well plate. Twenty-four h after seeding, FLIPR Ca2+ assay kit (Molecular Devices) dissolved in phosphate-buffered saline supplemented with 5 mM probenecid, pH 7.4 (f.c. 2.5 mM probenecid as loading buffer was added on the cell layer with no removal of medium) Downloaded from was added and measurements were initiated 90 min thereafter.

2+ Ca assays with human mGlu3 receptor expressing Gα16-HEK 293 cells were

performed similarly as for human mGlu2 expressing cells, with these exceptions: cells were jpet.aspetjournals.org seeded at a density of 10,000 cells/well and cell incubation with fluo-4-AM took place at

37°C instead of room temperature.

For the mGlu5 receptor, cells were seeded at 40,000 cells/well in PDL-coated 384- at ASPET Journals on September 30, 2021 well plates, and a day later, medium was removed before addition of a fluo-4-AM solution

(HBSS, 2.5 mM probenecid and ∼2 μM fluo-4-AM, pH 7.4). Measurements were performed

90 min later (while incubation at 37°C and 5% CO2).

For the human mGlu7 and 8 receptor, stably expressing Gα15-HEK cells were seeded at a density of 15,000 cells/well (PDL-coated 384-well plates) and incubated with FLIPR

Calcium 4 assay kit (dissolved in HBSS, 20 mM HEPES, 2.5 mM probenecid, pH 7.4) for 90 min at 25°C before measurement.

The ratio of peak versus basal fluorescent signals was used for data analysis.

[35S]GTPγS binding to membranes from CHO cells expressing the rat mGlu6 receptor and to membranes from L929sA cells expressing the human mGlu4 receptor.

Membrane preparations were performed as described above (see [35S]GTPγS binding).

Membranes, compound and glutamate were diluted in assay buffer (10 mM HEPES acid, 10

14

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

mM HEPES salt, pH 7.4, containing 100 mM NaCl, 0.3 mM MgCl2 and 10 μM GDP). Assay mixtures included 10 μg of membrane protein and were pre-incubated with compound alone or together with glutamate for 30 min at 37°C. [35S]GTPγS was added at a final concentration of 0.1 nM and incubated for another 30 min at 37°C. Reactions were terminated by rapid filtration through Unifilter-96 GF/B filter plates (Packard, Meriden, CT) using a 96-well

Packard filtermate harvester.

Selectivity of JNJ-40068782 in additional radioligand binding assays. JNJ-40068782 Downloaded from was tested at a concentration of 10 μM by CEREP (Celle L’Evescault, France) for its inhibition of radioligand binding to a battery of neurotransmitter and peptide receptors, ion jpet.aspetjournals.org channels and transporters.

Data analysis. at ASPET Journals on September 30, 2021 For both [35S]GTPγS and Ca2+ mobilization assays, data were processed using either the Lexis software interface developed at Janssen Research & Development or GraphPad

Prism (version 4.02) and were calculated as percentage of the control agonist challenge.

Sigmoid concentration-response curves plotting percentages of effect versus the log concentration of the test molecule were analysed using nonlinear regression analysis.

To further describe the signalling of glutamate in the presence of a PAM, the fold shift of the glutamate concentration-response was plotted in function of the concentration of modulator in GraphPad Prism (version 4.02) using nonlinear regression, interpolation was then used to allow estimates of concentrations needed for a 5-fold shift.

Inhibition [3H]JNJ-40068782 binding data were fitted to a one-site binding model

(GraphPad Prism version 4.02) and estimates of dissociation constants (Ki) were derived using the Cheng-Prusoff equation (Cheng and Prusoff, 1973). Saturation binding experiments

15

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R were analysed using nonlinear regression (one-site binding hyperbola; GraphPad Prism version 4.02).

In vivo studies.

All animal experiments were carried out in strict accordance with the Declaration of Helsinki and the European Communities Council Directive of 24th November 1986 (86/609/EEC) , and were approved by the animal care and use committee of Janssen Research and Downloaded from Development as well as the local ethical committee.

Pharmacokinetic analysis. Animals were given free access to food and water overnight prior to dosing, and throughout the study. Male Sprague-Dawley rats were dosed with a single jpet.aspetjournals.org oral dose of 10 mg/kg. Blood samples were taken at 0.5, 1, 2, 4, 8 and 24 h from the tail vein using EDTA as the anti-coagulant. Male Swiss mouse were dosed subcutaneously with a dose of 10 mg/kg, and blood samples were taken at 0.5, 1 and 1.5h after dosing. Plasma was at ASPET Journals on September 30, 2021 obtained following centrifugation, extracted using protein precipitation, and subsequently analysed for JNJ-40068782 using a liquid chromatography/tandem mass spectrometry qualified research method on an API4000 instrument (Applied Biosystems)

Sleep-wake architecture study in rat. Male Sprague Dawley rats (Charles River, France) weighing 250–300 g at the time of surgery were used. The effect of oral administration of

JNJ-40068782 (administered p.o. 2 h after light onset using a dose volume of 10 ml/kg) on sleep-wake organization in rats was determined as described in Ahnaou et al., 2009. In brief, sleep polysomnographic variables were determined in 32 adult, male Sprague Dawley rats

(n=8 each group), which were chronically implanted with electrodes for recording the cortical electroencephalogram (EEG), electrical neck muscle activity (EMG), and ocular movements (EOG). Polysomnographic data was scored with an automated sleep stages analyzer, scoring each 2-second epoch before averaging stages over 30-minutes periods.

16

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Sleep-wake state classifications were assigned based upon the combination of dynamics of five EEG frequency domains, integrated EMG, EOG, and body activity level: Active wake;

Passive wake; Rapid Eye Movement sleep (REM); Intermediate Stage (pre-REM transients); light non-REM sleep; and deep non-REM sleep. Different sleep-wake parameters like amount of time spent in each state were investigated over 20 post-administration hours.

Time spent in each sleep-wake state was expressed as a percentage of the recording period. A statistical analysis of the obtained data was carried out by a non-parametric analysis Downloaded from of variance of each 30-min period, followed by a Wilcoxon-Mann-Whitney rank sum test of comparisons with the control group. jpet.aspetjournals.org PCP-induced hyperlocomotion in mice. Test compound or solvent was administered alone (to monitor effects on spontaneous locomotion) or together with phencyclidine (PCP; 5 mg/kg) to male NMRI mice (22 ± 4 g and fasted overnight; Charles River Germany) 30 min at ASPET Journals on September 30, 2021 before measurement. Both JNJ-40068782 and PCP were injected subcutaneously (s.c.) and locomotor activity was measured for a period of 30 min. ED50 values (the doses inducing

50% responders to criterion) were determined as follows.

All-or-nothing criteria for significant (p < 0.05) effects were defined by analyzing a frequency distribution of a large series of historical control data obtained in solvent-treated animals. Based on the criteria obtained in this way, the ED50 and corresponding 95% confidence limits were determined according to the modified Spearman-Kaerber estimate, using theoretical probabilities instead of empirical ones. This modification allows determination of the ED50 and its confidence interval as a function of the slope of the log dose-response curve. Criterion for drug-induced decrease of spontaneous locomotion: total distance < 2,500 counts (4.1% false positives in controls; n = 410). Criterion for drug- induced inhibition of PCP-induced hyperlocomotion: total distance < 5,500 counts (3.9% false positives in controls; n = 154).

17

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Results

JNJ-40068782 acts as a selective positive allosteric modulator at the mGlu2 receptor.

Figure 2 shows the concentration-response curves for the glutamate-stimulated increase in [35S]GTPγS binding in absence and presence of JNJ-40068782, BINA, LY487379 or LY2607540. JNJ-40068782 like the other PAMs enhances the glutamate-stimulated increase in [35S]GTPγS. Interestingly, all compounds magnified the response produced by Downloaded from glutamate, shifting the glutamate concentration-effect curve both up-ward and left-ward.

While a concentration of 429 nM of BINA, 568 nM of JNJ-40068782 or 305 nM of

LY2607540 increases the glutamate potency (i.e. lower the glutamate EC50) by 5-fold, jpet.aspetjournals.org

LY487379 causes only a 2-3 fold shift in the glutamate concentration-response curve at the highest tested concentration of 1 μM. Maximal decreases in glutamate EC50 of 10-12 fold are

observed for the other compounds. at ASPET Journals on September 30, 2021

By quantifying the increase in response at a fixed glutamate concentration, for example the glutamate EC20, the potency (EC50) of JNJ-40068782 at the human mGlu2 receptor was estimated to be 143 nM (Table 1 and Figure 2B). This value is similar

(overlapping confidence intervals) to the potencies of BINA and LY487379 and LY2607540

(respective EC50 values of 89, 256 and 95 nM). The maximal response to glutamate was increased to 309 ± 23% upon addition of JNJ-40068782, which is similar to the potentiation seen with BINA and LY2607540 (221 ± 9 and 215 ± 7%, respectively) (Table 1). However,

LY487379 showed a more modest, 147 ± 9%, increase in glutamate Emax. Though both EC50 and Emax values slightly increased at the rat compared to the human mGlu2 receptor, we found a good correspondence between compound potencies obtained in the rat and human assays

(Table 1).

18

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

At higher concentrations, JNJ-40068782 also increased [35S]GTPγS binding in the absence of glutamate, suggesting that it acts as an ‘ago-potentiator’. Similar profiles were observed for BINA and LY2607540 but not LY487379. Interestingly, JNJ-40068782 along with the reference compounds acted as a pure mGlu2 receptor PAM at recombinant rat mGlu2 receptors (i.e. did not have any intrinsic activity in the absence of glutamate; Table 1).

Further profiling of JNJ-40068782 showed that it also facilitated glutamate-induced

Ca2+ mobilization at human recombinant mGlu2 receptors. In HEK293 cells co-expressing Downloaded from

Gα16, JNJ-40068782 potentiated the EC20 glutamate response with an EC50 value of 15 nM

(95% confidence interval 12 - 19 nM; n=30). Interestingly, in this assay JNJ-40068782 did

not increase the maximal response of glutamate (Emax = 119 ± 2%; also see Figure 2C). JNJ- jpet.aspetjournals.org

40068782 induced intracellular Ca2+ mobilization on its own (allosteric agonism) with an

EC50 of 618 nM (95% confidence interval 497 - 768 nM) and Emax of 81 ± 3% (n=25).

JNJ-40068782 did not show agonist, antagonist or PAM activity at the human mGlu3 at ASPET Journals on September 30, 2021 receptor or any of the group I or group III mGlu receptors up to 10 μM (data not shown).

Further profiling (CEREP) revealed 59% and 85% inhibition of h5HT2A (determined with

3 125 [ H]ketanserin) and hCCKA (determined with [ I]CCK-8s) receptor binding at 10 μM. The complete selectivity profile based on CEREP data is shown in Supplementary Table 1.

Modulation of [35S]GTPγS binding by JNJ-40068782 at native rat mGlu2 receptor.

Despite extensive washing of the rat brain sections, in the absence of exogenous glutamate, there remained a basal level of [35S]GTPγS binding that could be reduced by mGlu2/3 receptor antagonists (data not shown), suggesting that there was sufficient residual endogenous glutamate to produce a degree of mGlu2 and/or mGlu3 receptor activation. As a result, in rat brain sections glutamate produced much smaller responses than observed in the

19

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R recombinant human mGlu2 cell line, with, for example, 10 μM glutamate producing a maximal response of about 25% over basal.

The [35S]GTPγS binding to rat brain sections nevertheless does give a clear illustration that JNJ-40068782 can modulate the effects of glutamate at native rat brain mGlu2 receptors.

Hence, JNJ-40068782 was able to increase the [35S]GTPγS binding to rat brain slices in the absence of exogenous glutamate (Figure 3). Although this may imply that JNJ-40068782 has intrinsic direct agonist effects at native rat mGlu2 receptors (i.e., can activate mGlu2 Downloaded from receptors in the absence of glutamate), the possibility that this may be a modulation of endogenous glutamate that was not removed during washing of the slices cannot be excluded.

μ 35 γ

In the presence of 10 M glutamate, JNJ-40068782 produced a robust increase in [ S]GTP S jpet.aspetjournals.org binding, particularly in the cortex, striatum and hippocampus (Figure 3). Potentiation of glutamate by JNJ-40068782 was evident at the lowest concentration of the allosteric

modulator, and quantification of these data showed that whereas 10 μM glutamate or 1 μM at ASPET Journals on September 30, 2021

JNJ-40068782 alone increased [35S]GTPγS binding in the cortex by ∼10% and ∼20%, respectively, the combination led to a stimulation of ∼80%. In combination with glutamate, a maximal stimulation of ∼175% was detected after the addition of 10 or 100 μM JNJ-

40068782; in the absence of glutamate, 100 μM JNJ-40068782 increased the response to

125%. Hence, although the potency of JNJ-40068782 on [35S]GTPγS binding at native rat receptors is reduced relative to recombinant systems, presumably as a result of the complication of endogenous glutamate, the anatomical distribution of the responses (i.e., preferential stimulation of the binding in the cortex, striatum and hippocampus ) is consistent with an mGlu2 receptor-mediated response.

20

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

JNJ-40068782 does not affect [3H]LY341495 binding but increases [3H]DCG-IV binding.

To ascertain its allosteric character, JNJ-40068782 was tested for displacement of the prototypic orthosteric (i.e. glutamate site) mGlu2/3 receptor antagonist [3H]LY341495 binding to human mGlu2 receptor-expressing membranes. [3H]LY341495 binding to the human mGlu2 receptor was not inhibited upon the addition of JNJ-40068782 10 μM (Figure

4A). In contrast to the lack of effect of JNJ-40068782 on the binding of the antagonist radioligand, binding of the radiolabeled orthosteric agonist [3H]DCG-IV was increased up to Downloaded from 2.5-fold upon the addition of JNJ-40068782 at 3 μM (Figure 4B).

In order to assess whether JNJ-400068782 alters the apparent affinity of an agonist for jpet.aspetjournals.org the orthosteric binding site, saturation binding of the agonist [3H]DCG-IV was performed in the absence and presence of 3 μM JNJ-40068782. Figure 5A clearly demonstrates that there was a left-ward shift in the [3H]DCG-IV binding curve in the presence of JNJ-40068782. at ASPET Journals on September 30, 2021

Quantification of these data shows that in the absence of JNJ-40068782, the affinity of

[3H]DCG-IV for the orthosteric binding site was 240 ± 39 nM (average ± S.E.M. of 6 experiments) whereas with the PAM present, KD decreased about 8-fold, reflecting increased affinity (Table 2). Similar results were seen with other mGlu2 receptor PAMs. The number of binding sites, represented by the Bmax value, was unchanged. The linearity of the Scatchard analysis of the saturation binding data, illustrates that both in the absence and presence of

JNJ-40068782, the binding of [3H]DCG-IV is best described by a single binding site. In order to see whether the same effects were seen for the natural agonist glutamate, glutamate- mediated inhibition of [3H]DCG-IV binding was assessed in the absence and presence of 3

μM JNJ-40068782. In line with previous data, JNJ-40068782 not only increased the maximal binding, but also enhanced the affinity of glutamate, i.e. lowered the IC50 of glutamate by ∼5- fold (4.5- and 6.7-fold found in 2 independent experiments; Figure 5B).

21

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Binding of [3H]JNJ-40068782 to recombinant human mGlu2 receptors.

JNJ-40068782 was tritiated and used to characterize the binding to human recombinant mGlu2 receptors (Figure 6A). Binding was saturable and was best-fit to a single-site model defined by a KD of 12 ± 4 nM with a Bmax of 4.5 ± 1.9 pmol/mg protein (n=12). At a concentration of 10 nM, the level of specific binding was in the region of 70%. Interestingly, in the presence of agonist (either glutamate, DCG-IV or LY354740), the apparent affinity of Downloaded from 3 [ H]JNJ-40068782 was increased (average KD in the presence of 100 μM glutamate was 4.0 ±

1.2 nM; n=4), with the saturation curve being left-ward shifted (Figure 6B). In each of the 4

experiments where binding was measured in the absence or presence of glutamate, the Bmax jpet.aspetjournals.org value of [3H]JNJ-40068782 was increased upon addition of the agonist, with an average increase of 2-fold.

at ASPET Journals on September 30, 2021

The binding of [3H]JNJ-40068782 to human recombinant mGlu2 receptors was inhibited by JNJ-40068782 with a mean IC50 of 38 nM (95% confidence interval 27-49 nM, n=9, Figure 7A). In addition, BINA, LY483739 and LY2607540 inhibited [3H]JNJ-40068782 with respective IC50 values of 385 nM (95% confidence interval 217-681 nM, n=8), 116 nM

(95% confidence interval 78-171 nM, n=6) and 98 nM (95% confidence interval 44-216 nM, n=4). Though it is not clear whether the interaction between these molecules and JNJ-

40068782 is completely competitive, hill slopes of the inhibition curves are close to one and binding of [3H]JNJ-40068782 was fully displaced upon addition of compound (i.e. the same level of non-specific binding was observed for all – structurally unrelated – modulators), suggesting a competitive interaction. Hence, IC50 values were also converted to Ki values according to the Cheng-Prusoff equation (Ki was 20 nM for JNJ-40068782, 215 nM for

BINA, 65 nM for LY483739 and 55 nM for LY2607540). Additional studies showed that the

22

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

3 negative allosteric modulator RO4491533 also fully displaced [ H]JNJ-40068782 (IC50 of 16 nM (95% confidence interval 7-39 nM, n=4; Ki of 9 nM; data not shown).

Binding of [3H]JNJ-40068782 to rodent brain tissue.

3 [H]JNJ-40068782 bound with a similar affinity to receptors expressed in rat cortex; KD and Bmax were 10 ± 4 nM (c.f. 12 ± 4 nM at human recombinant mGlu2 receptors) and 14.7 ±

5.7 pmol/mg protein (n=6), respectively. IC50 values for displacing cortical mGlu2 receptors Downloaded from were 52 nM (95% confidence interval 34-78 nM, n=7), 617 nM (95% confidence interval

242-1571 nM, n=5), 102 nM (95% confidence interval 55-190 nM, n=5) and 79 nM (95%

confidence interval 43-143 nM, n=3) for JNJ-40068782, BINA, LY483739 and LY2607540 jpet.aspetjournals.org respectively (assuming a competitive interaction, Ki values are 26, 305, 51 and 39 nM, respectively), confirming that these PAMs also bind to native receptors (Figure 7B).

Using radioligand autoradiography, we examined [3H]JNJ-40068782 binding distributions in at ASPET Journals on September 30, 2021 rat brain sections in further detail (Figure 8A). High specific binding was observed in the hippocampal formation, granular layer of the cerebellum, striatum and cortex, corresponding very well with the expression pattern demonstrated using the mGlu2/3 receptor antagonist

[3H]LY341495. [3H]JNJ-40068782 binding site distribution was similar in wild type mice brain. Importantly, binding was lost in mGlu2 knock-out mice underlining the mGlu2- specificity of the radioligand (Figure 8B).

Effects of JNJ-40068782 on sleep-wake organization in the rat.

The preceding data collectively demonstrate that JNJ-40068782 not only binds to native mGlu2 receptors but also selectively potentiates the functional response at rat brain mGlu2 receptors. As JNJ-40068782 was shown to be brain-penetrant (with Cmax of ∼1400

23

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R ng/ml, and brain : plasma ratio of ∼0.4 after oral dosing of 10 mg/kg in rats), we set out to evaluate some of the in vivo functional effects of this novel mGlu2 receptor PAM.

We have previously shown that selective activation or positive allosteric modulation of the mGlu2 receptor in vivo affects the sleep-wake architecture of rats (Ahnaou et al.,

2009). To assess whether JNJ-40068782 has in vivo central activity, we evaluated sleep- wake organization of rats after oral dosing of JNJ-40068782.

The time course of vigilance states over 20 h following administration of vehicle and Downloaded from JNJ-40068782 is illustrated in Figure 9. Acute administration of JNJ-40068782 (3, 10 and 30 mg/kg, p.o.) significantly suppressed the time spent in REM sleep and intermediate stage,

whereas no major effects on the other sleep-wake stages were observed. Quantification of jpet.aspetjournals.org effect over a 4-h time period following compound administration revealed a dose-related reduction in time spent in REM sleep with a lowest active dose of 3 mg/kg p.o.,

corresponding to a plasma drug concentration of ∼420 ng/ml. REM sleep reduction was at ASPET Journals on September 30, 2021 accompanied by consistent lengthening in REM sleep onset latency (Figure 8, upper small bar graph panel in REM sleep stage).

Effects of JNJ-40068782 on PCP-induced hyperactivity in mice.

The effects of JNJ-40068782 on spontaneous locomotor activity was measured at doses ranging from 2.5-10 mg/kg s.c. (Figure 10A). The compound produced a modest reduction on spontaneous activity, with the total distance travelled dropping by 20% from

2974 cm in vehicle-treated mice to 2365 cm in animals receiving 20 mg/kg JNJ-40068782.

We subsequently evaluated whether JNJ-40068782 was able to mimic some of the effects of mGlu2/3 agonists in an animal model that is used to predict potential anti-psychotic activity. At a dose of 5 mg/kg (s.c.), PCP induces a profound increase in the total distance travelled by mice. Subcutaneous injection of JNJ-40068782 significantly attenuated the

24

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

increase in locomotor activity evoked by PCP, with an ED50 of 5.7 mg/kg s.c., corresponding to a plasma drug concentration of ∼1700 ng/ml (Figure 10B). Downloaded from jpet.aspetjournals.org at ASPET Journals on September 30, 2021

25

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Discussion

The pioneering work emanating from Eli Lilly using mGlu2/3 orthosteric (glutamate) binding site agonists such as LY404039 and LY354740 have defined the group II mGlu receptor subfamily as an attractive potential therapeutic target for anxiety disorders and schizophrenia

(Swanson et al., 2005; Fell et al., 2012). A key role of the mGlu2 receptor in mediating the pharmacological effects observed with mGlu2/3 receptor agonists is emphasized by data

demonstrating that compounds which selectively potentiate the effects of glutamate at mGlu2 Downloaded from receptors (mGlu2 PAMs) can reproduce some of the effects seen with the non-selective mGlu2/3 receptor agonists. Such compounds include CBiPES (Johnson et al., 2005), BINA jpet.aspetjournals.org (Galici et al., 2005, 2006) and LY487379 (Johnson et al., 2003; Schaffhauser et al., 2003;

Galici et al., 2005).

Our studies have provided new pharmacological tools to study the mGlu2 receptor PAM at ASPET Journals on September 30, 2021 concept, including JNJ-40068782 and [3H]JNJ-40068782. While the in vitro potencies for the reference mGlu2 receptor PAMs vary from lab to lab, presumably as a result of difference in receptor expression systems and/or assay methodologies, it nevertheless seems that

LY487379 is consistently less potent (with in vitro potency for the human mGlu2 receptor ranging from ∼250 nM to ∼1.5 μM) than BINA, LY2607540 and JNJ-40068782, all of which may be considered equipotent (with potencies ranging from ∼20 nM to 1 μM depending on the assay).

Using a [35S]GTPγS assay for mGlu2 receptors expressed in CHO cells, there does not seem to be any marked rat versus human species differences for the four above-mentioned PAMs although EC50 values were generally 2-fold lower and Emax 1.5–fold higher between rat and human mGlu2 data. Screening of over 100 additional compounds furthermore confirmed this pattern, with no examples of compounds being active in one species but not the other (data

26

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R not shown; correlation plots can be found in Supplementary Figure 1). JNJ-40068782 induced a receptor response even in the absence of agonist in some but not all in vitro assays, indicating that the functional effects of this molecule can be different depending on the functional read-out as well as the level of receptor expression and hence seem context- dependent. This is in line with the thinking that ligand ‘behaviours’ rather than inherent

‘properties’ can be described and that detection of agonist ‘behaviour’ depends on the ligand and sensitivity of the assay used (Langmead, 2012). It can also not be ruled out that the Downloaded from response can, at least in part, be attributed to the presence of endogenous glutamate.

It is not yet clear why JNJ-40068782 seems to be more potent in the Ca2+ assay compared to

35

the [ S]GTPγS binding assay. Moreover, the maximal response to glutamate was not jpet.aspetjournals.org increased when measuring changes in intracellular Ca2+ concentrations, in contrast to the

35 dramatic increase in response when measuring [ S]GTPγS accumulation. Several reasons

may underlie these differences including the use of different cellular backgrounds, potential at ASPET Journals on September 30, 2021 differences in receptor and/or G protein expression levels, the transient (Ca2+) versus end point ([35S]GTPγS) nature of the assay, potential fluorescent dye saturation, differences in equilibrium binding conditions, or compound solubility during the assay procedure. It can furthermore not be excluded that forced coupling of the mGlu2 receptor to the Ca2+ pathway affects ligand potency compared to using an assay where the receptor signals via a natural, endogenous protein.

JNJ-40068782 also enhanced native mGlu2 receptor signalling as shown using GTPγS autoradiography on rat brain slices.

Allosteric modulators may not only enhance the efficacy of the endogenous agonist by

“magnifying” the intracellular responses produced by the agonist but also by altering the apparent affinity of the agonist, or a mixture of the two (Conn et al., 2009). In the present

27

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R study, JNJ-40068782, along with BINA and LY487379, was able to increase the affinity of

3 agonists, with the KD for [ H]DCG-IV binding to recombinant human mGlu2 receptors decreasing by around 5-10-fold in the presence of mGlu2 PAMs. Moreover, and as expected, although JNJ-40068782 increased the affinity of agonists at the orthosteric binding site, it did not alter the binding of the orthosteric antagonist [3H]LY341495.

While our data thus far enabled to get an understanding of how PAMs can affect agonist Downloaded from signalling and binding, we set out to study effects on allosteric interaction by agonists with the use of selective mGlu2 receptor PAM radioligand binding. Radioligands that selectively

3 recognize an allosteric modulatory site have been described for the mGlu1 ([ H]R214127; jpet.aspetjournals.org

Lavreysen et al., 2003) and mGlu5 receptor ([3H]M-MPEP, Gasparini et al., 2002;

[3H]mPEPy, Cosford et al., 2003). Recently, a novel mGlu2/3 receptor agonist radioligand

was reported (Wright et al., 2012). However, [3H]JNJ-40068782 is the first selective mGlu2 at ASPET Journals on September 30, 2021 radioligand that is described extensively. JNJ-40068782 was selective for mGlu2 receptors as defined by CEREP profiling, where only a significant interaction at CCKA receptors, which are known to be only expressed in very discrete brain regions such as the interpeduncular nucleus and nucleus tractus solitarius (Hill et al., 1987), was observed at 10 μM of JNJ-

40068782. At relatively high concentrations, JNJ-40068782 also acted on the 5HT2A receptor

(IC50 ∼ 10 μM); it is, however, unlikely that it reaches sufficient amounts in vivo to trigger a

3 functional 5HT2A-related effect. Using [ H]ketanserin, we did not observe in vivo binding up to 30 mg/kg p.o. (data not shown); also, JNJ-40068782 did not affect sleep-wake behaviour in rats, while ample evidence exists for modulation of deep non-REM sleep via the 5HT2A receptor (Al-Shamma et al., 2010). Moreover, in rat brain [35S]GTPγS binding experiments, the preferential stimulation of binding in the cortex and striatum is again consistent with JNJ-

40068782 selectively modulating the function of mGlu2 receptors. Finally, the selectivity of

28

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R the radioligand for mGlu2 versus mGlu3 receptors was confirmed by the fact that binding was lost in mGlu2 but retained in mGlu3 receptor knockout mice brains.

[3H]JNJ-40068782 binding met all the requirements for a ligand well suited to study binding properties, pharmacology, and distribution of mGlu2 receptors. [3H]JNJ-40068782 showed saturable one-site binding, with its affinity (∼10 nM) being similar between cloned human receptors and native mGlu2 receptors in rat cortex. The structurally diverse PAMs BINA,

LY487379 and LY2607540 seem to share common determinants in the allosteric binding site Downloaded from since, both in recombinant cells and in brain tissue, they could all displace [3H]JNJ-40068782.

Interestingly, the mGlu2 receptor negative allosteric modulator RO4491533 was also able to inhibit the binding of JNJ-40068782 indicating that both negative and positive allosteric jpet.aspetjournals.org modulators share common determinants of binding within the allosteric modulatory site.

at ASPET Journals on September 30, 2021 [3H]JNJ-40068782 binding was used to get more insight in how orthosteric binding would influence binding to the allosteric 7TM domain. In line with the effect of JNJ-40068782 on agonist binding, demonstrated with both [3H]DCG-IV and glutamate, there was a reciprocal effect of orthosteric agonists (glutamate, DCG-IV and LY354740) enhancing binding affinity of [3H]JNJ-40068782. While agonists predominantly label the high affinity or G protein- coupled receptor state, an antagonist shows equal affinity for coupled and uncoupled receptors, or for both the high- and low-affinity states of the receptor. Our finding that the

3 3 Bmax for [ H]JNJ-40068782 was only about half of that of [ H]DCG-IV is interesting and may suggest that PAMs preferentially bind to certain receptor states that are not necessarily similar to those preferentially recognized by DCG-IV. Alternatively, JNJ-40068782 may bind to only one of the subunits within the mGlu2 homodimer, whereas DCG-IV likely binds to both, which might be consistent with previous reports that binding and closure of both extracellular domains within the mGlu receptor dimer is needed for full activity, whereas only a single

29

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R heptahelical domain is turned on upon receptor activation (Kniazeff et al., 2004; Hlavackova et al., 2005). Even more intriguing is the finding that in the presence of agonists (glutamate

3 or LY354740), the Bmax of [ H]JNJ-40068782 was consistently increased, opening the possibility of a changed receptor conformation allowing binding to both receptor subunits in the mGlu2 receptor dimer, upon which a ‘super-activated’ state may be imposed.

3

Because of its specificity, [ H]JNJ-40068782 proved to be suitable for investigation of mGlu2 Downloaded from receptor distribution in brain sections using radioligand autoradiography. These studies showed that the highest level of mGlu2 specific binding was present in the hippocampus, jpet.aspetjournals.org striatum, cortex and granular layer of the cerebellum. These results correspond to the binding of [3H]LY341495, and to the binding of [3H]LY354740 reported by Richards et al. (2005), where [3H]LY354740 was used under such conditions that it bound selectively to mGlu2 at ASPET Journals on September 30, 2021 receptors. The distribution of [3H]JNJ-40068782 binding was also comparable to the anatomical distribution of mGlu2 receptor mRNA and protein (Ohishi et al., 1993, 1998; Gu et al., 2008).

JNJ-40068782 proved to have appropriate pharmacological and pharmacokinetic properties to evaluate its effects in vivo. It is well-established that mGlu2 receptor modulation affects sleep-wake architecture in rats and mice (Feinberg et al., 2002; Ahnaou et al., 2009; Fell et al., 2010). Consistent with these data, we found that JNJ-40068782 dose-dependently decreased the amount of REM sleep in rats when dosed early in the light phase. The effects on sleep-wake organization in rats may well reflect the modulation of physiological glutamatergic signalling since it has been shown that glutamate levels are increased during

REM sleep in rat orbitofrontal and cerebral cortex (Lopez-Rodriguez et al., 2007 and Dash et al., 2009). Moreover, because of the translational nature of the sleep-wake EEG paradigm – polysomnography measurements in man may be used as a reproducible and sensitive

30

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R biomarker. Nevertheless, it remains to be determined to what extent the changes in REM sleep, although a reliable pharmacodynamic marker, have relevance for potential mGlu2- mediated therapeutic efficacy.

As regards to potential therapeutic indication for mGlu2 receptor modulation, JNJ-40068782 decreased PCP-induced hyperlocomotion, which is a model in which dopamine D2-related demonstrate robust efficacy. However, the doses at which JNJ-40068782 is active in this model correspond to plasma drug concentrations somewhat higher than those in Downloaded from the sleep-wake organization paradigm, although this may merely reflect the fact that greater drug concentrations may be required to demonstrate efficacy in a challenge model (PCP-

induced hyperactivity) compared to a more physiological situation (sleep-wake). While these jpet.aspetjournals.org findings provide more evidence that mGlu2 PAMs can mimic aspects of the pharmacology of mGlu2/3 receptor agonists, and hence have the potential to exhibit antipsychotic behavior,

studies are ongoing to provide more insight in the translation of compound activities in at ASPET Journals on September 30, 2021 preclinical animal models into relevant dose levels in a clinical setting.

Collectively, our data indicate that JNJ-40068782 is a selective PAM of native and recombinant rat and recombinant human mGlu2 receptors. It not only enhances the affinity of agonists, but also increases the maximal response. In the absence of exogenous glutamate,

JNJ-40068782 has a modest degree of intrinsic agonist efficacy. [3H]JNJ-40068782 represents an excellent novel radiotracer to study mGlu2 allosteric interaction, expression and distribution. With [3H]JNJ-40068782, we show for the first time that also agonists influence the affinity of PAMs. Finally, since JNJ-40068782 is active in vivo, it is a suitable pharmacological tool to increase our understanding how mGlu2 receptor modulation may affect diseases where glutamatergic transmission is disrupted.

31

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Acknowledgements

The authors wish to thank Stefan Pype and Claire Mackie for their continuous support;

Michel Mahieu and Heidi Huysmans for technical assistance; and Addex Therapeutics for their collaboration on the mGlu2 PAM discovery program, with special thanks to Francoise

Girard, Emmanuel Le Poul and Guillaume Duvey.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 30, 2021

32

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Authorship Contributions

Participated in research design: Lavreysen, Langlois, Ahnaou, Drinkenburg, te Riele,

Biesmans, Van der Linden, Peeters, Megens, Wintmolders, Macdonald, Lütjens,

Dautzenberg, Atack

Conducted experiments: Lavreysen, te Riele, Biesmans, Van der Linden, Peeters,

Wintmolders Downloaded from Contributed new reagents or analytic tools: Cid, Trabanco, Andrés

Performed data analysis: Lavreysen, Ahnaou, te Riele, Biesmans, Van der Linden, Peeters,

Megens, Wintmolders jpet.aspetjournals.org

Wrote or contributed to the writing of the manuscript: Lavreysen, Langlois, Ahnaou,

Drinkenburg, te Riele, Biesmans, Van der Linden, Peeters, Megens, Wintmolders, Cid,

Trabanco, Andrés, Macdonald, Lütjens, Dautzenberg, Atack at ASPET Journals on September 30, 2021

33

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

References

Ahnaou A, Dautzenberg FM, Geys H, Imogai H, Gibelin A, Moechars D, Steckler T and Drinkenburg WH

(2009) Modulation of group II metabotropic (mGlu2) elicits common changes in rat and

mice sleep-wake architecture. Eur J Pharmacol 603:62-72.

Al-Shamma HA, Anderson C, Chuang E, Luthringer R, Grottick AJ, Hauser E, Morgan M, Shanahan W,

Teegarden BR, Thomsen WJ and Behan D (2010) Nelotanserin, a novel selective human 5-

hydroxytryptamine2A inverse agonist for the treatment of insomnia. JPET 332: 281-290. Downloaded from

Auerbach BD, Osterweil EK, Bear MF (2011) Mutations causing syndromic autism define an axis of synaptic

pathophysiology. Nature 23:63-8.

Cheng Y and Prusoff WH (1973) Relationship between the inhibition constant (Ki) and the concentration of jpet.aspetjournals.org

inhibitor which causes 50 percent inhibition (IC50) of an enzymatic reaction. Biochem Pharmacol 22:3099-

3108.

Conn PJ, Lindsley CW, Jones C (2008) Activation of metabotropic glutamate receptors as a novel approach for at ASPET Journals on September 30, 2021 the treatment of schizophrenia. Trends Pharmacol Sc 30:25-31.

Conn PJ, Christopoulos A and Lindsley CW (2009) Allosteric modulators of GPCRs: a novel approach for the

treatment of CNS disorders. Nat Rev Drug Discov 8:41-54.

Cosford ND, Roppe J, Tehrani L, Schweiger EJ, Seiders TJ, Chaudary A, Rao S, Varney MA (2003) [3H]-

methoxymethyl-MTEP and [3H]-methoxy-PEPy: potent and selective radioligands for the metabotropic

glutamate subtype 5 (mGlu5) receptor. Bioorg Med Chem Lett 13:351-354.

Dash MB, Douglas CL, Vyazovskiy VV, Cirelli C, Tononi G (2009) Long-term homeostasis of extracellular

glutamate in the rat cerebral cortex across sleep and waking states. J Neurosci 29:620-629.

Dunayevich E, Erickson J, Levine L, Landbloom R, Schoepp DD and Tollefson GD (2008) Efficacy and

tolerability of an mGlu2/3 agonist in the treatment of generalized anxiety disorder. Neuropsychopharmacol

33:1603-1610.

Feinberg I, Campbell IG, Schoepp DD, Anderson K (2002) The selective group mGlu2/3 receptor agonist

LY379268 suppresses REM sleep and fast EEG in the rat. Pharmacol Biochem Behav 73:467-74.

Fell MJ, Svensson KA, Johnson BG and Schoepp DD (2008) Evidence for the role of metabotropic glutamate

(mGlu)2 not mGlu3 receptors in the preclinical antipsychotic pharmacology of the mGlu2/3 receptor

34

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

agonist (-)-(1R,4S,5S,6S)-4-amino-2-sulfonylbicyclo[3.1.0]hexane-4,6-dicarboxylic acid (LY404039). J

Pharmacol Exp Ther 326:209-217.

Fell MJ, Witkin JM, Falcone JF, Katner JS, Perry KW, Hart J, Rorick-Kehn L, Overshiner CD, Rasmussen K,

Chaney SF, Benvenga MJ, Li X, Marlow DL, Thompson LK, Luecke SK, Wafford KA, Seidel WF, Edgar

DM, Quets AT, Felder CC, Wang X, Heinz BA, Nikolayev A, Kuo MS, Mayhugh D, Khilevich A, Zhang

D, Ebert PJ, Eckstein JA, Ackermann BL, Swanson SP, Catlow JT, Dean RA, Jackson K, Tauscher-

Wisniewski S, Marek GJ, Schkeryantz JM, Svensson KA (2010) N-(4-((2-(trifluoromethyl)-3-hydroxy-4-

(isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4-carboxamide (THIIC), a novel Downloaded from metabotropic glutamate 2 potentiator with potential anxiolytic/antidepressant properties: in vivo profiling

suggests a link between behavioral and central nervous system neurochemical changes. J Pharmacol Exp

Ther 336:165-177. jpet.aspetjournals.org Fell MJ, McKinzie DL, Monn JA, Svensson, KA (2012) Group II metabotropic glutamate receptor agonists and

positive allosteric modulators as novel treatments for schizophrenia. Neuropharmacology 62: 1473-1483.

Galici R, Echemendia NG, Rodriguez AL and Conn PJ (2005) A selective allosteric potentiator of metabotropic

glutamate (mGlu) 2 receptors has effects similar to an orthosteric mGlu2/3 receptor agonist in mouse at ASPET Journals on September 30, 2021

models predictive of antipsychotic activity. J Pharmacol Exp Therap 315: 1181-1187.

Galici R, Jones CK, Hemstapat K, Nong Y, Echemendia NG, Williams LC, de Paulis T and Conn PJ (2006)

Biphenyl-Indanone A, a positive allosteric modulator of the metabotropic glutamate receptor Subtype 2,

has antipsychotic- and anxiolytic-Like effects in mice. J Pharmacol Exp Therap 318:173-185.

Gasparini F, Andres H, Flor PJ, Heinrich M, Inderbitzin W, Lingenhöhl K, Müller H, Munk VC, Omilusik K,

Stierlin C, Stoehr N, Vranesic I, Kuhn R (2002) [3H]-M-MPEP, a potent, subtype-selective radioligand for

the metabotropic glutamate receptor subtype 5. Bioorg Med Chem Lett 12:407-409.

Grillon C, Cordova J, Levine LR and Morgan III CA (2003). Anxiolytic effects of a novel group II metabotropic

glutamate receptor agonist (LY354740) in the fear-potentiated startle paradigm in humans.

Psychopharmacology 168:446–454.

Gu G, Lorrain DS, Wei H, Cole RL, Zhang X, Daggett LP, Schaffhauser HJ, Bristow LJ and Lechner SM (2008)

Distribution of metabotropic glutamate 2 and 3 receptors in the rat forebrain: Implication in emotional

responses and central disinhibition. Brain Res 1197:47-62.

35

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Happe HK, Bylund DB, Murrin LC (2001) Agonist-stimulated [35S]GTPgammaS autoradiography: optimization

for high sensitivity. Eur J Pharmacol 422:1-13.

Helton DR, Tizzano JP, Monn JA, Schoepp DD and Kallman MJ (1998) Anxiolytic and side-effect profile of

LY354740: a potent, highly selective, orally active agonist for group II metabotropic glutamate receptors. J.

Pharmacol Exp Ther 284:651-660.

Hill DR, Campbell NJ, Shaw TM, Woodruff GN (1987) Autoradiographic localization and biochemical

characterization of peripheral type CCK receptors in rat CNS using highly selective nonpeptide CCK

antagonists. J Neurosci 7:2967-2976. Downloaded from

Hlavackova V, Goudet C, Kniazeff J, Zikova A, Maurel D, Vol C, Trojanova J, Prézeau L, Pin J-P, Blahos J

(2005) Evidence for a single heptahelical domain being turned on upon activation of a dimeric GPCR. The

EMBO Journal 24:499-509. jpet.aspetjournals.org

Johnson MP, Baez M, Jagdmann GE Jr, Britton TC, Large TH, Callagaro DO, Tizzano JP, Monn JA and

Schoepp DD (2003) Discovery of allosteric potentiators for the metabotropic glutamate 2 receptor:

synthesis and subtype selectivity of N-(4-(2-methoxyphenoxy)phenyl)-N-(2,2,2- at ASPET Journals on September 30, 2021 trifluoroethylsulfonyl)pyrid-3-ylmethylamine. J Med Chem 46:3189-3192.

Johnson MP, Barda D, Britton TC, Emkey R, Hornback WJ, Jagdmann GE, McKinzie DL, Nisenbaum ES,

Tizzano JP and Schoepp DD (2005) Metabotropic glutamate 2 receptor potentiators: receptor modulation,

frequency-dependent synaptic activity, and efficacy in preclinical anxiety and psychosis model(s).

Psychopharmacol (Berl) 179:271-283.

Kew JN and Kemp JA (2005) Ionotropic and metabotropic glutamate receptor structure and pharmacology.

Psychopharmacology 179:4-29.

Kinon BJ, Zhang L, Millen BA, Osuntokun OO, Williams JE, Kollack-Walker S, Jackson K, Kryzhanovskaya L,

Jarkova N (2011) A multicenter, inpatient, phase 2, double-blind, placebo-controlled dose-ranging study of

LY2140023 monohydrate in patients with DSM-IV schizophrenia. J Clin Psychopharmacol 31: 349-355.

Kniazeff J, Bessis A-S, Maurel D, Ansanay H, Prézeau L, Pin J-P (2004) Closed state of both binding domains

of homodimeric mGlu receptors is required for full activity. Nature Struct & Mol Biol 11: 706-713.

Langmead CJ (2012) Ligand properties and behaviours in an allosteric age. Trends Pharmacol Sci 33: 621-622.

Lavreysen H, Janssen C, Bischoff F, Langlois X, Leysen JE and Lesage AS (2003) [3H]R214127: a novel high-

36

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

affinity radioligand for the mGlu1 receptor reveals a common binding site shared by multiple allosteric

antagonists. Mol Pharmacol 63:1082-1093.

Lavreysen H and Dautzenberg FM (2008) Therapeutic potential of group III metabotropic glutamate receptors.

Curr Med Chem 15:671-684.

Linden AM, Shannon H, Baez M, Yu JL, Koester A and Schoepp DD (2005) Anxiolytic-like activity of the

mGlu2/3 receptor agonist LY354740 in the elevated plus maze test is disrupted in metabotropic glutamate

receptor 2 and 3 knock-out mice. Psychopharmacol (Berl) 179:284-291.

Lopez-Rodriguez F, Medina-Ceja L, Wilson C, Jhung D, Morales-Villagran A (2007) Changes in extracellular Downloaded from

glutamate levels in rat orbitofrontal cortex during sleep and wakefulness. Archives of Medical Research

38:52-55.

Mezler M, Geneste H, Gault L and Marek GJ (2010) LY-2140023, a prodrug of the group II metabotropic jpet.aspetjournals.org

glutamate receptor agonist LY-404039 for the potential treatment of schizophrenia. Curr Opin Investig

Drugs 11:833-845.

Michelson D, Levine LR, Dellva MA, Mesters P, Schoepp DD, Dunayevich E (2005). Clinical studies with at ASPET Journals on September 30, 2021 mGlu2/3 receptor agonists: LY354740 compared with placebo in patients with generalized anxiety

disorder. Neuropharmacology 49(S1): 84–257.

Niswender CM, Conn PJ (2010) Metabotropic glutamate receptors: physiology, pharmacology, and disease

Annu Rev Pharmacol Toxicol 50:295–322.

Ohishi H, Shigemoto R, Nakanishi S and Mizuno N (1993) Distribution of the messenger RNA for a

metabotropic glutamate receptor, mGluR2, in the central nervous system of the rat. Neuroscience 53:1009-

1018.

Ohishi H, Neki A, Mizuno N (1998) Distribution of a metabotropic glutamate receptor, mGluR2, in the central

nervous system of the rat and mouse: an immunohistochemical study with a monoclonal antibody. Neurosci

Res 30:65-82.

Patil ST, Zhang L, Martenyi F, Lowe SL, Jackson KA, Andreev BV, Avedisova AS, Bardenstein LM, Gurovich

IY, Morozova MA, Mosolov SN, Neznanov NG, Reznik AM, Smulevich AB, Tochilov VA, Johnson BG,

Monn JA and Schoepp DD (2007) Activation of mGlu2/3 receptors as a new approach to treat

schizophrenia: a randomized Phase 2 clinical trial. Nat Med 13:1102-1107.

37

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Pinkerton AB, Cube RV, Hutchinson JH, James JK, Gardner MF, Rowe BA, Schaffhauser H, Rodriguez DE,

Campbell UC, Daggett LP, Vernier JM (2005) Allosteric potentiators of the metabotropic glutamate

receptor 2 (mGluR2). Part 3: Identification and biological activity of indanone containing mGluR2 receptor

potentiators. Bioorg Med Chem Lett 15:1565–1571.

Richards G, Messer J, Malherbe P, Pink R, Brockhaus M, Stadler H, Wichmann J, Schaffhauser H and Mutel V

(2005) Distribution and abundance of metabotropic glutamate receptor subtype 2 in rat brain revealed by

[3H]LY354740 binding in vitro and quantitative radioautography: correlation with the sites of synthesis,

expression, and agonist stimulation of [35S]GTPgammaS binding. J Comp Neurol 487:15-27. Downloaded from Rorick-Kehn LM, Johnson BG, Knitowski KM, Salhoff CR, Witkin JM, Perry KW, Griffey KI, Tizzano JP,

Monn JA, McKinzie DL and Schoepp DD (2007) In vivo pharmacological characterization of the

structurally novel, potent, selective mGlu2/3 receptor agonist LY404039 in animal models of psychiatric jpet.aspetjournals.org disorders. Psychopharmacol (Berl) 193:121-136.

Schaffhauser H, Rowe BA, Morales S, Chavez-Noriega LE, Yin R, Jachec C, Rao SP, Bain G, Pinkerton AB,

Vernier JM, Bristow LJ, Varney MA and Daggett LP (2003) Pharmacological characterization and

identification of amino acids involved in the positive modulation of metabotropic glutamate receptor at ASPET Journals on September 30, 2021

subtype 2. Mol Pharmacol 64:798–810.

Schoepp DD, Wright RA, Levine LR, Gaydos B and Potter WZ (2003) LY354740, an mGlu2/3 receptor agonist

as a novel approach to treat anxiety/stress. Stress 6:189–197.

Spooren WP, Gasparini F, van der Putten H, Koller M, Nakanishi S and Kuhn R (2000) Lack of effect of

LY314582 (a group 2 metabotropic glutamate receptor agonist) on phencyclidine-induced locomotor

activity in metabotropic glutamate receptor 2 knockout mice. Eur J Pharmacol 397:R1-2.

Spooren W, Ballard T, Gasparini F, Amalric M, Mutel V, Schreiber R (2003) Insight into the function of Group I

and Group II metabotropic glutamate (mGlu) receptors: behavioural characterization and implications for

the treatment of CNS disorders. Behav Pharmacol 14:257-77.

Steckler T, Lavreysen H, Oliveira AM, Aerts N, Van Craenendonck H, Prickaerts J, Megens A, Lesage AS.

(2005) Effects of mGlu1 receptor blockade on anxiety-related behaviour in the rat lick suppression test.

Psychopharmacology (Berl) 179:198-206.

38

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Swanson CJ, Bures M, Johnson MP, Linden AM, Monn JA and Schoepp DD (2005) Metabotropic glutamate

receptors as novel targets for anxiety and stress disorders. Nat Rev Drug Discov 4:131-144.

Tatarczyńska E, Kłodzińska A, Kroczka B, Chojnacka-Wójcik E, Pilc A (2001) The antianxiety-like effects of

antagonists of group I and agonists of group II and III metabotropic glutamate receptors after

intrahippocampal administration. Psychopharmacology (Berl) 158:94-9.

Tizzano JP, Griffey KI and Schoepp DD (2002) The anxiolytic action of mGlu2/3 receptor agonist, LY354740,

in the fear-potentiated startle model in rats is mechanistically distinct from diazepam. Pharmacol Biochem

Behav 73:367-374. Downloaded from

Varney MA, Gereau RW 4th (2002) Metabotropic glutamate receptor involvement in models of acute and

persistent pain: prospects for the development of novel analgesics. Curr Drug Targets CNS Neurol Disord

1:283-96. jpet.aspetjournals.org

Witkin JM, Marek GJ, Johnson BG and Schoepp DD (2007) Metabotropic glutamate receptors in the control of

mood disorders. CNS Neurol Disor Drug Targets 6:87-100.

Woolley ML, Pemberton DJ, Bate S, Corti C, Jones DNC (2008) The mGlu2 but not the mGlu3 receptor at ASPET Journals on September 30, 2021 mediates the actions of the mGluR2/3 agonist, LY379268, in mouse models predictive of antipsychotic

activity. Psychopharmacology 196:431-440.

Wright RA, Johnson BG, Zhang C, Salhoff C, Kingston AE, Calligaro DO, Monn JA, Schoepp DD, Marek GJ

(2012) CNS distribution of metabotropic glutamate 2 and 3 receptors: transgenic mice and [3H]LY459477

autoradiography. Neuropharmacology 2012 Jan 31 (Epub ahead of print).

39

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Figure Legends

Figure 1. Structure of JNJ-40068782. The asterix denotes the position of the tritium in [3H]JNJ-40068782.

Figure 2. [35S]GTPγS binding to hmGlu2 receptors expressed in CHO cells. A. Stimulation of [35S]GTPγS binding produced by glutamate in the absence and presence of various concentrations of JNJ-40068782; BINA;

LY487379; and LY2607540. Data are from a representative experiment. The experiment was repeated at least twice giving similar results. B. The concentration-effect curves for JNJ-40068782, BINA, LY487379 and

35 LY2607540 on the binding of [ S]GTPγS at an EC20-equivalent concentration of glutamate. Data are expressed Downloaded from as percentage of the maximal response to glutamate and are mean ± S.E.M of 6-13 experiments. C. The concentration-effect curves for JNJ-40068782 on the binding of [35S]GTPγS versus mobilization of Ca2+ at an

EC -equivalent concentration of glutamate (Glu) . Data are expressed as percentage of the maximal response to

20 jpet.aspetjournals.org glutamate and are mean ± S.E.M of 6 ([35S]GTPγS) or 30 (Ca2+) experiments.

Figure 3. Binding of [35S]GTPγS to rat brain sections revealed by quantitative autoradiography. A. Sagital rat at ASPET Journals on September 30, 2021 brain sections were incubated with various concentration of JNJ-40068782 in the absence or presence of 10 μM glutamate. The top left brain section represents, in absence of glutamate and JNJ-40068782, the basal

[35S]GTPγS binding. B. Quantification of the data represented in the images. Bar graphs represent the average

(n = 3 rats) percent stimulation by glutamate and/or PAM over basal binding.

Figure 4. [3H]LY341495 (A) and [3H]DCG-IV (B) binding in the presence of JNJ-40068782 and other mGlu2 receptor PAMs.

Figure 5. A. Saturation binding of the agonist [3H]DCG-IV to human recombinant mGlu2 receptors in the absence and presence of 3 μM JNJ-40068782. In the inset, the Scatchard analysis is shown. B. Displacement of

[3H]DCG-IV from the human mGlu2 receptor by glutamate in the absence and presence of JNJ-40068782. Data are from one representative experiment.

Figure 6. A. Binding of [3H]JNJ-40068782 to human recombinant mGlu2 receptors. A. Representative saturation curve showing the total and specific binding of [3H]JNJ-40068782, with specific binding being calculated as

40

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

total binding minus nonspecific binding. B. Saturation binding in the absence and presence of the orthosteric

(glutamate) binding site agonists glutamate (100 μM), DCG-IV (100 μM) and LY354740 (1 μM).

Figure 7. JNJ-40068782, BINA, LY487379 and LY2607540 displacement of [3H]JNJ-40068782 binding to

human recombinant (A) and rat cortical (B) mGlu2 receptors. Data are expressed as % specific binding ± S.E.M.

and are from 3-9 experiments.

Figure 8. A. Binding of 10 nM [3H]JNJ-40068782 to rat brain sections and comparison with the labelling of Downloaded from group II mGlu receptors with the mGlu2/3 receptor antagonist [3H]LY341495. B. Binding of 10 nM [3H]JNJ-

40068782 to wild-type versus mGlu2 knock-out mouse brain sections.

jpet.aspetjournals.org Figure 9. Percentage distribution of sleep-wake states for each h period of the 20 h recording session following oral administration of JNJ-40068782 (3, 10, 30 mg/kg) or vehicle. Open and dark areas in the abscissa axis indicate light and dark phase of the circadian time, respectively. Small bar charts indicate amounts of vigilance

states in minutes (plus S.E.M.) during the first 2 hours post-administration (n=8 for each group). Small bottom at ASPET Journals on September 30, 2021 right upper panel indicates REM sleep onset latency (ROL). Values are presented as means ± S.E.M. for each condition. Each symbol on curve plots indicate statistically significant difference (P<0.05) between vehicle and drug-dose injected groups. *indicates p<0.05: Wilcoxon-Mann-Whitney rank sum tests as compared to vehicle values.

Figure 10. Effect of JNJ-40068782 (0, 2.5, 5, 10 and 20 mg/kg, s.c.) on spontaneous locomotion (A) and PCP (B)

(5 mg/kg, s.c.; -0.5 h)-induced hyperlocomotion in mice. The dotted lines at 2500 and 5500 cm indicate the critical level for hypolocomotion and hyperlocomotion, respectively.

41

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Tables

Table 1. Functional activity of JNJ-40068782, BINA, LY487379 and LY2607540 at human and rat recombinant

mGlu2 receptors, in absence or presence of an EC20 concentration of glutamate. Values are an average of 6-13

experiments, %Emax values are indicated as mean ± S.E.M., EC50 values are geometric means with 95%

confidence intervals indicated in brackets.

absence of glutamate presence of EC20 glutamate

% Emax EC50 (nM) % Emax EC50 (nM) Downloaded from human mGlu2

JNJ-40068782 69 ± 9 925 (573 - 1493) 309 ± 23 143 (108-190) jpet.aspetjournals.org

BINA 60 ± 3 572 (141 – 2324) 221 ± 9 89 (48-162)

LY487379 20 ± 3 >50.000 147 ± 9 256 (193-337)

LY2607540 28 ± 2 720 (631 – 821) 215 ± 7 95 (77-117) at ASPET Journals on September 30, 2021 rat mGlu2

JNJ-40068782 31 ± 3.7 >30.000 444 ± 86 307 (287 – 327)

BINA 35 ± 4.9 >30.000 390 ± 63 138 (57-336)

LY487379 7.4 ± 3.9 >30.000 166 ± 14 501 (417-602)

LY2607540 13 ± 1.5 >30.000 273 ± 10 314 (251 – 392)

42

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. JPET#_204990R

Table 2: Characteristics of [3H]DCG-IV binding to human recombinant mGlu2 receptors in the absence and

presence of JNJ-40068782, BINA and LY487379. The KD and Bmax of [3H]DCG-IV in the absence of

compound is an average ± S.E.M. of 6 experiments. The same parameters measured in the presence of

compound come from 2 independent experiments (values from both experiments are given).

[3H]DCG-IV binding

KD (nM) Bmax (pmol/mg protein)

Control 240 ± 39 14.5 ± 2.8 Downloaded from

JNJ-40068782 (3 μM) 22 / 39 16.6 / 7.6

BINA (3 μM) 47 / 81 15.5 / 9.0 jpet.aspetjournals.org LY487379 (3 μM) 35 / 53 17.3 / 7.0

at ASPET Journals on September 30, 2021

43

JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from jpet.aspetjournals.org at ASPET Journals on September 30, 2021

Figure 1 O N N

N

* JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from jpet.aspetjournals.org at ASPET Journals on September 30, 2021 Figure 2 A B JNJ-40068782 BINA 4000 4000 350 + 10 µM + 10 µM JNJ-40068782 + 1 µM + 1 µM LY2607540 3500 3500 300 + 300 nM + 300 nM BINA + 100 nM + 100 nM LY487379 3000 3000 250 + 50 nM + 50 nM 2500 + 10 nM 2500 + 10 nM 200 + 5 nM + 5 nM 2000 control 2000 control cpm cpm 150 1500 1500 100

1000 1000 % 1 mM Glu response 50 500 500 0 0 0 -9 -8 -7 -6 -5 -4 -3 -2 -9 -8 -7 -6 -5 -4 -3 -2 -11 -10 -9 -8 -7 -6 -5 -4 Log [Glutamate], M Log [Glutamate], M Log [compound], M

LY487379 LY2607540 C 4000 4000 400 + 1 µM + 10 µM [35S]GTPγS binding + 1 µM 3500 + 500 nM 3500 350 Ca2+ assay + 300 nM + 300 nM + 100 nM 3000 + 100 nM 3000 300 + 50 nM + 50 nM 2500 2500 + 10 nM 250 + 10 nM + 5 nM 2000 control 2000 control 200 cpm cpm 150 1500 1500 100

1000 1000 % of maximal Glu effect 50 500 500 0 0 0 -9 -8 -7 -6 -5 -4 -3 -2 -9 -8 -7 -6 -5 -4 -3 -2 -12 -11 -10 -9 -8 -7 -6 -5 -4 Log [Glutamate], M Log [Glutamate], M Log [JNJ-40068782], M JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from jpet.aspetjournals.org at ASPET Journals on September 30, 2021

Figure 3 no glutamate 10 µM glutamate

no JNJ-40068782

1 µM JNJ-40068782

10 µM JNJ-40068782

100 µM JNJ-40068782

250 no glutamate 10 µM glutamate

200

150

100

S]GTPγS binding (% stimulation) 50 35 [

0 0 µM 1 µM 10 µM 100 µM Concentration of JNJ-40068782 JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from jpet.aspetjournals.org at ASPET Journals on September 30, 2021

Figure 4 A [3H]LY341495 B [3H]DCG-IV orthosteric mGlu2/3 antagonist orthosteric mGlu2/3 agonist 3500 30000

3000 25000 2500 20000 2000 15000 DPM DPM 1500 10000 1000 500 5000 0 0

control control 100 µM Glu1 µM BINA 3 µM BINA 10 µM LY487379 10 µM JNJ-40068782 10 µM LY341495 3 µM LY487379 3 µM JNJ-40068782 JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from jpet.aspetjournals.org at ASPET Journals on September 30, 2021

Figure 5 A 20.0 3 µM JNJ-40068782 no JNJ-40068782 17.5

15.0

12.5

10.0 0.08 7.5 0.06 0.04 5.0 0.02 2.5 0.00 Specific Binding (pmol/mg protein) 0 5 10 15 20 SB (pmol/mg protein) 0.0 0 100 200 300 400 500 600 [3H]DCG-IV, nM

25000 B control 3 µM JNJ-40068782 20000

15000 DPM 10000

5000

0 -10 -8 -6 -4 -2 [glutamate], Log (M) JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from jpet.aspetjournals.org at ASPET Journals on September 30, 2021

Figure 6 A 5.0 4.5 4.0 0.10 3.5 0.08 3.0 2.5 0.06 SB/F 2.0 0.04

1.5 0.02

1.0 0.00 0 1 2 3 4 5 0.5

Specific Binding (pmol/mg protein) SB (pmol/mg protein) 0.0 0 10 20 30 40 50 60 70 80 90 B [3H]JNJ-40068782, nM 10

8

6

4

2 control +100 µM glutamate +100 µM DCG-IV Specific Binding (pmol/mg protein) +1 µM LY354740 0 0 20 40 60 80 [3H]H]JNJ-40068782, nM JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from

Figure 7 A hmGlu2 jpet.aspetjournals.org 140 JNJ-40068782 BINA 120 LY487379 LY2607540 100 at ASPET Journals on September 30, 2021 80

60 % specific binding 40

20

0

-10 -9 -8 -7 -6 -5 Log [compound], M

B rat cortex 140 JNJ-40068782 BINA 120 LY487379 LY2607540 100

80

60 % specific binding 40

20

0

-11 -10 -9 -8 -7 -6 -5 -4 Log [compound], M JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from jpet.aspetjournals.org at ASPET Journals on September 30, 2021

Figure 8 A [3H]JNJ-40068782 [3H]LY341495 Hip non-specific binding Cx Cer Str

B wild-type mGlu2 KO

Str

Hip

Cer JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from jpet.aspetjournals.org at ASPET Journals on September 30, 2021 Figure 9 JNJ-40068782 (mg/kg) Vehicle 3 10 30

100 Active 25 Light wake sleep 80 20

60 15 40 30 40 30 10 20 20 10 20 10 5 0 0 0 0 0 1 2 3 4 5 6 7 8 9 1011121314151617181920 0 1 2 3 4 5 6 7 8 9 1011121314151617181920

16 Passive 80 Deep wake sleep 12 60

8 20 40 80 15 60 10 40 4 5 20 20 0 0 0 0 0 1 2 3 4 5 6 7 8 9 1011121314151617181920 0 1 2 3 4 5 6 7 8 9 1011121314151617181920 State amount (% recording time) ROL 4 Inter- 20 REM 200 160 * * mediate sleep 120 16 3 state 80 40 12 0 2 2.0 15 1.5 8 10 * 1.0 * 1 5 * 0.5 * 4 0.0 0 0 0 0 1 2 3 4 5 6 7 8 9 1011121314151617181920 0 1 2 3 4 5 6 7 8 9 1011121314151617181920 Time (# hours post-administration) JPET Fast Forward. Published on June 13, 2013 as DOI: 10.1124/jpet.113.204990 This article has not been copyedited and formatted. The final version may differ from this version. Downloaded from

Figure 10 A Spontaneous locomotion 20000 jpet.aspetjournals.org

15000

10000 at ASPET Journals on September 30, 2021 total distance (cm)

5000

0

0 JNJ-40068782 5 JNJ-40068782 2.5 JNJ-40068782 10 JNJ-4006878220 JNJ-40068782

B PCP-induced hyperlocomotion 20000

15000

10000 total distance (cm)

5000

0

veh/veh

0 JNJ-40068782 5 JNJ-40068782 2.5 JNJ-40068782 10 JNJ-4006878220 JNJ-40068782

PCP (5 mg/kg, s.c.) JPET#_204990R

Pharmacological characterization of JNJ-40068782, a new potent, selective and systemically active positive allosteric modulator of the mGlu2 receptor and its radioligand [3H]JNJ-40068782

Hilde Lavreysen, Xavier Langlois, Abdel Ahnaou, Wilhelmus Drinkenburg, Paula te Riele,

Ilse Biesmans, Ilse Van der Linden, Luc Peeters, Anton Megens, Cindy Wintmolders, José

Cid, Andrés A. Trabanco, Ignacio Andrés, Frank M. Dautzenberg, Robert Lütjens, Gregor

Macdonald, John Atack

JPET#_204990R

Supplemental Data

Supplemental Table 1

Interaction of JNJ-40068782 with a broad selection of other neurotransmitter targets (data from CEREP).

Species Target %inhibition at 10 M human 5-HT2A 59 ± 8 human 5-HT2B - human 5-HT1A - rat 5-HT1B - human 5-HT7 24 ± 2 human 5-HT3 - human 5-HT5a 10 ± 4 human 5-HT6 - rat alpha1 (non-selective) - rat alpha2 (non-selective) - human beta1 - human D2S - human H1 - human DAT 48 ± 8 human AT1 - rat BZD (central) 10 ± 2 human B2 - human CCK1 (CCKA) 85 ± 5 human D1 - human ETA - rat GABA (non-selective) - human GAL2 - human CXCR2 (IL-8B) - human CCR1 - human H2 25 ± 1 human MC4 - human MT1 (ML1A) 32 ± 6 human M1 12 ± 9 human M2 27 ± 9 human M3 - human NK2 10 ± 4 human NK3 - human Y1 - human Y2 - human A1 10 ± 5 human A2A - human A3 22 ± 3 human NTS1 (NT1) - human delta 2 (DOP) - guinea pig kappa (KOP) 25 ± 8 human mu (MOP) 17 ± 9 human NOP (ORL1) - mouse sst (non-selective) - human VPAC1 (VIP1) - human V1a - rat Ca2+ channel 13 ± 5 rat Na+ channel 26 ± 3 rat KV channel - rat SKCa channel - rat Cl- channel (GABA-gated) - human norepinephrine transporter 19 ± 7

Results are expressed as a percent inhibition of control specific binding by JNJ-40068782 at a concentration of 10 µM, average ± S.D. ‘-‘ indicates an inhibition of less than 10%

JPET#_204990R

Supplemental Figure 1

35 Correlation between [ S]GTPS data (pEC50 and Emax) obtained from the human versus rat mGlu2 receptor expressed in CHO cells (line of unity is indicated in blue)

50 S – pEC  S ]GTP 35 human mGlu2 PAM [ PAM mGlu2 human

rat mGlu2 PAM [35S ]GTPS – pEC 50

max S -E  S]GTP 35 human mGlu2 PAM [ PAM mGlu2 human

rat mGlu2 PAM [35S]GTP S -E  max