antidepressant and anxiolytic potential of the multimodal - ea3544

33
Antidepressant and anxiolytic potential of the multimodal antidepressant vortioxetine (Lu AA21004) assessed by behavioural and neurogenesis outcomes in mice q Jean-Philippe Guilloux a , Indira Mendez-David a , Alan Pehrson b , Bruno P. Guiard a , Christelle Repérant a , Sophie Orvoën a , Alain M. Gardier a , René Hen d, e , Bjarke Ebert c , Silke Miller b, 1 , Connie Sanchez b , Denis J. David a, * a Univ Paris-Sud, EA3544, Faculté de Pharmacie, Châtenay-Malabry F-92296, France b Lundbeck Research USA, 215 College Rd, Paramus, NJ 07652, USA c H. Lundbeck A/S, Medical Affairs Mood & Anxiety, Ottiliavej 9, DK-2500 Valby, Denmark d Department of Neuroscience, Columbia University, New York, NY 10032, USA e Department of Psychiatry, Columbia University, New York, NY 10032, USA article info Article history: Received 26 December 2012 Received in revised form 6 May 2013 Accepted 7 May 2013 Keywords: Multimodal antidepressant Anxiolytic Antidepressant Behaviour Mice Neurogenesis abstract Vortioxetine (Lu AA21004) is an investigational novel antidepressant with multimodal activity that functions as a 5-HT 3 , 5-HT 7 and 5-HT 1D receptor antagonist, 5-HT 1B receptor partial agonist, 5-HT 1A receptor agonist and inhibitor of the 5-HT transporter in vitro. Here we explore its anxiolytic and anti- depressant potential in adult mice. Vortioxetine was assessed in BalB/cJ@RJ mice using the open-eld and forced-swim tests (acute: p.o. 1 h, repeated: daily p.o. 21 days), and in 129S6/SvEvTac mice using the novelty suppressed feeding paradigm (acute: p.o. 1 h, sustained: daily p.o. 14 or 21 days). Fluoxetine and diazepam were controls. Acute and repeated dosing of vortioxetine produced more pronounced anxiolytic- and antidepressant-like activities than uoxetine. Vortioxetine signicantly increased cell proliferation and cell survival and stimulated maturation of immature granule cells in the subgranular zone of the dentate gyrus of the hippocampus after 21 days of treatment. After 14 days, a high dose of vortioxetine increased dendritic length and the number of dendrite intersections, suggesting that vor- tioxetine accelerates the maturation of immature neurons. Vortioxetine displays an antidepressant and anxiolytic prole following repeated administration associated with increased neurogenesis at several stages. Vortioxetine effects were observed at low levels of 5-HT transporter occupancy, suggesting an alternative mechanism of action to 5-HT reuptake inhibition. Ó 2013 The Authors. Published by Elsevier Ltd. All rights reserved. 1. Introduction Depression is a major psychiatric disease, with a z17% lifetime prevalence (Kessler et al., 2005). Rates of response to initial pharmacotherapy can vary from 30 to 60% depending on the studies while remission rates in the rst step of the STAR*D study was z37% (Guilloux et al., 2012; Rush et al., 2006). Side effects with Selective Serotonin Reuptake Inhibitors (SSRIs) are commonly re- ported during chronic treatment, notably insomnia, somnolence, dizziness, akathisia, and long-term sexual dysfunction (e.g., decreased libido, delayed ejaculation) (Hamon and Bourgoin, 2006). The modest efcacy of conventional antidepressants such as the selective serotonin (5-HT) reuptake inhibitors (SSRIs) calls for novel approaches to treat depression and anxiety disorders. Combinatorial pharmacological therapies, such as additional blockade of aminergic receptors in addition to monoamine trans- porter inhibition, have earlier been proposed to shorten the time to antidepressant effect and/or to increase efcacy in clinical studies (Artigas et al., 2006; Kennedy et al., 2011). The 5-HT 1A and the 5-HT 1B receptors were the rst serotonergic receptors targeted to treat anxiety and depression due to their q This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-No Derivative Works License, which per- mits non-commercial use, distribution, and reproduction in any medium, provided the original author and source are credited. * Corresponding author. Tel.: þ33 1 46 83 59 68; fax: þ33 1 46 83 53 55. E-mail addresses: [email protected] (J.-P. Guilloux), [email protected] (I. Mendez-David), [email protected] (A. Pehrson), [email protected] (B.P. Guiard), [email protected] (C. Repérant), [email protected] (S. Orvoën), [email protected] (A.M. Gardier), [email protected] (R. Hen), [email protected] (B. Ebert), [email protected] (S. Miller), [email protected] (C. Sanchez), [email protected] (D.J. David). 1 Present address: Amgen Inc., One Amgen Center Dr., MS 29-2-B, Thousand Oaks, CA 91320, USA. Contents lists available at SciVerse ScienceDirect Neuropharmacology journal homepage: www.elsevier.com/locate/neuropharm 0028-3908/$ e see front matter Ó 2013 The Authors. Published by Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.neuropharm.2013.05.014 Neuropharmacology 73 (2013) 147e159

Upload: others

Post on 11-Feb-2022

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Antidepressant and anxiolytic potential of the multimodal - EA3544

Antidepressant and anxiolytic potential of the multimodalantidepressant vortioxetine (Lu AA21004) assessed by behaviouraland neurogenesis outcomes in miceq

Jean-Philippe Guilloux a, Indira Mendez-David a, Alan Pehrson b, Bruno P. Guiard a,Christelle Repérant a, Sophie Orvoën a, Alain M. Gardier a, René Hen d,e, Bjarke Ebert c,Silke Miller b,1, Connie Sanchez b, Denis J. David a,*

aUniv Paris-Sud, EA3544, Faculté de Pharmacie, Châtenay-Malabry F-92296, Franceb Lundbeck Research USA, 215 College Rd, Paramus, NJ 07652, USAcH. Lundbeck A/S, Medical Affairs Mood & Anxiety, Ottiliavej 9, DK-2500 Valby, DenmarkdDepartment of Neuroscience, Columbia University, New York, NY 10032, USAeDepartment of Psychiatry, Columbia University, New York, NY 10032, USA

a r t i c l e i n f o

Article history:Received 26 December 2012Received in revised form6 May 2013Accepted 7 May 2013

Keywords:Multimodal antidepressantAnxiolyticAntidepressantBehaviourMiceNeurogenesis

a b s t r a c t

Vortioxetine (Lu AA21004) is an investigational novel antidepressant with multimodal activity thatfunctions as a 5-HT3, 5-HT7 and 5-HT1D receptor antagonist, 5-HT1B receptor partial agonist, 5-HT1Areceptor agonist and inhibitor of the 5-HT transporter in vitro. Here we explore its anxiolytic and anti-depressant potential in adult mice. Vortioxetine was assessed in BalB/cJ@RJ mice using the open-fieldand forced-swim tests (acute: p.o. 1 h, repeated: daily p.o. 21 days), and in 129S6/SvEvTac mice usingthe novelty suppressed feeding paradigm (acute: p.o. 1 h, sustained: daily p.o. 14 or 21 days). Fluoxetineand diazepam were controls. Acute and repeated dosing of vortioxetine produced more pronouncedanxiolytic- and antidepressant-like activities than fluoxetine. Vortioxetine significantly increased cellproliferation and cell survival and stimulated maturation of immature granule cells in the subgranularzone of the dentate gyrus of the hippocampus after 21 days of treatment. After 14 days, a high dose ofvortioxetine increased dendritic length and the number of dendrite intersections, suggesting that vor-tioxetine accelerates the maturation of immature neurons. Vortioxetine displays an antidepressant andanxiolytic profile following repeated administration associated with increased neurogenesis at severalstages. Vortioxetine effects were observed at low levels of 5-HT transporter occupancy, suggesting analternative mechanism of action to 5-HT reuptake inhibition.

! 2013 The Authors. Published by Elsevier Ltd. All rights reserved.

1. Introduction

Depression is a major psychiatric disease, with a z17% lifetimeprevalence (Kessler et al., 2005). Rates of response to initial

pharmacotherapy can vary from 30 to 60% depending on thestudies while remission rates in the first step of the STAR*D studywasz37% (Guilloux et al., 2012; Rush et al., 2006). Side effects withSelective Serotonin Reuptake Inhibitors (SSRIs) are commonly re-ported during chronic treatment, notably insomnia, somnolence,dizziness, akathisia, and long-term sexual dysfunction (e.g.,decreased libido, delayed ejaculation) (Hamon and Bourgoin,2006). The modest efficacy of conventional antidepressants suchas the selective serotonin (5-HT) reuptake inhibitors (SSRIs) callsfor novel approaches to treat depression and anxiety disorders.Combinatorial pharmacological therapies, such as additionalblockade of aminergic receptors in addition to monoamine trans-porter inhibition, have earlier been proposed to shorten the time toantidepressant effect and/or to increase efficacy in clinical studies(Artigas et al., 2006; Kennedy et al., 2011).

The 5-HT1A and the 5-HT1B receptors were the first serotonergicreceptors targeted to treat anxiety and depression due to their

q This is an open-access article distributed under the terms of the CreativeCommons Attribution-NonCommercial-No Derivative Works License, which per-mits non-commercial use, distribution, and reproduction in any medium, providedthe original author and source are credited.* Corresponding author. Tel.: þ33 1 46 83 59 68; fax: þ33 1 46 83 53 55.

E-mail addresses: [email protected] (J.-P. Guilloux),[email protected] (I. Mendez-David), [email protected] (A. Pehrson),[email protected] (B.P. Guiard), [email protected] (C. Repérant),[email protected] (S. Orvoën), [email protected] (A.M. Gardier),[email protected] (R. Hen), [email protected] (B. Ebert), [email protected](S. Miller), [email protected] (C. Sanchez), [email protected] (D.J. David).

1 Present address: Amgen Inc., One Amgen Center Dr., MS 29-2-B, ThousandOaks, CA 91320, USA.

Contents lists available at SciVerse ScienceDirect

Neuropharmacology

journal homepage: www.elsevier .com/locate/neuropharm

0028-3908/$ e see front matter ! 2013 The Authors. Published by Elsevier Ltd. All rights reserved.http://dx.doi.org/10.1016/j.neuropharm.2013.05.014

Neuropharmacology 73 (2013) 147e159

Page 2: Antidepressant and anxiolytic potential of the multimodal - EA3544

localization at the pre- and post-synaptic levels. Both receptorsmodulate serotonergic neurotransmission (Gingrich and Hen,2001; Guilloux et al., 2011). For instance, pindolol, a beta adreno-ceptor blocker with 5-HT1A receptor partial agonism, has shownsome efficacy in augmentation strategies with SSRIs; however, thelow doses used in clinical studies, its antagonistic action on 5-HT1Bheteroreceptors combined with its effects on post-synaptic 5-HT1Areceptors limits its efficacy (Guilloux et al., 2006;Whale et al., 2010;Martiny et al., 2012).

A link between the activity of antidepressant drugs and 5-HT3receptor function has been suggested since 5-HT3 receptor antag-onists administered alone exert antidepressant- and anxiolytic-likeeffects in preclinical settings (Costall and Naylor, 2004). Moreover,pretreatment with the 5-HT3 receptor antagonist ondansetronpotentiates the effects of antidepressant drugs in preclinical models(Redrobe and Bourin, 1997; Ramamoorthy et al., 2008). However,the preclinical observations were not confirmed in the few clinicalstudies that have been conducted. Thus, there is currently a weaksupport of 5-HT3 antagonism alone or in combination with SSRI inthe treatment of depression. Furthermore, whereas selective 5-HT3receptor antagonists are used routinely to attenuate nausea asso-ciated with chemotherapy, irradiation or cisplatin treatment, thereare only few studies conducted showing that 5-HT3 receptorantagonism reduces nausea in patients being treated with SSRIs(Bailey et al., 1995).

Early indications of an involvement of 5-HT7 receptors in mooddisorders came from a study showing down-regulation of 5-HT7receptor expression after chronic treatment with various antide-pressants (Mullins et al., 1999; for a review, see Mnie-Filali et al.,2011). Recent studies further support a role for 5-HT7 receptors intreating depression. Hence, SB-269970, a 5-HT7 receptor antago-nist, decreased immobility in both the tail suspension and forcedswim tests (Guscott et al., 2005; Hedlund et al., 2005; Faure et al.,2006; Wesolowska et al., 2006a, 2006b) and enhanced theantidepressant-like effect of citalopram (Bonaventure et al., 2007;Sarkisyan et al., 2010). In agreement with these pharmacologicaldata, 5-HT7 receptor knockout mice showed reduced immobility inboth the forced swim and the tail suspension tests (Hedlund et al.,2005). Thus, it appears that the efficacy of SSRIs may be enhancedby blocking feedback systems and modulating relevant receptors.

Vortioxetine (Lu AA21004; 1-[2-(2,4-dimethyl-phenylsulfanyl)-phenyl]-piperazine) is a novel investigational antidepressant withmultimodal activity. Vortioxetine acts as an inhibitor at the 5-HTtransporter (SERT, Ki ¼ 1.6 nM) in recombinant cells expressinghuman receptors or SERT and as a 5-HT3, 5-HT7 and 5-HT1D re-ceptor antagonist (Ki ¼ 3.7, 19 and 54 nM, respectively), a partialagonist at the 5-HT1B receptor (Ki ¼ 33 nM), an agonist at the 5-HT1A receptor (Ki ¼ 15 nM) (Bang-Andersen et al., 2011; Morket al., 2012; Westrich et al., 2012). In rats the binding affinitiesare Ki ¼ 1.1, 200, 3.7, 16 and 230 nM, for 5-HT3, 5-HT7, 5-HT1D, 5-HT1B, and 5-HT1A receptors, respectively and Ki ¼ 8.6 nM for theSERT (Mork et al., 2012; Westrich et al., 2012). In vivo, vortioxetineincreases the extracellular levels of 5-HT, noradrenaline (NA), anddopamine (DA) in rat prefrontal cortex and hippocampus (Morket al., 2012).

Although preclinical findings indicate that, acutely, vortioxetineproduces an antidepressant and anxiolytic profile (Mork et al.,2012), the behavioural consequences of chronic administrationhave not been described. To investigate the effects of chronic vor-tioxetine treatment, as well as to confirm its anxiolytic- andantidepressant-like activities, we assessed its behavioural effectsafter acute (1 h) or repeated (14 or 21 days) dosing using the openfield (OF) paradigm, the novelty suppressed feeding (NSF) para-digm and the mouse forced swim test (FST). OF and FST studieswere conducted in Balb/cJ mice that have been shown to display a

high basal anxiety- and depression-like behaviour (Belzung andGriebel, 2001). We confirmed the behavioural effects of 14 or 21days of treatment with vortioxetine in 129S6/SvEvTac mice in theNSF paradigm. As stimulation of hippocampal neurogenesis hasbeen suggested to underlie the delayed onset of therapeutic effi-cacy of SSRIs and tricyclic antidepressants (Duman et al., 1999;Malberg et al., 2000; Santarelli et al., 2003), we investigated theeffects of vortioxetine dosed for 14 or 21 days on cell proliferationand maturation/survival in the dentate gyrus in 129S6/SvEvTacmice.

2. Methods

2.1. Animals

One hundred and eighty BALB/cJ@Rj male mice, 7e8 weeks old (25e30 g, Centred’élevage Janvier, Le Genest-St-Isle, France) were used for the acute and repeateddosing experiments in the OF and FST. Eighty 129S6/SvEvTac male mice, 7e8 weeksold (25e30 g, Taconic Farms, Denmark) were used for the acute and repeated dosingNSF and the cell proliferation and survival/maturation study and Sholl analysis.

Mice were maintained under standard conditions (12/12 h light/dark cycle,lights on at 6AM, 22 # 1 $C, food and water ad libitum, 5 mice/cage). The protocolsinvolving animals and their care were conducted in conformity with the institu-tional guidelines that are in compliance with national and international laws andpolicies (Council directive # 87-848, October 19, 1987, Ministère de l’Agriculture etde la Forêt, Service Vétérinaire de la Santé et de la Protection Animale, permissions #92-256B to DJD).

2.2. Drugs and treatment

2.2.1. Acute studiesThree doses of vortioxetine (2.5, 5 and 10 mg/kg, free base dissolved in 10% b-

cyclodextrin, oral gavage, p.o.,) were used in the OF test, the NSF test and the FST. Theeffects of vortioxetine were compared to the vehicle control group (10% b-cyclo-dextrin) and also to a fluoxetine- (18 mg/kg p.o., (David et al., 2007)) and adiazepam-treated group (1.5 mg/kg, s.c. (David et al., 2007)). All doses were cor-rected for theweight of the salt. All treatments were administered 1 h before testing.

2.2.2. Chronic studiesTwo doses of vortioxetine (5 and 20 mg/kg/day, free base dissolved in 10% b-

cyclodextrin, oral gavage, p.o.) were tested in mice after 14 days of administration inthe NSF and 21 days of administration in the OF test, the NSF test and the FST. Themice were tested 24 h after the last dose. The effects of vortioxetine were comparedto a vehicle control group (10% b-cyclodextrin) and also to a fluoxetine-treated group(18 mg/kg/day p.o.).

2.3. Ex vivo SERT and 5-HT3 receptor occupancy assays

Brains frommice treated with vehicle, fluoxetine, or vortioxetine (1 h after acuteadministration or 24 h after the 14th or 21st injection) were flash frozen, sectionedcoronally using a cryostat, and then mounted on slides and frozen until use. Sliceswere 20 mm thick, and began at approximately þ1.2 mm anterior from bregma forSERT receptor occupancy or %2.7 mm posterior from bregma for 5-HT3 receptoroccupancy determination (Franklin and Paxinos, 2008). Slides were stored for atleast 24 h at %20 $C before use in autoradiography experiments.

2.3.1. Assessment of SERT occupancySlides were incubated at room temperature for 60 min in buffer (50 mM Trise

HCl, 150 mM NaCl, 5 mM KCl, pH ¼ 7.4) containing 4.5 nM [3H]-escitalopram.Nonspecific binding was determined using 1 mM escitalopram. Slides were washedbriefly in cold buffer, dried, and exposed in a Beta imager for 16 h. The region ofinterest (ROI) for the SERT assay included the lateral andmedial septum, the nucleusaccumbens and the olfactory tubercle. An example image of the ROI for the SERTassay can be found in Supplementary Fig. 2A.

2.3.2. Assessment of 5-HT3 receptor occupancySlides were preincubated for 5 min in a buffer consisting of 50 mM Tris and

150 mM NaCl. Slides were dried under a stream of air for 30e45 min. Subsequently,slides were incubated at room temperature for 60 min in buffer (50 mM TriseHCl,150 mM NaCl, 5 mM KCl, pH ¼ 7.4) containing 1 nM [3H]LY278584 (Perkin-Elmer,USA). Nonspecific binding was determined using 1 mM ondansetron. Slides werewashed briefly in cold buffer, dried, and exposed in a Beta imager for 24 h. The ROIfor the 5-HT3 receptor occupancy assay consisted of the hippocampus. An exampleimage for the 5-HT3 receptor occupancy assay can be found in SupplementaryFig. 2B.

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159148

Page 3: Antidepressant and anxiolytic potential of the multimodal - EA3544

2.4. Behavioural analysis

2.4.1. The open-field paradigmMotor activity was quantified in Plexiglas OF boxes 43 & 43 cm (MED associates,

Georgia, VT, USA) during a 10 min session (Popa et al., 2010). Two sets of 16 pulse-modulated infrared photobeams were placed on opposite walls 2.5 cm apart to re-cord xey ambulatory movements. Activity chambers were computer interfaced fordata sampling at 100 ms resolution. The centre was defined as a 32 & 32 cm centralarena. Dependent measures were: total time spent in the centre, the numbers ofentries into the centre and distance travelled in the centre divided by total distancetravelled. Overall motor activity was quantified as the total distance travelled (cm).

2.4.2. The novelty-suppressed feeding testThe NSF paradigm is a conflict test that elicits competing motivations: it does

not only measure anxiety components of behaviour, but also measures the animal’smotivation to eat the pellet. This implies that the NSF differs from anxiety-relatedtests such as the open field or the elevated plus maze paradigms. Briefly, animalswere food-deprived for 24 h prior to the test. Testing was performed in a50& 50& 20 cm box coveredwith bedding and illuminated by a 70W lamp. The NSFtest was carried out during a 5 min period as described by Santarelli et al. (2003). Atthe time of testing, a single pellet of food (regular chow)was placed on awhite paperplatform positioned in the centre of the box. Mice were tested individually afterplacing them in the corner of the box for 10 min. The latency to eating was timed.Immediately afterwards, the animal was transferred to its home cage and theamount of food consumed during the subsequent 5 min was measured, serving as acontrol for change in appetite as a possible confounding factor (home cage foodconsumption), because antidepressants are known to affect appetite.

2.4.3. The mouse forced swim testThe FST procedurewasmodified to enhance the sensitivity to detect the putative

antidepressant-like activity of drugs (Rainer et al., 2012; Porsolt et al., 1977). Briefly,mice were placed into clear plastic buckets, 20 cm in diameter and 23 cm deep, filledup to two-thirds with water at z24 $C. Automated scoring was done using theautomated X’PERT FST (Bioseb, Vitrolles, France). Dependent variables weremobility, swimming and climbing duration.

2.5. Immunohistochemistry

2.5.1. 5-Bromo-2-deoxyuridine (BrdU) labelling2.5.1.1. Proliferation study. Mice were administered BrdU (150 mg/kg i.p.) 2 h beforesacrifice and processed as described in David et al. (2009). BrdU-positive (BrdUþ)cells were counted using a BX51 microscope (Olympus, Germany).

2.5.1.2. Survival study. Mice were administered BrdU (150 mg/kg, i.p. b.i.d. for 3days) 4 weeks before sacrifice. We then proceeded as described by Xia et al. (2012).BrdUþ cells were counted under the microscope.

2.5.2. Doublecortin (DCX) labelling for maturation index studyThe immunohistochemistry protocol was adapted from David et al. (2009). DCX-

positive (DCXþ) cells were subcategorized according to their dendritic morphology:DCXþ cells and DCXþ cells with tertiary (or higher order) dendrites. The maturationindex was defined as the ratio of DCXþ cells possessing tertiary dendrites to the totalnumber of DCXþ cells.

2.6. Sholl analysis

For Sholl analysis, DCXþ cells with tertiary, relatively untruncated dendriticbranches were traced for each 35 mm hippocampal slice using Neurolucida software(MicroBrightField, Williston, VT) on an Olympus BX51 microscope equipped with amotorized stage device and &100 immersion oil objective. DCX immunohisto-chemistry was done to maximize the labelling of dendrites. Sholl analysis for den-dritic complexity was performed using the accompanying software (NeuroExplorer;MicroBrightField, version 10), calculating dendritic complexity including dendriticlength and number of intersections (branch points).

2.7. Statistical analysis

For all experiments except the NSF, a one-way ANOVA was performed and re-sults were expressed as mean # SEM values. When main effects were significant,treatment comparisons were analysed using either PLSD (behaviour) or Tukey’spost-hoc test. Unpaired t-test analysis was also used for planned comparisons. In theNSF test, a KaplaneMeier survival analysis was applied due to the non-normaldistribution of data, as described by Samuels and Hen (2012). Animals that didnot eat during the 10-min test period were statistically censored. The ManteleCoxlog-rank test was used to evaluate differences between experimental groups.Complete description of statistics can be found in the Supplementals(Supplementary Table 1). Linear correlation analyses of latency values in the NSF vs.number DCXþ cells with tertiary dendrites were performed using the Pearsoncorrelation coefficient “R”.

For ex vivo autoradiography experiments, surface radioactivity (expressed ascpm/mm2) was measured using Beta visionþ software version 2.0 (Biospace Lab,Paris, France). Radioactivity was quantified from a ROI defined a priori on the basis ofreceptor mapping experiments performed by this laboratory. This region remainedconsistent across each slice of tissue within a given assay. Specific binding wasdetermined by subtracting nonspecific binding from total binding. Specific bindingfor each brain was normalized to the average specific-bound radioactivity fromvehicle-treated individuals and expressed as a percentage of vehicle binding. Thesepercentages were subtracted from 100% to obtain percent receptor occupancy.

Differences were considered significant when p ' 0.05. All analyses were con-ducted using Statview 5.0 for IBM-compatible computers.

3. Results

3.1. SERT and 5-HT3 receptor occupancy after acute and chronictreatment with vortioxetine in BALB/cJ and 129Sv mice

Acute dosing of vortioxetine (5 mg/kg and 10 mg/kg, p.o., 1 h)resulted in 60e70% occupancy of the SERT for both strains at thetime of behavioural testing (Table 1). This was significantly lowerthan the approximately 90% SERT occupancy with fluoxetine(p < 0.01 and p < 0.05, compared to vortioxetine 5 and 10 mg/kg,respectively). Conversely, the same doses of vortioxetine resulted in>90% occupancy of the 5-HT3 receptors, while no detectable oc-cupancy was observed after fluoxetine administration (Table 2).

After chronic (daily for 14 or 21 days) administration of vorti-oxetine (5 or 20 mg/kg), practically no occupancy of the SERT wasmeasurable at the time of behavioural and neurogenic assessment,i.e., 24 h after the last dose in both strains, whereas fluoxetineoccupied >90% of the SERT at that time point (Table 1). Chronicdosing of vortioxetine, except in the high-dose BALB/cJ group,resulted in low non-significant occupancy of 5-HT3 receptors(Table 2).

3.2. Vortioxetine produces acute antidepressant and anxiolyticactivity in BALB/cJ and 129Sv mice

In the OF test in BALB/cJ mice (Fig. 1AeD), vortioxetine 2.5 and5 mg/kg induced an anxiolytic-like effect similar to that of diaz-epam (1 mg/kg), characterized by an increase in the time spent inthe centre (Fig. 1A: F5,62 ¼ 2.7, p < 0.05; p < 0.05 for vortioxetine2.5 mg/kg, p ¼ 0.053 for vortioxetine 5 mg/kg; p < 0.01 for diaz-epam). Vortioxetine and diazepam also increased the number ofentries (Fig. 1B p < 0.05 for vortioxetine 2.5 mg/kg or diazepam,unpaired t-test planned comparison) and the distance travelled inthe centre (Fig. 1C p < 0.01 for diazepam; p < 0.05 for vortioxetine2.5 and 5mg/kg, unpaired t-test planned comparison). Vortioxetineat a higher dose (10 mg/kg) as well as fluoxetine had no significanteffect on any of the measured parameters. Distance travelled in theperiphery (Fig. 1D), an index of locomotion in a non-stressfulenvironment, was unchanged for all treatments. Similarly, no ef-fect of treatment was observed on overall locomotor activity(Supplementary Fig. 3).

In the FST in BALB/cJ mice (Fig. 1EeG), vortioxetine (5 mg/kg)significantly increased the mobility duration (Fig. 1E p < 0.01, un-paired t-test planned comparison), and significantly increasedswimming (Fig. 1F p < 0.01, unpaired t-test planned comparison)and climbing behaviour (Fig. 1G p < 0.05, unpaired t-test plannedcomparison). Comparatively, fluoxetine also increased mobility(Fig. 1E p < 0.01, unpaired t-test planned comparison), apparentlythrough an increase of swimming duration only (Fig. 1F p < 0.05,unpaired t-test planned comparison).

The 5- but not the 10-mg/kg dose of vortioxetine had anantidepressant/anxiolytic-like effect in the NSF test in 129Sv micesimilar to that of diazepam (Fig. 2B: F5,54 ¼ 7.3, p< 0.0001; p< 0.01for diazepam or vortioxetine at 5 mg/kg). In contrast, fluoxetine

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159 149

Page 4: Antidepressant and anxiolytic potential of the multimodal - EA3544

(18 mg/kg) induced no significant effect on latency to eat the pelletand tended to increase latency (p ¼ 0.057). The results of the one-way ANOVA were confirmed by a KaplaneMeier survival analysisfollowed by a ManteleCox log-rank test (Fig. 2A, p < 0.01 fordiazepam and vortioxetine 5 mg/kg). Food consumption in thehome cage was monitored after the behavioural test and displayedno differences between groups (F5,54¼1.2, p¼ 0.34, SupplementaryFig. 4).

3.3. Chronic vortioxetine produced sustained antidepressant andanxiolytic activity in BALB/cJ and 129Sv mice

In BALB/cJ mice, chronic (21 days) administration of vortiox-etine, 5 mg/kg per day and test 24 h after the last dose, resulted inan anxiolytic-like activity in the OF test characterized by increasedtime spent in the centre (Fig. 3A: F3,48 ¼ 3.1, p < 0.05; p < 0.01 forvortioxetine 5 mg/kg), and increased number of entries (Fig. 3Bp < 0.05 for vortioxetine 5 mg/kg, unpaired t-test planned com-parison). Neither vortioxetine 20 mg/kg per day and test 24 h afterthe last dose or fluoxetine administration had a significant effect onthe number of entries, the time spent in the centre or distancetravelled. Distance travelled in the centre or in the periphery was

unchanged in all treatment groups (Fig. 3CeD). Similarly, no effectof treatment was observed on overall locomotor activity(Supplementary Fig. 5).

In the FST in BALB/cJ mice (Fig. 3EeG), chronic (21 days) vorti-oxetine, 5 mg/kg per day, but not 20 mg/kg per day, significantlyincreased the mobility duration (Fig. 3E: F3,42 ¼ 4.6, p < 0.01;p < 0.01 for vortioxetine 5 mg/kg). As in the acute study, the in-crease was due to a combination of increased duration of swim-ming (Fig. 3F: F3,42 ¼ 4.0, p < 0.05; p < 0.01 for vortioxetine 5 mg/kg) and climbing behaviour (Fig. 3G p < 0.05 for vortioxetine 5 mg/kg, unpaired t-test, planned comparison). Fluoxetine had no sig-nificant effect on any of the parameters measured in this test(p ¼ 0.69, p ¼ 0.49 and p ¼ 0.89, for mobility, swimming andclimbing, respectively).

Vortioxetine, 20 mg/kg per day and test 24 h after last dose,administered for 14 but not 21 days, induced a significantreduction in the latency to feed in the NSF test in 129Sv mice (14days: Fig. 4A: p < 0.01 KaplaneMeier survival analysis followedby a ManteleCox log-rank test, confirmed at the trend level(p ¼ 0.067) by one-way ANOVA in Fig. 4B. 21 days: p ¼ 0.37,Fig. 4D). At a lower dose (5 mg/kg per day) and test 24 h after thelast dose, vortioxetine reduced the latency to feed after 21 days of

Table 25-HT3 receptor occupancy in 129S6/SvEvTac and BALB/cJ mice after acute and chronic treatment. 5-HT3 receptor occupancy of vortioxetine and fluoxetine after acute (p.o., 1 h)or chronic (daily p.o. administration for 14 and 21 days andmeasurement 24 h after the last dose) in 129/SvEvTac and Balb/cJ ice. Data (n¼ 3e4 animals/group) are expressed inpercentage of occupancy (mean # SEM) and specific binding values in cpm/mm2 (mean # SEM).

Strain Treatment Duration

Acute 14 days 21 days

Occupancy (%) Specific bound(cpm/mm2)

Occupancy (%) Specific bound(cpm/mm2)

Occupancy (%) Specific bound(cpm/mm2)

129S6/SvEvTac Vehicle 0 # 4.2 0.09 # 0.004 0 # 6.8 0.1 # 0.01 0 # 11 0.11 # 0.01Fluoxetine %6.3 # 9.4 0.1 # 0.01 ND ND 8 # 9.3 0.097 # 0.01Vortioxetine 5 mg/kg/d 94 # 0.4***,### 0.005 # 0.0004***,### 3 # 13 0.1 # 0.01 13 # 10 0.092 # 0.01

20 mg/kg/d ND ND 8.2 # 8.4# 0.96 # 0.01# 19 # 9.8 0.085 # 0.01BALB/cJ Vehicle 0 # 7.6 0.22 # 0.02 ND ND 0 # 2.8 0.19 # 0.01

Fluoxetine 3.4 # 9.2 0.21 # 0.02 ND ND 12 # 3.2 0.16 # 0.01Vortioxetine 5 mg/kg/d 92 # 1.5***,### 0.016 # 0.003***,### ND ND %7 # 5.3 0.20 # 0.01

10 mg/kg/d 97 # 1.0***,### 0.005 # 0.002***,### ND ND ND ND20 mg/kg/d ND ND ND ND 42 # 15* 0.11 # 0.03*

***p < 0.0001, **p < 0.01 and *p < 0.05 when compared to the vehicle group, within strain and duration of treatment.###p < 0.0001, ##p < 0.01 and #p < 0.05 when compared to the fluoxetine group, within strain and duration of treatment.ND: not determined.Detailed statistical results are shown in Supplementary Table 1.

Table 1Serotonin transporter occupancy in 129S6/SvEvTac and BALB/cJ mice after acute and chronic treatment. Serotonin transporter occupancy of vortioxetine and fluoxetine afteracute (p.o., 1 h) or chronic (daily p.o. administration for 14 and 21 days and measurement 24 h after the last dose) in 129/SvEvTac and Balb/cJ mice. Data (n ¼ 3e4 animals/group) are expressed in percentage of occupancy (mean # SEM) and specific binding values in cpm/mm2 (mean # SEM).

Strain Treatment Duration

Acute 14 days 21 days

Occupancy (%) Specific bound(cpm/mm2)

Occupancy (%) Specific bound(cpm/mm2)

Occupancy (%) Specific bound(cpm/mm2)

129S6/SvEvTac Vehicle 0 # 4.0 11 # 0.4 0 # 5.9 9.7 # 0.6 0 # 5.4 10 # 0.6Fluoxetine 91 # 1.7*** 1 # 0.2*** ND ND 98 # 0.3*** 0.13 # 0.03***Vortioxetine 5 mg/kg/d 70 # 2.9***,## 3.2 # 0.3***,## 14 # 10 8.4 # 1 5.9 # 2.2### 9.6 # 0.2###

20 mg/kg/d ND ND %1.1 # 0.6 9.8 # 0.1 %0.4 # 2.9### 10 # 0.3###

BALB/cJ Vehicle 0 # 7.9 7.1 # 0.6 ND ND 0 # 3.1 7.7 # 0.2Fluoxetine 93 # 1.6*** 0.5 # 0.1*** ND ND 92 # 0.4*** 0.6 # 0.03***Vortioxetine 5 mg/kg/d 63 # 0.9***,## 2.6 # 0.1***,## ND ND 0.6 # 7.0### 7.6 # 0.5###

10 mg/kg/d 69 # 3.4***,# 2.17 # 0.2***,# ND ND ND ND20 mg/kg/d ND ND ND ND %5.9 # 4.1### 8.1 # 0.3###

***p < 0.0001, **p < 0.01 and *p < 0.05 when compared to the vehicle group, within strain and duration of treatment.###p < 0.0001, ##p < 0.01 and #p < 0.05 when compared to the fluoxetine group, within strain and duration of treatment.ND: not determined.Detailed statistical results are shown in Supplementary Table 1.

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159150

Page 5: Antidepressant and anxiolytic potential of the multimodal - EA3544

treatment (Fig. 4C: p < 0.01 using a KaplaneMeier survivalanalysis followed by a ManteleCox log-rank test, confirmed byone-way ANOVA in Fig. 4D: p < 0.05). Fluoxetine, 18 mg/kg perday and test 24 h after the last dose, administered for 14 or 21days failed to reduce the latency to eat significantly (p ¼ 0.22 andp ¼ 0.90 respectively). This lack of effect after 21 days of treat-ment has been previously observed (David et al., 2009), whilelonger duration of treatment induced reduction in latency in theNSF (Santarelli et al., 2003; Wang et al., 2008). Food consumptionin the home cage was monitored after the behavioural tests andno effect of vortioxetine on food consumption was observed afterboth 14 and 21 days of treatment (Supplementary Fig. 6AeB andSupplementary Table 1).

3.4. Effect of chronic (21 days) vortioxetine on the various steps ofadult hippocampal neurogenesis in 129Sv mice

Vortioxetine, 5 mg/kg per day and assessment 24 h after thelast dose, significantly increased the number of BrdUþ cells in thedentate gyrus of the hippocampus (Fig. 5A p < 0.05 for vortiox-etine 5 mg/kg per day, unpaired t-test planned comparison;Supplementary Fig. 7). Furthermore, the same dosing regimen alsoincreased the survival of newborn neurons (Fig. 5B p < 0.05 forvortioxetine 5 mg/kg, unpaired t-test planned comparison;Supplementary Fig. 8), an effect also seen with fluoxetine (18 mg/kg per day and assessment 24 h after the last dose) (p < 0.05,following PLSD post-hoc analysis). While the number of

Open Field

Dis

tanc

e in

per

iphe

ry (i

n cm

)

500

1000

1500

2000

0

Dis

tanc

e in

cen

ter (

in c

m)

** * *

100

200

300

400

0

Entri

es in

cen

ter * *

0

40

60

80

100

20

** * *

Tim

e in

the

cent

er (i

n se

c.)

0

40

60

80

20

A

B

C

D

Clim

bing

dur

atio

n (s

ec.) *

2

4

6

8

10

0

Swim

min

g du

ratio

n (s

ec.)

20

40

60

80

100

0

***

Mob

ility

(sec

.)

50

0

100

150

***

Forced Swimming TestE

F

G

Treatments

VehicleDiazepam 1.5 mg/kgFluoxetine 18 mg/kgVortioxetine 2.5 mg/kgVortioxetine 5 mg/kgVortioxetine 10 mg/kg

Fig. 1. Effects of acute vortioxetine treatment in the open field paradigm and the forced swim test in BALB/cJ@Rj mice. Behavioural effects of vortioxetine were studied after 1-h administration at doses of 2.5, 5 and 10 mg/kg, compared to those of vehicle, diazepam (1.5 mg/kg) and/or fluoxetine (10 mg/kg) in the open-field paradigm and the mouseforced swim test. Data are expressed as mean # SEM. Anxiety was expressed as total of the time spent in the centre (A) and the number of entries in the centre (B) for the entiresession. Locomotor activity was reported as total ambulatory distance travelled in the centre (C) and in the periphery (D) for the entire session. In the FST, mobility (E), swimming(F) and climbing (G) times were recorded. *p < 0.05; **p < 0.01 for drugs compared to the vehicle-treated group (n ¼ 10e12 animals/group).

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159 151

Page 6: Antidepressant and anxiolytic potential of the multimodal - EA3544

doublecortin-positive cells remained unchanged after all treat-ments (Fig. 5C, G), chronic vortioxetine (5 and 20 mg/kg per day)or fluoxetine increased the number of DCXþ cells with tertiarydendrites (Fig. 5D: F3,19 ¼ 3.3, p < 0.05 for fluoxetine and vorti-oxetine 5 and 20 mg/kg per day) and the maturation index(Fig. 5E: F3,19 ¼ 5.5, p < 0.05 for fluoxetine and vortioxetine 5 mg/kg per day; p < 0.0001 for vortioxetine 20 mg/kg per day). Thebehavioural measurements in the NSF correlated with maturationand branching of neurons in the adult hippocampus across alldrug-treated animals (R ¼ %0.44, p < 0.05).

In line with the significant effect of vortioxetine 20 mg/kg perday in the NSF test after 14 days of treatment (Fig. 4A), this dosingregimen also had an effect on some stages of neurogenesis.Compared to 21 days of treatment, the number of doublecortin-positive cells was unchanged (Fig. 6A, p > 0.05 for all treatment;Supplementary Fig. 9), but the number of DCXþ cells with tertiarydendrites was significantly increased (Fig. 6B: p < 0.05 for vorti-oxetine 20 mg/kg per day, unpaired t-test, planned comparison), aswas the maturation index (Fig. 6C: p < 0.05 for vortioxetine 20 mg/kg per day, unpaired t-test planned comparison). After 21 days,behavioural measurements in the NSF were correlated withmaturation and branching of neurons in the adult hippocampusacross all drug-treated animals (R ¼ %0.70, p < 0.01).

To further examine the effects of treatment on the dendriticmorphology of newborn cells, we performed Sholl analyses onDCXþ cells with tertiary dendrites (Fig. 6E). In the groups treatedchronically with vortioxetine or fluoxetine, DCXþ cells displayedincreased dendritic length (Fig. 6E: F3,12 ¼ 19.1, p < 0.001,p < 0.0001 for vortioxetine 20 mg/kg per day and p < 0.05 forfluoxetine) and increased number of intersections (Fig. 6F:F3,12 ¼ 17.3, p < 0.001, p < 0.0001 for vortioxetine 20 mg/kg per dayand p < 0.01 for fluoxetine).

4. Discussion

In this study performed in two different strains of mice, andusing various behavioural test associated with emotion-relateddisorders and/or antidepressant treatment response, we foundconverging evidence for anxiolytic and antidepressant effects ofvortioxetine, a novel antidepressant with multimodal activity, inpreclinical models.

4.1. Behavioural effects of vortioxetine

4.1.1. Acute antidepressant/anxiolytic activityAn acute dose of vortioxetine corresponding to 60e70% occu-

pancy of the SERT and >90% 5-HT3 receptor occupancy decreasedspontaneous (OF) and novelty-related (NSF) anxiety in naïve BALB/cJ and 129Sv mice, respectively, to the same extent as the benzo-diazepine, diazepam. In line with previous observations, an acutedose of the SSRI, fluoxetine, was inactive at a dose corresponding to>90% SERT occupancy (Holmes and Rodgers, 2003). These obser-vations support the hypothesis that the acute anxiolytic activity ofvortioxetine is mediated by a mechanism that does not only rely onoccupancy of the SERT. This is in line with the finding that vorti-oxetine has antidepressant effects in patients at doses where onlyapproximately 50% of the SERT are occupied as shown in PET im-aging studies (Areberg et al., 2012).

In both animal models, the anxiolytic effects of vortioxetinewere not statistically significant at the highest dose (10 mg/kg)tested. This biphasic doseeresponse relation was not observed inpreviously reported acute studies in rat and mouse models ofanxiety and depression, i.e., rat social interaction (significant ef-fect at 2, 4 and 8 mg/kg, p.o., corresponding to up to 50% SERToccupancy), rat conditioned fear (significant effect at 3.9 and

Late

ncy

to fe

ed (s

ec.)

** **

0

100

200

300

400B

Frac

tion

of a

nim

als

not e

atin

g

Time (in sec.)

0.2

0

0.4

0.6

0.8

1

0 100 200 300 400**

ATreatments

VehicleDiazepam 1.5 mg/kgFluoxetine 18 mg/kg

Vortioxetine 2.5 mg/kg

Vortioxetine 5 mg/kg

Vortioxetine 10 mg/kg

Veh DiazVortioxetine

5 10Flx 2.5

Fig. 2. Effects of acute vortioxetine treatment in the novelty suppressed feeding paradigm in 129/SvEvTac mice. The effects of vortioxetine were studied 1 h after administration atdoses of 2.5, 5 and 10 mg/kg, compared to vehicle, fluoxetine (18 mg/kg) and diazepam (1.5 mg/kg). Data are expressed as cumulative survival with percentage of animals that havenot eaten for over 10 min (A) and mean # SEM of latency time to feed (B). *p < 0.05; **p < 0.01 for vortioxetine compared to the vehicle-treated group (n ¼ 15e20 animals/group).

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159152

Page 7: Antidepressant and anxiolytic potential of the multimodal - EA3544

7.9 mg/kg, s.c., corresponding to approximately 50 and 90% SERToccupancy) (Mork et al., 2012) and in an unpublished marbleburying study in CD-1 mice (significant effects at 3.9 and 7.9 mg/kg, s.c.) (Sanchez, unpublished data). The underlying mechanismof the consistent biphasic dose response observed in BALB/cJ and129Sv mice in the different models is not readily explained.Indeed, the mechanism of action of vortioxetine is complex,involving modulation of 5-HT receptors and SERT activity,resulting in modulation of several neurotransmitter systems(Mork et al., 2012; Pehrson et al., 2012). Interestingly, 5-HT7 re-ceptor antagonists have shown a biphasic-response in severalanxiety paradigms (Wesolowska et al., 2006a, 2006b), while 5-HT3 receptor blockade and 5-HT1A receptor activation may beinvolved in dose-dependent anxiolytic effects (Zhang et al., 2001;Barrett and Gleeson, 1991). Moreover, significant strain differ-ences have been described with respect to stress sensitivity,neurotransmitter tone and responses to antidepressants; e.g.,

BALB/cJ and 129Sv mice were responsive to fluoxetine, whereasCD-1 and NIH-Swiss mice were not (Lucki et al., 2001). Thus,complex and oppositely directed interactions between neuro-transmitter systems and receptor mechanisms may be involved inthe net effects of vortioxetine.

In the FST, a paradigm designed for screening of potentialantidepressants (Rainer et al., 2012; Petit-Demouliere et al.,2005) an acute dose (5 mg/kg, p.o.) of vortioxetine correspond-ing to 60e70% SERT occupancy and >90% 5-HT3 receptor occu-pancy showed an antidepressant-like activity in BALB/cJ mice.However, while similar SERT and 5-HT3 occupancy was observedafter vortioxetine at both doses tested, only the lowest dose(5 mg/kg) induced a statistically significant antidepressant effectin the FST. Fluoxetine, as well as the highest dose of vortioxetineincreased the mobility only at the trend level (p < 0.07 andp < 0.09 respectively). An antidepressant-like effect was alsoobserved in NIH-Swiss mice (significant effect at 15.9 mg/kg, s.c.)

200

400

600

800

0Dis

tanc

e in

the

cent

er (i

n cm

)C

Dis

tanc

e in

per

iphe

ry (i

n cm

)

1000

2000

3000

4000

0

D

Entri

es in

cen

ter

B

0

100

150

200

25

250*

Tim

e in

the

cent

er (i

n se

c.)

0

100

150

200

50

A250 **

Forced Swimming TestOpen Field

Treatments

Vehicle

Fluoxetine 18 mg/kg/day

Vortioxetine 5 mg/kg/day

Vortioxetine 20 mg/kg/day

Clim

bing

dur

atio

n (s

ec.)

5

10

15

20

25

0

G

F

10

40

30

20

50

0

**

*

Swim

min

g du

ratio

n (s

ec.)

Mob

ility

(sec

.)

20

0

40

80E

60**

Fig. 3. Effects of chronic vortioxetine treatment in the open field paradigm and the forced swim test in BALB/cJ@Rj mice. Behavioural effects of vortioxetine were studied after 21days of administration at doses of 5 and 20 mg/kg/day, compared to those of vehicle and fluoxetine (18 mg/kg/day) in the open-field paradigm or the mouse forced swim test. Dataare expressed as mean # SEM. Anxiety was expressed as total of the time spent in the centre (A) and the number of entries in the centre (B) for the entire session. Locomotor activitywas reported as total ambulatory distance travelled in the centre (C) and in the periphery (D) for the entire session. In the FST, mobility (E), swimming (F) and climbing (G) timeswere recorded. *p < 0.05; **p < 0.01 for drugs compared to the vehicle-treated group (n ¼ 10e12 animals/group).

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159 153

Page 8: Antidepressant and anxiolytic potential of the multimodal - EA3544

(Sanchez, unpublished data). Here, vortioxetine increases bothclimbing and swimming behaviour, effects related to increases innoradrenergic and serotonergic neurotransmissions, respectively,whereas fluoxetine (>90% SERT occupancy) only affected swim-ming behaviour. These results corroborate microdialysis experi-ments showing increases of both neurotransmitters aftervortioxetine administration in rats (Mork et al., 2012). In thatstudy, immobility was significantly reduced by acute treatmentwith 7.8 mg/kg vortioxetine, corresponding to z90% SERT occu-pancy. However, Mork and colleagues observed no specific effectsof vortioxetine on swimming and climbing parameters in theFlinders Sensitive Line (FSL) rat model that was used in theirstudy. This discrepancy with our results may be due to the modelselected. Indeed, this line, selectively bred for high sensitivity tocholinergic agonism, also display changes in neurotransmittersystems, specifically a lower density of 5-HT1A receptors and ahigher density of 5-HT1B receptors in several brain regions (Nishiet al., 2009; Wegener et al., 2011). While the low affinity ofvortioxetine for rodent 5-HT1A receptors is unlikely to explain thediscrepancy between the findings of the present study in miceand the findings in FSL rats, its partial agonistic action at 5-HT1Breceptors may limit the increase in mobility in the FST, i.e., in amodel of elevated 5-HT levels, a 5-HT1B receptor partial agonisticeffect may block effects on mobility, as observed in other studies(Gardier et al., 2001; Guilloux et al., 2006, 2011).

4.1.2. Chronic antidepressant/anxiolytic activityRepeated daily dosing of vortioxetine (5 mg/kg, p.o.) for 14 days

or 21 days in 129Sv and 21 days in BALB/cJ mice resulted inantidepressant/anxiolytic-like effects. Indeed, anxiolytic and/orantidepressant-like effect was observed in the in the OF paradigm,the NSF test and the FST, whereas the same dosing regimen withfluoxetine (18 mg/kg, p.o.) was inactive. As in the acute studies, abiphasic dose response was observed, as vortioxetine 20 mg/kg perday was not active.

The behavioural testing took place 24 h after the last dose, atwhich time fluoxetine had >90% SERT occupancy, while vortiox-etine had comparatively low occupancies at SERT and 5-HT3 re-ceptors. Vortioxetine’s low occupancies 24 h after the final chronicdose can be explained by its half-life. Indeed, experiments per-formed in rats showed that the half-life of fluoxetine and nor-fluoxetine (a potent SERT inhibitor (Owens et al., 1997) andfluoxetine’s active metabolite) are about 5 and 15 h, respectively(Caccia et al., 1990). However, vortioxetine has a half-life around3.2 h (Mork et al., 2012) and does not have an active metabolite onits different targets (Areberg et al., 2012).

The lack of antidepressant/anxiolytic effect of fluoxetine after 21days dosing is compatible with previous experiences in BALB/cJ and129SVeV strains, where 28 days of fluoxetine treatment are neededto achieve an effect (Wang et al., 2008; Dulawa et al., 2004).Furthermore, in contrast to benzodiazepines, the delayed onset of

Late

ncy

to fe

ed (s

ec.)

0

100

300

200

400D

Frac

tion

of a

nim

als

not e

atin

g

Time (in sec.)

0.2

0

0.4

0.6

0.8

1

0 100 200 300 400

C

500 600 700

Late

ncy

to fe

ed (s

ec.)

0

50

100

150

200

250B

Frac

tion

of a

nim

als

not e

atin

g

Time (in sec.)

0.2

0

0.4

0.6

0.8

1

0 100 200 300 400

A

500 600 700** **

*

Treatments

VehicleFluoxetine 18 mg/kg/dayVortioxetine 5 mg/kg/dayVortioxetine 20 mg/kg/day

Veh FlxVortioxetine

5 mg/kg 20 mg/kg Veh FlxVortioxetine

5 mg/kg 20 mg/kg

Chronic treatment(14 days)

Chronic treatment(21 days)

p=0.067

Fig. 4. Effects of chronic vortioxetine treatment in the novelty suppressed feeding paradigm in 129/SvEvTac mice. The effects of vortioxetine were studied after 14 (AeB) or 21 (CeD) days of administration at doses of 5 and 20 mg/kg/day, compared to those of vehicle and fluoxetine (18 mg/kg). Data are expressed as cumulative survival with percentage ofanimals that have not eaten over 10 min (A & C) and mean # SEM of latency time to feed (B & D) *p < 0.05; **p < 0.01 for vortioxetine compared to the vehicle-treated group(n ¼ 15e20 animals/group).

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159154

Page 9: Antidepressant and anxiolytic potential of the multimodal - EA3544

anxiolytic effect of SSRIs in tests performed in mouse is consistentwith the delayed onset in their clinical anxiolytic efficacy (Bespalovet al., 2010).

In conclusion, the findings indicate that the antidepressant ac-tivity of vortioxetine is mediated by a mechanism different fromthat of an SSRI. Overall, these findings may indicate that vortiox-etine exerts its sustained antidepressant/anxiolytic activity by amechanism that is not related to a constant SERT inhibition overtime. Furthermore, vortioxetine is equally potent in two mousestrains with different 5-HT tone (Zhang et al., 2004), an effect that

has not been observed with SSRIs (Kulikov et al., 2011; Cervo et al.,2005). Here, the lack of strain-dependent effects would support thehypothesis that mechanisms that are independent of 5-HT syn-thesis or 5-HT tissue content may be involved.

4.2. Neurogenic effects of vortioxetine

Antidepressant action has been putatively associated withchanges in adult hippocampal neurogenesis (Airan et al., 2007;David et al., 2007, 2009; Santarelli et al., 2003; Wang et al., 2008).

100µm

R=-0.44 #

DCX+ cells with tertiary dendrites

Late

ncy

to fe

ed (i

n se

c)

200

100

300

400

00 500 1000 1500 25002000

F

Proliferation Survival

G

Mat

urat

ion

inde

x

0

0.1

0.2

0.3

0.4E

***

Tota

l DC

X+ cel

ls w

ithte

rtiar

y de

ndrit

es0

500

1000

1500

2000D

* * *

Tota

l DC

X+ Cel

ls

0

100020003000400050006000

C

BrdU

+ cel

ls

200

400

600

800

0

**

B

BrdU

+ clu

ster

s

0

100

200

300

400

500 *A

Treatments

Vehicle

Fluoxetine 18 mg/kg

Vortioxetine 5 mg/kg

Vortioxetine 20 mg/kg

Vortioxetine 20 mg/kg

Vortioxetine 5 mg/kg

Fluoxetine 18 mg/kg

Vehicle

Fig. 5. Neurogenic effects of chronic vortioxetine administration (21 days) in 129/SvEvTac mice. The effects of 21 days of treatment with vortioxetine (5 and 20 mg/kg/day) on cellproliferation (A), cell survival (B) or cell maturation (CeG) were compared to those of vehicle and fluoxetine. Maturationwas characterized by the total number of DCXþ cells (C), thenumber of DCXþ cells with tertiary dendrites (D) and the maturation index of newborn granule cells (E). Latency to feed in the NSF correlated with the number of DCXþ cells withtertiary dendrites under pharmacological stimulation (F). Representative illustrations of doublecortin staining following chronic administration (21 days) of either fluoxetine(18 mg/kg/d) or vortioxetine (5 or 20 mg/kg/d) (G). Data are expressed as mean # SEM. *p < 0.05, for effects of fluoxetine or vortioxetine compared to vehicle (n ¼ 4e10 animals pergroup). #p < 0.05 for R Pearson value of correlation of latency values with number of DCXþ cells with tertiary dendrites (n ¼ 16).

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159 155

Page 10: Antidepressant and anxiolytic potential of the multimodal - EA3544

Treatments

Vehicle

Fluoxetine 18 mg/kg/day

Vortioxetine 5 mg/kg/day

Vortioxetine 20 mg/kg/day

Vehicle

Fluoxetine 18 mg/kg/day

Vortioxetine 5 mg/kg/day

Vortioxetine 20 mg/kg/day

G

Schematic representation of DCX+ cells

Den

driti

c In

ters

ectio

n

Distance from soma (µm)0 50 100 150

0

1

2

3

4

5F

*** §§

Den

driti

c Le

ngth

(µm

)

Distance from soma (µm)

0

10

20

30

40

50

0 50 100 150

*** §E

Late

ncy

to fe

ed (s

ec)

0

100

200

300

0 500 1000 1500 2000DCX+ cells with tertiary dendrites

D

Mat

urat

ion

inde

x

0

0.1

0.2

0.3

0.4C*

Tota

l DC

X+ Cel

ls

0

1000

2000

3000

4000

5000

A B

Tota

l DC

X+ cel

ls w

ithte

rtiar

y de

ndrit

es

0

500

1000

1500

*

Fig. 6. Neurogenic effects of chronic vortioxetine administration (14 days) in 129/SvEvTac mice. The effects of 14-day treatment with vortioxetine (5 and 20 mg/kg/day) on cellmaturation (AeC) and dendritic complexity (EeG) were compared to those of vehicle and fluoxetine. Maturation was characterized by the total number of DCXþ cells (A), thenumber of DCXþ cells with tertiary dendrites (B) and the maturation index of newborn granule cells (C) (n ¼ 4e10 animals per group). Latency to feed in the NSF was correlatedwith number of DCXþ cells with tertiary dendrites under pharmacological stimulation (D) (p < 0.01, n ¼ 16). The effects of vortioxetine treatment on dendritic length (E) and thenumber of intersection (F) were measured (n ¼ 4e5 mice/group, 4e8 cells/mouse) using a Sholl analysis of DCXþ neurons (G). *p < 0.05, ***p < 0.0001 for effects of vortioxetinecompared to the vehicle-treated group (n ¼ 4e10 animals per group). xp < 0.05, xxp < 0.01 for effects of fluoxetine compared to the vehicle-treated group (n ¼ 4e10 animals pergroup). ##p < 0.01 for R Pearson value of correlation of latency values with number of DCXþ cells with tertiary dendrites.

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159156

Page 11: Antidepressant and anxiolytic potential of the multimodal - EA3544

However, not all antidepressant effects are related to neurogenesis(David et al., 2009) and increased neurogenesis by itself in naïveanimals does not induce antidepressant effects (Sahay et al., 2011).Thus, neurogenesis-related effects of antidepressants may berestricted to specific behaviours or symptoms that remain to befully detailed.

Among all stages implicated in neurogenesis, maturation ofyoung neurons is a crucial step for the functional integration ofyoung neurons into neural circuits. Studies in rodents have shownthat treatment with classical SSRIs such as fluoxetine and also novelantidepressants such as agomelatine can promote maturation ofpost-mitotic neurons (Rainer et al., 2012; Dagyte et al., 2010;Soumier et al., 2009; Wang et al., 2008). Here, we report thatchronic vortioxetine (5 mg/kg, p.o. per day for 21 days) not onlysignificantly increased cell proliferation within the hippocampus,but also increased the survival rate of BrdUþ cells and the numberof DCXþ cells with tertiary dendrites. Fluoxetine (18 mg/kg per dayp.o.) produced a similar effect in spite of its overall lack of effect inthe NSF test. Similarly, in spite of a lack of an overall effect of vor-tioxetine at 20mg/kg per day for 14 days in the NSF test, therewas asignificant increase in the number of DCXþ cells with tertiarydendrites, dendritic length and the number of dendritic in-tersections at this early time point. Interestingly, irrespective oftreatment and treatment duration (14 or 21 days), the latency tofeed vs. the number of DCXþ cells with tertiary dendrites werecorrelated. To our knowledge, this is the first time such a correla-tion has been observed.

It would be of interest to define which targets of vortioxetinecontribute most to its neurogenic effect. There is a large body ofevidence, including the present study, indicating that chronic SERTblockade is responsible for an increase in several steps of neuro-genesis. Pharmacological stimulation of 5-HT1A receptors increasescell proliferation, while its blockade induces opposite effects(Banasr et al., 2004; Klempin et al., 2010; Radley and Jacobs, 2002;Santarelli et al., 2003). 5-HT1B receptor blockade has no effect oncell proliferation within the hippocampus, but pharmacologicaldissection of auto- vs. hetero-receptors in rats suggests that acti-vation of the latter may contribute to cell proliferation (Banasr et al.,2004). Furthermore, activation by 5-HT1A receptor agonists in ro-dents increased cell survival in a time-dependent manner (Klempinet al., 2010; Soumier et al., 2010), while its blockade had oppositeeffects (Klempin et al., 2010), reinforcing the present results.

Unfortunately, no study has yet observed the effects of chronicpharmacological blockade or stimulation of 5-HT1B receptors oncell survival in the hippocampus in adult rodents.

Overall, while its precise mechanism of action remains to bedetailed, vortioxetine produces a robust neurogenic effect that iscompatible with its anxiolytic/antidepressant activity.

4.3. Comments and limitations

Here, we studied the effects of vortioxetine in selected strains ofmice depending on the readout measured. Adult male 129S6/SvEvTac mice were used for the NSF study because of their sensi-tivity to chronic antidepressants in this behavioural model(Santarelli et al., 2003), and because the rate of hippocampal neu-rogenesis allows for study antidepressant-induced neurogenesis.Male BALB/cJ mice were used for the FST studies because of theirhigh sensitivity to chronic antidepressant treatment in thesemodels (Belzung and Griebel, 2001; Dulawa et al., 2004), andbecause 129S6/SvEVTac mice do not behave adequately in thisparadigm.

Furthermore, neurogenesis results may differ depending on thestrain used. Indeed, 2 strains may utilize different cellular andmolecular machinery to mediate the neurogenic and behavioural

effects of chronic antidepressant treatment, and thus pro-neurogenic effects of vortioxetine observed here in 129S6/SvEV-Tac mice may differ in BALB/cJ mice. However, recent reports showthat female mice of the BALB/c and a 129 substrain (129SvJ) arequite similar onmeasures of basal adult neurogenesis including cellproliferation, survival, and neuronal differentiation, although the129SvJ mice show slightly less survival 4 weeks after BrdU injection(Kempermann et al., 1997).

Interestingly, the 5 mg/kg dose of vortioxetine had a significantimpact on animals’ behaviour after 21 days exposure, but not after14 days, while this was the opposite for the highest dose. Onepossible interpretation of these findings is that development oftolerance occurred at the highest dose administered. However, thisneeds to be confirmed in other strains of mice, or in appropriateanimal models of major depressive disorders.

Indeed, behavioural and neurogenic effects of vortioxetine needto be confirmed in other model. For instance, a putative effect ofvortioxetine after stress (unpredictable chronic mild stress [UCMS],social defeat) or neuroendocrine-induced alterations of animalbehaviour should be assessed, since thesemodels have shown goodpredictive validity. Furthermore, studies in genetic models lackingone or more of the targets involved in vortioxetine’s mechanism ofaction would help to dissect the contributions of the individualtargets to the overall effects.

5. Conclusion

Vortioxetine displays a sustained antidepressant and anxiolyticprofile associated with increased neurogenesis at several stages.The effect of vortioxetine was not associated with SERT occupancy,suggesting that its mechanism of action is different from that of anSSRI.

Conflict of interests

This work received financial support from Lundbeck and theTakeda Pharmaceutical Company, Ltd.

Denis David currently receives investigator-initiated researchsupport from Lundbeck and served as a consultant in the areas oftarget identification and validation and new compound develop-ment to Lundbeck and Servier in 2011e12.

Bruno Guiard currently receives investigator-initiated researchsupport fromNeurosearch and served as a consultant in the areas oftarget identification and validation and new compound develop-ment to Lundbeck and Servier in 2011e12.

Acknowledgements

This work was supported by the French Ministry of Higher Ed-ucation and Research and H Lundbeck. We thank the animal carefacility of the “Institut Fédératif de Recherche-IFR141” of the ParisSud University for their technical assistance.

Appendix A. Supplementary data

Supplementary data related to this article can be found at http://dx.doi.org/10.1016/j.neuropharm.2013.05.014.

References

Airan, R.D., Meltzer, L.A., Roy, M., Gong, Y., Chen, H., Deisseroth, K., 2007. High-speedimaging reveals neurophysiological links to behavior in an animal model ofdepression. Science 317, 819e823.

Areberg, J., Luntang-Jensen, M., Sogaard, B., Nilausen, D.O., 2012. Occupancy of theserotonin transporter after administration of Lu AA21004 and its relation to

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159 157

Page 12: Antidepressant and anxiolytic potential of the multimodal - EA3544

plasma concentration in healthy subjects. Basic Clin. Pharmacol. Toxicol. 110,401e404.

Artigas, F., Adell, A., Celada, P., 2006. Pindolol augmentation of antidepressantresponse. Curr. Drug Targets 7, 139e147.

Bailey, J.E., Potokar, J., Coupland, N., Nutt, D.J., 1995. The 5-HT3 antagonist ondan-setron reduces gastrointestinal side effects induced by a specific serotonin re-uptake inhibitor in man. J. Psychopharmacol. (Oxf.) 9, 137e141.

Banasr, M., Hery, M., Printemps, R., Daszuta, A., 2004. Serotonin-induced increasesin adult cell proliferation and neurogenesis are mediated through different andcommon 5-HT receptor subtypes in the dentate gyrus and the subventricularzone. Neuropsychopharmacology 29, 450e460.

Bang-Andersen, B., Ruhland, T., Jorgensen, M., Smith, G., Frederiksen, K.,Jensen, K.G., Zhong, H., Nielsen, S.M., Hogg, S., Mork, A., Stensbol, T.B., 2011.Discovery of 1-[2-(2,4-dimethylphenylsulfanyl)phenyl]piperazine (LuAA21004): a novel multimodal compound for the treatment of major depres-sive disorder. J. Med. Chem. 54, 3206e3221.

Barrett, J.E., Gleeson, S., 1991. Anxiolytic effects of 5-HT1A agonists, 5-HT3 antag-onists and benzodiazepines: conflict and drug discrimination studies. In:Rogers, R.J., Cooper, S.J. (Eds.), 5-HT1A Agonists, 5-HT3 Antagonists and Ben-zodiazepines: Their Comparative Behavioural Pharmacology. Wiley & Sons Ltd,Chichester, pp. 59e105.

Belzung, C., Griebel, G., 2001. Measuring normal and pathological anxiety-likebehaviour in mice: a review. Behav. Brain Res. 125, 141e149.

Bespalov, A.Y., van Gaalen, M.M., Gross, G., 2010. Antidepressant treatment inanxiety disorders. Curr. Top. Behav. Neurosci. 2, 361e390.

Bonaventure, P., Kelly, L., Aluisio, L., Shelton, J., Lord, B., Galici, R., Miller, K., Atack, J.,Lovenberg, T.W., Dugovic, C., 2007. Selective blockade of 5-hydroxytryptamine(5-HT)7 receptors enhances 5-HT transmission, antidepressant-like behavior,and rapid eye movement sleep suppression induced by citalopram in rodents.J. Pharmacol. Exp. Ther. 321, 690e698.

Caccia, S., Cappi, M., Fracasso, C., Garattini, S., 1990. Influence of dose and route ofadministration on the kinetics of fluoxetine and its metabolite norfluoxetine inthe rat. Psychopharmacology (Berl.) 100, 509e514.

Cervo, L., Canetta, A., Calcagno, E., Burbassi, S., Sacchetti, G., Caccia, S., Fracasso, C.,Albani, D., Forloni, G., Invernizzi, R.W., 2005. Genotype-dependent activity oftryptophan hydroxylase-2 determines the response to citalopram in a mousemodel of depression. J. Neurosci. 25, 8165e8172.

Costall, B., Naylor, R.J., 2004. 5-HT3 receptors. Current drug targets. CNS Neurol.Disord. 3, 27e37.

Dagyte, G., Trentani, A., Postema, F., Luiten, P.G., Den Boer, J.A., Gabriel, C., Mocaer, E.,Meerlo, P., Van der Zee, E.A., 2010. The novel antidepressant agomelatine nor-malizes hippocampal neuronal activity and promotes neurogenesis in chroni-cally stressed rats. CNS Neurosci. Ther. 16, 195e207.

David, D.J., Klemenhagen, K.C., Holick, K.A., Saxe, M.D., Mendez, I., Santarelli, L.,Craig, D.A., Zhong, H., Swanson, C.J., Hegde, L.G., Ping, X.I., Dong, D.,Marzabadi, M.R., Gerald, C.P., Hen, R., 2007. Efficacy of the MCHR1 antagonist N-[3-(1-{[4-(3,4-difluorophenoxy)phenyl]methyl}(4-piperidyl))-4-methylphenyl]-2-m ethylpropanamide (SNAP 94847) in mouse models ofanxiety and depression following acute and chronic administration is inde-pendent of hippocampal neurogenesis. J. Pharmacol. Exp. Ther. 321, 237e248.

David, D.J., Samuels, B.A., Rainer, Q., Wang, J.W., Marsteller, D., Mendez, I., Drew, M.,Craig, D.A., Guiard, B.P., Guilloux, J.P., Artymyshyn, R.P., Gardier, A.M., Gerald, C.,Antonijevic, I.A., Leonardo, E.D., Hen, R., 2009. Neurogenesis-dependent and-independent effects of fluoxetine in an animal model of anxiety/depression.Neuron 62, 479e493.

Dulawa, S.C., Holick, K.A., Gundersen, B., Hen, R., 2004. Effects of chronic fluoxetinein animal models of anxiety and depression. Neuropsychopharmacology 29,1321e1330.

Duman, R.S., Malberg, J., Thome, J., 1999. Neural plasticity to stress and antide-pressant treatment. Biol. Psychiatry 46, 1181e1191.

Faure, C., Mnie-Filali, O., Scarna, H., Debonnel, G., Haddjeri, N., 2006. Effects of the5-HT7 receptor antagonist SB-269970 on rat hormonal and temperature re-sponses to the 5-HT1A/7 receptor agonist 8-OH-DPAT. Neurosci. Lett. 404,122e126.

Franklin, K.B.J., Paxinos, G., 2008. The Mouse Brain in Stereotaxic Coordinates. Ac-ademic Press, Amsterdam; Boston.

Gardier, A.M., Trillat, A.C., Malagie, I., David, D., Hascoet, M., Colombel, M.C.,Jolliet, P., Jacquot, C., Hen, R., Bourin, M., 2001. [5-HT1B serotonin receptors andantidepressant effects of selective serotonin reuptake inhibitors]. C. R. Acad. Sci.III 324, 433e441.

Gingrich, J.A., Hen, R., 2001. Dissecting the role of the serotonin system in neuro-psychiatric disorders using knockout mice. Psychopharmacology (Berl.) 155, 1e10.

Guilloux, J.P., David, D.J., Guiard, B.P., Chenu, F., Reperant, C., Toth, M., Bourin, M.,Gardier, A.M., 2006. Blockade of 5-HT1A receptors by (þ/%)-pindolol potenti-ates cortical 5-HT outflow, but not antidepressant-like activity of paroxetine:microdialysis and behavioral approaches in 5-HT1A receptor knockout mice.Neuropsychopharmacology 31, 2162e2172.

Guilloux, J.P., David, D.J., Xia, L., Nguyen, H.T., Rainer, Q., Guiard, B.P., Reperant, C.,Deltheil, T., Toth, M., Hen, R., Gardier, A.M., 2011. Characterization of 5-HT(1A/1B)%/% mice: an animal model sensitive to anxiolytic treatments. Neurophar-macology 61, 478e488.

Guilloux, J.P., David, D.J., Samuels, B.A., David, I., Gardier, A.M., Guiard, B.P., 2012.Non-response to Initial Antidepressant Therapy. Psychology. InTech, Rijeka,Croatia.

Guscott, M., Bristow, L.J., Hadingham, K., Rosahl, T.W., Beer, M.S., Stanton, J.A.,Bromidge, F., Owens, A.P., Huscroft, I., Myers, J., Rupniak, N.M., Patel, S.,Whiting, P.J., Hutson, P.H., Fone, K.C., Biello, S.M., Kulagowski, J.J., McAllister, G.,2005. Genetic knockout and pharmacological blockade studies of the 5-HT7receptor suggest therapeutic potential in depression. Neuropharmacology 48,492e502.

Hamon, M., Bourgoin, S., 2006. Pharmacological profile of antidepressants: a likelybasis for their efficacy and side effects? Eur. Neuropsychopharmacol. 16, S625eS632.

Hedlund, P.B., Huitron-Resendiz, S., Henriksen, S.J., Sutcliffe, J.G., 2005. 5-HT7 re-ceptor inhibition and inactivation induce antidepressantlike behavior and sleeppattern. Biol. Psychiatry 58, 831e837.

Holmes, A., Rodgers, R.J., 2003. Prior exposure to the elevated plus-maze sensitizesmice to the acute behavioral effects of fluoxetine and phenelzine. Eur. J. Phar-macol. 459, 221e230.

Kennedy, S.H., Young, A.H., Blier, P., 2011. Strategies to achieve clinical effectiveness:refining existing therapies and pursuing emerging targets. J. Affect. Disord. 132(Suppl. 1), S21eS28.

Kempermann, G., Kuhn, H.G., Gage, F.H., 1997. Genetic influence on neurogenesis inthe dentate gyrus of adult mice. Proc. Natl. Acad. Sci. U. S. A. 94, 10409e10414.

Kessler, R.C., Berglund, P., Demler, O., Jin, R., Merikangas, K.R., Walters, E.E., 2005.Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in theNational Comorbidity Survey Replication. Arch. Gen. Psychiatry 62, 593e602.

Klempin, F., Babu, H., De Pietri Tonelli, D., Alarcon, E., Fabel, K., Kempermann, G.,2010. Oppositional effects of serotonin receptors 5-HT1a, 2, and 2c in theregulation of adult hippocampal neurogenesis. Front. Mol. Neurosci. 3.

Kulikov, A.V., Tikhonova, M.A., Osipova, D.V., Kulikov, V.A., Popova, N.K., 2011. As-sociation between tryptophan hydroxylase-2 genotype and the antidepressanteffect of citalopram and paroxetine on immobility time in the forced swim testin mice. Pharmacol. Biochem. Behav. 99, 683e687.

Lucki, I., Dalvi, A., Mayorga, A.J., 2001. Sensitivity to the effects of pharmacologicallyselective antidepressants in different strains of mice. Psychopharmacology(Berl.) 155, 315e322.

Malberg, J.E., Eisch, A.J., Nestler, E.J., Duman, R.S., 2000. Chronic antidepressanttreatment increases neurogenesis in adult rat hippocampus. J. Neurosci. 20,9104e9110.

Martiny, K., Lunde, M., Bech, P., Plenge, P., 2012. A short-term double-blind ran-domized controlled pilot trial with active or placebo pindolol in patients treatedwith venlafaxine for major depression. Nord. J. Psychiatry 66, 147e154.

Mnie-Filali, O., Faure, C., Lambas-Senas, L., El Mansari, M., Belblidia, H., Gondard, E.,Etievant, A., Scarna, H., Didier, A., Berod, A., Blier, P., Haddjeri, N., 2011. Phar-macological blockade of 5-HT7 receptors as a putative fast acting antidepres-sant strategy. Neuropsychopharmacology 36, 1275e1288.

Mork, A., Pehrson, A., Brennum, L.T., Nielsen, S.M., Zhong, H., Lassen, A.B., Miller, S.,Westrich, L., Boyle, N.J., Sanchez, C., Fischer, C.W., Liebenberg, N., Wegener, G.,Bundgaard, C., Hogg, S., Bang-Andersen, B., Stensbol, T.B., 2012. Pharmacologicaleffects of Lu AA21004: a novel multimodal compound for the treatment ofmajor depressive disorder. J. Pharmacol. Exp. Ther. 340, 666e675.

Mullins, U.L., Gianutsos, G., Eison, A.S., 1999. Effects of antidepressants on 5-HT7receptor regulation in the rat hypothalamus. Neuropsychopharmacology 21,352e367.

Nishi, K., Kanemaru, K., Diksic, M., 2009. A genetic rat model of depression, Flinderssensitive line, has a lower density of 5-HT(1A) receptors, but a higher density of5-HT(1B) receptors, compared to control rats. Neurochem. Int. 54, 299e307.

Owens, M.J., Morgan, W.N., Plott, S.J., Nemeroff, C.B., 1997. Neurotransmitter re-ceptor and transporter binding profile of antidepressants and their metabolites.J. Pharmacol. Exp. Ther. 283, 1305e1322.

Pehrson, A.L., Cremers, T., Betry, C., van der Hart, M.G., Jorgensen, L., Madsen, M.,Haddjeri, N., Ebert, B., Sanchez, C., 2012. Lu AA21004, a novel multimodal an-tidepressant, produces regionally selective increases of multipleneurotransmitters-A rat microdialysis and electrophysiology study. Eur. Neu-ropsychopharmacol. 23 (2), 133e145.

Petit-Demouliere, B., Chenu, F., Bourin, M., 2005. Forced swimming test in mice: areview of antidepressant activity. Psychopharmacology (Berl.) 177, 245e255.

Popa, D., Cerdan, J., Reperant, C., Guiard, B.P., Guilloux, J.P., David, D.J., Gardier, A.M.,2010. A longitudinal study of 5-HT outflow during chronic fluoxetine treatmentusing a new technique of chronic microdialysis in a highly emotional mousestrain. Eur. J. Pharmacol. 628, 83e90.

Porsolt, R.D., Bertin, A., Jalfre, M., 1977. Behavioral despair in mice: a primaryscreening test for antidepressants. Arch. Int. Pharmacodyn. Ther. 229, 327e336.

Radley, J.J., Jacobs, B.L., 2002. 5-HT1A receptor antagonist administration decreasescell proliferation in the dentate gyrus. Brain Res. 955, 264e267.

Rainer, Q., Xia, L., Guilloux, J.P., Gabriel, C., Mocaer, E., Hen, R., Enhamre, E.,Gardier, A.M., David, D.J., 2012. Beneficial behavioural and neurogenic effects ofagomelatine in a model of depression/anxiety. Int. J. Neuropsychopharmacol.15, 321e335.

Ramamoorthy, R., Radhakrishnan, M., Borah, M., 2008. Antidepressant-like effectsof serotonin type-3 antagonist, ondansetron: an investigation in behaviour-based rodent models. Behav. Pharmacol. 19, 29e40.

Redrobe, J.P., Bourin, M., 1997. Partial role of 5-HT2 and 5-HT3 receptors in theactivity of antidepressants in the mouse forced swimming test. Eur. J. Phar-macol. 325, 129e135.

Rush, A.J., Trivedi, M.H., Wisniewski, S.R., Nierenberg, A.A., Stewart, J.W., Warden, D.,Niederehe, G., Thase, M.E., Lavori, P.W., Lebowitz, B.D., McGrath, P.J.,Rosenbaum, J.F., Sackeim, H.A., Kupfer, D.J., Luther, J., Fava, M., 2006. Acute and

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159158

Page 13: Antidepressant and anxiolytic potential of the multimodal - EA3544

longer-term outcomes in depressed outpatients requiring one or severaltreatment steps: a STAR*D report. Am. J. Psychiatry 163, 1905e1917.

Sahay, A., Scobie, K.N., Hill, A.S., O’Carroll, C.M., Kheirbek, M.A., Burghardt, N.S.,Fenton, A.A., Dranovsky, A., Hen, R., 2011. Increasing adult hippocampal neu-rogenesis is sufficient to improve pattern separation. Nature 472, 466e470.

Samuels, B.A., Hen, R., 2012. Novelty-suppressed feeding in the mouse. In:Gould, T.D. (Ed.), Mood and Anxiety Related Phenotypes in Mice. Character-ization Using Behavioral Tests. Springer, pp. 107e121.

Santarelli, L., Saxe,M., Gross, C., Surget, A., Battaglia, F., Dulawa, S.,Weisstaub, N., Lee, J.,Duman, R., Arancio, O., Belzung, C., Hen, R., 2003. Requirement of hippocampalneurogenesis for the behavioral effects of antidepressants. Science 301, 805e809.

Sarkisyan, G., Roberts, A.J., Hedlund, P.B., 2010. The 5-HT(7) receptor as a mediatorand modulator of antidepressant-like behavior. Behav. Brain Res. 209, 99e108.

Soumier, A., Banasr, M., Goff, L.K., Daszuta, A., 2010. Region- and phase-dependenteffects of 5-HT(1A) and 5-HT(2C) receptor activation on adult neurogenesis. Eur.Neuropsychopharmacol. 20, 336e345.

Soumier, A., Banasr, M., Lortet, S., Masmejean, F., Bernard, N., Kerkerian-Le-Goff, L.,Gabriel, C., Millan, M.J., Mocaer, E., Daszuta, A., 2009. Mechanisms contributingto the phase-dependent regulation of neurogenesis by the novel antidepres-sant, agomelatine, in the adult rat hippocampus. Neuropsychopharmacology34, 2390e2403.

Wang, J.W., David, D.J., Monckton, J.E., Battaglia, F., Hen, R., 2008. Chronic fluoxetinestimulates maturation and synaptic plasticity of adult-born hippocampalgranule cells. J. Neurosci. 28, 1374e1384.

Wegener, G., Finger, B.C., Elfving, B., Keller, K., Liebenberg, N., Fischer, C.W.,Singewald, N., Slattery, D.A., Neumann, I.D., Mathe, A.A., 2011. Neuropeptide S

alters anxiety, but not depression-like behaviour in Flinders Sensitive Line rats:a genetic animal model of depression. Int. J. Neuropsychopharmacol., 1e13.

Wesolowska, A., Nikiforuk, A., Stachowicz, K., 2006a. Potential anxiolytic andantidepressant effects of the selective 5-HT7 receptor antagonist SB269970 after intrahippocampal administration to rats. Eur. J. Pharmacol.553, 185e190.

Wesolowska, A., Nikiforuk, A., Stachowicz, K., Tatarczynska, E., 2006b. Effect of theselective 5-HT7 receptor antagonist SB 269970 in animal models of anxiety anddepression. Neuropharmacology 51, 578e586.

Westrich, L., Pehrson, A., Zhong, H., Nielsen, S.M., Frederiksen, K., Stensbøl, T.B.,Boyle, N., Hentzer, M., Sanchez, C., 2012. In vitro and in vivo effects of themultimodal antidepressant vortioxetine (Lu AA21004) at human and rat tar-gets. Int. J. Psychiatry Clin. Pract. 16 (Suppl. 1), 47.

Whale, R., Terao, T., Cowen, P., Freemantle, N., Geddes, J., 2010. Pindolol augmen-tation of serotonin reuptake inhibitors for the treatment of depressive disorder:a systematic review. J. Psychopharmacol. (Oxf.) 24, 513e520.

Xia, L., Delomenie, C., David, I., Rainer, Q., Marouard, M., Delacroix, H., David, D.J.,Gardier, A.M., Guilloux, J.P., 2012. Ventral hippocampal molecular pathways andimpaired neurogenesis associated with 5-HT(1)A and 5-HT(1)B receptorsdisruption in mice. Neurosci. Lett. 521, 20e25.

Zhang, X., Beaulieu, J.M., Sotnikova, T.D., Gainetdinov, R.R., Caron, M.G., 2004.Tryptophan hydroxylase-2 controls brain serotonin synthesis. Science 305, 217.

Zhang, Z.J., Schmidt, D.E., de Paulis, T., Trivedi, B.L., Onaivi, E.S., Ebert, M.H.,Hewlett, W.A., 2001. Anxiolytic-like effects of DAIZAC, a selective high-affinity5-HT(3) receptor antagonist, in the mouse elevated plus-maze. Pharmacol.Biochem. Behav. 69, 571e578.

J.-P. Guilloux et al. / Neuropharmacology 73 (2013) 147e159 159

Page 14: Antidepressant and anxiolytic potential of the multimodal - EA3544

-- SUPPLEMENTARY INFORMATION --

Antidepressant and anxiolytic potential of the multimodal antidepressant

vortioxetine (Lu AA21004) assessed by behavioral and neurogenesis outcomes in

mice

Short Title: Antidepressant effects of vortioxetine

Authors: Jean-Philippe Guilloux1, Indira Mendez-David1, Alan Pehrson2, Bruno P. Guiard1,

Christelle Repérant1, Sophie Orvoën1, Alain M. Gardier1, René Hen4, Bjarke Ebert3, Silke

Miller2,*, Connie Sanchez2, Denis J. David1,§

1EA 3544, Lab. Neuropharmacologie, Faculté de Pharmacie, Université Paris-Sud XI, Châtenay-

Malabry F-92296, France. 2External Sourcing, Lundbeck Research USA, 215 College Road, Paramus, 07652 NJ, USA 3H. Lundbeck A/S, Medical Affairs Mood & Anxiety, Ottiliavej 9, DK-2500 Valby, Denmark 4Departments of Neuroscience and Psychiatry, Columbia University, New York, NY, 10032,

USA

§ To whom correspondence should be addressed. Dr Denis J. David Univ Paris-Sud, EA 3544, Faculté de Pharmacie, 5, rue JB Clément 92296 Châtenay-Malabry cedex France. Tel: +33.1.46.83.59.68 Fax: +33.1.46.83.53.55 E-mail: [email protected]

* Present address of Silke Miller: Silke Miller, PhD. Amgen Inc. One Amgen Center Dr. MS 29-2-B Thousand Oaks, CA 91320

Authors e-mail addresses J.P. Guilloux: [email protected] I. Mendez-David: [email protected] A. Pehrson: [email protected] B.P. Guiard: [email protected] C. Repérant: [email protected] S. Orvoën: [email protected]

A.M. Gardier: [email protected] R. Hen: [email protected] B. Ebert: [email protected] S. Miller: [email protected] C. Sanchez: [email protected] DJ. David: [email protected]

Page 15: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Table 1: Overall statistical results

Test

Parameter

Treatm

ent*g

roup

s*num

ber

Total*num

ber*o

f*sam

ples

One

3Way*ANOVA

*F3value

One

3Way*ANOVA

*P3value

Vortioxetin

e*(2.5*m

g/kg)*vs*V

eh

Vortioxetin

e*(5*m

g/kg)*vs*V

eh

Vortioxetin

e*(10*mg/kg)*vs*V

eh

Vortioxetin

e*(20*mg/kg)*vs*V

eh

Flx*vs*Veh

Diaz*vs*V

eh

Vortioxetin

e*(2.5*m

g/kg)*vs*F

lx

Vortioxetin

e*(5*m

g/kg)*vs*F

lx

Vortioxetin

e*(10*mg/kg)*vs*F

lx

Vortioxetin

e*(20*mg/kg)*vs*F

lx

Vortioxetin

e*(2.5*m

g/kg)*vs*D

iaz

Vortioxetin

e*(5*m

g/kg)*vs*D

iaz

Vortioxetin

e*(10*mg/kg)*vs*D

iaz

Table*1129S6/SvEvTac SERT-occupancy/specific-bound Acute 3 9 247.2 0.0001 NA 0.0001 NA NA 0.0001 NA NA 0.01 NA NA NA NA NA129S6/SvEvTac SERT-occupancy/specific-bound 14-days 3 9 1.484 0.299 NA >0.05 NA >0.05 NA NA NA NA NA NA NA NA NA129S6/SvEvTac SERT-occupancy/specific-bound 21-days 4 12 220.5 0.0001 NA >0.05 NA >0.05 0.0001 NA NA 0.0001 NA 0.0001 NA NA NA

Balb/cJ SERT-occupancy/specific-bound Acute 4 12 81.57 0.0001 NA 0.0001 0.0001 NA 0.0001 NA NA 0.01 0.05 NA NA NA NABalb/cJ SERT-occupancy/specific-bound 21-days 4 12 116.90 0.0001 NA >0.05 NA >0.05 0.0001 NA NA 0.0001 NA 0.0001 NA NA NA

Table*2129S6/SvEvTac 5GHT3-occupancy/specific-bound Acute 3 9 88.8 0.0001 NA 0.0001 NA NA >0.05 NA NA 0.0001 NA NA NA NA NA129S6/SvEvTac 5GHT3-occupancy/specific-bound 14-days 3 9 0.169 0.8482 NA >0.05 NA >0.05 NA NA NA NA NA NA NA NA NA129S6/SvEvTac 5GHT3-occupancy/specific-bound 21-days 4 12 0.65 0.6067 NA >0.05 NA >0.05 >0.05 NA NA >0.05 NA >0.05 NA NA NA

Balb/cJ 5GHT3-occupancy/specific-bound Acute 4 12 80.2 0.0001 NA 0.0001 0.0001 NA >0.05 NA NA 0.0001 0.0001 NA NA NA NABalb/cJ 5GHT3-occupancy/specific-bound 21-days 4 12 7.25 0.011 NA >0.05 NA <0.05 >0.05 NA NA >0.05 NA >0.05 NA NA NA

Figure*11A Open-Field Time-in-the-center 6 68 2.71 0.03 0.03 0.05 0.15 NA 0.58 0.04 0.03 0.08 0.31 NA 0.93 0.64 0.171B Open-Field Entries-in-the-center 6 68 1.67 0.16 0.04 0.68 0.38 NA 0.71 0.04 0.10 0.97 0.67 NA 0.81 0.09 0.121C Open-Field Distance-in-the-center 6 68 3.28 0.01 0.04 0.05 0.48 NA 0.37 0.01 0.02 0.01 0.15 NA 0.46 0.57 0.081D Open-Field Distance-in-periphery 6 68 2.16 0.07 0.85 0.42 0.15 NA 0.06 0.08 0.40 0.82 0.91 NA 0.85 0.42 0.47

Supplementary-Figure-3 Open-Field Total-Ambulatory-Distance 6 68 2.49 0.04 0.21 0.39 0.21 NA 0.80 0.01 0.16 0.30 0.16 NA 0.01 0.12 0.061E Forced-Swimming-Test Mobility 6 57 3.33 0.17 0.42 0.01 0.09 NA 0.07 NA 0.26 0.14 0.82 NA NA NA NA1F Forced-Swimming-Test Swimming 6 57 3.23 0.19 0.43 0.00 0.09 NA 0.06 NA 0.23 0.17 0.81 NA NA NA NA1G Forced-Swimming-Test Climbing 6 57 1.55 0.20 0.57 0.03 0.48 NA 0.48 NA 0.96 0.11 0.98 NA NA NA NA

Figure22B Novelty-Suppressed-Feeding Latency-to-feed 6 60 7.353 0.000 0.418 0.009 0.241 NA 0.057 0.007 0.263 0.000 0.003 NA 0.001 0.518 0.112

Supplementary-Figure-4 Novelty-Suppressed-Feeding Food-Consumption 6 60 1.169 0.336 0.990 0.046 0.271 NA 0.364 0.523 0.369 0.268 0.846 NA 0.530 0.169 0.642

Figure*33A Open-Field Time-in-the-center 4 52 3.090 0.035 NA 0.009 NA 0.680 0.110 NA NA 0.240 NA 0.230 NA NA NA3B Open-Field Entries-in-the-center 4 52 1.597 0.202 NA 0.060 NA 0.857 0.674 NA NA 0.123 NA 0.808 NA NA NA3C Open-Field Distance-in-the-center 4 52 1.213 0.315 NA 0.161 NA 0.991 0.226 NA NA 0.818 NA 0.214 NA NA NA3D Open-Field Distance-in-periphery 4 52 1.633 0.194 NA 0.087 NA 0.678 0.111 NA NA 0.241 NA 0.226 NA NA NA

Supplementary-Figure-5 Open-Field Total-Ambulatory-Distance 4 52 1.498 0.227 NA 0.040 NA 0.240 0.275 NA NA 0.289 NA 0.915 NA NA NA3E Forced-Swimming-Test Mobility 4 46 4.600 0.007 NA 0.006 NA 0.938 0.694 NA NA 0.003 NA 0.750 NA NA NA3F Forced-Swimming-Test Swimming 4 46 4.019 0.013 NA 0.114 NA 0.662 0.494 NA NA 0.003 NA 0.273 NA NA NA3G Forced-Swimming-Test Climbing 4 46 2.758 0.054 NA 0.047 NA 0.491 0.899 NA NA 0.075 NA 0.434 NA NA NA

Figure*44B Novelty-Suppressed-Feeding Latency-to-feed 4 74 3.410 0.020 NA 0.699 NA 0.067 0.213 NA NA 0.107 NA 0.002 NA NA NA

Supplementary-Figure-6A Novelty-Suppressed-Feeding Food-Consumption 4 74 7.830 0.000 NA 0.699 NA 0.961 0.000 NA NA 0.000 NA 0.000 NA NA NA4D Novelty-Suppressed-Feeding Latency-to-feed 4 74 2.040 0.110 NA 0.046 NA 0.368 0.896 NA NA 0.033 NA 0.301 NA NA NA

Supplementary-Figure-6B Novelty-Suppressed-Feeding Food-Consumption 4 74 3.280 0.025 NA 0.237 NA 0.672 0.078 NA NA 0.030 NA 0.250 NA NA NA

Figure*55A Proliferation BrdU-positive-cells-(proliferation) 4 31 1.760 0.170 NA 0.039 NA 0.105 0.157 NA NA 0.511 NA 0.988 NA NA NA5B Survival BrdU-positive-cells-(survival) 4 29 4.910 0.080 NA 0.037 NA 0.207 0.010 NA NA 0.198 NA 0.021 NA NA NA5C Maturation Total-DCX-Cells 4 22 0.368 0.776 NA 0.819 NA 0.696 0.512 NA NA 0.651 NA 0.320 NA NA NA5D Maturation DCX-cells-with-tertiary-dendrites 4 22 3.251 0.046 NA 0.031 NA 0.016 0.022 NA NA 0.687 NA 0.890 NA NA NA5E Maturation Maturation-Index 4 22 5.545 0.007 NA 0.017 NA 0.000 0.029 NA NA 0.981 NA 0.247 NA NA NA

Figure*66A Maturation Total-DCX-Cells 4 16 1.010 0.418 NA 0.220 NA 0.987 0.402 NA NA 0.721 NA 0.342 NA NA NA6B Maturation DCX-cells-with-tertiary-dendrites 4 16 3.730 0.042 NA 0.253 NA 0.125 0.800 NA NA 0.374 NA 0.075 NA NA NA6C Maturation Maturation-Index 4 16 0.891 0.470 NA 0.707 NA 0.162 0.494 NA NA 0.718 NA 0.513 NA NA NA6E Maturation Dendritic-Length 4 16 19.150 <0,0001 NA >0,05 NA <0,0001 <0,05 NA NA <0,01 NA <0,05 NA NA NA6F Maturation Dendritic-Intersection 4 16 17.350 <0.0001 NA >0,05 NA <0,0001 <0,01 NA NA <0,01 NA >0,05 NA NA NA

Page 16: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 1: Timeline of experiments

In acute studies (A), behavioral effects of acute diazepam (1.5 mg/kg), fluoxetine (18

mg/kg) or vortioxetine (2.5, 5 and 10 mg/kg) were monitored in the Open Field and

Forced Swimming Test in BALB/cJ@RJ mice and in the Novelty Suppressed Feeding

Test in 129S6/SvEvTac one hour after injection (p.o.). In chronic studies (B), behavioral

effects of chronic (14 or 21 days, p.o.) fluoxetine (18 mg/kg) or vortioxetine (5 and 20

mg/kg) were monitored in the Open Field and Forced Swimming Test in BALB/cJ@RJ

mice and in the Novelty Suppressed Feeding Test in 129S6/SvEvTac and then sacrified

for adult hippocampal neurogenesis study.

Page 17: Antidepressant and anxiolytic potential of the multimodal - EA3544

Behavior Analysis(OF, FST)

7 14 days0

Behavior Analysis(OF, FST)

7 14 21 days0

B. Chronic Studies

BALB/cJ@Rj

Drug administration Drug administration

Behavior Analysis(NSF)

Neurogenesis Analysis

7 14 days0

Behavior Analysis(NSF)

Neurogenesis Analysis

7 14 21 days0

129S6/SvEvTac

Drug administration Drug administration

Supplementary Figure 1

Behavior Analysis(OF or FST)

Drugadministration

A. Acute Studies

0 60 min

BALB/cJ@Rj

Behavior Analysis(NSF)

Drugadministration

0 60 min

129S6/SvEvTac

Page 18: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 2: Regions of Interest (ROIs) for the SERT and 5-HT3

receptor occupancy assays.

In the SERT occupancy assay (A), the ROI included the lateral septum (LS), medial

septum (MS), Nucleus Accumbens (NAc) and olfactory tubercle (Tu). The ROI for the 5-

HT3 receptor occupancy (B) assay consisted of the hippocampus (Hip).

Page 19: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 2

A

B

TotalBound

Non-SpecificBound

Page 20: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 3: Effects of acute vortioxetine treatment on

locomotor activity in the open field paradigm.

The effects of acute diazepam (1.5 mg/kg), fluoxetine (18 mg/kg) or vortioxetine (2.5, 5

and 10 mg/kg) on overall locomotor activity were measured during 30 minutes in the

open field paradigm, 1h after their administration (p.o.) (See Methods section). Data are

expressed as and mean ± SEM of distance travelled (n=10-12 animals/group). One way

ANOVA analysis showed a main effect of treatment on locomotor activity (F5,62=2.49,

p=0.04), and a significant effect of diazepam to increase total ambulatory distance.

Page 21: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 3

Tota

l Am

bula

tory

Dis

tanc

e (c

m)

0

250

500

750

1000

1250

1500

1750

2000

TreatmentsVehicle

Diazepam 1.5 mg/kg

Fluoxetine 18 mg/kg

Vortioxetine 2.5 mg/kg

Vortioxetine 5 mg/kg

Vortioxetine 10 mg/kg

Veh Diaz

**

Flx

Vortioxetine

5 mg/kg2.5 mg/kg 10 mg/kg

Page 22: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 4: Effects of acute vortioxetine treatment on food

consumption following the Novelty Supressed Feeding paradigm in

129/SvEvTac mice.

The effects of vortioxetine on the amount of food consumed during the subsequent 5

minutes following the behavioral test were measured after 1-hour administration at doses

of 2.5, 5 and 10 mg/kg, compared to vehicle-treated group and fluoxetine (18 mg/kg) and

diazepam (1.5 mg/kg). Data are expressed as and mean ± SEM of food consumed

normalized by mouse individual weight (n=15-20 animals/group). One way ANOVA

analysis showed no main effect of food consumption (F3,48=1.56, p=0.18).

Page 23: Antidepressant and anxiolytic potential of the multimodal - EA3544

Food

Con

sum

ptio

n (m

g/g

of m

ouse

wei

ght)

Supplementary Figure 4

0

1

2

3

Veh Diaz Flx

Vortioxetine

5 mg/kg2.5 mg/kg 10 mg/kg

TreatmentsVehicle

Diazepam 1.5 mg/kg

Fluoxetine 18 mg/kg

Vortioxetine 2.5 mg/kg

Vortioxetine 5 mg/kg

Vortioxetine 10 mg/kg

Page 24: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 5: Effects of chronic vortioxetine treatment on

locomotor activity in the open field paradigm.

The effects of chronic fluoxetine (18 mg/kg) or vortioxetine (5 and 20 mg/kg) on overall

locomotor activity were measured during 30 minutes in the open field paradigm. Data are

expressed as and mean ± SEM of distance travelled (n=10-12 animals/group). One way

ANOVA analysis showed no main effect of treatment on locomotor activity (F3,48=1.50,

p=0.23).

Page 25: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 5

Veh Flx

Vortioxetine

5 mg/kg 20 mg/kg0

500

1000

1500

2000

2500

3000

3500

4000

Tota

l Am

bula

tory

Dis

tanc

e (c

m)

TreatmentsVehicle

Fluoxetine 18 mg/kg/day

Vortioxetine 5 mg/kg/day

Vortioxetine 20 mg/kg/day

Page 26: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 6: Effects of chronic vortioxetine treatment on food

consumption following the Novelty Supressed Feeding paradigm in

129/SvEvTac mice.

The effects of vortioxetine on the amount of food consumed during the subsequent 5

minutes following the behavioral test were measured after 14 (A) or 21 (B) days-

administration at doses of 5 and 20 mg/kg, compared to vehicle-treated group and

fluoxetine (18 mg/kg). Data are expressed as and mean ± SEM of food consumed

normalized by mouse individual weight (n=15-20 animals/group). **p<0.01 for effects of

fluoxetine compare to the vehicle-treated group (n=15-20 animals/group). One way

ANOVA analysis showed no main effect of food consumption for day 14 (F3,74=7.83,

p=0.01) and main effect of food consumption day 21 (F3,74=3.23, p=0.02).

Page 27: Antidepressant and anxiolytic potential of the multimodal - EA3544

BB21 days

**Fo

od C

onsu

mpt

ion

(mg/

g of

mou

se w

eigh

t)

Veh Flx

Vortioxetine

5 mg/kg 20 mg/kg0

2

4

6

8

10

12

Food

Con

sum

ptio

n(m

g/g

of m

ouse

wei

ght)

A14 days

**

0

2

4

6

8

10

12

Supplementary Figure 6

TreatmentsVehicle

Fluoxetine 18 mg/kg/day

Vortioxetine 5 mg/kg/day

Vortioxetine 20 mg/kg/day

Veh Flx

Vortioxetine

5 mg/kg 20 mg/kg

Page 28: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 7: Effects of chronic vortioxetine treatment on cell

proliferation in the hippocampus of adult 129/SvEvTac mice.

Representative illustrations of BrdU immunoreactivity (10X magnification) in the dentate

gyrus of 129/SvEvTac mice following chronic administration (21 days) of either

fluoxetine (18 mg/kg/d) or vortioxetine (5 or 20 mg/kg/d).

Page 29: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 7

100 µm

Vehicle Fluoxetine 18 mg/kg/day

Vortioxetine 20 mg/kg/dayVortioxetine 5 mg/kg/day

Page 30: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 8: Effects of chronic vortioxetine treatment on cell

survival in the hippocampus of adult 129/SvEvTac mice.

Representative illustrations of BrdU immunoreactivity (10X magnification) in the dentate

gyrus of 129/SvEvTac mice following chronic administration (21 days) of either

fluoxetine (18 mg/kg/d) or vortioxetine (5 or 20 mg/kg/d).

Page 31: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 8

100 µm

Vehicle Fluoxetine 18 mg/kg/day

Vortioxetine 20 mg/kg/dayVortioxetine 5 mg/kg/day

Page 32: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 9: Effects of chronic vortioxetine treatment on

maturation of neurons in the hippocampus of adult 129/SvEvTac mice.!

Representative illustrations of doublecortin staining (10X magnification) following

chronic administration (14 days) of either fluoxetine (18 mg/kg/d) or vortioxetine (5 or

20 mg/kg/d).

Page 33: Antidepressant and anxiolytic potential of the multimodal - EA3544

Supplementary Figure 9

100 µm

Vehicle Fluoxetine 18 mg/kg/day

Vortioxetine 20 mg/kg/dayVortioxetine 5 mg/kg/day