Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Pharmacology and Experimental Therapeutics
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • My Cart
Journal of Pharmacology and Experimental Therapeutics

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Visit jpet on Facebook
  • Follow jpet on Twitter
  • Follow jpet on LinkedIn
Research ArticleNeuropharmacology
Open Access

Effects of the Glycine Transporter-1 Inhibitor Iclepertin (BI 425809) on Sensory Processing, Neural Network Function, and Cognition in Animal Models Related to Schizophrenia

Holger Rosenbrock, Cornelia Dorner-Ciossek, Riccardo Giovannini, Bernhard Schmid and Niklas Schuelert
Journal of Pharmacology and Experimental Therapeutics August 2022, 382 (2) 223-232; DOI: https://doi.org/10.1124/jpet.121.001071
Holger Rosenbrock
Department of CNS Discovery Research (H.R., C.D.-C., N.S.), Department of Medicinal Chemistry (R.G.), and Department of Drug Discovery Sciences (B.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Cornelia Dorner-Ciossek
Department of CNS Discovery Research (H.R., C.D.-C., N.S.), Department of Medicinal Chemistry (R.G.), and Department of Drug Discovery Sciences (B.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Riccardo Giovannini
Department of CNS Discovery Research (H.R., C.D.-C., N.S.), Department of Medicinal Chemistry (R.G.), and Department of Drug Discovery Sciences (B.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Bernhard Schmid
Department of CNS Discovery Research (H.R., C.D.-C., N.S.), Department of Medicinal Chemistry (R.G.), and Department of Drug Discovery Sciences (B.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Niklas Schuelert
Department of CNS Discovery Research (H.R., C.D.-C., N.S.), Department of Medicinal Chemistry (R.G.), and Department of Drug Discovery Sciences (B.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF + SI
  • PDF
Loading

Visual Overview

Figure
  • Download figure
  • Open in new tab
  • Download powerpoint

Abstract

N-methyl-D-aspartate (NMDA) receptor hypofunction leading to neural network dysfunction is thought to play an important role in the pathophysiology of cognitive impairment associated with schizophrenia (CIAS). Increasing extracellular concentrations of the NMDA receptor co-agonist glycine through inhibition of glycine transporter-1 (GlyT1) has the potential to treat CIAS by improving cortical network function through enhanced glutamatergic signaling. Indeed, the novel GlyT1 inhibitor iclepertin (BI 425809) improved cognition in a recent clinical study in patients with schizophrenia. The present study tested the ability of iclepertin to reverse deficits in auditory sensory processing and cortical network function induced by the uncompetetive NMDA receptor antagonist, MK-801, using electroencephalography (EEG) to measure auditory event-related potentials (AERPs) and 40 Hz auditory steady-state response (ASSR). In addition, improvements in memory performance with iclepertin were evaluated using the T-maze spontaneous alternation test in MK-801–treated mice and the social recognition test in naïve rats. Iclepertin reversed MK-801–induced deficits in the AERP readouts N1 amplitude and N1 gating, as well as reversing deficits in 40 Hz ASSR power and intertrial coherence. Additionally, iclepertin significantly attenuated an MK-801–induced increase in basal gamma power. Furthermore, iclepertin reversed MK-801–induced working memory deficits in mice and improved social recognition memory performance in rats. Overall, this study demonstrates that inhibition of GlyT1 is sufficient to attenuate MK-801–induced deficits in translatable EEG parameters relevant to schizophrenia. Moreover, iclepertin showed memory-enhancing effects in rodent cognition tasks, further demonstrating the potential for GlyT1 inhibition to treat CIAS.

SIGNIFICANCE STATEMENT Despite the significant patient burden caused by cognitive impairment associated with schizophrenia, there are currently no approved pharmacotherapies. In this preclinical study, the novel glycine transporter inhibitor iclepertin (BI 425809) reversed sensory processing deficits and neural network dysfunction evoked by inhibition of N-methyl-D-aspartate receptors and enhanced working memory performance and social recognition in rodents. These findings support previous clinical evidence for the procognitive effects of iclepertin.

Introduction

Patients with schizophrenia experience debilitating cognitive symptoms related to deficits in sensory processing and neural network function (O’Donnell et al., 2013; Javitt and Freedman, 2015; Schuelert et al., 2018; Shen et al., 2020); however, there are currently no approved pharmacotherapies targeting cognitive impairment associated with schizophrenia (CIAS) (Sinkeviciute et al., 2018). Thus, there is a need for better understanding of the mechanisms underlying sensory processing deficits and neural network dysfunction in CIAS to support the development of effective treatments.

Sensory processing deficits and neural network dysfunction in patients with schizophrenia can be assessed using neurophysiological biomarkers detectable with electroencephalography (EEG) (O’Donnell et al., 2013; Javitt and Freedman, 2015; Schuelert et al., 2018; Shen et al., 2020). Since the relevant sensory processes are phylogenetically conserved, these deficits have the potential to be modeled in preclinical studies (Bickel and Javitt, 2009; Rosen et al., 2015; Loiodice et al., 2021) for use as translatable biomarkers in the development of novel pharmacotherapies for CIAS (Javitt et al., 2008; Light and Swerdlow, 2015; Kim et al., 2020; Light et al., 2020). Patients with schizophrenia exhibit deficits in sensory gating, the process by which redundant or unnecessary stimuli are filtered out to prevent overload of higher-order processes (Shen et al., 2020). Deficits in auditory event-related potentials (AERPs) in paradigms such as the 40 Hz auditory steady-state response (ASSR), which demonstrates the ability of auditory circuits to support entrainment to temporally or continuous amplitude modulated tones, have been described in patients with schizophrenia (O’Donnell et al., 2013; Javitt and Freedman, 2015). Patients also exhibit disturbances in local field potential oscillations in the gamma frequency range (∼25–100 Hz), which are critical for coordinating network activity during typical brain functioning (Schuelert et al., 2018) and integral to several aspects of learning and memory (Uhlhaas and Singer, 2010; Cohen et al., 2015; Stefanescu and Shore, 2015).

N-methyl-D-aspartate (NMDA) receptors play a key role in cognition and sensory processing by providing excitatory input to gamma aminobutyric-acid (GABA) inhibitory interneurons, supporting the generation of gamma oscillations in cortical regions (Collingridge et al., 2013; Cohen et al., 2015). Thus, NMDA receptor hypofunction and GABAergic interneuron dysfunction are implicated in the pathophysiology of CIAS (Dauvermann et al., 2017; Uno and Coyle, 2019). NMDA receptor hypofunction is thought to disturb the excitatory/inhibitory (E/I) balance in prefrontal cortex (PFC) through impaired interneuron function resulting in disinhibition of pyramidal cells, leading to the disruption of gamma oscillations and impaired network connectivity in PFC (Gonzalez-Burgos and Lewis, 2012; Lisman, 2012; Cohen et al., 2015; Schoonover et al., 2020).

Postmortem studies of patients with schizophrenia reveal altered expression of NMDA receptor, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptor subunits, and proteins associated with clustering of both receptors (Beneyto and Meador-Woodruff, 2008; Vrajová et al., 2010; Forsyth and Lewis, 2017; Uno and Coyle, 2019). Large-scale genomic studies have demonstrated a strong link between schizophrenia and gene variants related to glutamatergic signaling, including the NMDA receptor complex (Kirov et al., 2012; Fromer et al., 2014; Schizophrenia Working Group of the Psychiatric Genomics Consortium, 2014; Forsyth and Lewis, 2017). Moreover, postmortem studies of patients with schizophrenia have found a reduced density of small interneurons and a 73% reduction in GABAergic interneurons expressing the NR2A NMDA receptor subunit in the cingulate cortex (Cohen et al., 2015). These data support a role for inhibitory interneuron dysfunction mediated by NMDA receptor hypofunction in the pathophysiology of CIAS (Dauvermann et al., 2017; Uno and Coyle, 2019).

The link between NMDA hypofunction, E/I imbalance, and CIAS is supported by the effects of noncompetitive NMDA receptor antagonists such as ketamine and phencyclidine, which induce psychosis and schizophrenia-like cognitive symptoms in healthy volunteers and exacerbate symptoms in patients with schizophrenia (Krystal et al., 1994; Moghaddam and Krystal, 2012). NMDA receptor antagonists also induce sensory deficits and neural network dysfunction in animal models and humans similar to those observed in patients. Exposure to NMDA receptor antagonists causes heterogeneous disturbances in gamma oscillations in rodents (Pinault, 2008; Hakami et al., 2009) and healthy volunteers (Hong et al., 2010; Ahnaou et al., 2017), which may impair synchrony and reduce signal-to-noise ratio in neural networks (Hakami et al., 2009). These findings are in line with reports describing atypical gamma oscillations in patients with schizophrenia compared with healthy controls (Cho et al., 2006; Hirano et al., 2015; Tanaka-Koshiyama et al., 2020), although the precise nature of these abnormalities varies across experimental paradigms (Molina et al., 2020).

Therefore, novel pharmacotherapies to ameliorate NMDA receptor hypofunction have the potential to reverse deficits in sensory processing, restore neural network function, and improve cognition (Collingridge et al., 2013). Examples of novel pharmacotherapies include NMDA receptor positive allosteric modulators (PAMs) that directly activate the NMDA receptor, D-amino acid oxidase (DAAO) inhibitors that increase the synaptic concentration of the NMDA receptor co-agonist D-serine, and kynurenine aminotransferase-2 (KAT-2) inhibitors that decrease the concentration of the endogenous competitive NMDA receptor antagonist kynurenic acid (Yoshida et al., 2019; Pei et al., 2021; Wu et al., 2021; Geoffroy et al., 2022). NMDA receptor PAMs and DAAO inhibitors have reached phase I and II clinical trials, respectively, whereas the KAT-2 inhibitors are still in preclinical development. Another novel strategy to ameliorate NMDA receptor hypofunction, which has progressed into phase III clinical trials, involves increasing the synaptic concentration of glycine, an obligatory co-agonist at NMDA receptors, by inhibition of glycine transporter-1 (GlyT1) (Harvey and Yee, 2013; Pinard et al., 2018; Pei et al., 2021; Wu et al., 2021). Synaptic glycine concentration is regulated by GlyT1, which is expressed both pre- and postsynaptically at glutamatergic synapses and on astrocytes (Zafra and Giménez, 2008; Harvey and Yee, 2013; Marques et al., 2020). Thus, increasing synaptic glycine concentration by inhibiting GlyT1 is thought to enhance NMDA receptor function (Depoortère et al., 2005; Alberati et al., 2012; Harvey and Yee, 2013; Ahnaou et al., 2020), which would be expected to normalize network function and therefore ameliorate cognitive impairment. Several GlyT1 inhibitors have been developed previously and evaluated in clinical trials for indications such as schizophrenia (positive, negative, or cognitive symptoms), obsessive-compulsive disorder, and addiction (Cioffi and Guzzo, 2016; Pei et al., 2021). One of the first GlyT1 inhibitors, the prototype inhibitor sarcosine, showed efficacy for the treatment of positive, negative, and cognitive symptoms of schizophrenia in small clinical trials (Tsai et al., 2004; Lane et al., 2010). However, development of sarcosine and other GlyT1 inhibitors has been later discontinued due to adverse effects, negative results in phase II, or undisclosed reasons (Cioffi and Guzzo, 2016; Pei et al., 2021). The most advanced GlyT1 inhibitor, bitopertin, showed promising phase II results for treatment of negative symptoms in schizophrenia, but efficacy on negative symptoms was not confirmed in larger phase III trials (Pinard et al., 2018).

Iclepertin is a novel GlyT1 inhibitor under development for treatment of CIAS. Previous studies have demonstrated that iclepertin is a potent and selective inhibitor of GlyT1 that shows functional target engagement by increased glycine levels in the cerebrospinal fluid (CSF) of rodents and healthy volunteers (Moschetti et al., 2018a,b; Rosenbrock et al., 2018). Furthermore, iclepertin has demonstrated procognitive effects in a phase II trial in patients with schizophrenia (Fleischhacker et al., 2021). Phase III trials investigating the efficacy and safety of iclepertin in patients with schizophrenia are currently underway (NCT04846868, NCT04846881, and NCT04860830). We report preclinical studies characterizing the effects of iclepertin on EEG biomarkers related to sensory processing and neural network function and on rodent cognition tasks related to schizophrenia.

Materials and Methods

Drugs

Iclepertin (BI 425809) was synthesized at Boehringer Ingelheim Research Italia (Milan, Italy). The molecular structure of iclepertin is shown in Fig. 1. The uncompetetive NMDA receptor antagonist, MK-801, was obtained from Sigma (Saint-Quentin-Fallavier, France).

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Structure of iclepertin.

Ethical Considerations

Procedures involving animals and their care were conducted in conformity with institutional and European Union guidelines (EEC Council Directive 86/609 and Directive 2010/63/EU) and were approved by the ethical committees of the respective regional councils.

In Vivo Pharmacokinetics of Iclepertin in Rats

To assess plasma exposure of iclepertin, male Han Wistar rats (Charles River, Sulzfeld; 8 weeks old, 250–270g, n = 3) were treated orally with iclepertin 2.6 mg/kg in a Natrosol (Ashland, Krefeld)/Tween-80 suspension (Dr. W. Kolb AG, Stuttgart) used as vehicle. Iclepertin was suspended under stirring in 0.5% Natrosol/0.01% Tween-80 in water and then sonicated for 10 minutes, resulting in a fine, slightly cloudy suspension. After retrobulbar puncture, blood was collected in ethylenediaminetetraacetic acid–containing tubes at 0, 0.25, 0.5, 1, 2, 4, 8, and 24 hours after administration of iclepertin. Samples were centrifuged at 2000 g for 10 minutes at 4°C, and the upper phase (plasma) was stored at −20°C. Iclepertin plasma concentrations were determined using liquid chromatography coupled to mass spectrometry. Iclepertin plasma concentrations were determined by liquid chromatography tandem-mass spectrometry (LC-MS/MS) using an API 4000 system (SCIEX). Plasma was precipitated using acetonitrile/water (80/20 v/v). After centrifugation at 2000 g for 10 minutes at 4°C, an aliquot of the upper phase was directly injected into the LC-MS/MS system. A linear calibration standard curve (13 concentrations) with 1/X weighting was used to determine iclepertin concentrations over 1–10,000 nmol/l.

EEG Biomarkers in the MK-801 Challenge Model in Mice

Animals

Male C57BL/6JRj mice (12–14 weeks old, 28–30 g, n = 21) (Janvier Laboratories, Le Genest-Saint-Isle, France) were group housed prior to surgery (four animals per cage) and single housed after surgery under standard light-dark conditions (12 hour/12 hour, lights on 06:00–18:00) with food and tap water available ad libitum. C57BL/6JRj mice were used as they are well suited for neurophysiological cortical network investigations using EEG techniques (Schuelert et al., 2018). Five epidural EEG electrodes were placed 10 days prior to recording (PFC: anterior posterior (AP) = +1.7 mm, medial lateral (ML): ± 1.0 mm; auditory cortex (AC): AP = −2.7 mm, ML: ± 4.0 mm; reference electrode placement: AP = −5.7 mm, ML: ± 0 mm). Blood samples were collected from satellite mice (n = 3 per dose group) to measure plasma drug exposure 60 minutes after subcutaneous administration.

Procedure

Mice were placed in a plexiglass cage in an attenuated sound box and allowed to adapt to their environment for 20 minutes before subcutaneous administration of iclepertin 0.3, 1, or 4 mg/kg or vehicle (20% HP-β-CD in water). Iclepertin was dissolved in 20% HP-β-CD in water and then sonicated for 10 minutes, resulting in a clear solution. After 15 minutes, all animals then received a subcutaneous dose of the MK-801 0.15 mg/kg or vehicle (saline). All injections had a total volume of 10 ml/kg. A crossover treatment schedule was used such that each mouse received all treatment conditions separated by a 3-day washout period. EEG, AERP, and auditory evoked oscillation recordings in PFC and AC began 15 minutes after administration of either MK-801 or vehicle, and were conducted as described previously (Schuelert et al., 2018).

EEG Analysis

EEG parameters were analyzed as previously described (Schuelert et al., 2018). Briefly, four recording channels (one AC channel and one PFC channel for each hemisphere) and one trigger channel (to identify the timing of tone presentations) were analyzed per animal. The sampling rate of recording channels and trigger channels was set to 1000 Hz. Any EEG recording segments with artifacts were removed. Analyzer 2 software (Brain Products GmbH, Munich, Germany) was used for data analysis.

EEG parameters of interest that were tested using a double-click protocol (Schuelert et al., 2018) included N1 amplitude, N1 gating, and basal and evoked gamma power. N1 (also known as N100) is a large negative potential elicited by sensory stimuli, which peaks approximately 100 milliseconds after stimulus onset in humans (Rosburg et al., 2008; Joos et al., 2014; Du et al., 2017) and 40 milliseconds after stimulus onset in rodents (Aguilar et al., 2021). In this study, N1 was defined as the maximum negative deflection between 30 and 50 milliseconds after the click. A representative trace of the N1 response is shown in Supplemental Fig. 1A. Sensory gating of N1 amplitude between paired click stimuli was measured using the ratio between the N1 amplitude of the response evoked by click 1 and the N1 amplitude of the response evoked by click 2 to evaluate the capacity of the sensory system to filter repeated stimuli. Basal gamma power (50–100 Hz) was defined as power outside of the auditory evoked response (3 seconds before click presentations), whereas evoked gamma power (35–80 Hz) was defined as the total event-related power (evoked plus basal power) minus the basal power. These frequency ranges were selected to demonstrate the largest effect of MK-801 compared with vehicle, thus providing greater sensitivity to detect any effects of iclepertin on MK-801–induced changes in basal and evoked gamma power (Supplemental Fig. 2). ASSR intertrial coherence (ITC) and ASSR evoked gamma power were assessed by presentation of a click train 2 seconds in length with a frequency of 40 Hz. The ASSR paradigm explores cortical capacity to support an entrained 40 Hz oscillation. ASSR ITC is a measure of the phase synchronization of brain oscillations in response to auditory stimuli across trials, indicating the ability of the sensory network to stereotypically process repeated input (Schuelert et al., 2018). A representative trace of the ASSR response is shown in Supplemental Fig. 1B. An ITC of 0 represents an absence of synchrony (no phase locking), whereas a score of 1 equates to perfect synchronization (fully phase-locked).

T-Maze Spontaneous Alternation Task in Mice

Animals

Male CD-1 mice (4–5 weeks old, 25–30g; Janvier Laboratories) were group housed (10 mice per cage) in a temperature-controlled room (21–22°C) on a reversed light-dark cycle (12 hour/12 hour, lights on: 17:30–05:30) with food and water available ad libitum. Satellite mice (n = 3 per dose group) were used for blood sample collection to measure plasma drug exposure 60 minutes after oral administration. CD1 mice were used as they are well suited for the T-maze due to their strong exploratory behavior, especially if tested when in their active phase under a reversed light cycle.

Procedure

Mice (n = 10 in each treatment group) were randomly assigned to receive iclepertin 0.005, 0.05, 0.15, 0.5, 1.5, or 4.5 mg/kg or vehicle (0.5% Natrosol/0.01% Tween-80) orally 60 minutes prior to the T-maze test, in addition to MK-801 0.1 mg/kg or vehicle (saline) administered subcutaneously 30 minutes after administration of iclepertin. Iclepertin was suspended under stirring in 0.5% Natrosol/0.01% Tween-80 in water and then sonicated for 10 minutes, resulting in a fine, slightly cloudy suspension. The T-maze spontaneous alternation task was performed at Neurofit SAS. (Illkirch, France) using a nontransparent T-maze as previously described (Rosenbrock et al., 2019). The experimental protocol comprised an individual session with one forced-choice trial proceeded by 14 free-choice trials. During the forced-choice trial, either the left or right arm of the T-maze was blocked to allow mice to explore the open arm. Thereafter, mice were allowed to enter either arm in the 14 free-choice trials. An error response was recorded if a mouse entered the same arm on two subsequent trials. A session ended when 14 free-choice trials had been-completed or 10 minutes had elapsed, whichever occurred first. All mice completed the 14 free-choice trials within the 10-minute time frame. Working memory performance was assessed by the percentage of successful alternation over the 14 free-choice trials.

Social Recognition Task in Rats

Animals

The social recognition task was performed in rats rather than mice due to species differences in social cognition; rats are more likely to seek the company of other rats and are less territorial and less aggressive in social contexts than mice. This makes the rat a more appropriate species to address social cognition preclinically (Kent Scientific Corporation, 2019; Kondrakiewicz et al., 2019). Adult male Wistar rats (12–15 weeks old, 300–330 g, n = 72) and juvenile males (23–25 days old, 50–70 g, n = 72) (Janvier, Le Genest-Saint-Isle, France) were group housed (three animals per cage) under standard light-dark conditions (12 hour/12 hour, lights on: 07:00–19:00) with food and water available ad libitum.

Procedure

Animals were randomly assigned to receive iclepertin 0.2 (n = 17), 0.6 (n = 18), or 1.8 mg/kg (n = 18) or vehicle (0.5% Natrosol/0.01% Tween-80; n = 18) administered orally 60 minutes prior to trial 1 (T1) and trial 2 (T2) with a volume of 5 ml/kg. Iclepertin was suspended under stirring in 0.5% Natrosol/0.01% Tween-80 in water and then sonicated for 10 minutes, resulting in a fine, slightly cloudy suspension. Only the adult rats were administered with iclepertin; the juvenile rats received no treatment. The social recognition task was performed as previously described (van der Staay et al., 2008). Briefly, rats were tested in a black square box with the floor covered with bedding material and under dimmed light conditions. A camera above the box recorded video footage for analysis using a DVD recorder. The social recognition test was performed across two consecutive days. On day 1 (T1), a cospecific cogender juvenile rat was placed into the testing arena with the adult rat, and they were allowed to interact for 3 minutes. At the end of T1, both rats were returned to their home cage. After 24 hours, the adult rat was reintroduced to the testing arena for another 30 minutes of habituation, whereas juveniles were kept singly housed. After habituation, the familiar juvenile (used in T1) was introduced to the testing arena with the adult rat and allowed to interact for 3 minutes (T2). All digitalized trials were analyzed by a trained observer blinded to the experimental conditions and treatment group. Sniffing and grooming of body parts, anogenital sniffing, and close following were scored. The difference in social investigation time between T1 and T2 was used as an index of social memory performance. Overall investigation time in T1 and T2 was also noted to detect nonspecific treatment effects on arousal or attention.

Statistical Analysis

EEG data were analyzed using a generalized linear model with factors of ‘animal,’ ‘day,’ and ‘treatment.’ Mean and standard error of the mean (S.E.M.) were calculated over all treatment days. To focus on the procognitive effects of iclepertin, only findings from PFC are reported in the main manuscript; EEG data from AC are shown in the supplement. One-way analysis of variance (ANOVA) was performed on the T-maze spontaneous alternation task data with Fisher’s Protected Least Significant Difference for pairwise comparisons. For the rat social recognition task, quantitative data are presented as mean and S.E.M. and were analyzed using two-way ANOVA with post hoc Bonferroni tests. The significance threshold was set at P < 0.05 for all analyses.

Results

In Vivo Pharmacokinetics of Iclepertin

Mean plasma concentration of iclepertin over 24 hours in rats after an oral dose of 2.6 mg/kg is shown in Fig. 2. The mean maximum concentration in plasma of 1010 nM was reached at 1.67 hours, and the resulting area under the concentration time-curves from time zero to infinity was 21 400 nM*h. In a previous study, the concentration of iclepertin in CSF, which was used as a surrogate for free brain concentration, was determined on average as 2% of the plasma concentration (Rosenbrock et al., 2018).

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

In vivo pharmacokinetics of iclepertin in rats. Mean ± S.E.M. plasma concentration of iclepertin over 24 hours in rats (n = 3) after an oral dose of 2.5 mg/kg.

Effects of Iclepertin on Sensory Processing and Cortical Network Function in Mice

In PFC, treatment with iclepertin 0.3 (P < 0.05), 1 (P < 0.01), or 4 mg/kg (P < 0.05) significantly reversed MK-801–induced deficits in N1 amplitude and N1 gating (Fig. 3, A and B), whereas iclepertin 0.3 (P < 0.01) or 1 mg/kg (P < 0.05) significantly reversed deficits in 40 Hz ASSR evoked gamma power (Fig. 3C). Iclepertin 1 mg/kg reversed deficits in 40 Hz ASSR ITC (P < 0.05; Fig. 3D) and reversed MK-801–induced increases in basal gamma power (P < 0.05; Fig. 3E). Similarly, iclepertin 0.3, 1, or 4 mg/kg significantly reversed the effects of MK-801 in AC for N1 amplitude, N1 gating, and basal gamma power (P < 0.01; Supplemental Fig. 3), but no MK-801-induced deficit compared with vehicle was observed in AC for ASSR evoked gamma power or ASSR ITC (Supplemental Fig. 3, C and D). No MK-801–induced deficits were observed in either brain region for evoked gamma power in the click paradigm; treatment with iclepertin 0.3, 1, or 4 mg/kg significantly increased evoked gamma power compared with vehicle in both PFC and AC (P < 0.05; Fig. 3F; Supplemental Fig. 3F), whereas iclepertin 0.3 or 1 mg/kg significantly increased evoked gamma power in PFC compared with MK-801 (P < 0.05; Fig. 3F).

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Effects of iclepertin on EEG measurements in PFC. The effects of orally administered iclepertin 0.3, 1, or 4 mg/kg on EEG measurements in PFC in mice after administration of MK-801 0.15 mg/kg or vehicle. Data are shown as mean ± S.E.M. (n = 21 for each treatment group). *P < 0.05, **P < 0.01, ***P < 0.005 compared with vehicle; #P < 0.05, ##P < 0.01 compared with MK-801.

Mean plasma exposures in satellite mice after subcutaneous administration of iclepertin 0.3, 1, and 4 mg/kg were 251, 842, and 2650 nM, respectively. Based on a rat CSF/plasma ratio of 0.02, these values correspond to estimated iclepertin CSF levels in the range of 1- to 10-fold of the GlyT1 half-maximal inhibitory concentration (IC50) (IC50 on GlyT1 = 5.2 nM; Rosenbrock et al., 2018).

Effects of Iclepertin on Working Memory in Mice

Administration of MK-801 0.1 mg/kg led to a significant reduction in spontaneous alternation (i.e., the percentage of trials in which correct alternation was observed) from a mean of 67.9% with vehicle to 40.7% with MK-801 (P < 0.05; Fig. 4), corresponding approximately to chance level (50% correct alternation). Oral treatment of mice with iclepertin at doses of 0.5–4.5 mg/kg led to a significant attenuation or reversal of the MK-801–induced reduction in spontaneous alternation compared with the MK-801–treated group (n = 10 animals per group; P < 0.05; Fig. 4). For the assessment of a potential procognitive compound in the T-maze spontaneous alternation test, a pharmacologically induced impairment of performance by an NMDA receptor antagonist such as MK-801 is needed to evaluate the drug effect. This is because naïve mice perform well in the test, with an average alternation of approximately 70% that is unlikely to be substantially further improved by the study drug.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Effects of iclepertin on working memory in mice. The effects of orally administered iclepertin 0.005, 0.05, 0.15, 0.5, 1.5, or 4.5 mg/kg on spontaneous alternation in the T-maze task in mice, after administration of subcutaneous MK-801 0.1 mg/kg or vehicle. Data are shown as mean ± S.E.M. (n = 10 in each treatment group). *P < 0.05 vs. MK-801 0.1 mg/kg/vehicle; #P < 0.05 vs. saline/vehicle.

Mean plasma exposures in satellite mice after oral administration of 0.5, 1.5, and 4.5 mg/kg were 51, 509, and 1523 nM, respectively, corresponding to estimated CSF levels of iclepertin in the range of 0.2- to 6-fold of the GlyT1 IC50.

Effects of Iclepertin on Social Recognition Memory in Rats

Treatment with iclepertin 0.2, 0.6, and 1.8 mg/kg, but not vehicle, improved social recognition memory as shown by the significant reduction in social investigation times between T1 and T2 for all dose groups (iclepertin 0.2, 0.6 mg/kg: P < 0.01; iclepertin 1.8 mg/kg: P < 0.001) (Fig. 5). As this is a test for assessment of long-term memory (episodic/recognition memory), a potential procognitive compound like iclepertin is tested without an additional pharmacological challenge since the rats are already compromised in their memory performance due to the 24-hour delay between T1 and T2. In addition, there were no significant differences in overall investigation time observed in T1 (P > 0.2), suggesting that there were no detectable nonspecific effects of iclepertin on behavior. Further, there were no overt effects on locomotion or other nonspecific effects of iclepertin observed during T1 and T2.

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Effects of iclepertin on social recognition in rats. The effects of orally administered iclepertin 0.2, 0.6, or 1.8 mg/kg on social investigation times in rats, compared with vehicle. Data are shown as mean ± S.E.M. (vehicle: n = 18; iclepertin 0.2 mg/kg: n = 17; iclepertin 0.6 mg/kg: n = 18; iclepertin 1.8 mg/kg: n = 18). **P < 0.01, ***P < 0.001. PO, orally administered.

Based on these findings as well as the previously reported rat CSF/plasma ratio of 0.02 and IC50 for inhibition of GlyT1 by iclepertin of 5.2 nM (Rosenbrock et al., 2018), the doses used in the social recognition test would be expected to result in plasma levels of iclepertin in the range of 180–1800 nM at T1 and in the range of 250–2500 nM at T2. These estimated values correspond to CSF levels of iclepertin in the range of 0.7- to 7-fold of the GlyT1 IC50 at T1 and in the range of 1- to 10-fold of the GlyT1 IC50 at T2. The increase in exposure at T2 of approximately 40% is due to residual exposure after administration at T1 (Fig. 2).

Discussion

Previous studies have demonstrated that the GlyT1 inhibitor iclepertin has shown high potency against GlyT1 (IC50 = 5.0 nM and 5.2 nM on human and rat GlyT1, respectively) and is highly selective, as demonstrated by a lack of activity toward GlyT2 (IC50 > 10 μM), or 102 other off-targets (IC50 > 10 μM) (Rosenbrock et al., 2018). Central target engagement of iclepertin was demonstrated by a dose-dependent increase of glycine in rat and human CSF (Rosenbrock et al., 2018), and the compound was generally well tolerated in single and multiple rising dose studies in healthy volunteers at doses and exposure levels expected to be clinically relevant (Moschetti et al., 2018a,b).

In these preclinical studies, iclepertin attenuated deficits in EEG parameters induced by the NMDA receptor antagonist MK-801 and improved working and episodic memory performance in rodent cognition tasks. With regard to quantitative EEG parameters, treatment with iclepertin reversed MK-801–induced deficits in N1 amplitude and N1 gating and significantly reversed an MK-801–induced increase in basal gamma power. Furthermore, iclepertin attenuated MK-801–induced deficits in ASSR ITC and ASSR evoked gamma power and also significantly attenuated an MK-801–induced increase in basal gamma power. Overall, these data suggest a normalization of E/I balance and improved signal-to-noise ratio, based principally on the normalization of N1 gating and basal gamma power alongside an increase in evoked gamma power after treatment with iclepertin.

With regard to cognition, iclepertin reversed MK-801–induced deficits in working memory in the spontaneous alternation task in mice and led to significant improvement of episodic memory function in social recognition in rats using a 24-hour forgetting paradigm (time interval between T1 and T2). Thus, this study demonstrates the ability of iclepertin to reverse deficits in clinically relevant EEG biomarkers and working memory in a rodent model of NMDA receptor hypofunction related to schizophrenia (Deserno et al., 2012; Gao et al., 2022). Iclepertin also improved recognition/episodic memory in naïve rats, a memory domain that is also impaired in patients with schizophrenia (Bonner-Jackson et al., 2005; Guo et al., 2019).

The main objective of the EEG part of the study was to test the ability of iclepertin to attenuate cortical network disturbances in a rodent model of NMDA receptor hypofunction related to schizophrenia. Thus, potential effects of iclepertin alone on EEG parameters were not explored. Nonetheless, a previous rodent EEG study using the GlyT1 inhibitor bitopertin showed similar attenuating effects on MK-801–induced EEG disturbances (Voehringer et al., 2019). In that study, effects of the GlyT1 inhibitor were also investigated without MK-801 challenge; no relevant effects on EEG were observed compared with vehicle, suggesting that iclepertin alone is unlikely to have relevant effects on EEG parameters. Disturbances in sensory processing and neural network function, thought to underlie CIAS, are commonly observed in patients with schizophrenia (O’Donnell et al., 2013; Javitt and Freedman, 2015; Shen et al., 2020). N1 (or N100) is an AERP associated with neurons in primary and association AC, and therefore pathology affecting the connectivity and projections of these areas may reduce N1 responses (Gonzalez-Heydrich et al., 2015). Deficits in auditory N1 amplitude are commonly found in patients with schizophrenia (Rosburg et al., 2008; Shen et al., 2020) and have also been observed in unaffected first-degree relatives of patients, suggesting that these deficits may reflect genetic predisposition for schizophrenia (Ahveninen et al., 2006; Force et al., 2008; Foxe et al., 2011). Patients with schizophrenia also exhibit an impaired ability to filter sensory stimuli, as evidenced by deficits in sensory gating such as decreased N1 gating (Rosburg et al., 2008; Shen et al., 2020). Our findings demonstrate similar MK-801–induced deficits in N1 amplitude and N1 gating in mice, supporting the translatability of our findings. These data are also consistent with a previous EEG study in rodents, in which low doses of MK-801 (0.03 mg/kg) increased N1 amplitude whereas higher doses of 0.1 and 0.3 mg/kg reduced N1 amplitude (Schuelert et al., 2018); this is also in line with reported findings in patients with schizophrenia (Rosburg et al., 2008; Mota et al., 2020; Shen et al., 2020).

Patients with schizophrenia also exhibit robust deficits in 40 Hz ASSR (Sivarao, 2015), including decreased ITC (Spencer et al., 2009; Mulert et al., 2011; O’Donnell et al., 2013; Hirano et al., 2015; Thuné et al., 2016) and reduced power (Spencer et al., 2009; O’Donnell et al., 2013; Thuné et al., 2016). These deficits are present from the first psychotic episode (Spencer et al., 2008; Wilson et al., 2008), and ITC deficits have been observed before onset in ultrahigh-risk individuals (Tada et al., 2016). In line with these observations, the current study demonstrated MK-801–induced deficits in ASSR evoked gamma power and ITC in PFC in mice. Further, comparable deficits in auditory evoked gamma power and ITC have been shown in a clinical trial in healthy volunteers treated with subanesthetic doses of the NMDA receptor antagonist ketamine (Curic et al., 2019).

The MK-801–induced increase in basal gamma power reported here has also been previously described in mice (Saunders et al., 2012; Hiyoshi et al., 2014) and is thought to represent electrophysiological “noise” that impairs detection and discrimination of transient stimuli or event-related changes in gamma oscillations (Sivarao, 2015; Schuelert et al., 2018). Patients with schizophrenia display similar abnormal increases in basal gamma power (Grent-’t-Jong et al., 2018) as well as deficits in evoked gamma power (Ferrarelli et al., 2008). It should be noted that in the present study, treatment with MK-801 only had an effect on ASSR evoked gamma power in PFC but not in the double-click paradigm. This is in contrast to previous preclinical studies, which have demonstrated an MK-801–induced reduction in evoked gamma power in addition to increased basal gamma power (Saunders et al., 2012; Sokolenko et al., 2019). This disparity may again be attributable to differences in the doses of MK-801 administered; in the present study mice received a 0.15-mg/kg dose of MK-801, whereas in previous studies the reduction in evoked gamma was observed at higher doses of MK-801 (0.3–1 mg/kg) (Saunders et al., 2012; Sokolenko et al., 2019).

Iclepertin showed a relatively consistent pharmacokinetic (PK) exposure-effect relationship in the current study. In all of the models used, efficacy could be achieved at CSF concentrations in the range of approximately 1- to 6-fold of the previously reported IC50 for inhibition of GlyT1 by iclepertin (Rosenbrock et al., 2018), and efficacy on working memory in the T-maze task was achieved even at lower iclepertin exposure levels. These findings are consistent with the results of phase I and II clinical trials (Moschetti et al., 2018a,b; Rosenbrock et al., 2018). In fact, data from a phase I study on functional target engagement in healthy volunteers demonstrated that after a single oral dose of iclepertin, CSF glycine was increased by 50%–60% at iclepertin CSF concentrations approximately equal to the GlyT1 IC50 (Rosenbrock et al., 2018). Furthermore, in the recent phase II trial in patients with schizophrenia, iclepertin improved cognition after 12 weeks of treatment at 10 mg and 25 mg doses (Fleischhacker et al., 2021), corresponding to mean steady-state postdose plasma concentrations of 246 and 529 nM (Fleischhacker et al., 2021). Based on the previously reported human CSF/plasma ratio of approximately 0.08 (Rosenbrock et al., 2018), patients exhibited improvements in cognitive function at estimated iclepertin CSF concentrations in the range of 3.5- to 8-fold of the GlyT1 IC50. Thus, the PK exposure-effect relationship in the rodent cognition tasks described here and the previously reported evidence of functional target engagement (Rosenbrock et al., 2018) (i.e., glycine increase in CSF) are in agreement with the PK exposure-effect relationship underlying cognitive improvement in patients with schizophrenia.

Overall, this study demonstrates EEG disturbances in animal models of NMDA receptor hypofunction, indicative of neural network dysfunction similar to those observed among patients with schizophrenia. Our data in this study demonstrate that iclepertin can reverse sensory and neural network deficits related to NMDA receptor hypofunction, indicating its potential for further development as a viable treatment option for CIAS. GlyT1 inhibitors have previously been shown to reverse MK-801–induced deficits in EEG parameters related to lower-frequency oscillations (Depoortère et al., 2005). To our knowledge the present study is the first to report positive effects of a GlyT1 inhibitor on AERPs in an animal model related to schizophrenia, evaluating basal and evoked gamma oscillations and ASSR.

An advantage of exploring the effects of treatment on EEG biomarkers in animal models is that for many relevant biomarkers, homologous or analogous processes can be studied in humans, meaning that EEG data are readily translatable (Javitt et al., 2008). Indeed, our findings are largely consistent with known deficits in patients with schizophrenia, supporting the clinical translatability of these EEG parameters as neurophysiological biomarkers of CIAS (Javitt et al., 2008; Light and Swerdlow, 2015; Kim et al., 2020; Light et al., 2020).

One potential limitation of this study is that the animal models used in this study do not capture all aspects of schizophrenia but instead reflect certain key features of the behavioral phenotype and pathophysiology associated with schizophrenia. Nonetheless, previous studies have shown that MK-801 administration in rodents induces a range of schizophrenia-like behavioral phenotypes, including hyper-locomotor activity, reduced sensorimotor gating, deficits in gamma oscillations and ASSR, decreased social interaction, and cognitive impairments including learning and memory deficits, supporting the validity of this model for drug development in schizophrenia (Bubeníková-Valesová et al., 2008; Svoboda et al., 2015; Schuelert et al., 2018; Lee and Zhou, 2019; Mabunga et al., 2019). It should be noted, however, that the current study assessed gamma oscillations at a higher frequency (basal: 50–100 Hz; evoked: 35–80 Hz) since the effects of MK-801 in rodents are most prominent in this frequency range (Ahnaou et al., 2017; Schuelert et al., 2018) compared with the lower frequencies (< 50 Hz) that are typically analyzed to explore deficits in patients with schizophrenia (Ferrarelli et al., 2008; Grent-’t-Jong et al., 2018).

In conclusion, this study provides additional evidence for the role of NMDA receptor hypofunction in neural network dysfunction and sensory processing deficits relevant to CIAS. Our data also demonstrate that the GlyT1 inhibitor iclepertin can reverse deficits in clinically relevant EEG parameters relating to sensory processing and neural network activity. In addition, the reported data demonstrate that iclepertin has procognitive effects in learning and memory tasks in rodents. The preclinical efficacy demonstrated in this study provides solid support for the development of iclepertin, which is currently being tested in phase III trials [Connex-1 (NCT04846868), Connex-2 (NCT04846881), and Connex-3 (NCT04860830)] as a potential treatment of CIAS.

Acknowledgments

The authors would like to thank Dr. E. Andriambeloson from Neurofit S.A.S. for his support regarding the T-maze study as well as A. Blasius, H. Assfalg, S. Weigele, and S. Schrade for their technical assistance. The sponsor was given the opportunity to review the manuscript for medical and scientific accuracy as well as intellectual property considerations. Editorial support in the form of initial preparation of the outline based on input from all authors, collation and incorporation of author feedback to develop subsequent drafts, assembling tables and figures, copyediting, and referencing was provided by Rebecca Dawson and Mark Condon, DPhil, of Fishawack Communications Ltd, part of Fishawack Health, and was funded by Boehringer Ingelheim International GmbH.

Authorship Contributions

Participated in research design: Rosenbrock, Dorner-Ciossek, Giovannini, Schmid, Schuelert.

Conducted experiments: Schuelert.

Contributed new reagents or analytic tools: Rosenbrock, Giovannini, Schmid, Schuelert.

Performed data analysis: Rosenbrock, Giovannini, Schmid, Schuelert.

Wrote or contributed to the writing of the manuscript: Rosenbrock, Dorner-Ciossek, Giovannini, Schmid, Schuelert.

Footnotes

    • Received December 22, 2021.
    • Accepted May 4, 2022.
  • This work was funded by Boehringer Ingelheim GmbH. The sponsor was given the opportunity to review the manuscript for medical and scientific accuracy as well as intellectual property considerations.

  • All authors are full-time employees of Boehringer Ingelheim.

  • Primary laboratory of origin: Boehringer Ingelheim International GmbH (Biberach an der Riss, Germany).

  • Part of this work has been previously published/presented in abstract form: Rosenbrock H, Giovannini R, Schmid B, Kramer G, Arban R, Dorner-Ciossek C (2016) Pharmacological characterization of the novel glycine transporter-1 inhibitor BI 425809 on target engagement in-vivo and in animal models related to cognitive symptoms of schizophrenia. Schizophrenia International Research Society Biennial Meeting; 2016 Apr 2–6; Florence, Italy. Schizophrenia International Research Society, Brentwood, TN.

  • https://doi.org/10.1124/jpet.121.001071.

  • ↵Embedded ImageThis article has supplemental material available at jpet.aspetjournals.org.

Abbreviations

AC
auditory cortex
AERP
auditory event-related potential
AP
anterior posterior
ASSR
auditory steady-state response
CIAS
cognitive impairment associated with schizophrenia
CSF
cerebrospinal fluid
EEG
electroencephalography
E/I
excitatory/inhibitory
GlyT1
glycine transporter-1
ITC
intertrial coherence
NMDA
N-methyl-D-aspartate
PFC
prefrontal cortex
PK
pharmacokinetic
T1
trial 1
T2
trial 2
  • Copyright © 2022 by The Author(s)

This is an open access article distributed under the CC BY Attribution 4.0 International license.

References

  1. ↵
    1. Aguilar DD,
    2. Radzik LK,
    3. Schiffino FL,
    4. Folorunso OO,
    5. Zielinski MR,
    6. Coyle JT,
    7. Balu DT, and
    8. McNally JM
    (2021) Altered neural oscillations and behavior in a genetic mouse model of NMDA receptor hypofunction. Sci Rep 11:9031.
    OpenUrl
  2. ↵
    1. Ahnaou A,
    2. Broadbelt T,
    3. Biermans R,
    4. Huysmans H,
    5. Manyakov NV, and
    6. Drinkenburg WHIM
    (2020) The phosphodiesterase-4 and glycine transporter-1 inhibitors enhance in vivo hippocampal theta network connectivity and synaptic plasticity, whereas D-serine does not. Transl Psychiatry 10:197.
    OpenUrl
  3. ↵
    1. Ahnaou A,
    2. Huysmans H,
    3. Van de Casteele T, and
    4. Drinkenburg WHIM
    (2017) Cortical high gamma network oscillations and connectivity: a translational index for antipsychotics to normalize aberrant neurophysiological activity. Transl Psychiatry 7:1285.
    OpenUrl
  4. ↵
    1. Ahveninen J,
    2. Jääskeläinen IP,
    3. Osipova D,
    4. Huttunen MO,
    5. Ilmoniemi RJ,
    6. Kaprio J,
    7. Lönnqvist J,
    8. Manninen M,
    9. Pakarinen S,
    10. Therman S, et al.
    (2006) Inherited auditory-cortical dysfunction in twin pairs discordant for schizophrenia. Biol Psychiatry 60:612–620.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Alberati D,
    2. Moreau JL,
    3. Lengyel J,
    4. Hauser N,
    5. Mory R,
    6. Borroni E,
    7. Pinard E,
    8. Knoflach F,
    9. Schlotterbeck G,
    10. Hainzl D, et al.
    (2012) Glycine reuptake inhibitor RG1678: a pharmacologic characterization of an investigational agent for the treatment of schizophrenia. Neuropharmacology 62:1152–1161.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Beneyto M and
    2. Meador-Woodruff JH
    (2008) Lamina-specific abnormalities of NMDA receptor-associated postsynaptic protein transcripts in the prefrontal cortex in schizophrenia and bipolar disorder. Neuropsychopharmacology 33:2175–2186.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Bickel S and
    2. Javitt DC
    (2009) Neurophysiological and neurochemical animal models of schizophrenia: focus on glutamate. Behav Brain Res 204:352–362.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Bonner-Jackson A,
    2. Haut K,
    3. Csernansky JG, and
    4. Barch DM
    (2005) The influence of encoding strategy on episodic memory and cortical activity in schizophrenia. Biol Psychiatry 58:47–55.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Bubeníková-Valesová V,
    2. Horácek J,
    3. Vrajová M, and
    4. Höschl C
    (2008) Models of schizophrenia in humans and animals based on inhibition of NMDA receptors. Neurosci Biobehav Rev 32:1014–1023.
    OpenUrlCrossRefPubMed
  10. ↵
    1. Cho RY,
    2. Konecky RO, and
    3. Carter CS
    (2006) Impairments in frontal cortical gamma synchrony and cognitive control in schizophrenia. Proc Natl Acad Sci USA 103:19878–19883.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Cioffi CL and
    2. Guzzo PR
    (2016) Inhibitors of glycine transporter-1: potential therapeutics for the treatment of CNS disorders. Curr Top Med Chem 16:3404–3437.
    OpenUrl
  12. ↵
    1. Cohen SM,
    2. Tsien RW,
    3. Goff DC, and
    4. Halassa MM
    (2015) The impact of NMDA receptor hypofunction on GABAergic neurons in the pathophysiology of schizophrenia. Schizophr Res 167:98–107.
    OpenUrlCrossRefPubMed
  13. ↵
    1. Collingridge GL,
    2. Volianskis A,
    3. Bannister N,
    4. France G,
    5. Hanna L,
    6. Mercier M,
    7. Tidball P,
    8. Fang G,
    9. Irvine MW,
    10. Costa BM, et al.
    (2013) The NMDA receptor as a target for cognitive enhancement. Neuropharmacology 64:13–26.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Curic S,
    2. Leicht G,
    3. Thiebes S,
    4. Andreou C,
    5. Polomac N,
    6. Eichler IC,
    7. Eichler L,
    8. Zöllner C,
    9. Gallinat J,
    10. Steinmann S, et al.
    (2019) Reduced auditory evoked gamma-band response and schizophrenia-like clinical symptoms under subanesthetic ketamine. Neuropsychopharmacology 44:1239–1246.
    OpenUrlCrossRef
  15. ↵
    1. Dauvermann MR,
    2. Lee G, and
    3. Dawson N
    (2017) Glutamatergic regulation of cognition and functional brain connectivity: insights from pharmacological, genetic and translational schizophrenia research. Br J Pharmacol 174:3136–3160.
    OpenUrl
  16. ↵
    1. Depoortère R,
    2. Dargazanli G,
    3. Estenne-Bouhtou G,
    4. Coste A,
    5. Lanneau C,
    6. Desvignes C,
    7. Poncelet M,
    8. Heaulme M,
    9. Santucci V,
    10. Decobert M, et al.
    (2005) Neurochemical, electrophysiological and pharmacological profiles of the selective inhibitor of the glycine transporter-1 SSR504734, a potential new type of antipsychotic. Neuropsychopharmacology 30:1963–1985.
    OpenUrlCrossRefPubMed
  17. ↵
    1. Deserno L,
    2. Sterzer P,
    3. Wüstenberg T,
    4. Heinz A, and
    5. Schlagenhauf F
    (2012) Reduced prefrontal-parietal effective connectivity and working memory deficits in schizophrenia. J Neurosci 32:12–20.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Du X,
    2. Choa FS,
    3. Summerfelt A,
    4. Rowland LM,
    5. Chiappelli J,
    6. Kochunov P, and
    7. Hong LE
    (2017) N100 as a generic cortical electrophysiological marker based on decomposition of TMS-evoked potentials across five anatomic locations. Exp Brain Res 235:69–81.
    OpenUrlCrossRef
  19. ↵
    1. Ferrarelli F,
    2. Massimini M,
    3. Peterson MJ,
    4. Riedner BA,
    5. Lazar M,
    6. Murphy MJ,
    7. Huber R,
    8. Rosanova M,
    9. Alexander AL,
    10. Kalin N, et al.
    (2008) Reduced evoked gamma oscillations in the frontal cortex in schizophrenia patients: a TMS/EEG study. Am J Psychiatry 165:996–1005.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Fleischhacker WW,
    2. Podhorna J,
    3. Gröschl M,
    4. Hake S,
    5. Zhao Y,
    6. Huang S,
    7. Keefe RSE,
    8. Desch M,
    9. Brenner R,
    10. Walling DP, et al.
    (2021) Efficacy and safety of the novel glycine transporter inhibitor BI 425809 once daily in patients with schizophrenia: a double-blind, randomised, placebo-controlled phase 2 study. Lancet Psychiatry 8:191–201.
    OpenUrl
  21. ↵
    1. Force RB,
    2. Venables NC, and
    3. Sponheim SR
    (2008) An auditory processing abnormality specific to liability for schizophrenia. Schizophr Res 103:298–310.
    OpenUrlCrossRefPubMed
  22. ↵
    1. Forsyth JK and
    2. Lewis DA
    (2017) Mapping the consequences of impaired synaptic plasticity in schizophrenia through development: an integrative model for diverse clinical features. Trends Cogn Sci 21:760–778.
    OpenUrl
  23. ↵
    1. Foxe JJ,
    2. Yeap S,
    3. Snyder AC,
    4. Kelly SP,
    5. Thakore JH, and
    6. Molholm S
    (2011) The N1 auditory evoked potential component as an endophenotype for schizophrenia: high-density electrical mapping in clinically unaffected first-degree relatives, first-episode, and chronic schizophrenia patients. Eur Arch Psychiatry Clin Neurosci 261:331–339.
    OpenUrlCrossRefPubMed
  24. ↵
    1. Fromer M,
    2. Pocklington AJ,
    3. Kavanagh DH,
    4. Williams HJ,
    5. Dwyer S,
    6. Gormley P,
    7. Georgieva L,
    8. Rees E,
    9. Palta P,
    10. Ruderfer DM, et al.
    (2014) De novo mutations in schizophrenia implicate synaptic networks. Nature 506:179–184.
    OpenUrlCrossRefPubMed
  25. ↵
    1. Gao WJ,
    2. Yang SS,
    3. Mack NR, and
    4. Chamberlin LA
    (2022) Aberrant maturation and connectivity of prefrontal cortex in schizophrenia-contribution of NMDA receptor development and hypofunction. Mol Psychiatry 27:731–743.
    OpenUrl
  26. ↵
    1. Gonzalez-Burgos G and
    2. Lewis DA
    (2012) NMDA receptor hypofunction, parvalbumin-positive neurons, and cortical gamma oscillations in schizophrenia. Schizophr Bull 38:950–957.
    OpenUrlCrossRefPubMed
  27. ↵
    1. Gonzalez-Heydrich J,
    2. Bosquet Enlow M,
    3. D’Angelo E,
    4. Seidman LJ,
    5. Gumlak S,
    6. Kim A,
    7. Woodberry KA,
    8. Rober A,
    9. Tembulkar S,
    10. Graber K, et al.
    (2015) Early auditory processing evoked potentials (N100) show a continuum of blunting from clinical high risk to psychosis in a pediatric sample. Schizophr Res 169:340–345.
    OpenUrl
  28. ↵
    1. Grent-’t-Jong T,
    2. Gross J,
    3. Goense J,
    4. Wibral M,
    5. Gajwani R,
    6. Gumley AI,
    7. Lawrie SM,
    8. Schwannauer M,
    9. Schultze-Lutter F,
    10. Navarro Schröder T, et al.
    (2018) Resting-state gamma-band power alterations in schizophrenia reveal E/I-balance abnormalities across illness-stages. eLife 7:e37799.
    OpenUrl
  29. ↵
    1. Guo JY,
    2. Ragland JD, and
    3. Carter CS
    (2019) Memory and cognition in schizophrenia. Mol Psychiatry 24:633–642.
    OpenUrlCrossRefPubMed
  30. ↵
    1. Hakami T,
    2. Jones NC,
    3. Tolmacheva EA,
    4. Gaudias J,
    5. Chaumont J,
    6. Salzberg M,
    7. O’Brien TJ, and
    8. Pinault D
    (2009) NMDA receptor hypofunction leads to generalized and persistent aberrant gamma oscillations independent of hyperlocomotion and the state of consciousness. PLoS One 4:e6755.
    OpenUrlCrossRefPubMed
  31. ↵
    1. Harvey RJ and
    2. Yee BK
    (2013) Glycine transporters as novel therapeutic targets in schizophrenia, alcohol dependence and pain. Nat Rev Drug Discov 12:866–885.
    OpenUrlCrossRefPubMed
  32. ↵
    1. Hirano Y,
    2. Oribe N,
    3. Kanba S,
    4. Onitsuka T,
    5. Nestor PG, and
    6. Spencer KM
    (2015) Spontaneous gamma activity in schizophrenia. JAMA Psychiatry 72:813–821.
    OpenUrl
  33. ↵
    1. Hiyoshi T,
    2. Kambe D,
    3. Karasawa J, and
    4. Chaki S
    (2014) Involvement of glutamatergic and GABAergic transmission in MK-801-increased gamma band oscillation power in rat cortical electroencephalograms. Neuroscience 280:262–274.
    OpenUrl
  34. ↵
    1. Hong LE,
    2. Summerfelt A,
    3. Buchanan RW,
    4. O’Donnell P,
    5. Thaker GK,
    6. Weiler MA, and
    7. Lahti AC
    (2010) Gamma and delta neural oscillations and association with clinical symptoms under subanesthetic ketamine. Neuropsychopharmacology 35:632–640.
    OpenUrlCrossRefPubMed
  35. ↵
    1. Javitt DC and
    2. Freedman R
    (2015) Sensory processing dysfunction in the personal experience and neuronal machinery of schizophrenia. Am J Psychiatry 172:17–31.
    OpenUrlCrossRefPubMed
  36. ↵
    1. Javitt DC,
    2. Spencer KM,
    3. Thaker GK,
    4. Winterer G, and
    5. Hajós M
    (2008) Neurophysiological biomarkers for drug development in schizophrenia. Nat Rev Drug Discov 7:68–83.
    OpenUrlCrossRefPubMed
  37. ↵
    1. Joos K,
    2. Gilles A,
    3. Van de Heyning P,
    4. De Ridder D, and
    5. Vanneste S
    (2014) From sensation to percept: the neural signature of auditory event-related potentials. Neurosci Biobehav Rev 42:148–156.
    OpenUrlCrossRefPubMed
  38. ↵
    1. Kim HK,
    2. Blumberger DM, and
    3. Daskalakis ZJ
    (2020) Neurophysiological biomarkers in schizophrenia-P50, mismatch negativity, and TMS-EMG and TMS-EEG. Front Psychiatry 11:795.
    OpenUrl
  39. ↵
    1. Kirov G,
    2. Pocklington AJ,
    3. Holmans P,
    4. Ivanov D,
    5. Ikeda M,
    6. Ruderfer D,
    7. Moran J,
    8. Chambert K,
    9. Toncheva D,
    10. Georgieva L, et al.
    (2012) De novo CNV analysis implicates specific abnormalities of postsynaptic signalling complexes in the pathogenesis of schizophrenia. Mol Psychiatry 17:142–153.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Kondrakiewicz K,
    2. Kostecki M,
    3. Szadzińska W, and
    4. Knapska E
    (2019) Ecological validity of social interaction tests in rats and mice. Genes Brain Behav 18:e12525.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Krystal JH,
    2. Karper LP,
    3. Seibyl JP,
    4. Freeman GK,
    5. Delaney R,
    6. Bremner JD,
    7. Heninger GR,
    8. Bowers Jr MB, and
    9. Charney DS
    (1994) Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry 51:199–214.
    OpenUrlCrossRefPubMed
  42. ↵
    1. Lane HY,
    2. Lin CH,
    3. Huang YJ,
    4. Liao CH,
    5. Chang YC, and
    6. Tsai GE
    (2010) A randomized, double-blind, placebo-controlled comparison study of sarcosine (N-methylglycine) and D-serine add-on treatment for schizophrenia. Int J Neuropsychopharmacol 13:451–460.
    OpenUrlCrossRefPubMed
  43. ↵
    1. Lee G and
    2. Zhou Y
    (2019) NMDAR hypofunction animal models of schizophrenia. Front Mol Neurosci 12:185.
    OpenUrl
  44. ↵
    1. Light GA,
    2. Joshi YB,
    3. Molina JL,
    4. Bhakta SG,
    5. Nungaray JA,
    6. Cardoso L,
    7. Kotz JE,
    8. Thomas ML, and
    9. Swerdlow NR
    (2020) Neurophysiological biomarkers for schizophrenia therapeutics. Biomarkers in Neuropsychiatry 2:100012 DOI: 10.1016/j.bionps.2020.100012.
    OpenUrl
  45. ↵
    1. Light GA and
    2. Swerdlow NR
    (2015) Future clinical uses of neurophysiological biomarkers to predict and monitor treatment response for schizophrenia. Ann N Y Acad Sci 1344:105–119.
    OpenUrlCrossRefPubMed
  46. ↵
    1. Lisman J
    (2012) Excitation, inhibition, local oscillations, or large-scale loops: what causes the symptoms of schizophrenia? Curr Opin Neurobiol 22:537–544.
    OpenUrlCrossRefPubMed
  47. ↵
    1. Loiodice S,
    2. Drinkenburg WH,
    3. Ahnaou A,
    4. McCarthy A,
    5. Viardot G,
    6. Cayre E,
    7. Rion B,
    8. Bertaina-Anglade V,
    9. Mano M,
    10. L’Hostis P, et al.
    (2021) Mismatch negativity as EEG biomarker supporting CNS drug development: a transnosographic and translational study. Transl Psychiatry 11:253.
    OpenUrl
  48. ↵
    1. Mabunga DFN,
    2. Park D,
    3. Ryu O,
    4. Valencia ST,
    5. Adil KJL,
    6. Kim S,
    7. Kwon KJ,
    8. Shin CY, and
    9. Jeon SJ
    (2019) Recapitulation of neuropsychiatric behavioral features in mice using acute low-dose MK-801 administration. Exp Neurobiol 28:697–708.
    OpenUrl
  49. ↵
    1. Marques BL,
    2. Oliveira-Lima OC,
    3. Carvalho GA,
    4. de Almeida Chiarelli R,
    5. Ribeiro RI,
    6. Parreira RC,
    7. da Madeira Freitas EM,
    8. Resende RR,
    9. Klempin F,
    10. Ulrich H, et al.
    (2020) Neurobiology of glycine transporters: from molecules to behavior. Neurosci Biobehav Rev 118:97–110.
    OpenUrl
  50. ↵
    1. Moghaddam B and
    2. Krystal JH
    (2012) Capturing the angel in “angel dust”: twenty years of translational neuroscience studies of NMDA receptor antagonists in animals and humans. Schizophr Bull 38:942–949.
    OpenUrlCrossRefPubMed
  51. ↵
    1. Molina JL,
    2. Thomas ML,
    3. Joshi YB,
    4. Hochberger WC,
    5. Koshiyama D,
    6. Nungaray JA,
    7. Cardoso L,
    8. Sprock J,
    9. Braff DL,
    10. Swerdlow NR, et al.
    (2020) Gamma oscillations predict pro-cognitive and clinical response to auditory-based cognitive training in schizophrenia. Transl Psychiatry 10:405.
    OpenUrl
  52. ↵
    1. Moschetti V,
    2. Desch M,
    3. Goetz S,
    4. Liesenfeld KH,
    5. Rosenbrock H,
    6. Kammerer KP,
    7. Wunderlich G, and
    8. Wind S
    (2018a) Safety, tolerability and pharmacokinetics of oral BI 425809, a glycine transporter 1 inhibitor, in healthy male volunteers: a partially randomised, single-blind, placebo-controlled, first-in-human study. Eur J Drug Metab Pharmacokinet 43:239–249.
    OpenUrl
    1. Moschetti V,
    2. Schlecker C,
    3. Wind S,
    4. Goetz S,
    5. Schmitt H,
    6. Schultz A,
    7. Liesenfeld KH,
    8. Wunderlich G, and
    9. Desch M
    (2018b) Multiple rising doses of oral BI 425809, a GlyT1 inhibitor, in young and elderly healthy volunteers: a randomised, double-blind, phase I study investigating safety and pharmacokinetics. Clin Drug Investig 38:737–750.
    OpenUrl
  53. ↵
    1. Mota A,
    2. Tomé D,
    3. Campos C, and
    4. Rocha N
    (2020) The auditory N1 in schizophrenia: comparative analysis with a monoaural stimuli paradigm. Psychiatr Danub 32:210–213.
    OpenUrl
  54. ↵
    1. Mulert C,
    2. Kirsch V,
    3. Pascual-Marqui R,
    4. McCarley RW, and
    5. Spencer KM
    (2011) Long-range synchrony of γ oscillations and auditory hallucination symptoms in schizophrenia. Int J Psychophysiol 79:55–63.
    OpenUrlCrossRefPubMed
  55. ↵
    1. O’Donnell BF,
    2. Vohs JL,
    3. Krishnan GP,
    4. Rass O,
    5. Hetrick WP, and
    6. Morzorati SL
    (2013) The auditory steady-state response (ASSR): a translational biomarker for schizophrenia. Suppl Clin Neurophysiol 62:101–112.
    OpenUrlCrossRefPubMed
  56. ↵
    1. Pei JC,
    2. Luo DZ,
    3. Gau SS,
    4. Chang CY, and
    5. Lai WS
    (2021) Directly and indirectly targeting the glycine modulatory site to modulate NMDA receptor function to address unmet medical needs of patients with schizophrenia. Front Psychiatry 12:742058.
    OpenUrl
  57. ↵
    1. Pinard E,
    2. Borroni E,
    3. Koerner A,
    4. Umbricht D, and
    5. Alberati D
    (2018) Glycine transporter type I (GlyT1) inhibitor, bitopertin: a journey from lab to patient. Chimia (Aarau) 72:477–484.
    OpenUrl
  58. ↵
    1. Pinault D
    (2008) N-methyl d-aspartate receptor antagonists ketamine and MK-801 induce wake-related aberrant gamma oscillations in the rat neocortex. Biol Psychiatry 63:730–735.
    OpenUrlCrossRefPubMed
  59. ↵
    1. Rosburg T,
    2. Boutros NN, and
    3. Ford JM
    (2008) Reduced auditory evoked potential component N100 in schizophrenia--a critical review. Psychiatry Res 161:259–274.
    OpenUrlCrossRefPubMed
  60. ↵
    1. Rosen AM,
    2. Spellman T, and
    3. Gordon JA
    (2015) Electrophysiological endophenotypes in rodent models of schizophrenia and psychosis. Biol Psychiatry 77:1041–1049.
    OpenUrlCrossRef
  61. ↵
    1. Rosenbrock H,
    2. Desch M,
    3. Kleiner O,
    4. Dorner-Ciossek C,
    5. Schmid B,
    6. Keller S,
    7. Schlecker C,
    8. Moschetti V,
    9. Goetz S,
    10. Liesenfeld KH, et al.
    (2018) Evaluation of pharmacokinetics and pharmacodynamics of BI 425809, a novel GlyT1 inhibitor: translational studies. Clin Transl Sci 11:616–623.
    OpenUrl
  62. ↵
    1. Rosenbrock H,
    2. Giovannini R,
    3. Schänzle G,
    4. Koros E,
    5. Runge F,
    6. Fuchs H,
    7. Marti A,
    8. Reymann KG,
    9. Schröder UH,
    10. Fedele E, et al.
    (2019) The novel phosphodiesterase 9A inhibitor BI 409306 increases cyclic guanosine monophosphate levels in the brain, promotes synaptic plasticity, and enhances memory function in rodents. J Pharmacol Exp Ther 371:633–641.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    1. Saunders JA,
    2. Gandal MJ, and
    3. Siegel SJ
    (2012) NMDA antagonists recreate signal-to-noise ratio and timing perturbations present in schizophrenia. Neurobiol Dis 46:93–100.
    OpenUrlCrossRefPubMed
  64. ↵
    Schizophrenia Working Group of the Psychiatric Genomics Consortium (2014) Biological insights from 108 schizophrenia-associated genetic loci. Nature 511:421–427.
    OpenUrlCrossRefPubMed
  65. ↵
    1. Schoonover KE,
    2. Dienel SJ, and
    3. Lewis DA
    (2020) Prefrontal cortical alterations of glutamate and GABA neurotransmission in schizophrenia: insights for rational biomarker development. Biomark Neuropsychiatry 3:100015.
    OpenUrlPubMed
  66. ↵
    1. Schuelert N,
    2. Dorner-Ciossek C,
    3. Brendel M, and
    4. Rosenbrock H
    (2018) A comprehensive analysis of auditory event-related potentials and network oscillations in an NMDA receptor antagonist mouse model using a novel wireless recording technology. Physiol Rep 6:e13782.
    OpenUrl
  67. ↵
    1. Shen CL,
    2. Chou TL,
    3. Lai WS,
    4. Hsieh MH,
    5. Liu CC,
    6. Liu CM, and
    7. Hwu HG
    (2020) P50, N100, and P200 auditory sensory gating deficits in schizophrenia patients. Front Psychiatry 11:868.
    OpenUrl
  68. ↵
    1. Sinkeviciute I,
    2. Begemann M,
    3. Prikken M,
    4. Oranje B,
    5. Johnsen E,
    6. Lei WU,
    7. Hugdahl K,
    8. Kroken RA,
    9. Rau C,
    10. Jacobs JD, et al.
    (2018) Efficacy of different types of cognitive enhancers for patients with schizophrenia: a meta-analysis. NPJ Schizophr 4:22.
    OpenUrl
  69. ↵
    1. Sivarao DV
    (2015) The 40-Hz auditory steady-state response: a selective biomarker for cortical NMDA function. Ann N Y Acad Sci 1344:27–36.
    OpenUrl
  70. ↵
    1. Sokolenko E,
    2. Hudson MR,
    3. Nithianantharajah J, and
    4. Jones NC
    (2019) The mGluR2/3 agonist LY379268 reverses NMDA receptor antagonist effects on cortical gamma oscillations and phase coherence, but not working memory impairments, in mice. J Psychopharmacol 33:1588–1599.
    OpenUrl
  71. ↵
    1. Spencer KM,
    2. Niznikiewicz MA,
    3. Nestor PG,
    4. Shenton ME, and
    5. McCarley RW
    (2009) Left auditory cortex gamma synchronization and auditory hallucination symptoms in schizophrenia. BMC Neurosci 10:85.
    OpenUrlCrossRefPubMed
  72. ↵
    1. Spencer KM,
    2. Salisbury DF,
    3. Shenton ME, and
    4. McCarley RW
    (2008) Gamma-band auditory steady-state responses are impaired in first episode psychosis. Biol Psychiatry 64:369–375.
    OpenUrlCrossRefPubMed
  73. ↵
    1. Stefanescu RA and
    2. Shore SE
    (2015) NMDA receptors mediate stimulus-timing-dependent plasticity and neural synchrony in the dorsal cochlear nucleus. Front Neural Circuits 9:75.
    OpenUrlCrossRefPubMed
  74. ↵
    1. Svoboda J,
    2. Stankova A,
    3. Entlerova M, and
    4. Stuchlik A
    (2015) Acute administration of MK-801 in an animal model of psychosis in rats interferes with cognitively demanding forms of behavioral flexibility on a rotating arena. Front Behav Neurosci 9:75.
    OpenUrl
  75. ↵
    1. Tada M,
    2. Nagai T,
    3. Kirihara K,
    4. Koike S,
    5. Suga M,
    6. Araki T,
    7. Kobayashi T, and
    8. Kasai K
    (2016) Differential alterations of auditory gamma oscillatory responses between pre-onset high-risk individuals and first-episode schizophrenia. Cereb Cortex 26:1027–1035.
    OpenUrlCrossRefPubMed
  76. ↵
    1. Tanaka-Koshiyama K,
    2. Koshiyama D,
    3. Miyakoshi M,
    4. Joshi YB,
    5. Molina JL,
    6. Sprock J,
    7. Braff DL, and
    8. Light GA
    (2020) Abnormal spontaneous gamma power is associated with verbal learning and memory dysfunction in schizophrenia. Front Psychiatry 11:832.
    OpenUrl
  77. ↵
    1. Thuné H,
    2. Recasens M, and
    3. Uhlhaas PJ
    (2016) The 40-Hz auditory steady-state response in patients with schizophrenia: a meta-analysis. JAMA Psychiatry 73:1145–1153.
    OpenUrl
  78. ↵
    1. Tsai G,
    2. Lane HY,
    3. Yang P,
    4. Chong MY, and
    5. Lange N
    (2004) Glycine transporter I inhibitor, N-methylglycine (sarcosine), added to antipsychotics for the treatment of schizophrenia. Biol Psychiatry 55:452–456.
    OpenUrlCrossRefPubMed
  79. ↵
    1. Uhlhaas PJ and
    2. Singer W
    (2010) Abnormal neural oscillations and synchrony in schizophrenia. Nat Rev Neurosci 11:100–113.
    OpenUrlCrossRefPubMed
  80. ↵
    1. Uno Y and
    2. Coyle JT
    (2019) Glutamate hypothesis in schizophrenia. Psychiatry Clin Neurosci 73:204–215.
    OpenUrl
  81. ↵
    1. van der Staay FJ,
    2. Rutten K,
    3. Bärfacker L,
    4. Devry J,
    5. Erb C,
    6. Heckroth H,
    7. Karthaus D,
    8. Tersteegen A,
    9. van Kampen M,
    10. Blokland A, et al.
    (2008) The novel selective PDE9 inhibitor BAY 73-6691 improves learning and memory in rodents. Neuropharmacology 55:908–918.
    OpenUrlCrossRefPubMed
  82. ↵
    1. Voehringer P,
    2. Schuelert N,
    3. Plano A,
    4. Dorner-Ciossek C, and
    5. Rosenbrock H
    (2019) Effects of the glycine transporter-1 inhibitor bitopertin on rodent electroencephalography and auditory event-related potential biomarkers related to schizophrenia. Eur Neuropsychopharmacol 29 (Suppl 6):S390 DOI: 10.1016/j.euroneuro.2019.09.558.
    OpenUrl
  83. ↵
    1. Vrajová M,
    2. Stastný F,
    3. Horácek J,
    4. Lochman J,
    5. Serý O,
    6. Peková S,
    7. Klaschka J, and
    8. Höschl C
    (2010) Expression of the hippocampal NMDA receptor GluN1 subunit and its splicing isoforms in schizophrenia: postmortem study. Neurochem Res 35:994–1002.
    OpenUrlCrossRefPubMed
  84. ↵
    1. Wilson TW,
    2. Hernandez OO,
    3. Asherin RM,
    4. Teale PD,
    5. Reite ML, and
    6. Rojas DC
    (2008) Cortical gamma generators suggest abnormal auditory circuitry in early-onset psychosis. Cereb Cortex 18:371–378.
    OpenUrlCrossRefPubMed
  85. ↵
    1. Wu K,
    2. Castellano D,
    3. Tian Q, and
    4. Lu W
    (2021) Distinct regulation of tonic GABAergic inhibition by NMDA receptor subtypes. Cell Rep 37:109960.
    OpenUrl
  86. ↵
    1. Zafra F and
    2. Giménez C
    (2008) Glycine transporters and synaptic function. IUBMB Life 60:810–817.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Journal of Pharmacology and Experimental Therapeutics: 382 (2)
Journal of Pharmacology and Experimental Therapeutics
Vol. 382, Issue 2
1 Aug 2022
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Journal of Pharmacology and Experimental Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Effects of the Glycine Transporter-1 Inhibitor Iclepertin (BI 425809) on Sensory Processing, Neural Network Function, and Cognition in Animal Models Related to Schizophrenia
(Your Name) has forwarded a page to you from Journal of Pharmacology and Experimental Therapeutics
(Your Name) thought you would be interested in this article in Journal of Pharmacology and Experimental Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Research ArticleNeuropharmacology

Iclepertin (BI 425809) in Schizophrenia-Related Models

Holger Rosenbrock, Cornelia Dorner-Ciossek, Riccardo Giovannini, Bernhard Schmid and Niklas Schuelert
Journal of Pharmacology and Experimental Therapeutics August 1, 2022, 382 (2) 223-232; DOI: https://doi.org/10.1124/jpet.121.001071

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Research ArticleNeuropharmacology

Iclepertin (BI 425809) in Schizophrenia-Related Models

Holger Rosenbrock, Cornelia Dorner-Ciossek, Riccardo Giovannini, Bernhard Schmid and Niklas Schuelert
Journal of Pharmacology and Experimental Therapeutics August 1, 2022, 382 (2) 223-232; DOI: https://doi.org/10.1124/jpet.121.001071
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Visual Overview
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Authorship Contributions
    • Footnotes
    • Abbreviations
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF + SI
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Glycine receptor modulation using monoclonal antibodies
  • D1 Agonist Versus Methylphenidate on PFC Working Memory
Show more Neuropharmacology

Similar Articles

Advertisement
  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About JPET
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Drug Metabolism and Disposition
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-0103 (Online)

Copyright © 2022 by the American Society for Pharmacology and Experimental Therapeutics