Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 1.
Published in final edited form as: Discov Med. 2012 Nov;14(78):335–343.

Emerging Approaches for Treatment of Schizophrenia: Modulation of Glutamatergic Signaling

Meredith J Noetzel 1, Carrie K Jones 1, P Jeffrey Conn 1
PMCID: PMC3787874  NIHMSID: NIHMS513987  PMID: 23200065

Abstract

Treatment options for schizophrenia that address all symptom categories (positive, negative, and cognitive) are lacking. Novel compounds that regulate signaling by the major excitatory neurotransmitter in the brain, glutamate, are emerging as a novel approach for the treatment of this disorder. Currently available medications ameliorate positive symptoms but do not have efficacy in reducing negative symptoms or cognitive disturbances. It is possible that agents that target glutamatergic signaling in the CNS could have efficacy in reducing all major symptom clusters, providing a more comprehensive treatment strategy, and also avoiding some of the adverse effects that are seen with currently available treatments. Three major approaches for targeting glutamate signaling are now advancing in preclinical and clinical development. First are inhibitors for a transporter for glycine termed GlyT1. Glycine is a co-agonist with glutamate for a specific subtype of glutamate receptor, termed the NMDA receptor, which is thought to be critically involved in brain circuits that are disrupted in schizophrenia patients. Inhibiting GlyT1 increases glycine levels and can selectively increase NMDA receptor signaling. Another promising approach is to increase activity of another family of glutamate receptors, termed metabotropic glutamate receptors (mGlus), which play important modulatory roles in brain circuits that are thought to be disrupted in schizophrenia patients. Activation of the group I (mGlu5) and the group II (mGlu2 and mGlu3) mGlus is hypothesized to normalize the disruption of aberrant signaling in these circuits. Novel drug-like molecules that increase activity of these receptors have robust efficacy in animal models that predict efficacy in treatment of schizophrenia. Early clinical studies provide some support for potential utility of these targets in reducing symptoms in schizophrenia patients. Clinical studies that are underway will provide further insights into the potential utility of these compounds in the treatment of multiple symptom domains in schizophrenia patients.

Introduction

Schizophrenia is a debilitating psychiatric illness that affects approximately 1% of the world’s population. The main symptoms associated with schizophrenia are grouped into three major symptom clusters that include positive symptoms, negative symptoms, and cognitive disturbances (Lewis and Lieberman, 2000). The positive symptoms include visual and auditory hallucinations, delusions, and thought disorder. The negative symptoms include social withdrawal and anhedonia. Cognitive impairments are characterized by disturbances in sensory information processing, attention, working memory, and executive functions (Nuechterlein et al., 2004). Current drug therapies for schizophrenia include typical (e.g., haloperidol and chlorpromazine) and atypical (e.g., olanzapine and clozapine) antipsychotics, all of which act as antagonists at D2 dopamine receptors and other monoamine receptor subtypes. Typical and atypical antipsychotics are at least partially effective in reducing the positive symptoms in most patients. However, some patients are refractory and these drugs are not effective in the treatment of the negative and cognitive symptoms (Lieberman et al., 2005; 2003). In addition, available antipsychotic medications induce metabolic syndrome, parkinsonian-like motor effects, and other adverse effects that are tied to their mechanism of action (Lieberman et al., 2005; 2003; Pramyothin and Khaodhiar, 2010). Thus, there is a tremendous need to develop new approaches for treating schizophrenia that provide greater efficacy in reducing all of the major symptoms associated with this disease and have fewer adverse effects.

Discovery of currently available treatment strategies for schizophrenia primarily focused on modulating signaling by dopamine, serotonin, and other monoamine systems. However, it is becoming increasingly clear that the pathophysiology underlying schizophrenia cannot be explained by simple changes in monoamine signaling but reflect more complex alterations in activity through key brain circuits that are critical for sensory, cognitive, and emotional processing (Lisman et al., 2008; Marek et al., 2010). While these brain circuits are modulated by dopamine and serotonin, the major excitatory and inhibitory neurotransmitters that are critical for signaling through these circuits are glutamate and GABA; these circuits can also be regulated by other neuromodulators, such as acetylcholine (for review, (Langmead et al., 2008).

Targeting Glutamatergic Signaling for the Treatment of Schizophrenia

Multiple studies over the past 6 decades reveal that selective antagonists at a specific subtype of receptor for the excitatory neurotransmitter glutamate termed the N-methyl-D-aspartate receptor (NMDA receptor) induce a syndrome in healthy individuals that includes positive, negative, and cognitive symptoms that are remarkably similar to those observed in schizophrenia patients. Furthermore, NMDA receptor antagonists exacerbate all three symptom clusters in individuals with schizophrenia (Conn et al., 2009a; Ghoneim et al., 1985; Krystal et al., 1994). In addition, multiple early studies with agents that increase NMDA receptor signaling, such as the amino acids glycine and serine, suggested that increasing NMDA receptor function may provide a benefit in reducing symptoms in schizophrenia patients. These observations have stimulated intense efforts aimed at developing novel approaches that could be used to increase NMDA receptor signaling. However, excessive activation of the NMDA receptor can induce severe toxicity, including epileptic seizure activity and neuronal death (Hirose and Chan, 1993). Thus, the major focus has been to develop agents that will have more subtle effects on NMDA receptor signaling and increase NMDA receptor responses to glutamate without inducing excessive activation of the NMDA receptor. In addition, major efforts have been focused on developing and understanding the impact of NMDA receptor antagonists on brain circuits that may be disrupted in schizophrenia patients and using this knowledge to identify novel targets to restore signaling balance in those brain circuits that are disrupted by NMDA receptor antagonists.

Glycine Transporter Inhibitors

While glutamate is viewed as the primary neurotransmitter responsible for activation of the NMDA receptor, activation of this receptor also requires binding of glycine, which serves as a co-agonist (Curras and Pallotta, 1996). Based on this observation, it is possible that increasing synaptic glycine levels could enhance NMDA receptor signaling but do so in a way that maintains a strict dependence of NMDA receptor activation on synaptic release of glutamate. Activators of the glycine site could be viewed as the most direct approaches to increasing NMDA receptor signaling in a manner that maintains activity dependence. Indeed, direct administration of glycine or serine, another amino acid agonist at the glycine site, was the first approach to increasing activation of NMDA receptors that was tested in clinical studies, and multiple studies have reported that glycine and serine can improve symptoms in schizophrenic patients (Coyle and Tsai, 2004; Heresco-Levy et al., 2004; Javitt, 2006; Lane et al., 2005). However, while encouraging, glycine is not optimized as a central nervous system (CNS)-active drug and suffers from a number of limitations, including poor pharmacokinetic profile and low brain exposure that compromise interpretation of clinical findings and make it unsuitable as a therapeutic agent. Indeed, other studies failed to confirm efficacy of glycine and D-serine (Buchanan et al., 2007; Weiser et al., 2012). It is unclear whether the conflicting data suggest that this mechanism will not provide robust efficacy in schizophrenia patients or if it is due to limitations in CNS exposure or differences in study design between the different trials.

Given the unfavorable properties of glycine, a preferable approach to direct administration of glycine would be to develop highly selective inhibitors of the glycine transporter, GlyT1. GlyT1 is expressed on glia and glutamatergic neurons of the forebrain, and is responsible for regulating glycine concentrations in glutamatergic synapses. Thus, blockade of GlyT1 could elevate synaptic glycine concentrations thereby facilitating NMDA receptor responses (for reviews, see Javitt, 2012; Moghaddam and Javitt, 2012). Early GlyT1 inhibitors were based on the chemical scaffold of the endogenous GlyT1 inhibitor, sarcosine. These agents have efficacy in rodent models that predict efficacy in reducing symptoms in schizophrenia patients (Kinney et al., 2003a) but also induce severe adverse effects, such as compulsive walking, respiratory distress, and eventually death (Perry et al., 2008; Yang and Svensson, 2008). Thus, most recent efforts have focused on selective GlyT1 inhibitors that are structurally unrelated to sarcosine. Multiple newer generation GlyT1 inhibitors have been developed that have robust efficacy in animal models that predict efficacy in the positive, negative, and cognitive symptoms observed in schizophrenia patients and are devoid of serious adverse effects at doses that have robust efficacy (Black et al., 2009; Boulay et al., 2010; Lindsley et al., 2006). Some of these newer GlyT1 inhibitors have now advanced into clinical development and are showing some promise in treatment of schizophrenia. While initiation of multiple clinical trials have been announced, data from the majority of these studies have not yet been reported and reports that are available only include abstracts and press releases, making it difficult to fully assess current clinical findings. However, a phase II study with a novel GlyT1 inhibitor developed by Roche, RG1678 (Pinard et al., 2010), administered as an add-on treatment in schizophrenic patients treated with atypical antipsychotic agents demonstrated significant improvement in negative symptoms (Hopkins, 2011; Umbricht et al., 2010). Interestingly, efficacy was observed at 10 and 30 mg dosage groups but not with a higher dose of 60 mg. While the safety profile for RG1678 was favorable with all three doses, this biphasic dose response relationship may present challenges in establishing appropriate doses in individual patients. Nevertheless, these clinical results are encouraging and led to initiation of a phase III clinical development program for this compound.

More recently, D’Souza et al. (2012) reported that another GlyT1 inhibitor, Org 25935, developed by Organon (now Merck) reversed some schizophrenia-like psychotic symptoms, perceptual alterations, and subjective effects of the NMDA receptor antagonist ketamine in healthy male subjects, providing further support for the hypothesis that GlyT1 inhibitors may provide a novel approach to the treatment of schizophrenia. However, Szegedi et al. (2011) and Dogterom et al. (2011) presented unpublished findings from Merck suggesting that ORG 25935 does not provide further improvement in negative symptoms in patients treated with atypical antipsychotic agents. Thus, while some early data are encouraging, it will be important to fully evaluate emerging clinical data from studies assessing the potential efficacy of GlyT1 inhibitors in schizophrenia patients.

Metabotropic Glutamate Receptor mGlu5

In addition to signaling through the NMDA receptor and other glutamate-gated cation channels, glutamate can also modulate or fine-tune activity in brain circuits by actions on a family of G-protein coupled glutamate receptors termed metabotropic glutamate (mGlu) receptors (Niswender and Conn, 2010). Eight different subtypes of mGlu receptors, termed mGlu1 - mGlu8 exist in the mammalian brain where they play multiple roles in regulating CNS function. Interestingly, one mGlu subtype, mGlu5, has emerged as a closely associated signaling partner with NMDA receptors and may play an integral role in regulating NMDA receptor function in a variety of forebrain regions. NMDA receptors physically interact with mGlu5 via binding to scaffolding proteins (Ehlers, 1999) and functionally interact via a reciprocal positive feedback system in which mGlu5 potentiates NMDA receptor currents (Attucci et al., 2001; Awad et al., 2000; Doherty et al., 2000; Mannaioni et al., 2001; Marino and Conn, 2002; Ugolini et al., 1999). NMDA receptors also regulate responses to activation of mGlu5 through activation of the protein phosphatase calcineurin or a protein kinase (Alagarsamy et al., 1999; Alagarsamy et al., 2005). Thus, selective activators of mGlu5 may provide a viable approach to increasing NMDA receptor function for the treatment of symptoms associated with schizophrenia. Consistent with this hypothesis, mGlu5 knock-out mice display a behavioral phenotype resembling some behavioral effects of NMDA receptor antagonists (Brody et al., 2004; Kinney et al., 2003b). Furthermore, selective mGlu5 antagonists potentiate the psychotomimetic effects of NMDA receptor antagonists in animal models (Brody et al., 2004; Campbell et al., 2004; Kinney et al., 2003b). Taken together, these findings led to the hypothesis that mGlu5 may represent a viable target for the development of novel agents for treatment of schizophrenia (Conn et al., 2009a). Promising evidence in animal models provides support for this hypothesis, such that selective activators of mGlu5 have been shown to have potential for efficacy in treatment of positive and negative symptoms of schizophrenia and may also have cognitive-enhancing effects (Conn et al., 2009a).

Unfortunately, it has been extremely difficult to develop selective agonists of mGlu5 that have suitable properties for use as therapeutic agents. The glutamate binding site is highly conserved across the eight mGlu subtypes (Conn and Pin, 1997; Niswender and Conn, 2010), making it difficult to develop selective glutamate-site ligands. However, there has been tremendous progress in developing highly selective mGlu5 activators that do not act at the conserved glutamate binding site but instead act as positive allosteric modulators (PAMs) of mGlu5 (for review, see Stauffer, 2011). These prototypical mGlu5 PAMs do not activate the receptor directly but potentiate responses to glutamate through binding at an allosteric site. Progress in optimizing mGlu5 PAMs for use in animal studies has been rapid and these compounds have robust activity in a number of animal models that provide support for the hypothesis that selective mGlu5 PAMs have potential as a novel therapeutics for the treatment of schizophrenia. For instance, several structurally distinct mGlu5 PAMs have efficacy in multiple rodent models that are traditionally used to predict efficacy in reducing positive symptoms in schizophrenia patients (Epping-Jordan et al., 2005; Kinney et al., 2005; Liu et al., 2008; Rodriguez et al., 2010; Schlumberger et al., 2009a; Xiong et al., 2010; Vinson and Conn, 2012). These include models such as amphetamine-induced hyper-locomotion and apomorphine-induced disruption of sensory motor gating that reflect increases in dopaminergic signaling and are responsive to existing typical and atypical antipsychotic agents. However, unlike currently marketed antipsychotics, mGlu5 PAMs do not inhibit dopamine and other monoamine receptors (Kinney et al., 2005). This suggests that these agents modulate circuits that are important for the pathophysiology underlying schizophrenia and that these same circuits are also modulated by dopaminergic systems. Especially encouraging is the finding that mGlu5 PAMs belonging to multiple chemical scaffolds have efficacy in these models and these compounds span a range of chemical series that are known to only have activity at mGlu5. In addition to demonstrating efficacy in animal models that predict effects on positive symptoms, early studies suggest that mGlu5 PAMs may have effects in animal models that are relevant to the negative symptoms in schizophrenia. Specifically, one of the first systemically active mGlu5 PAMs, CDPPB, attenuates the ability of an NMDA receptor antagonist, MK-801, to induce deficits in sucrose preference in rats (Vardigan et al., 2010). Finally, abundant evidence from animal studies now supports the hypothesis that mGlu5 PAMs may have efficacy in improving multiple domains of cognitive function. Selective mGlu5 PAMs enhance forms of synaptic plasticity that are thought to provide a cellular mechanism for specific forms of cognitive function (Ayala et al., 2009; Noetzel et al., 2012) and mGlu5 PAMs enhance multiple forms of cognitive function in animal models that are thought to reflect specific domains of cognition that are disrupted in schizophrenia patients (Ayala et al., 2009; Chan et al., 2008; Clifton et al., 2012; Darrah et al., 2008; Gastambide et al., 2012; Horio et al., 2012; Liu et al., 2008; Uslaner et al., 2009).

While the studies outlined above are encouraging, to date no mGlu5 PAMs have been advanced into clinical studies. As with most novel targets for CNS disorders, there are clear challenges that must be overcome before mGlu5 PAMs could be used for the treatment of schizophrenia. For instance, Parmentier-Batteur et al. (2012) recently reported that repeated administration of the mGlu5 PAM CDPPB may lead to desensitization and a diminished response. It is not clear whether this represents a pharmacodynamic tolerance that will be seen with all mGlu5 PAMs or if this will impact efficacy with chronic dosing. However, the possibility that efficacy could be lost with chronic administration of mGlu5 PAMs warrants further investigation. More importantly, recent studies suggest that some mGlu5 PAMs may also have severe target-dependent adverse effects, including induction of behavioral convulsions and excitotoxicity (Parmentier-Batteur et al., 2011; Rook et al., 2012). If these adverse effects are common to all mGlu5 PAMs, this could prevent these agents from advancing to clinical development. However, some mGlu5 PAMs have been shown to have efficacy without inducing obvious adverse effects. Furthermore, a recent study suggests that some mGlu5 PAMs can directly activate mGlu5 as allosteric agonists (Noetzel et al., 2012; Rook et al., 2012). Interestingly, closely related mGlu5 PAMs that differ in terms of allosteric agonist activity also differ in their propensity to induce these adverse effects. Thus, mGlu5 PAMs that have been optimized to have robust agonist activity induce severe adverse effects including behavioral convulsions, whereas closely related mGlu5 PAMs with no observable agonist activity exhibit in vivo efficacy in animal models predictive of antipsychotic activity but do not induce seizure activity or other observable adverse effects. It is likely that multiple factors can contribute to the safety profiles of mGlu5 PAMs. However, these data suggest that relatively subtle differences in the properties of different mGlu5 PAMs can dramatically influence the overall in vivo profile of these compound. Understanding the potential liabilities of mGlu5 PAMs and the factors that influence different in vivo profiles of these compounds will be critical for advancing mGlu5 PAMs into clinical testing in patients suffering from schizophrenia.

Group II Metabotropic Glutamate Receptors

In addition to mGlu5, major efforts have been focused on selective activation of two other mGlu subtypes, mGlu2 and mGlu3, as a novel approach for treatment of schizophrenia. The mGlu2 and mGlu3 receptor subtypes are closely related in terms of primary sequence, signaling, and pharmacological profile, and together these subtypes are referred to as the group II mGlu receptors. Studies dating back to the 1990s reveal that highly selective group II mGlu agonists have robust activity in a range of animal models predictive of antipsychotic activity (Lorrain et al., 2003; Marek et al., 2000; Moghaddam and Adams, 1998; Schoepp and Marek, 2002). These group II mGlu agonists have similar agonist activity at mGlu2 and mGlu3, but are highly selective for the group II mGlus relative to all other mGlu subtypes. Unlike mGlu5 PAMs and GlyT1 inhibitors, mGlu2/3 agonists do not directly potentiate NMDA receptor currents in most neuronal populations. Instead, these agents are thought to act at a circuit level to restore signaling balance of transmission through forebrain circuits that are disrupted by NMDA receptor antagonists and are thought to be disrupted in schizophrenia patients (Battaglia et al., 1997; Lovinger and McCool, 1995; Moghaddam et al., 1997; Moghaddam and Adams, 1998). For example, NMDA receptor antagonists and other psychotomimetic agents increase activity at synapses of glutamatergic projections from the thalamus to the prefrontal cortex (PFC). This increase in spontaneous activity is thought to reflect a loss of normal inhibitory control of transmission to the PFC and has been postulated to play a critical role in the pathophysiology of schizophrenia. Activation of group II mGlu receptors can dramatically reduce excessive excitatory transmission at these synapses in the PFC and thus may provide a mechanism by which these agents could provide antipsychotic effects.

Early clinical studies provided exciting support for the hypothesis that group II mGlu agonists could provide efficacy in the treatment of schizophrenia. In a phase II trial, LY2140023, a selective group II mGlu agonist developed by Eli Lilly, improved ratings for positive and negative symptoms in patients suffering from schizophrenia (Patil et al., 2007). Furthermore, there were no major liabilities associated with current medications, including sedation, amnesic symptoms, withdrawal upon discontinuation of the drug, prolactin elevation, extrapyramidal symptoms, or weight gain. Unfortunately, subsequent studies have not provided a clear confirmation of the efficacy of mGlu2/3 agonists in the treatment of schizophrenia. For instance, in a larger multicenter trial, Kinon et al. (2011) reported that LY2140023 did not differentiate from placebo on standard scales of positive and negative symptoms. However, the placebo response in this study was higher than expected and the positive comparator, olanzapine, also failed to significantly separate from placebo. Thus, the results from this trial were inconclusive and did not provide a clear interpretation in terms of the potential efficacy of the group II mGlu receptor agonist. Unfortunately, further clinical studies of the effects of group II mGlu agonists have not been published in the peer reviewed literature. However, Eli Lilly recently announced that they had terminated development of LY2140023 based on a third clinical study in which they failed to observe efficacy in reducing symptoms of schizophrenia. In this latest study, the placebo response was reported to be within normal range and the positive comparator showed clear efficacy in separating from placebo (Carroll, 2012).

While the results of the Eli Lilly trial are disappointing, a possibility remains that future agents that target group II mGlu receptors will provide more reliable results. It is likely that subsequent clinical studies will shift from mGlu2/3 agonists to focus on novel PAMs that are highly selective for mGlu2 and have no activity at mGlu3. As discussed above for mGlu5 PAMs, these compounds do not activate mGlu2 directly but bind to a site distinct from the glutamate binding site to increase responses of mGlu2 to glutamate. Multiple mGlu2 PAMs have been identified and these compounds have similar effects to those of mGlu2/3 agonists on excitatory transmission in the PFC (Benneyworth et al., 2007) and in animal models that predict antipsychotic efficacy (Duplantier et al., 2009; Galici et al., 2005; 2006; Govek et al., 2005; Johnson et al., 2003; Pinkerton et al., 2005). While it is not known whether mGlu2 PAMs will offer advantages over mGlu2/3 agonists, it is conceivable that selectively targeting mGlu2 will provide more reliable efficacy. Also, it is possible that chronic administration of traditional agonists leads to receptor desensitization and this could reduce the clinical response to the agonists. Thus, selective mGlu2 PAMs may provide an alternative approach that could have advantages to mGlu2/3 agonists. At present, clinical data using mGlu2 PAMs have not been reported. However, Addex Pharmaceuticals and Johnson and Johnson have advanced a novel mGlu2 PAM termed ADX71149 into phase II efficacy studies in schizophrenia patients.

Summary

Current treatment strategies for schizophrenia primarily address the positive symptoms of the disorder. This leaves a large unmet need in the treatment of the negative and cognitive symptoms. Evidence suggests that targeting aberrant glutamatergic signaling may provide a new approach that could be used to treat all three symptom clusters in schizophrenia; a considerable improvement over the current therapeutics. The coming years will be instrumental in determining the feasibility of modulating glutamate signaling as therapeutic treatments for schizophrenia using compounds that act as GlyT1 inhibitors, mGlu5 PAMs and/or mGlu2/3 agonists or PAMs. Other targets may also be viable approaches to address glutamatergic tone for the treatment of schizophrenia; AMPA receptor expression is decreased while D-amino acid oxidase (DAAO) activity is elevated in schizophrenia, raising the possibility of these proteins as possible targets (for review, see de Bartolomeis et al., 2012). Interestingly, AMPAkines are allosteric modulators of AMPA receptors, increasing channel opening, which may compensate for the lower expression levels (de Bartolomeis et al., 2012). Inhibitors are sought for DAAO, thereby decreasing the degradation of D-serine, mediated by this enzyme (Madeira et al., 2008). There are multiple avenues for glutamate based therapies for the treatment of schizophrenia. It will be important to continue to explore these possibilities to ultimately improve patient outcomes.

Footnotes

Disclosure

P.J.C. is a consultant for Seaside Therapeutic and Karuna Pharmaceuticals and receives research support from Bristol-Myers Squibb. C.K.J. receives research support from Bristol-Myers Squibb. P.J.C. and C.K.J. are inventors on multiple composition of matter patents protecting allosteric modulators of mGlus and other GPCRs.

References

  1. Alagarsamy S, Marino MJ, Rouse ST, Gereau RWT, Heinemann SF, Conn PJ. Activation of NMDA receptors reverses desensitization of mGluR5 in native and recombinant systems. Nat Neurosci. 1999;2(3):234–240. doi: 10.1038/6338. [DOI] [PubMed] [Google Scholar]
  2. Alagarsamy S, Saugstad J, Warren L, Mansuy IM, Gereau RWT, Conn PJ. NMDA-induced potentiation of mGluR5 is mediated by activation of protein phosphatase 2B/calcineurin. Neuropharmacology. 2005;49(Suppl 1):135–145. doi: 10.1016/j.neuropharm.2005.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Attucci S, Carla V, Mannaioni G, Moroni F. Activation of type 5 metabotropic glutamate receptors enhances NMDA responses in mice cortical wedges. Br J Pharmacol. 2001;132(4):799–806. doi: 10.1038/sj.bjp.0703904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Awad H, Hubert GW, Smith Y, Levey AI, Conn PJ. Activation of metabotropic glutamate receptor 5 has direct excitatory effects and potentiates NMDA receptor currents in neurons of the subthalamic nucleus. J Neurosci. 2000;20(21):7871–7879. doi: 10.1523/JNEUROSCI.20-21-07871.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Ayala JE, Chen Y, Banko JL, Sheffler DJ, Williams R, Telk AN, Watson NL, Xiang Z, Zhang Y, Jones PJ, Lindsley CW, Olive MF, Conn PJ. mGluR5 positive allosteric modulators facilitate both hippocampal LTP and LTD and enhance spatial learning. Neuropsychopharmacology. 2009;34(9):2057–2071. doi: 10.1038/npp.2009.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Battaglia G, Monn JA, Schoepp DD. In vivo inhibition of veratridine-evoked release of striatal excitatory amino acids by the group II metabotropic glutamate receptor agonist LY354740 in rats. Neurosci Lett. 1997;229(3):161–164. doi: 10.1016/s0304-3940(97)00442-4. [DOI] [PubMed] [Google Scholar]
  7. Benneyworth MA, Xiang Z, Smith RL, Garcia EE, Conn PJ, Sanders-Bush E. A selective positive allosteric modulator of metabotropic glutamate receptor subtype 2 blocks a hallucinogenic drug model of psychosis. Mol Pharmacol. 2007;72(2):477–484. doi: 10.1124/mol.107.035170. [DOI] [PubMed] [Google Scholar]
  8. Black MD, Varty GB, Arad M, Barak S, De Levie A, Boulay D, Pichat P, Griebel G, Weiner I. Procognitive and antipsychotic efficacy of glycine transport 1 inhibitors (GlyT1) in acute and neurodevelopmental models of schizophrenia: latent inhibition studies in the rat. Psychopharmacology. 2009;202(1-3):385–396. doi: 10.1007/s00213-008-1289-2. [DOI] [PubMed] [Google Scholar]
  9. Boulay D, Bergis O, Avenet P, Griebel G. The glycine transporter-1 inhibitor SSR103800 displays a selective and specific antipsychotic-like profile in normal and transgenic mice. Neuropsychopharmacology. 2010;35(2):416–427. doi: 10.1038/npp.2009.144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Brody SA, Dulawa SC, Conquet F, Geyer MA. Assessment of a pre-pulse inhibition deficit in a mutant mouse lacking mGlu5 receptors. Mol Psychiatry. 2004;9(1):35–41. doi: 10.1038/sj.mp.4001404. [DOI] [PubMed] [Google Scholar]
  11. Buchanan RW, Javitt DC, Marder SR, Schooler NR, Gold JM, Mcmahon RP, Heresco-Levy U, Carpenter WT. The Cognitive and Negative Symptoms in Schizophrenia Trial (CONSIST): the efficacy of glutamatergic agents for negative symptoms and cognitive impairments. Am J Psychiatry. 2007;164(10):1593–1602. doi: 10.1176/appi.ajp.2007.06081358. [DOI] [PubMed] [Google Scholar]
  12. Campbell UC, Lalwani K, Hernandez L, Kinney GG, Conn PJ, Bristow LJ. The mGluR5 antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) potentiates PCP-induced cognitive deficits in rats. Psychopharmacology. 2004;175(3):310–318. doi: 10.1007/s00213-004-1827-5. [DOI] [PubMed] [Google Scholar]
  13. Carroll J. Strike three: Bad data bury Eli Lilly’s late-stage schizophrenia drug. Fierce Biotech. 2012 Aug 29; [Google Scholar]
  14. Chan MH, Chiu PH, Sou JH, Chen HH. Attenuation of ketamine-evoked behavioral responses by mGluR5 positive modulators in mice. Psychopharmacology. 2008;198(1):141–148. doi: 10.1007/s00213-008-1103-1. [DOI] [PubMed] [Google Scholar]
  15. Clifton NE, Morisot N, Girardon S, Millan MJ, Loiseau F. Enhancement of social novelty discrimination by positive allosteric modulators at metabotropic glutamate 5 receptors: adolescent administration prevents adult-onset deficits induced by neonatal treatment with phencyclidine. Psychopharmacology (Berl) 2012 Sep 16; doi: 10.1007/s00213-012-2845-3. epub ahead of print. [DOI] [PubMed] [Google Scholar]
  16. Conn PJ, Lindsley CW, Jones CK. Activation of metabotropic glutamate receptors as a novel approach for the treatment of schizophrenia. Trends Pharmacol Sci. 2009a;30(1):25–31. doi: 10.1016/j.tips.2008.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Conn PJ, Pin JP. Pharmacology and functions of metabotropic glutamate receptors. Annu Rev Pharmacol Toxicol. 1997;37:205–237. doi: 10.1146/annurev.pharmtox.37.1.205. [DOI] [PubMed] [Google Scholar]
  18. Coyle JT, Tsai G. The NMDA receptor glycine modulatory site: a therapeutic target for improving cognition and reducing negative symptoms in schizophrenia. Psychopharmacology. 2004;174(1):32–38. doi: 10.1007/s00213-003-1709-2. [DOI] [PubMed] [Google Scholar]
  19. Curras MC, Pallotta BS. Single-channel evidence for glycine and NMDA requirement in NMDA receptor activation. Brain Res. 1996;740(1-2):27–40. doi: 10.1016/s0006-8993(96)00845-1. [DOI] [PubMed] [Google Scholar]
  20. D’souza DC, Singh N, Elander J, Carbuto M, Pittman B, De Haes JU, Sjogren M, Peeters P, Ranganathan M, Schipper J. Glycine transporter inhibitor attenuates the psychotomimetic effects of ketamine in healthy males: preliminary evidence. Neuropsychopharmacology. 2012;37(4):1036–1046. doi: 10.1038/npp.2011.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Darrah JM, Stefani MR, Moghaddam B. Interaction of N-methyl-D-aspartate and group 5 metabotropic glutamate receptors on behavioral flexibility using a novel operant set-shift paradigm. Behav Pharmacol. 2008;19(3):225–234. doi: 10.1097/FBP.0b013e3282feb0ac. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. De Bartolomeis A, Sarappa C, Magara S, Iasevoli F. Targeting glutamate system for novel antipsychotic approaches: relevance for residual psychotic symptoms and treatment resistant schizophrenia. E J Pharmacol. 2012;682(1-3):1–11. doi: 10.1016/j.ejphar.2012.02.033. [DOI] [PubMed] [Google Scholar]
  23. Dogterom P, Thomson F, Hargreaves R, Hamill T, Sur C, Uslaner J, Eddins D, Vardigan JD, Jayaraman S, Morrow J, Kleijn H, Schipper J, Ereshefsky L, Jhee S, Schoepp DD. Characterization of the Relationship between Target Occupancy/Modulation and Preclinical/Clinical Responses for the Glycine Transporter1 (GlyT1) Inhibitor Org 25935; Proceedings of the American College of Neuropsychopharmacology Annual Meeting; Hawaii, USA. 2011.p. p361. [Google Scholar]
  24. Doherty AJ, Palmer MJ, Bortolotto ZA, Hargreaves A, Kingston AE, Ornstein PL, Schoepp DD, Lodge D, Collingridge GL. A novel, competitive mGlu(5) receptor antagonist (LY344545) blocks DHPG-induced potentiation of NMDA responses but not the induction of LTP in rat hippocampal slices. Br J Pharmacol. 2000;131(2):239–244. doi: 10.1038/sj.bjp.0703574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Duplantier AJ, Efremov I, Candler J, Doran AC, Ganong AH, Haas JA, Hanks AN, Kraus KG, Lazzaro JT, Jr., Lu J, Maklad N, Mccarthy SA, O’sullivan TJ, Rogers BN, Siuciak JA, Spracklin DK, Zhang L. 3-Benzyl-1,3-oxazolidin-2-ones as mGluR2 positive allosteric modulators: Hit-to lead and lead optimization. Bioorg Med Chem Lett. 2009;19(9):2524–2529. doi: 10.1016/j.bmcl.2009.03.032. [DOI] [PubMed] [Google Scholar]
  26. Ehlers MD. Synapse structure: glutamate receptors connected by the shanks. Curr Biol. 1999;9(22):R848–850. doi: 10.1016/s0960-9822(00)80043-3. [DOI] [PubMed] [Google Scholar]
  27. Epping-Jordan MP, Nayak S, Derouet F, Dominguez H, Bessis AS, Le Poul E, Ludwig B, Mutel V, Poli SM, Rocher JP. In vivo characterization of mGluR5 positive allosteric modulators as novel treatments for schizophrenia and cognitive dysfunction. Neuropharmacology. 2005;49(Suppl 1):243. [Google Scholar]
  28. Galici R, Echemendia NG, Rodriguez AL, Conn PJ. A selective allosteric potentiator of metabotropic glutamate (mGlu) 2 receptors has effects similar to an orthosteric mGlu2/3 receptor agonist in mouse models predictive of antipsychotic activity. J Pharmacol Exp Ther. 2005;315(3):1181–1187. doi: 10.1124/jpet.105.091074. [DOI] [PubMed] [Google Scholar]
  29. Galici R, Jones CK, Hemstapat K, Nong Y, Echemendia NG, Williams LC, De Paulis T, Conn PJ. Biphenyl-indanone A, a positive allosteric modulator of the metabotropic glutamate receptor subtype 2, has antipsychotic- and anxiolytic-like effects in mice. J Pharmacol Exp Ther. 2006;318(1):173–185. doi: 10.1124/jpet.106.102046. [DOI] [PubMed] [Google Scholar]
  30. Gastambide F, Cotel MC, Gilmour G, O’neill MJ, Robbins TW, Tricklebank MD. Selective remediation of reversal learning deficits in the neurodevelopmental MAM model of schizophrenia by a novel mGlu5 positive allosteric modulator. Neuropsychopharmacology. 2012;37(4):1057–1066. doi: 10.1038/npp.2011.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Ghoneim MM, Hinrichs JV, Mewaldt SP, Petersen RC. Ketamine: behavioral effects of subanesthetic doses. J Clin Psychopharmacol. 1985;5(2):70–77. [PubMed] [Google Scholar]
  32. Govek SP, Bonnefous C, Hutchinson JH, Kamenecka T, Mcquiston J, Pracitto R, Zhao LX, Gardner MF, James JK, Daggett LP, Rowe BA, Schaffhauser H, Bristow LJ, Campbell UC, Rodriguez DE, Vernier JM. Benzazoles as allosteric potentiators of metabotropic glutamate receptor 2 (mGluR2): efficacy in an animal model for schizophrenia. Bioorg Med Chem Lett. 2005;15(18):4068–4072. doi: 10.1016/j.bmcl.2005.06.017. [DOI] [PubMed] [Google Scholar]
  33. Heresco-Levy U, Ermilov M, Lichtenberg P, Bar G, Javitt DC. High-dose glycine added to olanzapine and risperidone for the treatment of schizophrenia. Biol Psychiatry. 2004;55(2):165–171. doi: 10.1016/s0006-3223(03)00707-8. [DOI] [PubMed] [Google Scholar]
  34. Hirose K, Chan PH. Blockade of glutamate excitotoxicity and its clinical applications. Neurochem Res. 1993;18(4):479–483. doi: 10.1007/BF00967252. [DOI] [PubMed] [Google Scholar]
  35. Hopkins CR. ACS Chemical Neuroscience Molecule Spotlight on RG1678. ACS Chem Neurosci. 2011;2(12):685–686. doi: 10.1021/cn200108z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Horio M, Fujita Y, Hashimoto K. Therapeutic effects of metabotropic glutamate receptor 5 positive allosteric modulator CDPPB on phencyclidine-induced cognitive deficits in mice. Fundam Clin Pharmacol. 2012 May 2; doi: 10.1111/j.1472-8206.2012.01045.x. epub ahead of print. [DOI] [PubMed] [Google Scholar]
  37. Javitt DC. Is the glycine site half saturated or half unsaturated? Effects of glutamatergic drugs in schizophrenia patients. Curr Opin Psychiatry. 2006;19(2):151–157. doi: 10.1097/01.yco.0000214340.14131.bd. [DOI] [PubMed] [Google Scholar]
  38. Javitt DC. Glycine transport inhibitors in the treatment of schizophrenia. Handb Exp Pharmacol. 2012;(213):367–399. doi: 10.1007/978-3-642-25758-2_12. [DOI] [PubMed] [Google Scholar]
  39. Johnson MP, Baez M, Jagdmann GE, Jr., Britton TC, Large TH, Callagaro DO, Tizzano JP, Monn JA, Schoepp DD. Discovery of allosteric potentiators for the metabotropic glutamate 2 receptor: synthesis and subtype selectivity of N-(4-(2-methoxyphenoxy)phenyl)-N-(2,2,2-trifluoroethylsulfonyl)pyrid-3-ylmethylamine. J Med Chem. 2003;46(15):3189–3192. doi: 10.1021/jm034015u. [DOI] [PubMed] [Google Scholar]
  40. Kinney GG, Burno M, Campbell UC, Hernandez LM, Rodriguez D, Bristow LJ, Conn PJ. Metabotropic glutamate subtype 5 receptors modulate locomotor activity and sensorimotor gating in rodents. J Pharmacol Exp Ther. 2003b;306(1):116–123. doi: 10.1124/jpet.103.048702. [DOI] [PubMed] [Google Scholar]
  41. Kinney GG, O’Brien JA, Lemaire W, Burno M, Bickel DJ, Clements MK, Chen TB, Wisnoski DD, Lindsley CW, Tiller PR, Smith S, Jacobson MA, Sur C, Duggan ME, Pettibone DJ, Conn PJ, Williams DL., Jr. A novel selective positive allosteric modulator of metabotropic glutamate receptor subtype 5 has in vivo activity and antipsychotic-like effects in rat behavioral models. J Pharmacol Exp Ther. 2005;313(1):199–206. doi: 10.1124/jpet.104.079244. [DOI] [PubMed] [Google Scholar]
  42. Kinney GG, Sur C, Burno M, Mallorga PJ, Williams JB, Figueroa DJ, Wittmann M, Lemaire W, Conn PJ. The glycine transporter type 1 inhibitor N-[3-(4′-fluorophenyl)-3-(4′-phenylphenoxy)propyl]sarcosine potentiates NMDA receptor-mediated responses in vivo and produces an antipsychotic profile in rodent behavior. J Nurosci. 2003a;23(20):7586–7591. doi: 10.1523/JNEUROSCI.23-20-07586.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Kinon BJ, Zhang L, Millen BA, Osuntokun OO, Williams JE, Kollack-Walker S, Jackson K, Kryzhanovskaya L, Jarkova N. A multicenter, inpatient, phase 2, double-blind, placebo-controlled dose-ranging study of LY2140023 monohydrate in patients with DSM-IV schizophrenia. J Clin Psychopharmacol. 2011;31(3):349–355. doi: 10.1097/JCP.0b013e318218dcd5. [DOI] [PubMed] [Google Scholar]
  44. Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB, Jr, Charney DS. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994;51(3):199–214. doi: 10.1001/archpsyc.1994.03950030035004. [DOI] [PubMed] [Google Scholar]
  45. Lane HY, Chang YC, Liu YC, Chiu CC, Tsai GE. Sarcosine or D-serine add-on treatment for acute exacerbation of schizophrenia: a randomized, double-blind, placebo-controlled study. Arch Gen Psychiatry. 2005;62(11):1196–1204. doi: 10.1001/archpsyc.62.11.1196. [DOI] [PubMed] [Google Scholar]
  46. Langmead CJ, Watson J, Reavill C. Muscarinic acetylcholine receptors as CNS drug targets. Pharmacol Ther. 2008;117(2):232–243. doi: 10.1016/j.pharmthera.2007.09.009. [DOI] [PubMed] [Google Scholar]
  47. Lieberman JA, Stroup TS, Mcevoy JP, Swartz MS, Rosenheck RA, Perkins DO, Keefe RS, Davis SM, Davis CE, Lebowitz BD, Severe J, Hsiao JK. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med. 2005;353(12):1209–1223. doi: 10.1056/NEJMoa051688. [DOI] [PubMed] [Google Scholar]
  48. Lieberman JA, Tollefson G, Tohen M, Green AI, Gur RE, Kahn R, Mcevoy J, Perkins D, Sharma T, Zipursky R, Wei H, Hamer RM. Comparative efficacy and safety of atypical and conventional antipsychotic drugs in first-episode psychosis: a randomized, double-blind trial of olanzapine versus haloperidol. Am J Psychiatry. 2003;160(8):1396–1404. doi: 10.1176/appi.ajp.160.8.1396. [DOI] [PubMed] [Google Scholar]
  49. Lindsley CW, Zhao Z, Leister WH, O’brien J, Lemaire W, Williams DL, Jr, Chen TB, Chang RS, Burno M, Jacobson MA, Sur C, Kinney GG, Pettibone DJ, Tiller PR, Smith S, Tsou NN, Duggan ME, Conn PJ, Hartman GD. Design, synthesis, and in vivo efficacy of glycine transporter-1 (GlyT1) inhibitors derived from a series of [4-phenyl-1-(propylsulfonyl)piperidin-4-yl]methyl benzamides. ChemMedChem. 2006;1(8):807–811. doi: 10.1002/cmdc.200600097. [DOI] [PubMed] [Google Scholar]
  50. Lisman JE, Coyle JT, Green RW, Javitt DC, Benes FM, Heckers S, Grace AA. Circuit-based framework for understanding neurotransmitter and risk gene interactions in schizophrenia. Trends Neurosci. 2008;31(5):234–242. doi: 10.1016/j.tins.2008.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Liu F, Grauer S, Kelley C, Navarra R, Graf R, Zhang G, Atkinson PJ, Popiolek M, Wantuch C, Khawaja X, Smith D, Olsen M, Kouranova E, Lai M, Pruthi F, Pulicicchio C, Day M, Gilbert A, Pausch MH, Brandon NJ, et al. ADX47273 [S-(4-fluoro-phenyl)-{3-[3-(4-fluoro-phenyl)-[1,2,4]-oxadiazol-5-yl]-piper idin-1-yl}-methanone]: a novel metabotropic glutamate receptor 5-selective positive allosteric modulator with preclinical antipsychotic-like and procognitive activities. J Pharmacol Exp Ther. 2008;327(3):827–839. doi: 10.1124/jpet.108.136580. [DOI] [PubMed] [Google Scholar]
  52. Lorrain DS, Baccei CS, Bristow LJ, Anderson JJ, Varney MA. Effects of ketamine and N-methyl-D-aspartate on glutamate and dopamine release in the rat prefrontal cortex: modulation by a group II selective metabotropic glutamate receptor agonist LY379268. Neuroscience. 2003;117(3):697–706. doi: 10.1016/s0306-4522(02)00652-8. [DOI] [PubMed] [Google Scholar]
  53. Lovinger DM, Mccool BA. Metabotropic glutamate receptor-mediated presynaptic depression at corticostriatal synapses involves mGLuR2 or 3. J Neurophysiol. 1995;73(3):1076–1083. doi: 10.1152/jn.1995.73.3.1076. [DOI] [PubMed] [Google Scholar]
  54. Madeira C, Freitas ME, Vargas-Lopes C, Wolosker H, Panizzutti R. Increased brain D-amino acid oxidase (DAAO) activity in schizophrenia. Schizophr Res. 2008;101(1-3):76–83. doi: 10.1016/j.schres.2008.02.002. [DOI] [PubMed] [Google Scholar]
  55. Mannaioni G, Marino MJ, Valenti O, Traynelis SF, Conn PJ. Metabotropic glutamate receptors 1 and 5 differentially regulate CA1 pyramidal cell function. J Neurosci. 2001;21(16):5925–5934. doi: 10.1523/JNEUROSCI.21-16-05925.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Marek GJ, Behl B, Bespalov AY, Gross G, Lee Y, Schoemaker H. Glutamatergic (N-methyl-D-aspartate receptor) hypofrontality in schizophrenia: too little juice or a miswired brain? Mol Pharmacol. 2010;77(3):317–326. doi: 10.1124/mol.109.059865. [DOI] [PubMed] [Google Scholar]
  57. Marek GJ, Wright RA, Schoepp DD, Monn JA, Aghajanian GK. Physiological antagonism between 5-hydroxytryptamine(2A) and group II metabotropic glutamate receptors in prefrontal cortex. J Pharmacol Exp Ther. 2000;292(1):76–87. [PubMed] [Google Scholar]
  58. Marino MJ, Conn PJ. Direct and indirect modulation of the N-methyl D-aspartate receptor. Curr Drug Targets. 2002;1(1):1–16. doi: 10.2174/1568007023339544. [DOI] [PubMed] [Google Scholar]
  59. Moghaddam B, Adams B, Verma A, Daly D. Activation of glutamatergic neurotransmission by ketamine: a novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. J Neurosci. 1997;17(8):2921–2927. doi: 10.1523/JNEUROSCI.17-08-02921.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Moghaddam B, Adams BW. Reversal of phencyclidine effects by a group II metabotropic glutamate receptor agonist in rats. Science. 1998;281(5381):1349–1352. doi: 10.1126/science.281.5381.1349. [DOI] [PubMed] [Google Scholar]
  61. Moghaddam B, Javitt D. From revolution to evolution: the glutamate hypothesis of schizophrenia and its implication for treatment. Neuropsychopharmacology. 2012;37(1):4–15. doi: 10.1038/npp.2011.181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Niswender CM, Conn PJ. Metabotropic glutamate receptors: physiology, pharmacology, and disease. Ann Rev Pharmacol Toxicol. 2010;50:295–322. doi: 10.1146/annurev.pharmtox.011008.145533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Noetzel MJ, Rook JM, Vinson PN, Cho HP, Days E, Zhou Y, Rodriguez AL, Lavreysen H, Stauffer SR, Niswender CM, Xiang Z, Daniels JS, Jones CK, Lindsley CW, Weaver CD, Conn PJ. Functional impact of allosteric agonist activity of selective positive allosteric modulators of metabotropic glutamate receptor subtype 5 in regulating central nervous system function. Mol Pharmacol. 2012;81(2):120–133. doi: 10.1124/mol.111.075184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Nuechterlein KH, Barch DM, Gold JM, Goldberg TE, Green MF, Heaton RK. Identification of separable cognitive factors in schizophrenia. Schizophr Res. 2004;72(1):29–39. doi: 10.1016/j.schres.2004.09.007. [DOI] [PubMed] [Google Scholar]
  65. Parmentier-Batteur S, Hutson PH, Menzel K, Mattson BA, Uslaner JH, Forest T, Zhang XF, O’brien JA, Magliaro BC, Huszar SL, Hugues A, Lodge KE, Jacobson MA, Stump CA, Tynebor RM, Anthony NJ, Williams TA. Preclinical profile of a novel metabotropic glutamate receptor 5 positive allosteric modulator: from efficacy to mechanism-based neurotoxicity; Proceedings of the 7th International Metabotropic Glutamate Receptors Meeting; Taormina, Italy. 2011.p. 12. [Google Scholar]
  66. Parmentier-Batteur S, O’brien JA, Doran S, Nguyen SJ, Flick RB, Uslaner JM, Chen H, Finger EN, Williams TM, Jacobson MA, Hutson PH. Differential effects of the mGluR5 positive allosteric modulator CDPPB in the cortex and striatum following repeated administration. Neuropharmacology. 2012;62(3):1453–1460. doi: 10.1016/j.neuropharm.2010.11.013. [DOI] [PubMed] [Google Scholar]
  67. Patil ST, Zhang L, Martenyi F, Lowe SL, Jackson KA, Andreev BV, Avedisova AS, Bardenstein LM, Gurovich IY, Morozova MA, Mosolov SN, Neznanov NG, Reznik AM, Smulevich AB, Tochilov VA, Johnson BG, Monn JA, Schoepp DD. Activation of mGlu2/3 receptors as a new approach to treat schizophrenia: a randomized Phase 2 clinical trial. Nat Med. 2007;13(9):1102–1107. doi: 10.1038/nm1632. [DOI] [PubMed] [Google Scholar]
  68. Perry KW, Falcone JF, Fell MJ, Ryder JW, Yu H, Love PL, Katner J, Gordon KD, Wade MR, Man T, Nomikos GG, Phebus LA, Cauvin AJ, Johnson KW, Jones CK, Hoffmann BJ, Sandusky GE, Walter MW, Porter WJ, Yang L, et al. Neurochemical and behavioral profiling of the selective GlyT1 inhibitors ALX5407 and LY2365109 indicate a preferential action in caudal vs. cortical brain areas. Neuropharmacology. 2008;55(5):743–754. doi: 10.1016/j.neuropharm.2008.06.016. [DOI] [PubMed] [Google Scholar]
  69. Pinard E, Alanine A, Alberati D, Bender M, Borroni E, Bourdeaux P, Brom V, Burner S, Fischer H, Hainzl D, Halm R, Hauser N, Jolidon S, Lengyel J, Marty HP, Meyer T, Moreau JL, Mory R, Narquizian R, Nettekoven M, et al. Selective GlyT1 inhibitors: discovery of [4-(3-fluoro-5-trifluoromethylpyridin-2-yl)piperazin-1-yl][5-methanesulfonyl-2-((S)-2,2,2-trifluoro-1-methylethoxy)phenyl]methanone (RG1678), a promising novel medicine to treat schizophrenia. J Med Chem. 2010;53(12):4603–4614. doi: 10.1021/jm100210p. [DOI] [PubMed] [Google Scholar]
  70. Pinkerton AB, Cube RV, Hutchinson JH, James JK, Gardner MF, Rowe BA, Schaffhauser H, Rodriguez DE, Campbell UC, Daggett LP, Vernier JM. Allosteric potentiators of the metabotropic glutamate receptor 2 (mGlu2). Part 3: Identification and biological activity of indanone containing mGlu2 receptor potentiators. Bioorg Med Chem Lett. 2005;15(6):1565–1571. doi: 10.1016/j.bmcl.2005.01.077. [DOI] [PubMed] [Google Scholar]
  71. Pramyothin P, Khaodhiar L. Metabolic syndrome with the atypical antipsychotics. Curr Opin Endocrinol Diabetes Obes. 2010;17(5):460–466. doi: 10.1097/MED.0b013e32833de61c. [DOI] [PubMed] [Google Scholar]
  72. Rodriguez AL, Grier MD, Jones CK, Herman EJ, Kane AS, Smith RL, Williams R, Zhou Y, Marlo JE, Days EL, Blatt TN, Jadhav S, Menon UN, Vinson PN, Rook JM, Stauffer SR, Niswender CM, Lindsley CW, Weaver CD, Conn PJ. Discovery of novel allosteric modulators of metabotropic glutamate receptor subtype 5 reveals chemical and functional diversity and in vivo activity in rat behavioral models of anxiolytic and antipsychotic activity. Mol Pharmacol. 2010;78(6):1105–1123. doi: 10.1124/mol.110.067207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Rook JM, Noetzel Meredith J., Pouliot Wendy A., Bridges Thomas M., Vinson Paige N., Cho Hyekyung P., Zhou Ya, Gogliotti Rocco D., Manka Jason T., Gregory Karen J., Stauffer Shaun R., Edward Dudek F, Xiang Zixiu, Niswender Colleen M., Scott Daniels J, Jones Carrie K., Lindsley Craig W., Conn PJ. Unique Signaling Profiles o fPositive Allosteric Modulators of Metabotropic Glutamate Receptor Subtype 5 Determine Differences in In Vivo Activity. Biol Psychiatry. 2012 doi: 10.1016/j.biopsych.2012.09.012. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Schlumberger C, Pietraszek M, Gravius A, Klein KU, Greco S, More L, Danysz W. Comparison of the mGlu(5) receptor positive allosteric modulator ADX47273 and the mGlu(2/3) receptor agonist LY354740 in tests for antipsychotic-like activity. Eur J Pharmacol. 2009a;623(1-3):73–83. doi: 10.1016/j.ejphar.2009.09.006. [DOI] [PubMed] [Google Scholar]
  75. Schoepp DD, Marek GJ. Preclinical pharmacology of mGlu2 / 3 receptor agonists: novel agents for schizophrenia? Curr Drug Targets. 2002;1:215–225. doi: 10.2174/1568007024606177. [DOI] [PubMed] [Google Scholar]
  76. Stauffer SR. Progress toward positive allosteric modulators of the metabotropic glutamate receptor subtype 5 (mGlu(5)) ACS Chem Neurosci. 2011;2(8):450–470. doi: 10.1021/cn2000519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Szegedi A, Jansen W, Karson C, Schipper J, Schoemaker J. Adjunctive treatment with the selective glycine uptake inhibitor Org25935 in persistent negative symptoms of schizophrenia: results from the GIANT trial; Proceedings of the American College of Neuropsychopharmacology Annual Meeting; Hawaii, USA. 2011.p. 258. [Google Scholar]
  78. Ugolini A, Corsi M, Bordi F. Potentiation of NMDA and AMPA responses by the specific mGluR5 agonist CHPG in spinal cord motoneurons. Neuropharmacology. 1999;38(10):1569–1576. doi: 10.1016/s0028-3908(99)00095-7. [DOI] [PubMed] [Google Scholar]
  79. Umbricht D, Yoo Kisook, Youssef Eriene, Dorflinger Ernest, Martin-Facklam Meret, Bausch Alexander, Arrowsmith Richard, Alberati Daniela, Marder Stephen, Santarelli L. Glycine transporter type 1 (GLYT1) inhibitor RG1678: positive results of the proof-of-concept study for the treatment of negative symptoms in schizophrenia; Proceedings of the ACNP 49th Annual Meeing; Miami Beach, Florida, USA. 2010.p. 277. [Google Scholar]
  80. Uslaner JM, Parmentier-Batteur S, Flick RB, Surles NO, Lam JS, McNaughton CH, Jacobson MA, Hutson PH. Dose-dependent effect of CDPPB, the mGluR5 positive allosteric modulator, on recognition memory is associated with GluR1 and CREB phosphorylation in the prefrontal cortex and hippocampus. Neuropharmacology. 2009;57(5-6):531–538. doi: 10.1016/j.neuropharm.2009.07.022. [DOI] [PubMed] [Google Scholar]
  81. Vardigan JD, Huszar SL, McNaughton CH, Hutson PH, Uslaner JM. MK-801 produces a deficit in sucrose preference that is reversed by clozapine, D-serine, and the metabotropic glutamate 5 receptor positive allosteric modulator CDPPB: relevance to negative symptoms associated with schizophrenia? Pharmacol Biochem Behav. 2010;95(2):223–229. doi: 10.1016/j.pbb.2010.01.010. [DOI] [PubMed] [Google Scholar]
  82. Vinson PN, Conn PJ. Metabotropic glutamate receptors as therapeutic targets for schizophrenia. Neuropharmacology. 2012;62(3):1461–1472. doi: 10.1016/j.neuropharm.2011.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Weiser M, Heresco-Levy U, Davidson M, Javitt DC, Werbeloff N, Gershon AA, Abramovich Y, Amital D, Doron A, Konas S, Levkovitz Y, Liba D, Teitelbaum A, Mashiach M, Zimmerman Y. A multicenter, add-on randomized controlled trial of low-dose d-serine for negative and cognitive symptoms of schizophrenia. J Clin Psychiatry. 2012;73(6):e728–e734. doi: 10.4088/JCP.11m07031. [DOI] [PubMed] [Google Scholar]
  84. Xiong H, Brugel TA, Balestra M, Brown DG, Brush KA, Hightower C, Hinkley L, Hoesch V, Kang J, Koether GM, Mccauley JP, Jr, Mclaren FM, Panko LM, Simpson TR, Smith RW, Woods JM, Brockel B, Chhajlani V, Gadient RA, Spear N, et al. 4-aryl piperazine and piperidine amides as novel mGluR5 positive allosteric modulators. Bioorg Med Chem Lett. 2010;20(24):7381–7384. doi: 10.1016/j.bmcl.2010.10.036. [DOI] [PubMed] [Google Scholar]
  85. Yang CR, Svensson KA. Allosteric modulation of NMDA receptor via elevation of brain glycine and D-serine: the therapeutic potentials for schizophrenia. Pharmacol Ther. 2008;120(3):317–332. doi: 10.1016/j.pharmthera.2008.08.004. [DOI] [PubMed] [Google Scholar]

RESOURCES