Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jan 1.
Published in final edited form as: Neurobiol Dis. 2013 Sep 27;61:10.1016/j.nbd.2013.09.013. doi: 10.1016/j.nbd.2013.09.013

Development of allosteric modulators of GPCRs for treatment of CNS disorders

Hilary Highfield Nickols 1, P Jeffrey Conn 2
PMCID: PMC3875303  NIHMSID: NIHMS532411  PMID: 24076101

Abstract

The discovery of allosteric modulators of G protein-coupled receptors (GPCRs) provides a promising new strategy with potential for developing novel treatments for a variety of central nervous system (CNS) disorders. Traditional drug discovery efforts targeting GPCRs have focused on developing ligands for orthosteric sites which bind endogenous ligands. Allosteric modulators target a site separate from the orthosteric site to modulate receptor function. These allosteric agents can either potentiate (positive allosteric modulator, PAM) or inhibit (negative allosteric modulator, NAM) the receptor response and often provide much greater subtype selectivity than do orthosteric ligands for the same receptors. Experimental evidence has revealed more nuanced pharmacological modes of action of allosteric modulators, with some PAMs showing allosteric agonism in combination with positive allosteric modulation in response to endogenous ligand (ago-potentiators) as well as “bitopic” ligands that interact with both the allosteric and orthosteric sites. Drugs targeting the allosteric site allow for increased drug selectivity and potentially decreased adverse side effects. Promising evidence has demonstrated potential utility of a number of allosteric modulators of GPCRs in multiple CNS disorders, including neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease, as well as psychiatric or neurobehavioral diseases such as anxiety, schizophrenia, and addiction.

Keywords: Allosteric modulator, CNS, Drug discovery, GPCR, Metabotropic glutamate receptor, Muscarinic acetylcholine receptor

INTRODUCTION

There is an urgent need for development of new therapeutic agents for treatment of debilitating CNS diseases, which affect millions of people worldwide. While discovery and development of new therapeutic agents is challenging for all therapeutic areas, CNS drug discovery efforts have been especially challenging and have a very high attrition rate (Kola and Landis, 2004). The GPCRs have been among the most fruitful targets for developing drugs for treatment of CNS disorders, as well as range of other human disease states. Many current clinical therapeutic agents act by targeting this important receptor class and downstream signaling pathways (Allen and Roth, 2011) (Melancon et al., 2012) (Fang et al., 2003). However, some major subfamilies of GPCRs have proven intractable in drug discovery efforts because of a difficulty in achieving high subtype selectivity and drug-like properties, including high CNS exposure, that are critical for advancing novel agents for treatment of neurological and psychiatric disorders. Historically, drug discovery efforts targeting GPCRs have focused on development of traditional agonists and antagonists that interact with the orthosteric neurotransmitter binding site to either mimic or block the action of the endogenous neurotransmitter or agonist. While this has been fruitful, there are many instances where the high conservation of the orthosteric binding site across related receptors prevents development of subtype selective agents. Also, developing drug candidates based on the chemical scaffolds of the endogenous ligand may raise challenges in establishing appropriate profiles in terms of pharmacokinetic properties or brain exposure.

In recent years, advances in the development of allosteric modulators of GPCRs1 has emerged as a promising new approach for developing therapeutic agents that may be useful for treatment of CNS disorders. Allosteric modulators of GPCRs bind to sites that are separate from the orthosteric binding site of the endogenous ligand and are often less highly conserved than the orthosteric site (Conn et al., 2009a). For some GPCRs, this has allowed optimization of allosteric modulators that achieve much greater subtype selectivity than is possible with traditional orthosteric ligands. In addition, allosteric modulators have other potential advantages, including ability to develop agents that have functional selectivity, allowing for potential targeting of select downstream signaling pathways, and a greater diversity of chemical scaffolds that can facilitate efforts to optimize pharmacokinetic and other drug-like properties of potential drug candidates. The surge in development of allosteric agents has revealed a varied repertoire of drug activities, including PAMs and NAMs as well as agents with combined allosteric agonist and PAM activity and neutral ligands, termed silent allosteric modulators (SAMs) that bind to the allosteric site but do not potentiate or inhibit responses to the endogenous agonist (see (Melancon et al., 2012) (Conn et al., 2009a) and (Niswender and Conn, 2010) for reviews). In addition, allosteric agonists with a bitopic binding mode (binds to both the allosteric site and the orthosteric site) have been identified (Digby et al., 2012a; Lebon et al., 2009; Spalding et al., 2002). These varied modes of action provide tools for experimental investigation into GPCR structure and function. To date, there is a wide variety of allosteric modulators that are showing promise for potential treatment of CNS diseases (Conn et al., 2009d). Among these, some of the most advanced and well understood include allosteric modulators of the metabotropic glutamate (mGlu) receptors and the muscarinic acetylcholine receptors (mAChRs). For instance, allosteric modulators of specific subtypes of mGlu receptors have potential utility in treatment of schizophrenia, autistic spectrum disorders, and Parkinson’s disease (Morin et al., 2013b). Positive allosteric modulators of the M1 and M4 muscarinic receptors show promising applications in both Alzheimer’s disease and schizophrenia. This is an exciting time in CNS drug discovery with several allosteric modulator candidates moving from preclinical models into clinical development.

ALLOSTERIC MODULATORS OF GPCRS

GPCRs, also called seven transmembrane spanning receptors (7TMRs), represent the largest family of cell surface receptors and are the targets of intense drug discovery efforts. While a number of available drugs on the market target GPCR signaling pathways, overall less than 20% of GPCRs are targeted (Allen and Roth, 2011). Ubiquitous receptors, these seven transmembrane-spanning proteins transduce extracellular signals for ligands as diverse as ions, photons of light, odorants and peptides into intracellular signaling cascades. Over 800 human GPCRs have been identified to date with five major families (and multiple subfamilies) based on their amino acid sequences (Katritch et al., 2013). Despite intense drug discovery and development efforts, clinically useful drugs do not exist for the large majority of these receptors. As noted above, the orthosteric binding site within GPCR subfamilies are often highly conserved, making the development of subtype specific ligands difficult. Of the orthosteric ligands developed, many of those with the highest subtype selectivity are antagonists.

Discovery and optimization of novel highly selective allosteric modulators of GPCRs has opened exciting new opportunities for development of highly selective drug candidates for specific GPCR subtypes that were intractable using traditional approaches. While the major advances in discovery of allosteric modulators of GPCRs have only occurred over the past decade, the principle of targeting allosteric sites on neurotransmitter receptors that act as ligand-gated ion channels has a long history and has been highly successful in developing agents for treatment of CNS disorders (Melancon et al., 2012). The classic example of an allosteric modulator is the benzodiazepine class, which are positive allosteric modulators at the GABAA receptors (Mohler et al., 2002) these agents provide effective treatment of anxiety, sleep, and seizure disorders without inducing the adverse side effects that would be observed with direct-acting GABAA receptor agonists. Allosteric modulators have been discovered for GPCRs, enzymes, and other ion channels (Bogoyevitch and Fairlie, 2007; Burford et al., 2013; Conn et al., 2009a; Hogg et al., 2005; Kenakin and Miller, 2010; Lewis et al., 2008; Scott et al., 2009).

As for the ligand-gated ion channels, GPCRs occupied by allosteric ligands can be modulated in a positive or negative manner (Conn et al., 2009a). The interaction of the receptor with an allosteric modulator can result in multiple pharmacological effects: affinity modulation, impacting the association-dissociation rate of the orthosteric ligand; efficacy modulation, affecting orthosteric ligand-induced downstream signaling responses; and agonism/inverse agonism, affecting receptor signaling in a positive or negative manner either in the presence or absence of orthosteric ligand. Ternary complex models that use cooperativity to quantify the allosteric effects are useful conceptually, but have been difficult to fit to experimental data. Using the operational model of allosterism, it is possible of to quantify allosteric drug properties for use in drug development efforts. The advantages of allosteric modulators include: receptor selectivity with subtype selectivity and selective cooperativity at a given subtype as well as receptor activity dependence which maintains spatial and temporal activity dependence of endogenous signaling for those ligands which show efficacy only in the presence of the endogenous orthosteric ligand (Conn et al., 2009a) (Lewis et al., 2008).

Allosteric agents have the potential to show differential effects on downstream signaling pathways, termed functional selectivity (biased agonism, stimulus trafficking). For example, mGlu5 activates both intracellular calcium mobilization and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling in rat cortical astrocytes. An early example of functional selectivity of allosteric modulators of GPCRs came with studies comparing the effects of the mGlu5 PAMs DFB and CPPHA in native rat cortical astrocytes. While both showed similar positive modulatory effects on DHPG-induced intracellular calcium transients, their effects on ERK1/2 signaling differed (Zhang et al., 2005). There are now multiple examples in which allosteric modulators have been identified that have differential effects on coupling of the GPCR to different signaling pathways (Niswender et al., 2010b) (Noetzel et al., 2013) (Sheffler and Conn, 2008) (Maj et al., 2003) (Sachpatzidis et al., 2003) (Digby et al., 2012a; Kenakin, 2010; Mathiesen et al., 2005; Wei et al., 2003). Leveraging the functional selectivity of allosteric modulators of GPCRs provides a potential opportunity to develop agents that selectively target GPCR signaling pathways critical for therapeutic efficacy without modulating signaling pathways that lead to adverse effects.

METABOTROPIC GLUTAMATE RECEPTORS

Metabotropic glutamate receptors (mGlu) represent promising drug targets for a variety of psychiatric and neurodegenerative CNS disorders. Glutamate, the major excitatory neurotransmitter in the CNS, signals through both ionotropic and metabotropic glutamate receptors. Metabotropic glutamate receptors modulate cell excitability and synaptic transmission, as opposed to eliciting fast synaptic responses, which are mediated by ionotropic glutamate receptors. The metabotropic glutamate receptor (mGlu) family, which couple to intracellular second messengers through heterotrimeric G-proteins, includes eight members that serve neuromodulatory roles within the CNS. Members of family 3 (or Class C) GPCRs, mGlu receptors are characterized by a large extracellular “venus flytrap” N-terminal region, which serves as the glutamate (orthosteric) binding site. mGlu receptors are divided into three subgroups according to agonist binding, signaling transduction pathways, and sequence homology. Group I, which includes mGlu1 and mGlu5 receptors, are coupled to Gq/11 and mediate IP3/Ca2+ signal transduction (Abe et al., 1992). Group II (mGlu2,3) and Group III (mGlu4,6,7,8) negatively couple to adenylyl cyclase and other effector systems through Gi/o proteins. While there is abundant sequence homology within receptor subgroups at the orthosteric binding site, allosteric ligands bind to a unique topographically distinct site within the transmembrane domain. The allosteric site contains a higher level of sequence diversity between receptor subtypes as compared to the orthosteric site, allowing for greater subtype selectivity of allosteric ligands (Christopoulos and Kenakin, 2002; Conn et al., 2009a). mGlu receptors are expressed in neurons and glial cells, including astrocytes, oligodendrocytes, and microglia. In neurons, mGlu1 and mGlu5 are expressed postsynaptically, modulating cell excitability and post-synaptic efficacy whereas mGlu2,3,4,7,8, are predominately expressed presynaptically, where they can regulate neurotransmitter release (Conn and Pin, 1997). The CNS therapeutic targets for mGlus that have received the most attention include Parkinson’s disease, Fragile X syndrome/autism spectrum disorders, schizophrenia, cognition, addiction, depression, anxiety and pain.

Parkinson’s disease

Parkinson’s disease (PD) is a chronic neurodegenerative disorder characterized by motor symptoms of rigidity, bradykinesia, tremor, rigidity, postural instability and gait disturbance (Jankovic, 2008). Associated non-motor symptoms include cognitive decline, mood and sleep disturbance (Chaudhuri et al., 2006). The incidence of PD in patient’s older than 55 years is approximately 1% worldwide, creating a substantial disease burden in the aging world population. The pathology observed in PD patients includes progressive degeneration of dopamine neurons in the substantia nigra pars compacta (SNc) with resulting dysfunction of the basal ganglia-thalamocortical motor circuit (Figure 1) (Dickerson and Conn, 2012; Johnson et al., 2009; Wichmann and DeLong, 1996; Zhang et al., 2005). Currently available pharmacological treatments for PD are aimed at dopamine replacement using L-DOPA or dopamine receptor agonists. While initially effective, dopamine-replacement strategies have undesired side effects including dyskinesia and have decreased efficacy over time as the disease progresses (Prashanth et al., 2011). The basal ganglia deep brain nuclei are central to the control of motor function, and the group III mGlus, including mGlu4 (Bradley et al., 1999), are expressed in neurons of different basal ganglia nuclei. Dopamine depletion associated with PD in the nigrostriatal pathway leads to hyperactivity of inhibitory projections from the striatum to the globus pallidus, the first synapse in the basal ganglia “indirect pathway” (Hirsch et al., 2000). This glutamatergic overactivity of the indirect pathway is thought to contribute to the motor dysfunction (Blandini et al., 2000) and DA neuronal loss (Greenamyre and O’Brien, 1991; Przedborski, 2005) in PD patients. Clinical and preclinical studies in PD patients and animal models of PD suggest that decreasing the pathologic overactivity of this indirect pathway may be beneficial in reducing the motor symptoms associated with PD. The Group III mGlus (mGlu4, mGlu7, mGlu8) are expressed presynaptically at different synapses in the basal ganglia circuit and are promising targets for PD (Conn et al., 2005). Of particular relevance, mGlu4 is expressed presynaptically at the striato-pallidal synapse and reduces GABAergic, transmission, which is overactive in PD patients following loss of dopamine neurons. The action of mGlu4 PAMs are hypothesized to act by reducing activity within the indirect pathway (Johnson et al., 2009). Interestingly, the mGlu4 subtype of mGlu receptor is expressed in presynaptic terminals of striato-pallidal projections and activation of mGlu4 with the group III selective agonist L-AP4 decreases transmission at the striato-pallidal synapse by inhibiting neurotransmitter release from presynaptic terminals (Matsui and Kita, 2003) (Valenti et al., 2005) (Valenti et al., 2003b) (Bogenpohl et al., 2013). This effect was absent in mGlu4 knockout mice, confirming a critical role for mGlu4. Additionally, L-AP4 and other mGlu4 receptor agonists reverse motor symptoms in preclinical rodent models of PD (Valenti et al., 2003b) (MacInnes et al., 2004) (Sibille et al., 2007) (Konieczny et al., 2007).

Figure 1.

Figure 1

Basal ganglia circuit. The basal ganglia are a collection of interconnected deep gray subcortical nuclei that are essential in the control of motor function. The major input nucleus of the basal ganglia is the striatum (caudate and putamen). The cerebral cortex sends stimulatory inputs to the striatum, including primary motor cortex. The major output nuclei include the substantia nigra pars reticulata (SNr) and the internal globus pallidus (GPi). Two major parallel pathways project from the striatum to the major output nuclei of the basal ganglia. The direct pathway contains inhibitory projections from the striatum to the output nuclei. GABAergic neurons in the striatum that express D1 dopamine receptors project to the SNr and GPi. The indirect pathway is polysynaptic, and ends in excitatory projections to the output nuclei from the subthalamic nucleus (STN). GABAergic neurons in the striatum that express D2 dopamine receptors project to the external globus pallidus (GPe), and subsequently the GPe sends inhibitory projections to the STN. The result of disruption of the indirect pathway is disinhibition of the STN, and net increased excitatory stimulation to the output nuclei. The balanced activity between the direct pathway inhibition of the output nuclei and the indirect pathway excitation is essential for the normal control of motor activity. The action of dopamine in the striatum decreases transmission through the indirect pathway and increases transmission through the direct pathway. Loss of dopamine neurons in the substantia nigra pars compacta (SNc) in PD leads to decreased inhibitory tone in the direct pathway with subsequent increased stimulatory tone in the indirect pathway, increased GABAergic tone at the thalamus and reduced excitation of the motor cortex. Metabotropic glutamate receptors are expressed throughout the basal ganglia. The Group I receptors (mGlu1 and mGlu5) are expressed in multiple locations in the basal ganglia. They are located post-synaptically, and inhibit the basal ganglia response to dopamine. Group II receptors (mGlu2/3) are located presynaptically to the cortico-striatal, STN-SNr, and STN-SNc synapses. Of the Group III receptors, mGlu4 is known to modulate corticostriatal, STN-SNr, as well as intrastriatal GABAergic synapses. Cholinergic systems modulate basal ganglia function. Of primary importance, tonically active cholinergic interneurons in the striatum release acetylcholine to modulate basal ganglia and motor function. They act project to neighboring medium spiny neurons in the striatum. Muscarinic acetylcholine receptors are expressed in the basal ganglia. M1, M2, M4 receptors are expressed in the striatum, with M5 receptors expressed on dopaminergic neurons in the ventral tegmental area (VTA) and SNc. M4 receptors colocalize with D1 expressing neurons in the striatum. The development of subtype specific allosteric modulators of muscarinic ACh receptors will allow for further characterization of their location and function in the basal ganglia circuit. Adapted from Dickerson and Conn 2012, Johnson et al. 2009, Conn et al. 2005, and Wichmann and DeLong, 1996.

The discovery of the mGlu4-selective PAMs, including PHCCC (Maj et al., 2003) (Marino et al., 2003) and multiple more highly optimized agents (Niswender et al., 2008) (East et al., 2010b; Jones et al., 2012a; Jones et al., 2011) (Jimenez et al., 2012) (Celanire and Campo, 2012) (Bennouar et al., 2013) has provided new tools that have allowed systematic studies aimed at testing the hypothesis that mGlu4 PAMs may have utility in reducing motor symptoms in PD patients (Dickerson and Conn, 2012; Engers et al., 2009; Hopkins et al., 2009; Konieczny et al., 2007). Interestingly, multiple structurally diverse mGlu4-selective PAMs have robust efficacy in reducing motor disability in a range of rodent models, including haloperidol-induced catalepsy and both unilateral and bilateral lesions of DA neurons using 6-OHDA (Bennouar et al., 2013; Engers et al., 2010; Jones et al., 2012a) (Le Poul et al., 2012; MacInnes et al., 2004) (Taylor et al., 2003) (Lopez et al., 2007; Marino et al., 2003). Furthermore, the systemically active mGlu4 PAMs VU0364770 (Jones et al., 2012a) and ADX88178 (Celanire and Campo, 2012) potentiate the antiparkinsonian activity of L-DOPA, showing potential as L-DOPA sparing agents.

In addition to providing symptomatic relief in PD patients, early data suggest that mGlu4 PAMs may also show disease modifying effects by decreasing the degeneration of substantia nigra neurons. Thus, the mGlu4 PAM PHCCC reduces DA cell death in MPTP-treated WT mice, while showing no protective effect in mGlu4 knockout mice (Battaglia et al., 2006). While the precise mechanism of this neuroprotective effect is not clear, it could be mediated by a combination of reduced excitation of DA neurons (Valenti et al., 2003a), anti-inflammatory effects of mGlu4 activation (Fallarino et al., 2010; Taylor et al., 2003), and other potential neuroprotective actions on DA neurons (Battaglia et al., 2006; Bruno et al., 2000). Additionally, the mGlu4 PAM PHCCC as well as the Group III agonist ACPT-1 show a decrease in rodent models of neuropathic pain (Goudet et al., 2008), which may be effective for the non-motor symptoms of PD. Thus, mGlu4 PAMs have potential for both symptomatic and disease-modifying treatment for PD patients. While mGlu4 PAMs have not advanced to clinical testing, multiple pharmaceutical companies and academic/industry partnerships are making exciting progress that may allow direct testing of this hypothesis in clinical studies (Robichaud et al., 2011).

PD patients chronically treated with L-DOPA require increasing doses to maintain efficacy and progressively develop L-DOPA-induced dyskinesia (LID) (Marsden and Parkes, 1977) (Fabbrini et al., 2007; Jenner, 2008; Meissner et al., 2011). Amantadine, an NMDAR antagonist, is currently the only drug to show clinical efficacy in LID, and the adverse effects include cognitive impairment. Although the exact mechanism resulting in LID is not established, with decreased dopamine in the basal ganglia in PD, mGlu5 is hypothesized to be involved in compensatory mechanisms and L-DOPA-induced motor complications. mGlu5 is expressed in the striatum and basal ganglia (Shigemoto et al., 1993), and basal ganglia levels are increased in 6-OHDA lesioned rats (Pellegrino et al., 2007), as well as in parkinsonian primates with LID (Ouattara et al., 2010; Samadi et al., 2008). Preclinical rodent models of PD show efficacy with mGlu5 NAMs to decrease LID (Dekundy et al., 2006; Levandis et al., 2008; Mela et al., 2007; Rylander et al., 2009). Fenobam, an mGlu5 NAM, also decreased LID in both rodent and primate models of PD (Rylander et al., 2010). The addition of MPEP to L-DOPA treatment in parkinsonian MPTP lesioned non-human primates substantially decreased LID both in dyskinetic (Morin et al., 2010) and de novo lesioned animals (Morin et al., 2013a). Additionally, basal ganglia [3H]ABP688 specific binding (mGlu5) was significantly less in primates treated with MPEP combined with L-DOPA compared to L-DOPA treated animals (Morin et al., 2013b). These studies suggest that mGlu5 NAMs may be useful as adjunct treatments to L-DOPA for PD. Currently, the mGlu5 NAMs AFQ056 (Mavoglurant) and ADX48621 (Dipraglurant) (Rylander et al., 2010) are in phase IIa clinical studies for treatment of LID in PD. Other possible therapeutic targets for mGlu5 NAMs in addition to FXS/autism spectrum disorders and LID, include gastroesophageal reflux disease (GERD) (Zerbib et al., 2010) (Keywood et al., 2009), migraine, and anxiety/stress disorders (Swanson et al., 2005).

It is important to note that administration of mGlu5 NAMs may be associated with adverse effects. For instance, the mGlu5 NAM MPEP exacerbates PCP-induced psychotomimetic and cognition impairment in animal models (Brody et al., 2004a) (Campbell et al., 2004) and early clinical studies suggest the possibility that mGlu5 NAMs could have psychotomimetic effects in humans (Friedmann CTH, 1980; Itil TM, 1978; Pecknold et al., 1982a). This may be mediated by inhibition of mGlu5-induced regulation of the NMDA subtype of glutamate receptor (Awad et al., 2000; Doherty et al., 2000; Henry et al., 2002; Kinney et al., 2003; Pisani et al., 2001) and the established psychotomimetic effect of manipulations that inhibit NMDA receptor function (Lahti et al., 1995; Malhotra et al., 1997). Interestingly, most mGlu5 NAMs have inverse agonist activity, which may contribute to this side effect profile (Porter et al., 2005b). However, recent studies have shown that it is possible to develop mGlu5 NAMs with weak negative cooperativity that only partially block glutamate activation of mGlu5 with full occupancy of the receptor (Rodriguez et al., 2005a). While in vivo studies with these partial allosteric antagonists have not been performed, it is possible that these agents could provide clinical efficacy while minimizing adverse effects associated with full blockade or inverse agonist activity at mGlu5.

Of interest, A2A adenosine receptors are also expressed in the striatopallidal neurons and form oligomers with the D2 dopamine receptor. A2A receptor antagonists are pro-dopaminergic, and therefore have the potential to reduce the symptoms associated with dopamine depletion in PD (Kulisevsky and Poyurovsky, 2012). The A2A receptor antagonist preladenant (SCH412384) delays haloperidol–induced extrapyramidal symptom onset in non-human primates (Varty et al., 2008). Therefore, the development of A2A NAMs would provide a valuable tool for the study of dyskinesia associated with PD and movement disorders.

In addition to mGlu4 PAMs and mGlu5 NAMs, the development of mGlu2 and mGlu8 PAMs may be useful for Parkinson’s disease therapy. The Group II mGlus are located presynaptically on glutamatergic axon terminals in the substantia nigra pars reticulata (SNr), potentially modulating excitatory neurotransmission (Bradley et al., 2000). Administration of group II agonists, by either the intracerbroventicular or the intranigral route, results in a reversal of akinesia in reserpine-treated rats (Dawson et al., 2000; Murray et al., 2002). Treatment of rat midbrain slices with the selective agonist LY379268 leads to long-term depression (LTD) of excitatory postsynaptic current (EPSC) amplitude in GABAergic SNr neurons. This effect was absent in mGlu2 but not mGlu3 knockout mice, indicating that activation of mGlu2 is essential for induction of LTD in the SNr, with possible application of mGlu2 agonism for treatment of the motor symptoms of PD (Johnson et al., 2011). Non-selective group III agonists are effective in preclinical PD models. The mGlu8 agonist DCPG (Thomas et al., 2001), administered by intracerebroventricular route, showed robust reversal of prolonged, but not acute, haloperidol-induced catalepsy and reserpine-induced akinesia (Johnson et al., 2013). Further, DCPG administration decreased forelimb use asymmetry in unilateral 6-OHDA lesioned rats. This evidence supports a role for mGlu8 agonism in potential PD treatment. Therefore, the development of mGlu2 and mGlu8 PAMs may provide therapeutic benefit in PD.

Fragile X syndrome and autism spectrum disorders

Fragile X syndrome (FXS) is an X-linked monogenic disorder, and is the most common form of human inherited intellectual disability and inherited cause of autism (Santoro et al., 2012). The brains of patient’s with FXS appear normal on gross examination, yet microscopically the dendritic spines demonstrate an elongate immature phenotype. Patients with FXS have poor motor coordination, tactile hypersensitivity, loose bowel movements, and an increased incidence of epilepsy. These individuals have mental disability that includes attention deficit hyperactivity, obsessive-compulsive behaviors and labile mood. FXS is most often caused by a trinucleotide repeat expansion (CGG) in 5′ untranslated region of the Fragile X mental retardation 1 (FMR1) gene, leading to hypermethylation and vastly decreasing or silencing the expression of the fragile X mental retardation protein (FMRP). FMRP represses translation of specific mRNAs (Bear et al., 2004) and is located in the postsynaptic region of glutamatergic synapses. Interestingly, FMRP inhibits translation of key proteins in the CNS that are stimulated by mGlu1 and mGlu5 (Bhakar et al., 2012). FMRP plays a critical role in long-term depression and other forms of synaptic plasticity. The absence of FMRP expression results in increased constitutive mGlu5 signaling and subsequent “excessive” mGlu5-mediated protein synthesis in post-synaptic dendrites with resulting dysregulation of synaptic function. Generation of Fmr1 knockout mice with decreased mGlu5 expression (50%) showed improvement in many rodent phenotypes associated with Fragile X, including rescue of neuronal spine density, supporting increased activity of mGlu5 as a key component in disease development (Dolen et al., 2007). The mammalian target of rapamycin (mTOR) pathway and ERK pathway are implicated in coupling of mGlu5 to the translational complex (Bhakar et al., 2012a). The mGlu5 NAMs MPEP and fenobam improve fragile X phenotypes in animal models of FXS. Furthermore, chronic pharmacological mGlu5 inhibition with CTEP in the Fmr1 knockout mouse corrected many features of fragile X in adult mice (Michalon et al., 2012). These findings highlight the importance of mGlu5 in FXS, and raise the possibility that constitutive activity of mGlu5 may be important in FXS. Therefore, the inverse agonist activity of mGlu5 NAMs, such as is observed for MPEP, may be important for mGlu5 NAM efficacy in this disease. Further studies comparing compounds with and without inverse agonist activity will determine the importance of mGlu5 constitutive activity in FXS. These findings show promise for chronic mGlu5 NAM treatment of patients with the FXS phenotype. A number of mGlu5 NAMs are now being investigated in clinical studies for efficacy in treatment of FXS as well as other indications (Bhakar et al., 2012a; Emmitte, 2013). In a small clinical trial of 30 fragile X patients, the mGlu5 NAM AFQ056 (Novartis) showed improvement in patients with full methylation of the FMR1 promoter region, demonstrating that epigenetic modification of the promoter may determine responsiveness to mGlu5 NAMs (Jacquemont et al., 2011; van Bon et al., 2011). Using lymphoblastoid cell lines from FXS patients, treatment with AFQ056 did not induce demethylation of the FMR1 gene promoter and levels of FMR1 mRNA remained constant (Tabolacci et al., 2012)

In addition to potential utility in treatment of FXS, recent studies raise the possibility that mGlu5 NAMs may also be useful for treatment of a broader range of autistic spectrum disorders, including idiopathic autism (Silverman et al., 2012). However, preclinical studies in mice bearing mutations that lead to tuberous sclerosis, another developmental autistic spectrum disorder suggest that mGlu5 NAMs could exacerbate symptoms and that mGlu5 PAMs could have therapeutic effects. Interestingly, tuberous sclerosis complex (TSC) patients often have associated symptoms similar to Fragile X patients, including epilepsy, autism spectrum disorders, and mental disability (Tsai and Sahin, 2011). TSC mice treated with an mGlu5 PAM showed reversal of cognitive defects, supporting a potential role for mGlu5 in TSC (Auerbach et al., 2011). Thus, it will be critical to carefully consider specific patient populations and develop a more complete understanding of the potential impact of mGlu5 modulators in different childhood developmental disorders. FXS and TSC serve as valuable models to understand the neurobiology behind genetically complex developmental brain disorders the potential imact of different genotypes on therapeutic response to mGlu5 modulators (Krueger and Bear, 2011).

In addition to potential utility of mGlu5 modulators, it is important to note that the early studies suggest that the signaling by both mGlu5 and the closely related mGlu1, are equally impacted by mutations that lead to FXS (Bhakar et al., 2012b). Thus, mGlu1 NAMs could also provide efficacy in FXS patients. While mGlu1 has received less attention than mGlu5 as a potential target for treatment of FXS, Thomas et al. (Thomas et al., 2012) recently reported that selective mGlu1 NAMs have robust efficacy in reversing multiple symptoms in FXS mice. Thus, it will be important to understand the potential for selective NAMs for both subtypes in treatment of FXS and related disorders.

Schizophrenia and anxiety disorders

Schizophrenia is a debilitating psychiatric disorder affecting approximately 1% of the population across the globe. The manifestation of this disorder includes a triad of symptom clusters: positive symptoms, negative symptoms, and cognitive impairment (Kim et al., 2009). Current therapies for the treatment of psychosis (positive symptoms) in schizophrenia patients focus on blockade of the D2 dopamine receptors, and are severely limited by poor efficacy as well as adverse side effects (extrapyramidal motor symptoms and metabolic syndrome and sexual dysfunction). These combined effects limit the successful therapeutic window for the D2 antagonists, with patients frequently switching drugs to maintain effective treatment of symptoms. Additionally, some D2 antagonists only show partial efficacy in some patients. The finding that NMDA receptor channel blockers, including PCP, ketamine, and dizocilpine (MK-801), induce psychosis in human volunteers, led to the hypoglutamatergic theory of schizophrenia (Javitt, 1987). In addition, the administration of NMDA receptor antagonists to schizophrenic patients exacerbates both cognitive and psychotic symptoms (Lahti et al., 1995; Malhotra et al., 1997). Clinical trials using glycine co-agonists, which enhance NMDA receptor function, in combination with standard antipsychotic therapy show efficacy in decreasing negative symptoms and increasing cognition in schizophrenic patients (Coyle and Tsai, 2004).

There is a large body of evidence that Group II (mGlu2 and mGlu 3) agonists or mGlu2 PAMs provide effective action against the positive symptoms, while mGlu5 PAMs may have efficacy in reducing all symptoms clusters schizophrenia patients (Herman et al., 2012). The mGlu2/3 agonists LY354740 and LY379268 have robust efficacy in multiple rodent models of antipsychotic-like activity (Chaki et al., 2013) for both schizophrenia (Conn et al., 2008; Schoepp and Marek, 2002) and anxiolytic disorders (Schoepp et al., 2003; Swanson et al., 2005). The antipsychotic-like activity is likely to be mediated, at least in part by reduced glutamate release from presynaptic terminals on projections from the thalamus to the prefrontal cortex in rodents (Cartmell et al., 1999a; Moghaddam, 2004) (Marek, 2010). Further, an early clinical study with a selective mGlu2/3 agonist showed promising effects in a phase II clinical trial in patients with schizophrenia (Patil et al., 2007). Unfortunately, this efficacy has not been reliably observed in subsequent clinical studies (Kinon et al., 2011) (Hopkins, 2013) (Adams et al., 2013). Experiments using mGlu2 and mGlu3 knockout mice provide strong evidence that the antipsychotic-like effects of mGlu2/3 agonists in rodent models are mediated by activation of mGlu2 (Fell et al., 2008). However, it has not been possible to develop orthosteric agonists that are highly selective for mGlu2 relative to mGlu3.

Efforts to develop selective mGlu2 PAMs have been highly successful and multiple studies reveal that mGlu2-selective PAMs have robust efficacy in rodent models of antipsychotic activity that are similar to those observed with mGlu2/3 agonists (Cartmell et al., 1999b; Galici et al., 2006; Lorrain et al., 2003; Moghaddam and Adams, 1998) (Benneyworth et al., 2007). ADX71149, a highly selective mGlu2 PAM that is now in clinical development for treatment of schizophrenia and anxious depression, met the primary objectives of safety and tolerability in phase I studies and advanced to phase IIa clinical testing. While the results of this important study have not yet been published, preliminary reports from part B of the Phase IIa study suggest that ADX71149 may have efficacy in reducing negative symptoms in a subgroup of schizophrenia patients (http://www.addextherapeutics.com/investors/press-releases/news-details/article/addex-reports-top-line-data-from-a-successful-phase-2a-clinical-study-with-adx71149-in-schizophrenia/).

Activation of mGlu4 with positive allosteric modulators may also provide a promising therapeutic avenue for schizophrenia and the development of antipsychotic agents. The Group III mGlu4-preferring orthosteric agonist LSP1-2111 showed dose-dependent inhibition of amphetamine-induced hyperactivity (Wieronska et al., 2012) and reversal of MK-801 induced deficits in novel object recognition in rats (Wieronska et al., 2013), indicating potential effects on both the positive and cognitive symptoms of schizophrenia. Further, recent studies with mGlu4 PAMs show promising results in animal models that are used to predict antipsychotic effects (Slawinska et al., 2013a; Slawinska et al., 2013b). For instance, the brain-penetrant mGlu4 PAMs Lu AF21934 and Lu AF32615 showed dose-dependent reduction of amphetamine-induced hyperactivity and antagonism of 2,5-dimethoxy-4-iodoamphetamine (DOI)-induced head twitch tests in wild type but not mGlu4 −/− mice, supporting a key role for mGlu4 in brain circuits involved in these behavioral models (Slawinska et al., 2013a). The mechanism of action for mGlu4 in rodent models of antipsychotic activity are not known but are likely to be unrelated to the antiparkinsonian effects of mGlu4 PAMs. Recent studies suggest that antipsychotic actions of mGlu4 PAMs may parallel the effects of mGlu2/3 receptor agonists and mGlu2 PAMs. For instance, activation of mGlu4 reduces transmission at the same thalamo-cortical terminals in the prefrontal cortex that are modulated by activation of mGlu2 (Zhang and Marek, 2007). In addition, activation of mGlu4 reduces excitatory transmission in midbrain dopamine neurons (Valenti et al., 2005) and this could contribute to the antipsychotic-like effects of mGlu4 agonists or PAMs.

In addition to mGlu2-selective PAMs, recent efforts suggest that mGlu5-selective PAMs may have potential as a novel approach for treatment of schizophrenia. Activation of mGlu5 receptors is known to enhance NMDA receptor function in multiple cell populations and has excitatory effects that may work in concert with NMDA receptors to increase activity in forebrain circuits that are thought to be important for the psychotomimetic effects of NMDA receptor antagonists (Brody et al., 2004b; Kinney et al., 2003; Lecourtier et al., 2007). Additionally, mGlu5 receptors interact physically with NMDA receptors through intracellular scaffolding proteins (Niswender and Conn, 2010). As noted above, antagonists of NMDA receptors induce positive and negative symptoms, as well as deficits in cognitive function in humans that are similar to those observed in schizophrenia patients (Adler et al., 1998; Halberstadt, 1995; Krystal et al., 1994; Malhotra et al., 1996; Newcomer et al., 1999; Parwani et al., 2005; Rowland, 2005). A large body of clinical and preclinical studies have led to the hypothesis that reduced activity of NMDA subtypes of glutamate receptors or brain circuits that are regulated by NMDA receptors may play an important role in the pathophysiology underlying schizophrenia (Conn et al., 2009d; Field et al., 2011; Nicoletti et al., 2011). Furthermore, multiple studies suggest that agents that enhancing NMDA receptor signaling could provide efficacy in reducing the symptoms associated with schizophrenia (Coyle, 2006; Lindsley et al., 2006). Based on this, and the clear role of mGlu5 acting in tandem with NMDA receptors to regulate transmission through glutamatergic circuits in forebrain regions, selective activators of mGlu5 have been raised as a potential novel treatment strategy for schizophrenia that may have efficacy in reducing both psychotic and negative symptoms as well as providing pro-cognitive activity (Conn et al., 2009c). Consistent with this, mGlu5 KO mice show disrupted prepulse inhibition (PPI), a model of sensory motor gating shown to be disrupted in schizophrenic patients (Brody et al., 2004a) (Kinney et al., 2003). In addition, the mGlu5 NAM MPEP potentiates PCP-induced psychotomimetic and cognition impairment in animal models (Brody et al., 2004a) (Campbell et al., 2004). Based on these important findings, major efforts were launched to develop highly selective mGlu5 PAMs. These efforts have yielded a large number of structurally diverse highly selective mGlu5 PAMs and multiple studies have demonstrated that mGlu5-selective PAMs have robust efficacy in rodent models of schizophrenia that predict efficacy in reducing both positive and negative symptoms (Conn et al., 2009d; Liu et al., 2008b; Schlumberger et al., 2010; Schlumberger et al., 2009). In addition, mGlu5 PAMs potentiate multiple forms of synaptic plasticity that are thought to underlie specific aspects of learning and memory (Ayala et al., 2009; Liu et al., 2008a; Rosenbrock et al., 2010; Stefani and Moghaddam, 2010a), and improve cognitive function in multiple animal models (Darrah et al., 2008; Stefani and Moghaddam, 2010b; Vardigan et al., 2010). These exciting studies suggest that mGlu5 PAMs have the potential to provide a fundamental advance in schizophrenia therapy that could have efficacy in treatment of all major symptom clusters of the disease.

While the more subtle approach to modulation of glutamatergic function using mGlu5 PAMs has the potential to provide less adverse effect liability than would be observed with direct agonists of mGlu5 or NMDA receptors, recent studies reveal that some mGlu5 PAMs induce severe seizure activity (Rook et al., 2012) and excitotoxicity leading to cell death in the auditory cortex, hippocampus, and other forebrain regions (Parmentier-Batteur et al., 2013). However, other mGlu5 PAMs are well tolerated and it is clear that all mGlu5 PAMs do not have the same adverse effect liability (Rook et al., 2013b). This suggests that mGlu5 PAMs likely differ in their effects on mGlu5 signaling and understanding these differences and the mechanistic underpinnings of mGlu5 PAM-mediated toxicity will be critical for fully developing mGlu5 PAMs as potential therapeutic agents. One property that has been shown to play an important role in determining whether specific mGlu5 PAMs will induce seizures and behavioral convulsions is the presence or absence of allosteric agonist activity (Rook et al., 2013b). Of note, allosteric agonism is context dependent and can be influenced by differences in receptor expression. For example, Rook et al (2013) recently reported that the mGlu5 PAM VU0424465 showed intrinsic agonist activity both in cell lines and in native systems, while a group of closely related compounds displayed agonist activity only in overexpressing cell lines and others showed no detectable allosteric agonist activity in any cell lines or native systems examined. Interestingly, VU0424465 showed adverse effects, including epileptiform activity, while the related compounds that were devoid of allosteric agonist activity in native systems did not (Rook et al., 2013a). These adverse effects are dose-dependent and showed increased severity over time. These findings highlight the importance of selecting mGlu5 PAMs lacking detectable intrinsic (glutamate-independent) agonist activity to avoid glutamate-independent receptor activity and the associated side effects. Also, as discussed above, mGlu5 PAMs and other GPCR allosteric modulators can regulate specific aspects of mGlu5 signaling without affecting others (Noetzel et al., 2013; Zhang et al., 2005). It is possible that some mGlu5 PAMs have greater effects on signaling pathways that are involved in the adverse effect liability and that those that are biased towards other pathways could provide efficacy without the severe adverse effect liability. In addition to these different properties, recent studies suggest that hepatic metabolism of some mGlu5 PAMs can yield metabolites that have robust activities that differ from activity of the parent compound and can contribute to the adverse effect liability (Bridges et al., 2013). In future studies, it will be critical to gain a clear understanding of the mechanisms underlying the different safety profiles of different mGlu5 PAMs. Interestingly, in a developmental model of schizophrenia (neonatal PCP-induced cognition impairment), administration of mGlu5 PAMs in adolescence prevented the appearance of delayed cognitive deficits in adult rats. Further, mGlu5 PAM administration reversed the delay-induced impairment in adult rats, as evaluated by social novelty discrimination (Clifton et al., 2013). These findings suggest the exciting possibility of a preventative role for mGlu5 PAM treatment in the development of schizophrenia, and further work will be important to evaluate these findings.

Addiction

Glutamatergic neurotransmission is hypothesized to play a key role in both establishment and maintenance of drug addiction (Nicoletti et al., 2011). Negative allosteric modulators of Group I (mGlu1 and mGlu5) mGlu receptors have potential as therapeutic agents for treatment of addictive disorders (Achat-Mendes et al., 2012; Bird and Lawrence, 2009). The mGlu5 receptors are highly expressed in the mesolimbic areas, regions central to the brain reward system. Mutant mGlu5 null mice do not self-administer cocaine or exhibit locomotor-stimulating effects, despite normal levels of dopamine in the nucleus accumbens, supporting a role for mGlu5 in addiction (Chiamulera et al., 2001). MPEP and MTEP treatment (mGlu5 NAMs) show efficacy in cocaine abuse rodent and non-human primate models (Kenny et al., 2005; Kumaresan et al., 2009; Lee et al., 2005; Platt et al., 2008). Additionally, the novel mGlu5 NAM VU0463841 shows activity in a rat model of cocaine addiction, with dose-dependent reduction of cocaine place preference and cocaine self-administration (Amato et al., 2013). Recently, mGlu1 antagonism (mGlu1 antagonist JNJ16259685 ) was reported to inhibit cocaine-induced conditioned place preference (CPP) through inhibition of protein synthesis in the ventral tegemental areas (VTA) (Yu et al., 2013).

In addition to Group I antagonists, experimental evidence suggests a potential role for mGlu2 in addictive disorders. Cocaine reinforcement is elevated in mGlu2 knockout mice, supporting the concept that mGlu2 negatively regulates the drug reward system (Morishima et al., 2005). mGlu2/3 agonism is associated with decreased nicotine and cocaine self-administration (Adewale et al., 2006; Liechti and Markou, 2007), providing opportunity for the development of mGlu2/3 (likely mGlu2-selective) PAMs for addiction.

Major Depressive Disorder (MDD)

Major depressive disorder (MDD) represents one of the most common forms of mental illness and is a significant financial and social burden (Chaki et al., 2013). Monoamine oxidase inhibitors and tricyclic antidepressants are the current mainstays of therapy for depression and show slow onset of action as well as poor efficacy. A subgroup of patients are resistant to therapy, termed treatment resistant depression (TRD). Ketamine, a noncompetitive (orthosteric) NMDA receptor antagonist, has shown considerable efficacy for treatment-resistant depression in a double blind placebo controlled trial (Zarate et al., 2006). Additional evidence of the role of hyperfunction of the glutamatergic system in depression stems from the observation that glutamate levels in plasma and limbic brains areas are elevated in depressed patients (Sanacora et al., 2004). Modulation of glutamatergic transmission, particularly with mGlu2/3 agonists (Feinberg et al., 2002; Fell et al., 2011), mGlu2/3 antagonists (Campo et al., 2011b; Chaki et al., 2004; Palucha-Poniewiera et al., 2010), and mGlu5 antagonists (Belozertseva et al., 2007; Li et al., 2006; Palucha et al., 2005) (Campo et al., 2011a; Pilc et al., 2013) (Chaki et al., 2013) show promising effects as potential treatments for depression. Recently, the novel mGlu5 NAM GRN-529 showed efficacy in multiple rodent models of depression, including those relevant to anxiety and pain, symptoms often associated with treatment resistant depression (Hughes et al., 2013). Furthermore, fenobam had efficacy in reducing anxiety in a clinical proof-of-concept study (Pecknold et al., 1982b; Porter et al., 2005a). Currently, the mGlu5 antagonist RG7090 is in a phase II study for adjunctive therapy in patients with major depressive disorder (MDD) (www.clinicaltrials.gov).

Additional CNS therapeutic areas for mGlu allosteric modulators

The allosteric modulators of mGlu receptors have many other potential uses including pain, stress/anxiety disorders, and movement disorders (see Table 1) (Nicoletti et al., 2011). Neuropathic pain is a chronic condition that leads to allodynia and hyperalgesia (Schkeryantz et al., 2007). The mGlu1 receptor is expressed in CNS regions essential to nociceptive processing as well as in afferent nociceptive nerve terminals (Martin et al., 1992). mGlu1 knockout mice show decreased pain sensitivity (Schkeryantz et al., 2007) and administration of the mGlu1-selective NAM JNJ16259685 (Lavreysen et al., 2004) is reported to show efficacy in a rodent neuropathic pain model (formalin hyperalgesia) (Mabire et al., 2005). Additionally, mGlu1-selective antagonists showed in vivo activity in the spinal nerve ligation test (Bennett et al., 2012). Taken together, the development of mGlu1-selective NAMs (Annoura H, 1996; Mabire et al., 2005) and efficacy in preclinical models are promising for the potential treatment of neuropathic pain. mGlu5 NAMs show analgesic efficacy in preclinical models of neuropathic pain (Kumar et al., 2010; Montana et al., 2009). Agonists of mGlu 2/3 receptors also show analgesic effects in models of chronic and neuropathic pain, but show tolerance after chronic treatment (Jones et al., 2005).

Table 1.

Potential application of metabotropic (mGlu) and muscarinic (mACh) allosteric modulators in CNS diseases

Receptor MOA CNS disease applications Compounds
mGlu1 NAM Neuropathic pain, FXS, Anxiety/Stress disorders, Addiction CPCCOEt (Annoura H 1996), JNJ16259685 (Lavreysen, Pereira et al. 2004; Thomas, Bui et al. 2012)
PAM Ro67-7476 (Knoflach, Mutel et al. 2001), VU71(Hemstapat, de Paulis et al. 2006), Ro67-4853 (Wichmann, Bleicher et al. 2002), VU48 (Hemstapat, de Paulis et al. 2006)
mGlu5 NAM Addiction, Anxiety, Chronic pain, Depression, FXS (autism spectrum disorders), Migraine, PD-LID, MPEP (Gasparini, Lingenhohl et al. 1999), MTEP (Cosford, Tehrani et al. 2003), CTEP (Lindemann, Jaeschke et al. 2011), Fenobam (Porter, Jaeschke et al. 2005), AFQ056 (Jacquemont, Curie et al. 2011); M-5MPEP (partial NAM), Br-5MPEPy (partial NAM)(Rodriguez, Nong et al. 2005); GRN-529 (Hughes, Neal et al. 2013), VU046381 (Amato, Felts et al. 2013), RG7090 (RO4917523, antagonist, in clinical trials, FXS, http://clinicaltrials.gov/ct2/results?term=RO4917523)
PAM Anxiety disorders, Huntington’s disease, Schizophrenia, TSC ADX47273 (Liu, Grauer et al. 2008),
VU0360172 (Rodriguez, Grier et al. 2010), VU29(Ayala, Chen et al. 2009), LSN2463359 (Gastambide, Gilmour et al. 2013; Gilmour, Broad et al. 2013)
mGlu2/3 NAM Depression RO4432717 (Goeldner, Ballard et al. 2013), MNI-137 (Hemstapat, Da Costa et al. 2007)
mGlu2-selective PAM Addiction, AD, Anxiety disorders, Depression, Schizophrenia BINA (Galici, Jones et al. 2006; Benneyworth, Xiang et al. 2007), LY487379 (Johnson, Baez et al. 2003), ADX71149 (Hashimoto, Malchow et al. 2013)
NAM Depression MNI-167, RO4988546, RO5488608, RO4491533, RO4491533
mGlu3-selective NAM Depression VU0463597/ML-289 (Sheffler, Wenthur et al. 2012)
mGlu4 PAM Neuroinflammation, Neuroprotection, PD, Schizophrenia PHCCC (Maj, Bruno et al. 2003; Marino, Williams et al. 2003), VU0155041 (PAM/allosteric agonist) (Niswender, Johnson et al. 2008), VU0364770 (Jones, Bubser et al. 2012), ADX88178 (Celanire and Campo 2012), Lu AF21934 (Slawinska, Wieronska et al. 2013; Slawinska, Wieronska et al. 2013), Lu AF32615 (East, Bamford et al. 2010)
mGlu7 NAM Anxiety, Depression MMPIP (Hikichi, Murai et al. 2010; Niswender, Johnson et al. 2010) and ADX71743 (Kalinichev, Rouillier et al. 2013)
Allosteric Agonist Anxiety, Depression, PD AMN082 (Ugolini, Large et al. 2008)
mGlu8 agonist Parkinson’s disease, Anxiety DCPG (Thomas, Wright et al. 2001)
M1 PAM AD, Addiction, Movement disorders, Neuropathic pain, PD, Schizophrenia Brucine (Lazareno, Birdsall et al. 1999), BQCA (Shirey, Brady et al. 2009), PQCA (Uslaner, Eddins et al. 2013), ML-137 (Bridges, Lewis et al. 2010), ML-169/ VU0405652 (Bridges, Reid et al. 2010)
Allosteric agonist AD, Movement disorders AC-42 (Spalding, Trotter et al. 2002), N-desmethylclozapine (Sur, Mallorga et al. 2003), TBPB (Jones, Brady et al. 2008), 77-LH-28-1(Langmead, Austin et al. 2008), VU0184670, VU0357017 (Lebois, Bridges et al. 2010; Digby, Noetzel et al. 2012), VU0364572 (Digby, Utley et al. 2012)
M4 M4-selective Antagonist Dystonia, PD Tropicamide (Betz, McLaughlin et al. 2007)
PAM AD, Addiction, Movement disorders, Neuropathic pain, OCD, PD, Schizophrenia Thiochrome (Lazareno, Dolezal et al. 2004), VU152099 (Brady, Jones et al. 2008), VU152100 (Brady, Jones et al. 2008), LY2033928 (Chan, McKinzie et al. 2008), ML173 (Bridges, Niswender et al. 2010), ML293 (Sheffler, Wenthur et al. 2012)
M5 NAM Addiction, Anxiety disorders, Schizophrenia N/A
PAM Anxiety disorders, ADHA, PD, Schizophrenia, VU0238429 (Bridges, Marlo et al. 2009) (Bridges, LeBois et al. 2010)

In addition to mGlu4, subtype-selective allosteric modulators for the other Group III mGlu receptors (mGlu7 and mGlu8) may also have potential for treatment of CNS disorders. Recently, the mGlu7 NAMs MMPIP (Hikichi et al., 2010) and ADX71743 (Kalinichev et al., 2013) have shown potential efficacy in animal models for treatment of anxiety and depression, and provide excellent tool compounds to further elucidate the role of mGlu7 in CNS diseases. As described above, the mGlu8 agonist DCPG (Thomas et al., 2001) showed efficacy in preclinical models of PD (Johnson et al., 2013), and the development of mGlu8 selective PAMs may provide novel therapeutics for PD patients. Recently, mGlu5 PAMs showed neuroprotective effects in the BACHD mouse model of Huntington’s disease (Doria et al., 2013). The continued development of mGlu allosteric modulators provides promising tools for the investigation of the role of individual receptor subtypes in CNS disease and the creation of novel therapeutics for an array of CNS disorders.

MUSCARINIC ACETYLCHOLINE RECEPTORS

Allosteric modulators of muscarinic receptors show promise as potential therapies for a number of CNS disorders, including Alzheimer’s disease and schizophrenia. Other target areas of benefit may include neuropathic and chronic pain, epilepsy, sleep disorders, Parkinson’s disease, and movement disorders (Conn et al., 2009b). Acetylcholine signals through both muscarinic and nicotinic receptors. Muscarinic acetylcholine receptors are widely expressed in the CNS and include five subtypes (M1-M5)(see (Langmead et al., 2008c) for review). These Family A GPCRs respond to the endogenous agonist acetylcholine and are further subdivided into two groups based on signaling pathways. The M1, M3, and M5 receptors signal through Gq/11, activating PLCβ leading to increased intracellular calcium. The M2 and M4 receptors signal through Gi/o proteins and inhibit adenylate cyclase. Also, M2 and M4 signaling through Giβγ subunits modulates ion channel activity. Muscarinic acetylcholine receptors (mAChRs) are expressed throughout the CNS, including targets for cholinergic interneurons in the striatum as well as targets of projections from the medial septum and hindbrain nuclei in the, midbrain, neocortex and limbic areas important for learning and memory. Both the M1 and M4 receptors are associated with learning, memory and cognition (Hasselmo, 2006; Hasselmo and Giocomo, 2006), and drugs specific for these subtypes may be useful in treatment of the cognitive symptoms associated with schizophrenia and Alzheimer’s disease. The orthosteric acetylcholine (ACh) binding site is highly conserved, thwarting attempts to create an orthosteric drug with true subtype selectivity. Drugs developed for the orthosteric site lack subtype specificity and result in dose-dependent adverse side effects (bradycardia, sweating, salivation and gastrointestinal distress) from activation of the peripheral M2 and M3 mAChR subtypes (Heinrich et al., 2009).

Alzheimer’s disease

Alzheimer’s disease represents a major public health problem as the world population ages in the coming decades. The features of this progressive neurodegenerative disease include neuronal loss, behavioral and cognitive changes and decreased cerebral blood flow (Bartus et al., 1982; Hanyu et al., 2003). Acetylcholine receptor agonists and acetylcholinesterase (AChE) inhibitors improve the cognitive symptoms in Alzheimer disease patients (Feldman, 2002; Grossberg, 2002) and AChE inhibitors are among the few medications available for treatment of AD. These cholinergic agents have limited efficacy and induce peripheral side effects, which limit their use (Lockhart et al., 2009). Studies have suggested that activation of muscarinic acetylcholine receptors may prove useful in the treatment of multiple symptoms in the spectrum of both Alzheimer’s disease and schizophrenia (Langmead et al., 2008b). The muscarinic receptor orthosteric agonist xanomaline (M1 and M4 selective) has been shown to be effective in reducing the psychotic symptoms in patients with both schizophrenia and AD. A phase III placebo-controlled clinical trial showed that xanomeline robustly decreased psychotic symptoms in patients with Alzheimer’s disease (Bodick et al., 1997a; Bodick et al., 1997b). While improved cognition was observed for those patients who completed the trial (high dose vs. placebo), the end-point analysis was not statistically significant for cognitive improvement. The main limiting side effect of xanomeline involved gastrointestinal associated symptoms (Bodick et al., 1997b). This study demonstrates that muscarinic agonists have potential in treating both the cognitive and behavioral aspects of Alzheimer’s disease.

Both M1 and M4 allosteric agonists and PAMs would be desirable drug candidates for therapeutic development. The M1 receptor is expressed in high levels in the CNS, particularly the cortex, hippocampus, and striatum (Levey et al., 1995; Levey et al., 1991). Efforts have focused on the development of M1 specific ligands for AD, as M1 is thought to be the critical subtype for cognition, attention, and sensory processing (Fisher, 2008; Langmead et al., 2008b; Robinson et al., 2011). Additionally, M1 knockout mice show specific cognitive deficits (Anagnostaras et al., 2003; Miyakawa et al., 2001). For M1 selective orthosteric agonists, the lack of true selectivity for M1, coupled with high receptor reserve of M2 and M4 in native tissues, results in functional activity at multiple receptor subtypes, with unacceptable side effect profiles. For example, the orthosteric agonist AF267, thought to have subtype specificity for M1, showed activity at both M3 and M5 receptors (Jones et al., 2008). Current M1/M4 selective muscarinic orthosteric ligands show intolerable side effects related to their lack of muscarinic receptor subtype selectivity. The development of muscarinic allosteric modulators has opened a therapeutic window for treating CNS disorders with the potential to minimize peripheral muscarinic side effects.

Brucine, the first M1 PAM identified, provided proof-of-concept for the development of M1 subtype specific ligands (Lazareno et al., 1998). A functional screening approach identified novel M1 PAMs, including VU0090157 and VU0029767, that showed pure PAM activity (Marlo et al., 2009) and could differentially regulate coupling to different signaling pathways. The systemically active M1 PAM BQCA induces an increase in activation of the prefrontal cortex (PFC) and improves in PFC-dependent cognitive function in a transgenic mouse model of PD (Shirey et al., 2009). In addition, BQCA has activity in other models that suggest possible efficacy in improving cognitive function and reducing psychotic symptoms (Ma et al., 2009). Recently, a newer M1 PAM, PQCA was shown to improve cognitive function in non-human primates, including improvements in the object retrieval detour task in rhesus macaques. Further, PQCA treatment increased frontal cortical cerebral blood flow at parallel drug concentrations, providing a potential translational biomarker for future studies (Uslaner et al., 2013). The continued development and optimization of M1 PAMs, such as ML137, is providing exciting tools for investigative efforts into the role of M1 in CNS disorders (Melancon et al., 2013; Poslusney et al., 2013).

Additionally, M1 selective allosteric agonists have been developed, including AC-42 (Spalding et al., 2002), N-desmethylclozapine (Sur et al., 2003), TBPB (Jones et al., 2008) and 77-LH-28-1 (Langmead et al., 2008a). The M1 allosteric agonist AC260584 shows antipsychotic activity in preclinical models (Vanover et al., 2008), but also is active at the 5-HT2A serotonin receptor, D2 dopamine receptor and α1A adrenergic receptor (Heinrich et al., 2009). The recently developed selective allosteric agonists VU0357017 and VU0364572 showed robust efficacy in a rodent hippocampal dependent learning paradigm (Lebois et al., 2010). A number of M1 allosteric agonists show bitopic binding (binds to both an allosteric site and the orthosteric site), including 77-LH-28-1 (Lebon et al., 2009), AC-42 (Spalding et al., 2002), and VU0364572 (Digby et al., 2012b). These allosteric agonists provided valuable tools for the investigation of M1 in AD, but are limited by activity at other GPCRs and show complicated pharmacology with their bitopic mode of action. Furthermore, selectivity of these compounds is based on functional selectivity and issues with high receptor reserve have made it difficult to maintain high selectivity while optimizing for maximal M1 potency and efficacy while achieving drug-like properties of highly selective M1 allosteric agonists (Digby et al., 2012b).

In addition to acute actions on cognitive function, activation of M1 reduces the pathological processing of amyloid precursor protein (APP) associated with Alzheimer’s disease. TBPB, a systemically active M1 allosteric agonist, shows decreased production of β APP (Jones et al., 2008) and the M1 agonist AF267B shifts APP towards the non-amyloidogenic pathway (Caccamo et al., 2006) (Jones et al., 2008). Additionally, M1 agonists show evidence of decreasing CSF Aβ42 levels in AD patients (Fisher, 2008; Heinrich et al., 2009). Therefore, drugs that activate AChRs may have disease modifying properties in AD as well as improve cognitive function.

Schizophrenia

The finding that the M1/ M4 preferring orthosteric agonist xanomeline has antipsychotic-like effects in AD patients raises the question of whether M1 and/or M4 activation could also have efficacy in reducing psychotic symptoms in patients suffering from schizophrenia. The surprising finding of antipsychotic efficacy of xanomeline in AD patients prompted a Phase II clinical trial to evaluate antipsychotic efficacy of xanomeline in schizophrenia patients (Shekhar et al., 2008). Interestingly, xanomeline improved positive, negative, and cognitive symptoms in patients suffering from schizophrenia (Shekhar et al., 2008). This finding is especially interesting in light of previous studies showing that muscarinic receptor antagonists worsen symptoms in schizophrenic patients (Tandon et al., 1991) and produce psychotic symptoms in some individuals not suffering from schizophrenia or related disorders (Osterholm and Camoriano, 1982). Furthermore, analysis of postmortem brain samples from schizophrenic patients show decreased levels of M1/ M4 receptor binding in key brain regions implicated in the disease, including the prefrontal cortex, superior temporal gyrus, hippocampus, and dorsal striatum (Zavitsanou et al., 2004) (Crook et al., 2000; Dean et al., 2002; Deng and Huang, 2005). Taken together, these data raise the exciting possibility that M1 and/or M4 PAMs may also have potential utility in treatment of schizophrenia.

The M4 receptor is expressed in numerous areas of the brain including the cortex, striatum, and hippocampus (Levey et al., 1995). M4 colocalizes with the D1 receptor in the striatum (Ince et al., 1997), suggesting a balance between cholinergic and dopaminergic neurotransmission. Mutant mice with knockout of M4 in D1 dopamine expressing cells (D1-M4-KO) do not show an antipsychotic response to xanomeline in rodent models of schizophrenia, highlighting the potential importance of M4 receptors in schizophrenia (Dencker et al., 2011). Xanomeline treatment attenuates amphetamine-induced hyperactivity in M1 knockout mice, and completely inhibits the effects of amphetamine in M4 knockout mice, suggesting a greater role for M4 in this process (Woolley et al., 2009). Development of the initial M4 PAM thiochrome showed proof-of-concept for designing compounds with M4 subtype selectivity (Lazareno et al., 2004). The discovery of a highly selective M4 PAM, VU0010010 (Shirey et al., 2008) and subsequent development of centrally active M4 PAMs (VU0152099, VU0152100, and VU0448088) demonstrated reversal of amphetamine-induced hyperlocomotor activity in rats (Brady et al., 2008; Le et al., 2013). The M4 PAM LY2033298 also showed effect in rodent models of antipsychotic efficacy, including conditioned avoidance responding and prepulse inhibition (Chan et al., 2008). LY2033298 potentiated of the effect of oxotremorine (nonselective muscarinic agonist)-mediated inhibition of conditioned avoidance responding, indicative of antipsychotic properties (Leach et al., 2010). The effect was reduced in M4 knockout mice, supporting a potential role for M4 in models of schizophrenia. Interestingly, LY2033298 can have allosteric agonist activity under specific conditions as well (Chan et al., 2008; Nawaratne et al., 2010). These data provide strong support for the hypothesis that the antipsychotic effects of xanomeline are in part mediated by activation of M4 and that highly selective M4 PAMs may provide a novel approach to development of antipsychotic agents. In addition to potential efficacy in patients suffering from schizophrenia, M4 PAMs could also provide efficacy in reducing the psychotic symptoms observed in patients suffering from AD and other neurodegenerative disorders.

Based on the discussion of M1 PAM actions in models of AD above, it is possible that M1 PAMs could also provide efficacy in improving cognitive function in schizophrenia patients. Interestingly, schizophrenic patients with an M1 genetic polymorphism (CHRM1) had more correct responses with less perseverative errors in the Wisconsin Card Sorting Test (Liao et al., 2003). In total, the data generated thus far favors a prominent role of M4 in psychotic symptoms and M1 as a major contributor to specific domains of cognitive function. However, further studies are needed to develop a full understanding of the respective roles of M1 and M4 in the clinical efficacy of xanomeline and it is likely that M1 activity also contributes to the antipsycotic efficacy and M4 to the cognition-enhancing effects. The development of highly selective positive allosteric modulators for M1 and M4 receptors provides the tools needed to develop this understanding. Furthermore, M1 and M4 PAMs provide exciting new therapeutic opportunities to achieve subtype specificity with minimal peripheral muscarinic side effects and shows promise for the identification of novel therapeutics for both Alzheimer’s disease and schizophrenia. Multiple companies are now focusing effort on discovery and development of selective M1 PAMs for treatment of schizophrenia and Alzheimer’s disease. The Vanderbilt Center for Neuroscience Drug Discovery has now partnered with Astrazeneca to advance M4 PAMs into clinical development (Jones et al., 2012b). Hopefully, these and efforts by other companies will provide clinical assessment of the potential utility of M1 and M4 PAMs in AD and schizophrenia in the coming years.

M1/M4 agonists also have potential therapeutic applications in addiction and chronic neuropathic pain. The muscarinic receptors in the brain play a key role in the development of addiction (Sofuoglu and Mooney, 2009; Williams and Adinoff, 2008). M1/M4 muscarinic agonists mediate attenuation of cocaine self-administration and cocaine discriminative stimulus, which is abolished in M1/M4 knockout mice (Thomsen et al., 2010; Thomsen et al., 2012). Therefore, M1 and/or M4 agonism is under investigation in the treatment of drug dependence and addiction. M1/M4 agonists show efficacy in chronic inflammatory and neuropathic pain, with xanomeline showing analgesic effects in a chronic inflammatory neuropathic pain rodent model (Martino et al., 2011).

M5 Modulators

The M5 muscarinic receptor subtype comprises less than 2% of the CNS muscarinic receptors, and is expressed in both the cerebrovascular system and midbrain dopamine neurons (Weiner et al., 1990b). The M5 receptor is the sole muscarinic subtype detected in the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) (Vilaro et al., 1990; Weiner et al., 1990a), where it is coexpressed with D2 dopamine receptors, suggesting a role for M5 in the modulation of dopaminergic transmission (Weiner et al., 1990a). Modulation of dopaminergic function using M5 PAMs or NAMs has been raised as having potential utility in treatment of addiction (Raffa, 2009) and could also have utility for treatment of other disorders that involved changes in dopaminergic function, including schizophrenia, and attention-deficit hyperactivity disorder (ADHD). The combination of the genes for M5 (CHRM5) and α7 nicotinic acetylcholine receptor show linkage with schizophrenia susceptibility in humans (De Luca et al., 2004). Additionally, the M5 knockout mouse shows decreased amphetamine-induced hyperlocomotion (Wang et al., 2004). The localization of the M5 receptor to dopaminergic midbrain neurons and association with a preclinical model of schizophrenia is encouraging for research into the role of M5 in schizophrenia. Studies of M5 knockout mice suggest that M5 may be target in AD due to cerebrovascular tone regulation (Yamada et al., 2001) as well as cognitive function. Until recently, no selective M5 ligands existed, making it difficult to fully evaluate the potential utility of M5 modulators in animal models related to these disorders. However, the recent discovery of highly selective M5 PAMs (Bridges et al., 2009) (Bridges et al., 2010a) provides novel tools for the investigation of the role of M5 in CNS disorders.

Potential utility of M4 NAMs in treatment of Parkinson’s disease and dystonia

Historically anticholinergic agents were used as treatments in PD, beginning with the deadly nightshade Atropa belladonna. The concept of striatal dopaminergic-cholinergic antagonism developed with findings that with decreased dopamine in the striatum there is evidence of increased acetylcholine levels in PD (Salamone et al., 2001). Anticholinergics in PD act through muscarinic receptors, and acetylcholinesterase inhibitors (such as physostigmine) are known to worsen PD symptoms (Bourke and Druckenbrod, 1998; Ott and Lannon, 1992). Drug-induced tremulous jaw movements serves as a recent model for the resting tremor associated with Parkinson’s disease. Tremulous jaw movements induced by the muscarinic antagonist pilocarpine are robustly decreased in M4 knockout mice, suggesting that the M4 subtype plays a key role (Salamone et al., 2001). Additionally, the M4-preferring antagonist tropicamide inhibits tremulous jaw movements in a rodent model of Parkinson’s disease (Betz et al., 2007). Therefore, there is therapeutic potential for use of M4 NAMs to control tremor associated with PD. M4-targeted therapies have potential applications in other movement disorders, including dystonia and Huntington’s disease, where there is evidence of cholinergic dysfunction (Pisani et al., 2007; Smith et al., 2006). Also, muscarinic antagonists are among the few agents that can provide efficacy in treatment of generalized dystonia (Jankovic, 2006; Martella et al., 2009). It is possible that highly selective NAMs for M4 or another mAChR subtype could provide antidystonic efficacy without the adverse effects observed with non-selective mAChR antagonists.

OPTIMIZATION OF ALLOSTERIC MODULATORS AS DRUG CANDIDATES

Development and optimization of allosteric modulators of GPCRs as drug candidates presents multiple challenges (Conn, 2012; Klein et al., 2013; Melancon et al., 2012). The success of lead compound development for a number of allosteric modulators of GPCRs has established a set of drug optimization strategies for this class. With the nonlinear or “flat/shallow” nature of SAR for many members of this drug class, designing libraries around a central core by focusing on “islands” of constituents has led to successful lead optimization (Nawaratne et al., 2010; Wood et al., 2011). Also, it is critical to focus attention on both SAR of potency and cooperativity of allosteric modulators as well as differential effects on different aspects of receptor signaling (Wootten et al., 2013). Attention to principals emerging from medicinal chemistry optimization of allosteric modulators increases the opportunities for advancing successful drug candidates in this class. Once a hit compound is identified via high throughput screening, chemical tractability should be established through the synthesis of appropriate focused iterative compound libraries to generate lead compounds. The use of systematic fluorine substitution strategies has been successful for identifying fluorinated cores tolerant of change, and proved successful for M1 PAM development (Yang et al., 2010) (Bridges et al., 2010d; Reid et al., 2011). Therefore, fluorine substitution may prove a good first tier strategy for optimization of hit compounds. Selecting hit compounds with the combination of both tractable SAR and encouraging physicochemical properties has proven successful (Kenakin and Miller, 2010). It is essential to fully characterize lead compounds using radioligand binding to establish in vitro receptor interaction. Running multiple (secondary/parallel) functional screens (such as GTPγ S if primary screening performed was FLIPR calcium assay) will help to avoid stimulus bias in cases where biased ligands are not desired. In addition, allosteric modulators may show differential signaling depending on the cellular context used for screening (Niswender et al., 2010b). The use of in vitro native systems, such as brain slice electrophysiology, supports biological relevance of lead compounds. It is also important to note that the choice of orthosteric compound is critical for allosteric modulator screening and optimization efforts, as the cooperativity between the allosteric modulator and orthosteric site can change depending on the orthosteric probe ligand, a concept termed “probe dependence” (Kenakin, 2005; May and Christopoulos, 2003). Use of the endogenous orthosteric ligand increases the likelihood of physiological relevance. Screening assays using non-native agonists should be interpreted with the caveat of probe-dependent pharmacology. Additionally, allosteric sites vary across species and may lead to differences between in vitro screens and in vivo animal model pharmacology. While cell line systems that allow relatively high throughput are critical for screening compounds in a chemistry program, it is critical to me mindful that there are instances in which there is a disconnect between effects of allosteric modulators in cell based assays and in biologically relevant native systems and in vivo assays. The differences in pharmacology observed in cell based assays as compared to native systems can stem from undetected stimulus bias, or context-dependence of allosteric modulator action and can make allosteric modulator optimization especially challenging. This can be further complicated by “molecular switches” in which subtle changes to the structure of a compound changes the mode of pharmacology of allosteric modulators (i.e. mode switching with PAM to NAM/SAM conversion with subtle structural changes) and/or changes the receptor subtype selectivity can confound lead compound development (Utley et al., 2011) (Gregory et al., 2013) (Bhagwanth et al., 2012; Melancon et al., 2012; Wood et al., 2011). The screening assays used to identify lead compounds have a limited ability to detect weak or partial allosteric modulators, which may prove to be useful compounds for further development (Christopoulos and Kenakin, 2002). This is especially important when metabolites of administered allosteric modulators have pharmacological activity that is different from that of the parent compound and thereby confound interpretation of in vivo effects of systemically administered allosteric modulators (Bridges et al., 2013). It is recommended to use compounds of the same chemotype and demonstrate competitive interactions with test compounds for both ex vivo occupancy studies and in vivo imaging studies. In summary, the drug discovery effort for allosteric modulators has provided valuable insight into optimal methods for hit-to-lead development. While the complex pharmacology of mode switching, functional bias and bitopic ligand activity complicate the discovery process for allosteric ligands, they provide novel insight into the function of GPCRs. In fact, functional selectivity may become an asset for allosteric modulators targeting disease states linked to a specific signaling pathway.

FUTURE OPPORTUNITIES

Allosteric modulators of GPCRs represent exciting drug candidates for the treatment of an array of CNS disorders. In addition to the metabotropic and muscarinic allosteric modulators detailed above, drugs have been developed targeting a number of other GPCRs (see (Conn et al., 2009a) for review and AlloSteric database (http://mdl.shsmu.edu.cn/ASD/) (Huang et al., 2011). Several drugs have successfully entered the marketplace, providing proof of concept for allosteric modulators as clinical therapeutics. Cinacalcet (Sensipar), a PAM for the calcium sensing receptor (CaSR), is currently used to treat patients with hyperparathyroidism (Davey et al., 2012) (Nemeth et al., 2004). Maraviroc (Selzentry), a NAM of chemokine receptor 5 (CCR5) (Garcia-Perez et al., 2011), prevents HIV-1 cellular entry. In addition to GPCRs, allosteric modulators have also been developed for enzymes including allosteric kinase (Akt) inhibitors (Lindsley et al., 2005) and phospholipase D (PLD) (Lavieri et al., 2010; Scott et al., 2009).

Advantages of allosteric modulators of GPCRs include subtype selectivity and functional selectivity. With the emerging literature on signaling bias, it may be possible to tailor allosteric development to target specific downstream receptor pathways, avoiding undesirable side effects elicited by parallel signaling pathways. Due to the functional selectivity possible with allosteric ligands, it is crucial to incorporate multiple functional assays into drug screening paradigms. Probing multiple pathways will allow for early detection of pathway dependent allosteric modulation (Conn et al., 2009a). The development and utilization of the operational model of allosterism allows for characterization and quantification of allosteric drug properties. In addition, it may be possible to utilize partial or silent allosteric modulators to avoid adverse effect liability. PAM ligands may also exhibit allosteric agonist activity, termed ago-potentiators, a feature that may prove advantageous in certain CNS diseases. As a part of drug development in the neurodegenerative and psychiatric area, there is increased emphasis on early demonstration of in vivo target engagement using microPET imaging in non-human primate models, with development of radioactive ligands critical to this effort (Lee and Farde, 2006; Marik J, 2011). This PET data is especially important in the absence of specific disease biomarkers, and can help with in vivo dose selection to avoid under or overdosing. The surge in efforts to develop allosteric modulators of GPCRs and the collaboration between industry and academics paints a promising picture for the effort to develop effective treatments for CNS diseases.

Acknowledgments

PJC is supported by grants NIMH, NINDS, and the Michael J. Fox Foundation. The authors would like to thank the members of the VCNDD for useful input and discussions related to preparation of this review.

Footnotes

1

ABBREVIATIONS: 5MPEP, 5-methyl-6-(phenylethynyl)-pyridine; 6-OHDA, 6-hydroxydopamine; 7TMR, seven transmembrane receptor; 77-LH-28-1, 1-[3-(4-butyl-1-piperidinyl)propyl]-3,4-dihydro-2(1H)-quinolinone; AC-42, 4-n-butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]-piperidine; AChE, acetylcholinesterase; ACPT-1, (1S,3R,4S)-1-aminocyclo-pentane-1,3,4-tricarboxylic acid); AD, Alzheimer’s disease; ADX71743, (+)-6-(2,4-dimethylphenyl)-2-ethyl-6,7-dihydrobenzo[d]oxazol-4(5H)-one; AFQ056, (3aS,5S,7aR)-methyl 5-hydroxy-5-(m-tolylethynyl)octahydro-1H-indole-1-carboxylate; APP, amyloid precursor protein; BINA, potassium 30-([(2-cyclopentyl-6-7-dimethyl-1-oxo-2,3-dihydro-1H-inden-5yl)oxy]methyl)biphenyl l-4-carboxylate; BQCA; benzylquinolone carboxylic acid; CDPPB, 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide; CFMMC, 3-cyclohexyl-5-fluoro-6-methyl-7-(2-morpholin-4-ylethoxy)-4H-chromen-4-one; CNS, central nervous system; CPPHA, N-[4-chloro-2[(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)methyl]phenyl]-2-hydrobenzamide; CTEP, 2-chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-1Himidazol-4-yl)ethynyl)pyridine; DA, dopamine; DFB, [(3-fluorophenyl)methylene]hydrazone-3-fluorobenzaldehyde; DHPG, dihydroxyphenylglycine; ERK1/2, extracellular signal-regulated kinase 1/2; FMRP, fragile X mental retardation protein; FTIDC, 4-[1-(2-fluoropyridin-3-yl)-5-methyl-1H-1,2,3-triazol-4-yl]-N-isopropyl-N-methyl-3,6-dihydropyridine-1(2H)-carboxamide; FXS, Fragile X Syndrome; GABA, γ-aminobutyric acid; JNJ16259685, (3,4-dihydro-2H-pyrano[2,3]b quinolin-7-yl)(cis-4-methoxycyclohexyl) methanone; L-AP4, L-(+)-2-amino-4-phosphonobutyric acid; L-DOPA, L-3,4-dihydroxyphenylalanine; Lu AF21934, (1S,2S)-N1-(3,4-dichlorophenyl)cyclohexane-1,2-dicarboxamide; Lu AF32615, (4-((E)-styryl)-pyrimidin-2-ylamine; mGlu, metabotropic glutamate receptor; M-5MPEP, 2-(2-(3-methoxyphenyl)ethynyl)-5-methylpyridine; MMPIP, 6-(4-methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo[ 4,5-c]pyridin-4(5H)-one; MPEP, 2-methyl-6-(phenylethynyl)-pyridine; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; MTEP, 3[(2-methyl-1,3-thiazol-4-yl)ethylnyl]pyridine; NAM, negative allosteric modulator; NMDA, N-methyl-D-aspartate; PAM, positive allosteric modulator; PCP, Phencyclidine; PD, Parkinson’s disease; PD-LID, Parkinson’s disease levodopa-induced dyskinesia; PET, positron emission tomography; PHCCC, N-phenyl-7-(hydroxylimino)cyclopropa[b]chromen-1a-carboxamide; PQCA, (1-((4-cyano-4-(pyridine-2-yl)piperidine-1-yl)methyl-4-oxo-4 H-quinolizine-3-carboxylic acid); SAM, silent allosteric modulator; SIB-1757, 6-Methyl-2-(phenylazo)-3-pyridinol; SIB-1893, 2-Methyl-6-(2-phenylethenyl)pyridine; TBPB, 1-(1′-2-methylbenzyl)-1,4′-bipiperidin-4-yl)-1H-benzo[d]imidazol-2(3H)-one.

DISCLOSURES

P.J.Conn is a consultant for Karuna Pharmaceuticals and receives research support from Bristol-Meyers Squibb and Astrazeneca. P.J. Conn is an inventor on multiple composition of matter patents protecting allosteric modulators of GPCRs.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Abe T, et al. Molecular characterization of a novel metabotropic glutamate receptor mGluR5 coupled to inositol phosphate/Ca2+ signal transduction. J Biol Chem. 1992;267:13361–8. [PubMed] [Google Scholar]
  2. Achat-Mendes C, et al. Antagonism of metabotropic glutamate 1 receptors attenuates behavioral effects of cocaine and methamphetamine in squirrel monkeys. J Pharmacol Exp Ther. 2012;343:214–24. doi: 10.1124/jpet.112.196295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Adams DH, et al. A long-term, phase 2, multicenter, randomized, open-label, comparative safety study of pomaglumetad methionil (LY2140023 monohydrate) versus atypical antipsychotic standard of care in patients with schizophrenia. BMC Psychiatry. 2013;13:143. doi: 10.1186/1471-244X-13-143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Adewale AS, et al. Pharmacological stimulation of group ii metabotropic glutamate receptors reduces cocaine self-administration and cocaine-induced reinstatement of drug seeking in squirrel monkeys. J Pharmacol Exp Ther. 2006;318:922–31. doi: 10.1124/jpet.106.105387. [DOI] [PubMed] [Google Scholar]
  5. Adler CM, et al. Effects of ketamine on thought disorder, working memory, and semantic memory in healthy volunteers. Biol Psychiatry. 1998;43:811–6. doi: 10.1016/s0006-3223(97)00556-8. [DOI] [PubMed] [Google Scholar]
  6. Allen JA, Roth BL. Strategies to discover unexpected targets for drugs active at G protein-coupled receptors. Annu Rev Pharmacol Toxicol. 2011;51:117–44. doi: 10.1146/annurev-pharmtox-010510-100553. [DOI] [PubMed] [Google Scholar]
  7. Amato RJ, et al. Substituted 1-Phenyl-3-(pyridin-2-yl)urea Negative Allosteric Modulators of mGlu: Discovery of a New Tool Compound VU0463841 with Activity in Rat Models of Cocaine Addiction. ACS Chem Neurosci. 2013 doi: 10.1021/cn400070k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Anagnostaras SG, et al. Selective cognitive dysfunction in acetylcholine M1 muscarinic receptor mutant mice. Nature neuroscience. 2003;6:51–8. doi: 10.1038/nn992. [DOI] [PubMed] [Google Scholar]
  9. Annoura H, FA, Uesugi M, Tatsuoka T, Horikawa Y. A novel class of antagonists for metabotropic glutamate receptors, 7-(ydroxyimino)cyclopropa[b]chromen-1a-carboxylates. Bioorganic & medicinal chemistry letters. 1996;6:763–766. [Google Scholar]
  10. Auerbach BD, et al. Mutations causing syndromic autism define an axis of synaptic pathophysiology. Nature. 2011;480:63–8. doi: 10.1038/nature10658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Awad H, et al. Activation of metabotropic glutamate receptor 5 has direct excitatory effects and potentiates NMDA receptor currents in neurons of the subthalamic nucleus. J Neurosci. 2000;20:7871–9. doi: 10.1523/JNEUROSCI.20-21-07871.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Ayala JE, et al. mGluR5 positive allosteric modulators facilitate both hippocampal LTP and LTD and enhance spatial learning. Neuropsychopharmacology. 2009;34:2057–71. doi: 10.1038/npp.2009.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bartus RT, et al. The cholinergic hypothesis of geriatric memory dysfunction. Science. 1982;217:408–14. doi: 10.1126/science.7046051. [DOI] [PubMed] [Google Scholar]
  14. Battaglia G, et al. Pharmacological activation of mGlu4 metabotropic glutamate receptors reduces nigrostriatal degeneration in mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. J Neurosci. 2006;26:7222–9. doi: 10.1523/JNEUROSCI.1595-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bear MF, et al. The mGluR theory of fragile X mental retardation. Trends Neurosci. 2004;27:370–7. doi: 10.1016/j.tins.2004.04.009. [DOI] [PubMed] [Google Scholar]
  16. Belozertseva IV, et al. Antidepressant-like effects of mGluR1 and mGluR5 antagonists in the rat forced swim and the mouse tail suspension tests. European neuropsychopharmacology : the journal of the European College of Neuropsychopharmacology. 2007;17:172–9. doi: 10.1016/j.euroneuro.2006.03.002. [DOI] [PubMed] [Google Scholar]
  17. Bennett CE, et al. Fused tricyclic mGluR1 antagonists for the treatment of neuropathic pain. Bioorganic & medicinal chemistry letters. 2012;22:1575–8. doi: 10.1016/j.bmcl.2011.12.131. [DOI] [PubMed] [Google Scholar]
  18. Benneyworth MA, et al. A selective positive allosteric modulator of metabotropic glutamate receptor subtype 2 blocks a hallucinogenic drug model of psychosis. Mol Pharmacol. 2007;72:477–84. doi: 10.1124/mol.107.035170. [DOI] [PubMed] [Google Scholar]
  19. Bennouar KE, et al. Synergy between L-DOPA and a novel positive allosteric modulator of metabotropic glutamate receptor 4: implications for Parkinson’s disease treatment and dyskinesia. Neuropharmacology. 2013;66:158–69. doi: 10.1016/j.neuropharm.2012.03.022. [DOI] [PubMed] [Google Scholar]
  20. Betz AJ, et al. The muscarinic receptor antagonist tropicamide suppresses tremulous jaw movements in a rodent model of parkinsonian tremor: possible role of M4 receptors. Psychopharmacology (Berl) 2007;194:347–59. doi: 10.1007/s00213-007-0844-6. [DOI] [PubMed] [Google Scholar]
  21. Bhagwanth S, et al. Transformation of Pro-Leu-Gly-NH(2) peptidomimetic positive allosteric modulators of the dopamine D(2) receptor into negative modulators. ACS Chem Neurosci. 2012;3:274–84. doi: 10.1021/cn200096u. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Bhakar AL, et al. The pathophysiology of fragile X (and what it teaches us about synapses) Annu Rev Neurosci. 2012a;35:417–43. doi: 10.1146/annurev-neuro-060909-153138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Bhakar AL, et al. The pathophysiology of fragile X (and what it teaches us about synapses) Annual review of neuroscience. 2012b;35:417–43. doi: 10.1146/annurev-neuro-060909-153138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Bird MK, Lawrence AJ. Group I metabotropic glutamate receptors: involvement in drug-seeking and drug-induced plasticity. Curr Mol Pharmacol. 2009;2:83–94. doi: 10.2174/1874467210902010083. [DOI] [PubMed] [Google Scholar]
  25. Blandini F, et al. Functional changes of the basal ganglia circuitry in Parkinson’s disease. Prog Neurobiol. 2000;62:63–88. doi: 10.1016/s0301-0082(99)00067-2. [DOI] [PubMed] [Google Scholar]
  26. Bodick NC, et al. Effects of xanomeline, a selective muscarinic receptor agonist, on cognitive function and behavioral symptoms in Alzheimer disease. Arch Neurol. 1997a;54:465–73. doi: 10.1001/archneur.1997.00550160091022. [DOI] [PubMed] [Google Scholar]
  27. Bodick NC, et al. The selective muscarinic agonist xanomeline improves both the cognitive deficits and behavioral symptoms of Alzheimer disease. Alzheimer Dis Assoc Disord. 1997b;11(Suppl 4):S16–22. [PubMed] [Google Scholar]
  28. Bogenpohl J, et al. Metabotropic glutamate receptor 4 in the basal ganglia of parkinsonian monkeys: ultrastructural localization and electrophysiological effects of activation in the striatopallidal complex. Neuropharmacology. 2013;66:242–52. doi: 10.1016/j.neuropharm.2012.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Bogoyevitch MA, Fairlie DP. A new paradigm for protein kinase inhibition: blocking phosphorylation without directly targeting ATP binding. Drug Discov Today. 2007;12:622–33. doi: 10.1016/j.drudis.2007.06.008. [DOI] [PubMed] [Google Scholar]
  30. Bourke D, Druckenbrod RW. Possible association between donepezil and worsening Parkinson’s disease. Ann Pharmacother. 1998;32:610–1. doi: 10.1345/aph.17355. [DOI] [PubMed] [Google Scholar]
  31. Bradley SR, et al. Activation of group II metabotropic glutamate receptors inhibits synaptic excitation of the substantia Nigra pars reticulata. J Neurosci. 2000;20:3085–94. doi: 10.1523/JNEUROSCI.20-09-03085.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Bradley SR, et al. Immunohistochemical localization of subtype 4a metabotropic glutamate receptors in the rat and mouse basal ganglia. J Comp Neurol. 1999;407:33–46. [PubMed] [Google Scholar]
  33. Brady AE, et al. Centrally active allosteric potentiators of the M4 muscarinic acetylcholine receptor reverse amphetamine-induced hyperlocomotor activity in rats. J Pharmacol Exp Ther. 2008;327:941–53. doi: 10.1124/jpet.108.140350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Bridges TM, et al. The antipsychotic potential of muscarinic allosteric modulation. Drug News Perspect. 2010a;23:229–40. doi: 10.1358/dnp.2010.23.4.1416977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Bridges TM, et al. Probe Reports from the NIH Molecular Libraries Program. Bethesda (MD): 2010b. Discovery and development of the a highly selective M1 Positive Allosteric Modulator (PAM) [PubMed] [Google Scholar]
  36. Bridges TM, et al. Discovery of the first highly M5-preferring muscarinic acetylcholine receptor ligand, an M5 positive allosteric modulator derived from a series of 5-trifluoromethoxy N-benzyl isatins. J Med Chem. 2009;52:3445–8. doi: 10.1021/jm900286j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Bridges TM, et al. Probe Reports from the NIH Molecular Libraries Program. Bethesda (MD): 2010c. Discovery of a Highly Selective in vitro and in vivo M4 Positive Allosteric Modulator (PAM) Series with Greatly Improved Human Receptor Activity. [PubMed] [Google Scholar]
  38. Bridges TM, et al. Chemical lead optimization of a pan Gq mAChR M1, M3, M5 positive allosteric modulator (PAM) lead. Part II: development of a potent and highly selective M1 PAM. Bioorg Med Chem Lett. 2010d;20:1972–5. doi: 10.1016/j.bmcl.2010.01.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Bridges TM, et al. Probe Reports from the NIH Molecular Libraries Program. Bethesda (MD): 2010e. Discovery and development of a second highly selective M1 Positive Allosteric Modulator (PAM) [PubMed] [Google Scholar]
  40. Bridges TM, et al. Biotransformation of a Novel Positive Allosteric Modulator of mGlu5 Contributes to Seizures in Rats Involving a Receptor Agonism-Dependent Mechanism. Drug Metab Dispos. 2013 doi: 10.1124/dmd.113.052084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Brody SA, et al. Effect of antipsychotic treatment on the prepulse inhibition deficit of mGluR5 knockout mice. Psychopharmacology (Berl) 2004a;172:187–95. doi: 10.1007/s00213-003-1635-3. [DOI] [PubMed] [Google Scholar]
  42. Brody SA, et al. Assessment of a prepulse inhibition deficit in a mutant mouse lacking mGlu5 receptors. Mol Psychiatry. 2004b;9:35–41. doi: 10.1038/sj.mp.4001404. [DOI] [PubMed] [Google Scholar]
  43. Bruno V, et al. Selective activation of mGlu4 metabotropic glutamate receptors is protective against excitotoxic neuronal death. J Neurosci. 2000;20:6413–20. doi: 10.1523/JNEUROSCI.20-17-06413.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Burford NT, et al. Discovery of positive allosteric modulators and silent allosteric modulators of the mu-opioid receptor. Proc Natl Acad Sci U S A. 2013;110:10830–5. doi: 10.1073/pnas.1300393110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Caccamo A, et al. M1 receptors play a central role in modulating AD-like pathology in transgenic mice. Neuron. 2006;49:671–82. doi: 10.1016/j.neuron.2006.01.020. [DOI] [PubMed] [Google Scholar]
  46. Campbell UC, et al. The mGluR5 antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) potentiates PCP-induced cognitive deficits in rats. Psychopharmacology (Berl) 2004;175:310–8. doi: 10.1007/s00213-004-1827-5. [DOI] [PubMed] [Google Scholar]
  47. Campo B, et al. Characterization of an mGluR2/3 negative allosteric modulator in rodent models of depression. Journal of neurogenetics. 2011a;25:152–66. doi: 10.3109/01677063.2011.627485. [DOI] [PubMed] [Google Scholar]
  48. Campo B, et al. Characterization of an mGluR2/3 negative allosteric modulator in rodent models of depression. J Neurogenet. 2011b;25:152–66. doi: 10.3109/01677063.2011.627485. [DOI] [PubMed] [Google Scholar]
  49. Cartmell J, et al. The metabotropic glutamate 2/3 receptor agonists LY354740 and LY379268 selectively attenuate phencyclidine versus d-amphetamine motor behaviors in rats. J Pharmacol Exp Ther. 1999a;291:161–70. [PubMed] [Google Scholar]
  50. Cartmell J, et al. The metabotropic glutamate 2/3 receptor agonists LY354740 and LY379268 selectively attenuate phencyclidine versus d-amphetamine motor behaviors in rats. The Journal of pharmacology and experimental therapeutics. 1999b;291:161–70. [PubMed] [Google Scholar]
  51. Celanire S, Campo B. Recent advances in the drug discovery of metabotropic glutamate receptor 4 (mGluR4) activators for the treatment of CNS and non-CNS disorders. Expert Opin Drug Discov. 2012;7:261–80. doi: 10.1517/17460441.2012.660914. [DOI] [PubMed] [Google Scholar]
  52. Chaki S, et al. mGlu2/3 and mGlu5 receptors: potential targets for novel antidepressants. Neuropharmacology. 2013;66:40–52. doi: 10.1016/j.neuropharm.2012.05.022. [DOI] [PubMed] [Google Scholar]
  53. Chaki S, et al. MGS0039: a potent and selective group II metabotropic glutamate receptor antagonist with antidepressant-like activity. Neuropharmacology. 2004;46:457–67. doi: 10.1016/j.neuropharm.2003.10.009. [DOI] [PubMed] [Google Scholar]
  54. Chan WY, et al. Allosteric modulation of the muscarinic M4 receptor as an approach to treating schizophrenia. Proc Natl Acad Sci U S A. 2008;105:10978–83. doi: 10.1073/pnas.0800567105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Chaudhuri KR, et al. Non-motor symptoms of Parkinson’s disease: diagnosis and management. Lancet Neurol. 2006;5:235–45. doi: 10.1016/S1474-4422(06)70373-8. [DOI] [PubMed] [Google Scholar]
  56. Chiamulera C, et al. Reinforcing and locomotor stimulant effects of cocaine are absent in mGluR5 null mutant mice. Nature neuroscience. 2001;4:873–4. doi: 10.1038/nn0901-873. [DOI] [PubMed] [Google Scholar]
  57. Christopoulos A, Kenakin T. G protein-coupled receptor allosterism and complexing. Pharmacological reviews. 2002;54:323–74. doi: 10.1124/pr.54.2.323. [DOI] [PubMed] [Google Scholar]
  58. Clifton NE, et al. 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. 2013;225:579–94. doi: 10.1007/s00213-012-2845-3. [DOI] [PubMed] [Google Scholar]
  59. Conn PJ, et al. Metabotropic glutamate receptors in the basal ganglia motor circuit. Nat Rev Neurosci. 2005;6:787–98. doi: 10.1038/nrn1763. [DOI] [PubMed] [Google Scholar]
  60. Conn PJ, et al. Allosteric modulators of GPCRs: a novel approach for the treatment of CNS disorders. Nat Rev Drug Discov. 2009a;8:41–54. doi: 10.1038/nrd2760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Conn PJ, et al. Subtype-selective allosteric modulators of muscarinic receptors for the treatment of CNS disorders. Trends Pharmacol Sci. 2009b;30:148–55. doi: 10.1016/j.tips.2008.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Conn PJ, Kuduk SD, Doller D. Drug Design Strategies for GPCR Allosteric Modulators. In: Desai IMC, editor. Annual Reports In Medicinal Chemistry. Academic Press; Burlington: 2012. pp. 441–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Conn PJ, et al. Activation of metabotropic glutamate receptors as a novel approach for the treatment of schizophrenia. Trends in pharmacological sciences. 2009c;30:25–31. doi: 10.1016/j.tips.2008.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Conn PJ, et al. Activation of metabotropic glutamate receptors as a novel approach for the treatment of schizophrenia. Trends Pharmacol Sci. 2009d;30:25–31. doi: 10.1016/j.tips.2008.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Conn PJ, Pin JP. Pharmacology and functions of metabotropic glutamate receptors. Annu Rev Pharmacol Toxicol. 1997;37:205–37. doi: 10.1146/annurev.pharmtox.37.1.205. [DOI] [PubMed] [Google Scholar]
  66. Conn PJ, et al. Schizophrenia: moving beyond monoamine antagonists. Mol Interv. 2008;8:99–107. doi: 10.1124/mi.8.2.7. [DOI] [PubMed] [Google Scholar]
  67. Cosford ND, et al. 3-[(2-Methyl-1,3-thiazol-4-yl)ethynyl]-pyridine: a potent and highly selective metabotropic glutamate subtype 5 receptor antagonist with anxiolytic activity. Journal of medicinal chemistry. 2003;46:204–6. doi: 10.1021/jm025570j. [DOI] [PubMed] [Google Scholar]
  68. Coyle JT. Glutamate and schizophrenia: beyond the dopamine hypothesis. Cell Mol Neurobiol. 2006;26:365–84. doi: 10.1007/s10571-006-9062-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Coyle JT, Tsai G. The NMDA receptor glycine modulatory site: a therapeutic target for improving cognition and reducing negative symptoms in schizophrenia. Psychopharmacology. 2004;174:32–8. doi: 10.1007/s00213-003-1709-2. [DOI] [PubMed] [Google Scholar]
  70. Crook JM, et al. Decreased muscarinic receptor binding in subjects with schizophrenia: a study of the human hippocampal formation. Biol Psychiatry. 2000;48:381–8. doi: 10.1016/s0006-3223(00)00918-5. [DOI] [PubMed] [Google Scholar]
  71. Darrah JM, et al. 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;19:225–34. doi: 10.1097/FBP.0b013e3282feb0ac. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Davey AE, et al. Positive and negative allosteric modulators promote biased signaling at the calcium-sensing receptor. Endocrinology. 2012;153:1232–41. doi: 10.1210/en.2011-1426. [DOI] [PubMed] [Google Scholar]
  73. Dawson L, et al. The group II metabotropic glutamate receptor agonist, DCG-IV, alleviates akinesia following intranigral or intraventricular administration in the reserpine-treated rat. Br J Pharmacol. 2000;129:541–6. doi: 10.1038/sj.bjp.0703105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. De Luca V, et al. Linkage of M5 muscarinic and alpha7-nicotinic receptor genes on 15q13 to schizophrenia. Neuropsychobiology. 2004;50:124–7. doi: 10.1159/000079102. [DOI] [PubMed] [Google Scholar]
  75. Dean B, et al. Decreased muscarinic1 receptors in the dorsolateral prefrontal cortex of subjects with schizophrenia. Mol Psychiatry. 2002;7:1083–91. doi: 10.1038/sj.mp.4001199. [DOI] [PubMed] [Google Scholar]
  76. Dekundy A, et al. Effects of group I metabotropic glutamate receptors blockade in experimental models of Parkinson’s disease. Brain Res Bull. 2006;69:318–26. doi: 10.1016/j.brainresbull.2005.12.009. [DOI] [PubMed] [Google Scholar]
  77. Dencker D, et al. Involvement of a subpopulation of neuronal M4 muscarinic acetylcholine receptors in the antipsychotic-like effects of the M1/M4 preferring muscarinic receptor agonist xanomeline. J Neurosci. 2011;31:5905–8. doi: 10.1523/JNEUROSCI.0370-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Deng C, Huang XF. Decreased density of muscarinic receptors in the superior temporal gyrusin schizophrenia. J Neurosci Res. 2005;81:883–90. doi: 10.1002/jnr.20600. [DOI] [PubMed] [Google Scholar]
  79. Dickerson JW, Conn PJ. Therapeutic potential of targeting metabotropic glutamate receptors for Parkinson’s disease. Neurodegener Dis Manag. 2012;2:221–232. doi: 10.2217/nmt.12.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Digby GJ, et al. Novel allosteric agonists of M1 muscarinic acetylcholine receptors induce brain region-specific responses that correspond with behavioral effects in animal models. J Neurosci. 2012a;32:8532–44. doi: 10.1523/JNEUROSCI.0337-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Digby GJ, et al. Chemical modification of the M(1) agonist VU0364572 reveals molecular switches in pharmacology and a bitopic binding mode. ACS Chem Neurosci. 2012b;3:1025–36. doi: 10.1021/cn300103e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Doherty AJ, et al. 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;131:239–44. doi: 10.1038/sj.bjp.0703574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Dolen G, et al. Correction of fragile X syndrome in mice. Neuron. 2007;56:955–62. doi: 10.1016/j.neuron.2007.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Doria J, et al. Metabotropic glutamate receptor 5 positive allosteric modulators are neuroprotective in a mouse model of Huntington’s disease. Br J Pharmacol. 2013;169:909–21. doi: 10.1111/bph.12164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. East SP, et al. An orally bioavailable positive allosteric modulator of the mGlu4 receptor with efficacy in an animal model of motor dysfunction. Bioorganic & medicinal chemistry letters. 2010a;20:4901–5. doi: 10.1016/j.bmcl.2010.06.078. [DOI] [PubMed] [Google Scholar]
  86. East SP, et al. An orally bioavailable positive allosteric modulator of the mGlu4 receptor with efficacy in an animal model of motor dysfunction. Bioorg Med Chem Lett. 2010b;20:4901–5. doi: 10.1016/j.bmcl.2010.06.078. [DOI] [PubMed] [Google Scholar]
  87. Emmitte KA. mGlu5 negative allosteric modulators: a patent review (2010–2012) Expert Opin Ther Pat. 2013;23:393–408. doi: 10.1517/13543776.2013.760544. [DOI] [PubMed] [Google Scholar]
  88. Engers DW, et al. Probe Reports from the NIH Molecular Libraries Program. Bethesda (MD): 2010. Discovery of a novel metabotropic glutamate receptor 4 (mGlu4) positive allosteric modulator (PAM) extended probe: Characterization of ML292, a potent and selective mGlu4 PAM which produces efficacy alone or in combination with L-DOPA in preclinical rodent models of Parkinson’s disease. [PubMed] [Google Scholar]
  89. Engers DW, et al. Synthesis and evaluation of a series of heterobiarylamides that are centrally penetrant metabotropic glutamate receptor 4 (mGluR4) positive allosteric modulators (PAMs) J Med Chem. 2009;52:4115–8. doi: 10.1021/jm9005065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Fabbrini G, et al. Levodopa-induced dyskinesias. Mov Disord. 2007;22:1379–89. doi: 10.1002/mds.21475. quiz 1523. [DOI] [PubMed] [Google Scholar]
  91. Fallarino F, et al. Metabotropic glutamate receptor-4 modulates adaptive immunity and restrains neuroinflammation. Nat Med. 2010;16:897–902. doi: 10.1038/nm.2183. [DOI] [PubMed] [Google Scholar]
  92. Fang Y, et al. G protein-coupled receptor microarrays for drug discovery. Drug Discov Today. 2003;8:755–61. doi: 10.1016/s1359-6446(03)02779-x. [DOI] [PubMed] [Google Scholar]
  93. Feinberg I, et al. The selective group mGlu2/3 receptor agonist LY379268 suppresses REM sleep and fast EEG in the rat. Pharmacol Biochem Behav. 2002;73:467–74. doi: 10.1016/s0091-3057(02)00843-2. [DOI] [PubMed] [Google Scholar]
  94. Feldman H. Treating Alzheimer’s disease with cholinesterase inhibitors: what have we learned so far? Int Psychogeriatr. 2002;14(Suppl 1):3–5. doi: 10.1017/s1041610203008639. [DOI] [PubMed] [Google Scholar]
  95. Fell MJ, et al. Evidence for the role of metabotropic glutamate (mGlu)2 not mGlu3 receptors in the preclinical antipsychotic pharmacology of the mGlu2/3 receptor agonist (-)-(1R,4S,5S,6S)-4-amino-2-sulfonylbicyclo[3.1.0]hexane-4,6-dicarboxylic acid (LY404039) J Pharmacol Exp Ther. 2008;326:209–17. doi: 10.1124/jpet.108.136861. [DOI] [PubMed] [Google Scholar]
  96. Fell MJ, et al. N-(4-((2-(trifluoromethyl)-3-hydroxy-4-(isobutyryl)phenoxy)methyl)benzyl)-1-methy l-1H-imidazole-4-carboxamide (THIIC), a novel metabotropic glutamate 2 potentiator with potential anxiolytic/antidepressant properties: in vivo profiling suggests a link between behavioral and central nervous system neurochemical changes. J Pharmacol Exp Ther. 2011;336:165–77. doi: 10.1124/jpet.110.172957. [DOI] [PubMed] [Google Scholar]
  97. Field JR, et al. Targeting glutamate synapses in schizophrenia. Trends Mol Med. 2011;17:689–98. doi: 10.1016/j.molmed.2011.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Fisher A. M1 muscarinic agonists target major hallmarks of Alzheimer’s disease--the pivotal role of brain M1 receptors. Neurodegener Dis. 2008;5:237–40. doi: 10.1159/000113712. [DOI] [PubMed] [Google Scholar]
  99. Friedmann CTH, DL, Ciccone PE, Rubin RT. Phase II double blind controlled study of a new anxiolytic, fenobam (McN-3377) vs placebo. Curr Ther Res. 1980;27:144–151. [Google Scholar]
  100. Galici R, et al. 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;318:173–85. doi: 10.1124/jpet.106.102046. [DOI] [PubMed] [Google Scholar]
  101. Garcia-Perez J, et al. Allosteric model of maraviroc binding to CC chemokine receptor 5 (CCR5) J Biol Chem. 2011;286:33409–21. doi: 10.1074/jbc.M111.279596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Gasparini F, et al. 2-Methyl-6-(phenylethynyl)-pyridine (MPEP), a potent, selective and systemically active mGlu5 receptor antagonist. Neuropharmacology. 1999;38:1493–503. doi: 10.1016/s0028-3908(99)00082-9. [DOI] [PubMed] [Google Scholar]
  103. Gastambide F, et al. The mGlu(5) positive allosteric modulator LSN2463359 differentially modulates motor, instrumental and cognitive effects of NMDA receptor antagonists in the rat. Neuropharmacology. 2013;64:240–7. doi: 10.1016/j.neuropharm.2012.07.039. [DOI] [PubMed] [Google Scholar]
  104. Gilmour G, et al. In vitro characterisation of the novel positive allosteric modulators of the mGlu(5) receptor, LSN2463359 and LSN2814617, and their effects on sleep architecture and operant responding in the rat. Neuropharmacology. 2013;64:224–39. doi: 10.1016/j.neuropharm.2012.07.030. [DOI] [PubMed] [Google Scholar]
  105. Goeldner C, et al. Cognitive impairment in major depression and the mGlu2 receptor as a therapeutic target. Neuropharmacology. 2013;64:337–46. doi: 10.1016/j.neuropharm.2012.08.001. [DOI] [PubMed] [Google Scholar]
  106. Goudet C, et al. Group III metabotropic glutamate receptors inhibit hyperalgesia in animal models of inflammation and neuropathic pain. Pain. 2008;137:112–24. doi: 10.1016/j.pain.2007.08.020. [DOI] [PubMed] [Google Scholar]
  107. Greenamyre JT, O’Brien CF. N-methyl-D-aspartate antagonists in the treatment of Parkinson’s disease. Arch Neurol. 1991;48:977–81. doi: 10.1001/archneur.1991.00530210109030. [DOI] [PubMed] [Google Scholar]
  108. Gregory KJ, et al. Probing the metabotropic glutamate receptor 5 (mGlu(5)) positive allosteric modulator (PAM) binding pocket: discovery of point mutations that engender a “molecular switch” in PAM pharmacology. Mol Pharmacol. 2013;83:991–1006. doi: 10.1124/mol.112.083949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Grossberg GT. The ABC of Alzheimer’s disease: behavioral symptoms and their treatment. Int Psychogeriatr. 2002;14(Suppl 1):27–49. doi: 10.1017/s1041610203008652. [DOI] [PubMed] [Google Scholar]
  110. Halberstadt AL. The phencyclidine-glutamate model of schizophrenia. Clin Neuropharmacol. 1995;18:237–49. doi: 10.1097/00002826-199506000-00004. [DOI] [PubMed] [Google Scholar]
  111. Hanyu H, et al. Effect of age on regional cerebral blood flow patterns in Alzheimer’s disease patients. J Neurol Sci. 2003;209:25–30. doi: 10.1016/s0022-510x(02)00456-2. [DOI] [PubMed] [Google Scholar]
  112. Hashimoto K, et al. Glutamate modulators as potential therapeutic drugs in schizophrenia and affective disorders. European archives of psychiatry and clinical neuroscience. 2013 doi: 10.1007/s00406-013-0399-y. [DOI] [PubMed] [Google Scholar]
  113. Hasselmo ME. The role of acetylcholine in learning and memory. Curr Opin Neurobiol. 2006;16:710–5. doi: 10.1016/j.conb.2006.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Hasselmo ME, Giocomo LM. Cholinergic modulation of cortical function. J Mol Neurosci. 2006;30:133–5. doi: 10.1385/JMN:30:1:133. [DOI] [PubMed] [Google Scholar]
  115. Heinrich JN, et al. Pharmacological comparison of muscarinic ligands: historical versus more recent muscarinic M1-preferring receptor agonists. Eur J Pharmacol. 2009;605:53–6. doi: 10.1016/j.ejphar.2008.12.044. [DOI] [PubMed] [Google Scholar]
  116. Hemstapat K, et al. A novel family of potent negative allosteric modulators of group II metabotropic glutamate receptors. The Journal of pharmacology and experimental therapeutics. 2007;322:254–64. doi: 10.1124/jpet.106.117093. [DOI] [PubMed] [Google Scholar]
  117. Hemstapat K, et al. A novel class of positive allosteric modulators of metabotropic glutamate receptor subtype 1 interact with a site distinct from that of negative allosteric modulators. Mol Pharmacol. 2006;70:616–26. doi: 10.1124/mol.105.021857. [DOI] [PubMed] [Google Scholar]
  118. Henry SA, et al. The mGluR5 antagonist MPEP, but not the mGluR2/3 agonist LY314582, augments PCP effects on prepulse inhibition and locomotor activity. Neuropharmacology. 2002;43:1199–209. doi: 10.1016/s0028-3908(02)00332-5. [DOI] [PubMed] [Google Scholar]
  119. Herman EJ, et al. Metabotropic glutamate receptors for new treatments in schizophrenia. Handb Exp Pharmacol. 2012:297–365. doi: 10.1007/978-3-642-25758-2_11. [DOI] [PubMed] [Google Scholar]
  120. Hikichi H, et al. Effects of a novel metabotropic glutamate receptor 7 negative allosteric modulator, 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazonolo[4,5-c]pyridin-4(5H)-one (MMPIP), on the central nervous system in rodents. Eur J Pharmacol. 2010;639:106–14. doi: 10.1016/j.ejphar.2009.08.047. [DOI] [PubMed] [Google Scholar]
  121. Hirsch EC, et al. Metabolic effects of nigrostriatal denervation in basal ganglia. Trends Neurosci. 2000;23:S78–85. doi: 10.1016/s1471-1931(00)00021-5. [DOI] [PubMed] [Google Scholar]
  122. Hogg RC, et al. Allosteric modulation of ligand-gated ion channels. Biochem Pharmacol. 2005;70:1267–76. doi: 10.1016/j.bcp.2005.06.010. [DOI] [PubMed] [Google Scholar]
  123. Hopkins CR. Is there a path forward for mGlu(2) positive allosteric modulators for the treatment of schizophrenia? ACS Chem Neurosci. 2013;4:211–3. doi: 10.1021/cn400023y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Hopkins CR, et al. mGluR4-positive allosteric modulation as potential treatment for Parkinson’s disease. Future Med Chem. 2009;1:501–13. doi: 10.4155/fmc.09.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Huang Z, et al. ASD: a comprehensive database of allosteric proteins and modulators. Nucleic Acids Res. 2011;39:D663–9. doi: 10.1093/nar/gkq1022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Hughes ZA, et al. Negative allosteric modulation of metabotropic glutamate receptor 5 results in broad spectrum activity relevant to treatment resistant depression. Neuropharmacology. 2013;66:202–14. doi: 10.1016/j.neuropharm.2012.04.007. [DOI] [PubMed] [Google Scholar]
  127. Ince E, et al. Differential expression of D1 and D2 dopamine and m4 muscarinic acetylcholine receptor proteins in identified striatonigral neurons. Synapse. 1997;27:357–66. doi: 10.1002/(SICI)1098-2396(199712)27:4<357::AID-SYN9>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
  128. Itil TM, SB, Huque M, Mukhopadhyay S, Blasucci DNKT, Ciccone PE. The clinical and quantitative EEG effects and plasma levels of fenobam (McN-3377) in subjects with anxiety: an open rising dose tolerance and efficacy study. Curr Ther Res. 1978:708–724. [Google Scholar]
  129. Jacquemont S, et al. Epigenetic modification of the FMR1 gene in fragile X syndrome is associated with differential response to the mGluR5 antagonist AFQ056. Sci Transl Med. 2011;3:64ra1. doi: 10.1126/scitranslmed.3001708. [DOI] [PubMed] [Google Scholar]
  130. Jankovic J. Treatment of dystonia. Lancet Neurol. 2006;5:864–72. doi: 10.1016/S1474-4422(06)70574-9. [DOI] [PubMed] [Google Scholar]
  131. Jankovic J. Parkinson’s disease: clinical features and diagnosis. J Neurol Neurosurg Psychiatry. 2008;79:368–76. doi: 10.1136/jnnp.2007.131045. [DOI] [PubMed] [Google Scholar]
  132. Javitt DC. Negative schizophrenic symptomatology and the PCP (phencyclidine) model of schizophrenia. The Hillside journal of clinical psychiatry. 1987;9:12–35. [PubMed] [Google Scholar]
  133. Jenner P. Molecular mechanisms of L-DOPA-induced dyskinesia. Nat Rev Neurosci. 2008;9:665–77. doi: 10.1038/nrn2471. [DOI] [PubMed] [Google Scholar]
  134. Jimenez HN, et al. 4-(1-Phenyl-1H-pyrazol-4-yl)quinolines as novel, selective and brain penetrant metabotropic glutamate receptor 4 positive allosteric modulators. Bioorg Med Chem Lett. 2012;22:3235–9. doi: 10.1016/j.bmcl.2012.03.032. [DOI] [PubMed] [Google Scholar]
  135. Johnson KA, et al. Glutamate receptors as therapeutic targets for Parkinson’s disease. CNS Neurol Disord Drug Targets. 2009;8:475–91. doi: 10.2174/187152709789824606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Johnson KA, et al. The metabotropic glutamate receptor 8 agonist (S)-3,4-DCPG reverses motor deficits in prolonged but not acute models of Parkinson’s disease. Neuropharmacology. 2013;66:187–95. doi: 10.1016/j.neuropharm.2012.03.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Johnson KA, et al. Activation of group II metabotropic glutamate receptors induces long-term depression of excitatory synaptic transmission in the substantia nigra pars reticulata. Neurosci Lett. 2011;504:102–6. doi: 10.1016/j.neulet.2011.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Johnson MP, et al. 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. Journal of medicinal chemistry. 2003;46:3189–92. doi: 10.1021/jm034015u. [DOI] [PubMed] [Google Scholar]
  139. Jones CK, et al. Novel selective allosteric activator of the M1 muscarinic acetylcholine receptor regulates amyloid processing and produces antipsychotic-like activity in rats. J Neurosci. 2008;28:10422–33. doi: 10.1523/JNEUROSCI.1850-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Jones CK, et al. The metabotropic glutamate receptor 4-positive allosteric modulator VU0364770 produces efficacy alone and in combination with L-DOPA or an adenosine 2A antagonist in preclinical rodent models of Parkinson’s disease. J Pharmacol Exp Ther. 2012a;340:404–21. doi: 10.1124/jpet.111.187443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Jones CK, et al. Muscarinic and nicotinic acetylcholine receptor agonists and allosteric modulators for the treatment of schizophrenia. Neuropsychopharmacology. 2012b;37:16–42. doi: 10.1038/npp.2011.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Jones CK, et al. Analgesic effects of the selective group II (mGlu2/3) metabotropic glutamate receptor agonists LY379268 and LY389795 in persistent and inflammatory pain models after acute and repeated dosing. Neuropharmacology. 2005;49(Suppl 1):206–18. doi: 10.1016/j.neuropharm.2005.05.008. [DOI] [PubMed] [Google Scholar]
  143. Jones CK, et al. Discovery, synthesis, and structure-activity relationship development of a series of N-4-(2,5-dioxopyrrolidin-1-yl)phenylpicolinamides (VU0400195, ML182): characterization of a novel positive allosteric modulator of the metabotropic glutamate receptor 4 (mGlu(4)) with oral efficacy in an antiparkinsonian animal model. J Med Chem. 2011;54:7639–47. doi: 10.1021/jm200956q. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Kalinichev M, et al. ADX71743, a potent and selective negative allosteric modulator of metabotropic glutamate receptor 7: in vitro and in vivo characterization. The Journal of pharmacology and experimental therapeutics. 2013;344:624–36. doi: 10.1124/jpet.112.200915. [DOI] [PubMed] [Google Scholar]
  145. Katritch V, et al. Structure-function of the G protein-coupled receptor superfamily. Annu Rev Pharmacol Toxicol. 2013;53:531–56. doi: 10.1146/annurev-pharmtox-032112-135923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Kenakin T. New concepts in drug discovery: collateral efficacy and permissive antagonism. Nature reviews. Drug discovery. 2005;4:919–27. doi: 10.1038/nrd1875. [DOI] [PubMed] [Google Scholar]
  147. Kenakin T. G protein coupled receptors as allosteric proteins and the role of allosteric modulators. J Recept Signal Transduct Res. 2010;30:313–21. doi: 10.3109/10799893.2010.503964. [DOI] [PubMed] [Google Scholar]
  148. Kenakin T, Miller LJ. Seven transmembrane receptors as shapeshifting proteins: the impact of allosteric modulation and functional selectivity on new drug discovery. Pharmacol Rev. 2010;62:265–304. doi: 10.1124/pr.108.000992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Kenny PJ, et al. Metabotropic glutamate 5 receptor blockade may attenuate cocaine self-administration by decreasing brain reward function in rats. Psychopharmacology. 2005;179:247–54. doi: 10.1007/s00213-004-2069-2. [DOI] [PubMed] [Google Scholar]
  150. Keywood C, et al. A proof-of-concept study evaluating the effect of ADX10059, a metabotropic glutamate receptor-5 negative allosteric modulator, on acid exposure and symptoms in gastro-oesophageal reflux disease. Gut. 2009;58:1192–9. doi: 10.1136/gut.2008.162040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Kim DH, et al. Building a better antipsychotic: receptor targets for the treatment of multiple symptom dimensions of schizophrenia. Neurotherapeutics. 2009;6:78–85. doi: 10.1016/j.nurt.2008.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Kinney GG, et al. Metabotropic glutamate subtype 5 receptors modulate locomotor activity and sensorimotor gating in rodents. J Pharmacol Exp Ther. 2003;306:116–23. doi: 10.1124/jpet.103.048702. [DOI] [PubMed] [Google Scholar]
  153. Kinon BJ, et al. 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;31:349–55. doi: 10.1097/JCP.0b013e318218dcd5. [DOI] [PubMed] [Google Scholar]
  154. Klein MT, et al. Approaches for probing allosteric interactions at 7 transmembrane spanning receptors. Prog Mol Biol Transl Sci. 2013;115:1–59. doi: 10.1016/B978-0-12-394587-7.00001-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Knoflach F, et al. Positive allosteric modulators of metabotropic glutamate 1 receptor: characterization, mechanism of action, and binding site. Proc Natl Acad Sci U S A. 2001;98:13402–7. doi: 10.1073/pnas.231358298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov. 2004;3:711–5. doi: 10.1038/nrd1470. [DOI] [PubMed] [Google Scholar]
  157. Konieczny J, et al. Neuroscience. 2007c;145:611–20. doi: 10.1016/j.neuroscience.2006.12.006. [DOI] [PubMed] [Google Scholar]
  158. Krueger DD, Bear MF. Toward fulfilling the promise of molecular medicine in fragile X syndrome. Annual review of medicine. 2011;62:411–29. doi: 10.1146/annurev-med-061109-134644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Krystal JH, et al. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994;51:199–214. doi: 10.1001/archpsyc.1994.03950030035004. [DOI] [PubMed] [Google Scholar]
  160. Kulisevsky J, Poyurovsky M. Adenosine A2A-receptor antagonism and pathophysiology of Parkinson’s disease and drug-induced movement disorders. European neurology. 2012;67:4–11. doi: 10.1159/000331768. [DOI] [PubMed] [Google Scholar]
  161. Kumar N, et al. Metabotropic glutamate receptors (mGluRs) regulate noxious stimulus-induced glutamate release in the spinal cord dorsal horn of rats with neuropathic and inflammatory pain. Journal of neurochemistry. 2010;114:281–90. doi: 10.1111/j.1471-4159.2010.06761.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Kumaresan V, et al. Metabotropic glutamate receptor 5 (mGluR5) antagonists attenuate cocaine priming- and cue-induced reinstatement of cocaine seeking. Behavioural brain research. 2009;202:238–44. doi: 10.1016/j.bbr.2009.03.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Lahti AC, et al. Ketamine activates psychosis and alters limbic blood flow in schizophrenia. Neuroreport. 1995;6:869–72. doi: 10.1097/00001756-199504190-00011. [DOI] [PubMed] [Google Scholar]
  164. Langmead CJ, et al. Characterization of a CNS penetrant, selective M1 muscarinic receptor agonist, 77-LH-28-1. Br J Pharmacol. 2008a;154:1104–15. doi: 10.1038/bjp.2008.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Langmead CJ, et al. Muscarinic acetylcholine receptors as CNS drug targets. Pharmacol Ther. 2008b;117:232–43. doi: 10.1016/j.pharmthera.2007.09.009. [DOI] [PubMed] [Google Scholar]
  166. Langmead CJ, et al. Muscarinic acetylcholine receptors as CNS drug targets. Pharmacology & therapeutics. 2008c;117:232–43. doi: 10.1016/j.pharmthera.2007.09.009. [DOI] [PubMed] [Google Scholar]
  167. Lavieri RR, et al. Design, synthesis, and biological evaluation of halogenated N-(2-(4-oxo-1-phenyl-1,3,8-triazaspiro[4.5]decan-8-yl)ethyl)benzamides: discovery of an isoform-selective small molecule phospholipase D2 inhibitor. J Med Chem. 2010;53:6706–19. doi: 10.1021/jm100814g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Lavreysen H, et al. Metabotropic glutamate 1 receptor distribution and occupancy in the rat brain: a quantitative autoradiographic study using [3H]R214127. Neuropharmacology. 2004;46:609–19. doi: 10.1016/j.neuropharm.2003.11.014. [DOI] [PubMed] [Google Scholar]
  169. Lazareno S, et al. Allosteric effects of four stereoisomers of a fused indole ring system with 3H-N-methylscopolamine and acetylcholine at M1-M4 muscarinic receptors. Life Sci. 1999;64:519–26. doi: 10.1016/s0024-3205(98)00596-7. [DOI] [PubMed] [Google Scholar]
  170. Lazareno S, et al. Thiochrome enhances acetylcholine affinity at muscarinic M4 receptors: receptor subtype selectivity via cooperativity rather than affinity. Mol Pharmacol. 2004;65:257–66. doi: 10.1124/mol.65.1.257. [DOI] [PubMed] [Google Scholar]
  171. Lazareno S, et al. Subtype-selective positive cooperative interactions between brucine analogues and acetylcholine at muscarinic receptors: radioligand binding studies. Mol Pharmacol. 1998;53:573–89. doi: 10.1124/mol.53.3.573. [DOI] [PubMed] [Google Scholar]
  172. Le Poul E, et al. A potent and selective metabotropic glutamate receptor 4 positive allosteric modulator improves movement in rodent models of Parkinson’s disease. J Pharmacol Exp Ther. 2012;343:167–77. doi: 10.1124/jpet.112.196063. [DOI] [PubMed] [Google Scholar]
  173. Le U, et al. Discovery of a selective M(4) positive allosteric modulator based on the 3-amino-thieno[2,3-b]pyridine-2-carboxamide scaffold: development of ML253, a potent and brain penetrant compound that is active in a preclinical model of schizophrenia. Bioorg Med Chem Lett. 2013;23:346–50. doi: 10.1016/j.bmcl.2012.10.073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Leach K, et al. Molecular mechanisms of action and in vivo validation of an M4 muscarinic acetylcholine receptor allosteric modulator with potential antipsychotic properties. Neuropsychopharmacology. 2010;35:855–69. doi: 10.1038/npp.2009.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Lebois EP, et al. Discovery and characterization of novel subtype-selective allosteric agonists for the investigation of M(1) receptor function in the central nervous system. ACS Chem Neurosci. 2010;1:104–121. doi: 10.1021/cn900003h. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Lebon G, et al. Mutagenic mapping suggests a novel binding mode for selective agonists of M1 muscarinic acetylcholine receptors. Mol Pharmacol. 2009;75:331–41. doi: 10.1124/mol.108.050963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Lecourtier L, et al. Positive allosteric modulation of metabotropic glutamate 5 (mGlu5) receptors reverses N-Methyl-D-aspartate antagonist-induced alteration of neuronal firing in prefrontal cortex. Biol Psychiatry. 2007;62:739–46. doi: 10.1016/j.biopsych.2006.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Lee B, et al. Attenuation of behavioral effects of cocaine by the Metabotropic Glutamate Receptor 5 Antagonist 2-Methyl-6-(phenylethynyl)-pyridine in squirrel monkeys: comparison with dizocilpine. The Journal of pharmacology and experimental therapeutics. 2005;312:1232–40. doi: 10.1124/jpet.104.078733. [DOI] [PubMed] [Google Scholar]
  179. Lee CM, Farde L. Using positron emission tomography to facilitate CNS drug development. Trends in pharmacological sciences. 2006;27:310–6. doi: 10.1016/j.tips.2006.04.004. [DOI] [PubMed] [Google Scholar]
  180. Levandis G, et al. Systemic administration of an mGluR5 antagonist, but not unilateral subthalamic lesion, counteracts l-DOPA-induced dyskinesias in a rodent model of Parkinson’s disease. Neurobiol Dis. 2008;29:161–8. doi: 10.1016/j.nbd.2007.08.011. [DOI] [PubMed] [Google Scholar]
  181. Levey AI, et al. Expression of m1-m4 muscarinic acetylcholine receptor proteins in rat hippocampus and regulation by cholinergic innervation. J Neurosci. 1995;15:4077–92. doi: 10.1523/JNEUROSCI.15-05-04077.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Levey AI, et al. Identification and localization of muscarinic acetylcholine receptor proteins in brain with subtype-specific antibodies. J Neurosci. 1991;11:3218–26. doi: 10.1523/JNEUROSCI.11-10-03218.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Lewis JA, et al. Allosteric modulation of kinases and GPCRs: design principles and structural diversity. Curr Opin Chem Biol. 2008;12:269–80. doi: 10.1016/j.cbpa.2008.02.014. [DOI] [PubMed] [Google Scholar]
  184. Li X, et al. Metabotropic glutamate 5 receptor antagonism is associated with antidepressant-like effects in mice. The Journal of pharmacology and experimental therapeutics. 2006;319:254–9. doi: 10.1124/jpet.106.103143. [DOI] [PubMed] [Google Scholar]
  185. Liao DL, et al. Association of muscarinic m1 receptor genetic polymorphisms with psychiatric symptoms and cognitive function in schizophrenic patients. Neuropsychobiology. 2003;48:72–6. doi: 10.1159/000072880. [DOI] [PubMed] [Google Scholar]
  186. Liechti ME, Markou A. Interactive effects of the mGlu5 receptor antagonist MPEP and the mGlu2/3 receptor antagonist LY341495 on nicotine self-administration and reward deficits associated with nicotine withdrawal in rats. European journal of pharmacology. 2007;554:164–74. doi: 10.1016/j.ejphar.2006.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Lindemann L, et al. CTEP: a novel, potent, long-acting, and orally bioavailable metabotropic glutamate receptor 5 inhibitor. The Journal of pharmacology and experimental therapeutics. 2011;339:474–86. doi: 10.1124/jpet.111.185660. [DOI] [PubMed] [Google Scholar]
  188. Lindsley CW, et al. Progress towards validating the NMDA receptor hypofunction hypothesis of schizophrenia. Curr Top Med Chem. 2006;6:771–85. doi: 10.2174/156802606777057599. [DOI] [PubMed] [Google Scholar]
  189. Lindsley CW, et al. Allosteric Akt (PKB) inhibitors: discovery and SAR of isozyme selective inhibitors. Bioorg Med Chem Lett. 2005;15:761–4. doi: 10.1016/j.bmcl.2004.11.011. [DOI] [PubMed] [Google Scholar]
  190. Liu F, et al. ADX47273 [S-(4-fluoro-phenyl)-{3-[3-(4-fluoro-phenyl)-[1,2,4]-oxadiazol-5-yl]-piperidin-1-yl}-methanone]: a novel metabotropic glutamate receptor 5-selective positive allosteric modulator with preclinical antipsychotic-like and procognitive activities. The Journal of pharmacology and experimental therapeutics. 2008a;327:827–39. doi: 10.1124/jpet.108.136580. [DOI] [PubMed] [Google Scholar]
  191. Liu F, et al. ADX47273 [S-(4-fluoro-phenyl)-{3-[3-(4-fluoro-phenyl)-[1,2,4]-oxadiazol-5-yl]-piperidin-1-yl}-methanone]: a novel metabotropic glutamate receptor 5-selective positive allosteric modulator with preclinical antipsychotic-like and procognitive activities. J Pharmacol Exp Ther. 2008b;327:827–39. doi: 10.1124/jpet.108.136580. [DOI] [PubMed] [Google Scholar]
  192. Lockhart IA, et al. Safety and tolerability of donepezil, rivastigmine and galantamine for patients with Alzheimer’s disease: systematic review of the ‘real-world’ evidence. Dement Geriatr Cogn Disord. 2009;28:389–403. doi: 10.1159/000255578. [DOI] [PubMed] [Google Scholar]
  193. Lopez S, et al. Targeting group III metabotropic glutamate receptors produces complex behavioral effects in rodent models of Parkinson’s disease. J Neurosci. 2007;27:6701–11. doi: 10.1523/JNEUROSCI.0299-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Lorrain DS, et al. Group II mGlu receptor activation suppresses norepinephrine release in the ventral hippocampus and locomotor responses to acute ketamine challenge. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2003;28:1622–32. doi: 10.1038/sj.npp.1300238. [DOI] [PubMed] [Google Scholar]
  195. Ma L, et al. Selective activation of the M1 muscarinic acetylcholine receptor achieved by allosteric potentiation. Proc Natl Acad Sci U S A. 2009;106:15950–5. doi: 10.1073/pnas.0900903106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Mabire D, et al. Synthesis, structure-activity relationship, and receptor pharmacology of a new series of quinoline derivatives acting as selective, noncompetitive mGlu1 antagonists. Journal of medicinal chemistry. 2005;48:2134–53. doi: 10.1021/jm049499o. [DOI] [PubMed] [Google Scholar]
  197. MacInnes N, et al. Activation of group III metabotropic glutamate receptors in selected regions of the basal ganglia alleviates akinesia in the reserpine-treated rat. Br J Pharmacol. 2004;141:15–22. doi: 10.1038/sj.bjp.0705566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Maj M, et al. (-)-PHCCC, a positive allosteric modulator of mGluR4: characterization, mechanism of action, and neuroprotection. Neuropharmacology. 2003;45:895–906. doi: 10.1016/s0028-3908(03)00271-5. [DOI] [PubMed] [Google Scholar]
  199. Malhotra AK, et al. Ketamine-induced exacerbation of psychotic symptoms and cognitive impairment in neuroleptic-free schizophrenics. Neuropsychopharmacology. 1997;17:141–50. doi: 10.1016/S0893-133X(97)00036-5. [DOI] [PubMed] [Google Scholar]
  200. Malhotra AK, et al. NMDA receptor function and human cognition: the effects of ketamine in healthy volunteers. Neuropsychopharmacology. 1996;14:301–7. doi: 10.1016/0893-133X(95)00137-3. [DOI] [PubMed] [Google Scholar]
  201. Marek GJ. Metabotropic glutamate2/3 (mGlu2/3) receptors, schizophrenia and cognition. Eur J Pharmacol. 2010;639:81–90. doi: 10.1016/j.ejphar.2010.02.058. [DOI] [PubMed] [Google Scholar]
  202. Marik JSBS, Williams S-P, van Bruggen N. New imaging paradigms in drug development: the PET imaging approach. In: Beckmann WBN, Lam K, Timmerman H, editors. Drug Discovery Today: Technologies. Elsevier; 2011. p. 2011. [DOI] [PubMed] [Google Scholar]
  203. Marino MJ, et al. Allosteric modulation of group III metabotropic glutamate receptor 4: a potential approach to Parkinson’s disease treatment. Proc Natl Acad Sci U S A. 2003;100:13668–73. doi: 10.1073/pnas.1835724100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Marlo JE, et al. Discovery and characterization of novel allosteric potentiators of M1 muscarinic receptors reveals multiple modes of activity. Mol Pharmacol. 2009;75:577–88. doi: 10.1124/mol.108.052886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Marsden CD, Parkes JD. Success and problems of long-term levodopa therapy in Parkinson’s disease. Lancet. 1977;1:345–9. doi: 10.1016/s0140-6736(77)91146-1. [DOI] [PubMed] [Google Scholar]
  206. Martella G, et al. Impairment of bidirectional synaptic plasticity in the striatum of a mouse model of DYT1 dystonia: role of endogenous acetylcholine. Brain. 2009;132:2336–49. doi: 10.1093/brain/awp194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Martin LJ, et al. Cellular localization of a metabotropic glutamate receptor in rat brain. Neuron. 1992;9:259–70. doi: 10.1016/0896-6273(92)90165-a. [DOI] [PubMed] [Google Scholar]
  208. Martino G, et al. The M1/M4 preferring agonist xanomeline is analgesic in rodent models of chronic inflammatory and neuropathic pain via central site of action. Pain. 2011;152:2852–60. doi: 10.1016/j.pain.2011.09.017. [DOI] [PubMed] [Google Scholar]
  209. Mathiesen JM, et al. Identification of indole derivatives exclusively interfering with a G protein-independent signaling pathway of the prostaglandin D2 receptor CRTH2. Molecular pharmacology. 2005;68:393–402. doi: 10.1124/mol.104.010520. [DOI] [PubMed] [Google Scholar]
  210. Matsui T, Kita H. Activation of group III metabotropic glutamate receptors presynaptically reduces both GABAergic and glutamatergic transmission in the rat globus pallidus. Neuroscience. 2003;122:727–37. doi: 10.1016/j.neuroscience.2003.08.032. [DOI] [PubMed] [Google Scholar]
  211. May LT, Christopoulos A. Allosteric modulators of G-protein-coupled receptors. Current opinion in pharmacology. 2003;3:551–6. doi: 10.1016/s1471-4892(03)00107-3. [DOI] [PubMed] [Google Scholar]
  212. Meissner WG, et al. Priorities in Parkinson’s disease research. Nat Rev Drug Discov. 2011;10:377–93. doi: 10.1038/nrd3430. [DOI] [PubMed] [Google Scholar]
  213. Mela F, et al. Antagonism of metabotropic glutamate receptor type 5 attenuates l-DOPA-induced dyskinesia and its molecular and neurochemical correlates in a rat model of Parkinson’s disease. J Neurochem. 2007;101:483–97. doi: 10.1111/j.1471-4159.2007.04456.x. [DOI] [PubMed] [Google Scholar]
  214. Melancon BJ, et al. Allosteric modulation of seven transmembrane spanning receptors: theory, practice, and opportunities for central nervous system drug discovery. J Med Chem. 2012;55:1445–64. doi: 10.1021/jm201139r. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Melancon BJ, et al. Isatin replacements applied to the highly selective, muscarinic M1 PAM ML137: continued optimization of an MLPCN probe molecule. Bioorg Med Chem Lett. 2013;23:412–6. doi: 10.1016/j.bmcl.2012.11.092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Michalon A, et al. Chronic pharmacological mGlu5 inhibition corrects fragile X in adult mice. Neuron. 2012;74:49–56. doi: 10.1016/j.neuron.2012.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Miyakawa T, et al. Hyperactivity and intact hippocampus-dependent learning in mice lacking the M1 muscarinic acetylcholine receptor. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2001;21:5239–50. doi: 10.1523/JNEUROSCI.21-14-05239.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Moghaddam B. Targeting metabotropic glutamate receptors for treatment of the cognitive symptoms of schizophrenia. Psychopharmacology (Berl) 2004;174:39–44. doi: 10.1007/s00213-004-1792-z. [DOI] [PubMed] [Google Scholar]
  219. Moghaddam B, Adams BW. Reversal of phencyclidine effects by a group II metabotropic glutamate receptor agonist in rats. Science. 1998;281:1349–52. doi: 10.1126/science.281.5381.1349. [DOI] [PubMed] [Google Scholar]
  220. Mohler H, et al. A new benzodiazepine pharmacology. J Pharmacol Exp Ther. 2002;300:2–8. doi: 10.1124/jpet.300.1.2. [DOI] [PubMed] [Google Scholar]
  221. Montana MC, et al. The metabotropic glutamate receptor subtype 5 antagonist fenobam is analgesic and has improved in vivo selectivity compared with the prototypical antagonist 2-methyl-6-(phenylethynyl)-pyridine. The Journal of pharmacology and experimental therapeutics. 2009;330:834–43. doi: 10.1124/jpet.109.154138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Morin N, et al. Effect of the metabotropic glutamate receptor type 5 antagonists MPEP and MTEP in parkinsonian monkeys. Neuropharmacology. 2010;58:981–6. doi: 10.1016/j.neuropharm.2009.12.024. [DOI] [PubMed] [Google Scholar]
  223. Morin N, et al. MPEP, an mGlu5 receptor antagonist, reduces the development of L-DOPA-induced motor complications in de novo parkinsonian monkeys: biochemical correlates. Neuropharmacology. 2013a;66:355–64. doi: 10.1016/j.neuropharm.2012.07.036. [DOI] [PubMed] [Google Scholar]
  224. Morin N, et al. Chronic treatment with MPEP, an mGlu5 receptor antagonist, normalizes basal ganglia glutamate neurotransmission in l-DOPA-treated parkinsonian monkeys. Neuropharmacology. 2013b;73C:216–231. doi: 10.1016/j.neuropharm.2013.05.028. [DOI] [PubMed] [Google Scholar]
  225. Morishima Y, et al. Enhanced cocaine responsiveness and impaired motor coordination in metabotropic glutamate receptor subtype 2 knockout mice. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:4170–5. doi: 10.1073/pnas.0500914102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Murray TK, et al. Evaluation of the mGluR2/3 agonist LY379268 in rodent models of Parkinson’s disease. Pharmacol Biochem Behav. 2002;73:455–66. doi: 10.1016/s0091-3057(02)00842-0. [DOI] [PubMed] [Google Scholar]
  227. Nawaratne V, et al. Structural determinants of allosteric agonism and modulation at the M4 muscarinic acetylcholine receptor: identification of ligand-specific and global activation mechanisms. J Biol Chem. 2010;285:19012–21. doi: 10.1074/jbc.M110.125096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Nemeth EF, et al. Pharmacodynamics of the type II calcimimetic compound cinacalcet HCl. J Pharmacol Exp Ther. 2004;308:627–35. doi: 10.1124/jpet.103.057273. [DOI] [PubMed] [Google Scholar]
  229. Newcomer JW, et al. Ketamine-induced NMDA receptor hypofunction as a model of memory impairment and psychosis. Neuropsychopharmacology. 1999;20:106–18. doi: 10.1016/S0893-133X(98)00067-0. [DOI] [PubMed] [Google Scholar]
  230. Nicoletti F, et al. Metabotropic glutamate receptors: from the workbench to the bedside. Neuropharmacology. 2011;60:1017–41. doi: 10.1016/j.neuropharm.2010.10.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. Niswender CM, Conn PJ. Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu Rev Pharmacol Toxicol. 2010;50:295–322. doi: 10.1146/annurev.pharmtox.011008.145533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Niswender CM, et al. Context-dependent pharmacology exhibited by negative allosteric modulators of metabotropic glutamate receptor 7. Molecular pharmacology. 2010a;77:459–68. doi: 10.1124/mol.109.058768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Niswender CM, et al. Context-dependent pharmacology exhibited by negative allosteric modulators of metabotropic glutamate receptor 7. Mol Pharmacol. 2010b;77:459–68. doi: 10.1124/mol.109.058768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Niswender CM, et al. Discovery, characterization, and antiparkinsonian effect of novel positive allosteric modulators of metabotropic glutamate receptor 4. Mol Pharmacol. 2008;74:1345–58. doi: 10.1124/mol.108.049551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Noetzel MJ, et al. A novel metabotropic glutamate receptor 5 positive allosteric modulator acts at a unique site and confers stimulus bias to mGlu5 signaling. Mol Pharmacol. 2013;83:835–47. doi: 10.1124/mol.112.082891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Osterholm RK, Camoriano JK. Transdermal scopolamine psychosis. JAMA. 1982;247:3081. [PubMed] [Google Scholar]
  237. Ott BR, Lannon MC. Exacerbation of parkinsonism by tacrine. Clin Neuropharmacol. 1992;15:322–5. doi: 10.1097/00002826-199208000-00008. [DOI] [PubMed] [Google Scholar]
  238. Ouattara B, et al. Effect of L-Dopa on metabotropic glutamate receptor 5 in the brain of parkinsonian monkeys. J Neurochem. 2010;113:715–24. doi: 10.1111/j.1471-4159.2010.06635.x. [DOI] [PubMed] [Google Scholar]
  239. Palucha-Poniewiera A, et al. On the mechanism of the antidepressant-like action of group II mGlu receptor antagonist, MGS0039. Psychopharmacology (Berl) 2010;212:523–35. doi: 10.1007/s00213-010-1978-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Palucha A, et al. Potential antidepressant-like effect of MTEP, a potent and highly selective mGluR5 antagonist. Pharmacology, biochemistry, and behavior. 2005;81:901–6. doi: 10.1016/j.pbb.2005.06.015. [DOI] [PubMed] [Google Scholar]
  241. Parmentier-Batteur S, et al. Mechanism based neurotoxicity of mGlu5 positive allosteric modulators - Development challenges for a promising novel antipsychotic target. Neuropharmacology. 2013 doi: 10.1016/j.neuropharm.2012.12.003. [DOI] [PubMed] [Google Scholar]
  242. Parwani A, et al. The effects of a subanesthetic dose of ketamine on verbal memory in normal volunteers. Psychopharmacology (Berl) 2005;183:265–74. doi: 10.1007/s00213-005-0177-2. [DOI] [PubMed] [Google Scholar]
  243. Patil ST, et al. Activation of mGlu2/3 receptors as a new approach to treat schizophrenia: a randomized Phase 2 clinical trial. Nat Med. 2007;13:1102–7. doi: 10.1038/nm1632. [DOI] [PubMed] [Google Scholar]
  244. Pecknold JC, et al. Treatment of anxiety using fenobam (a nonbenzodiazepine) in a double-blind standard (diazepam) placebo-controlled study. J Clin Psychopharmacol. 1982a;2:129–33. [PubMed] [Google Scholar]
  245. Pecknold JC, et al. Treatment of anxiety using fenobam (a nonbenzodiazepine) in a double-blind standard (diazepam) placebo-controlled study. Journal of clinical psychopharmacology. 1982b;2:129–33. [PubMed] [Google Scholar]
  246. Pellegrino D, et al. Modulation of dopaminergic and glutamatergic brain function: PET studies on parkinsonian rats. J Nucl Med. 2007;48:1147–53. doi: 10.2967/jnumed.106.037796. [DOI] [PubMed] [Google Scholar]
  247. Pilc A, et al. Glutamate-based antidepressants: preclinical psychopharmacology. Biol Psychiatry. 2013;73:1125–32. doi: 10.1016/j.biopsych.2013.01.021. [DOI] [PubMed] [Google Scholar]
  248. Pisani A, et al. Re-emergence of striatal cholinergic interneurons in movement disorders. Trends Neurosci. 2007;30:545–53. doi: 10.1016/j.tins.2007.07.008. [DOI] [PubMed] [Google Scholar]
  249. Pisani A, et al. Metabotropic glutamate receptor 5 mediates the potentiation of N-methyl-D-aspartate responses in medium spiny striatal neurons. Neuroscience. 2001;106:579–87. doi: 10.1016/s0306-4522(01)00297-4. [DOI] [PubMed] [Google Scholar]
  250. Platt DM, et al. Attenuation of cocaine self-administration in squirrel monkeys following repeated administration of the mGluR5 antagonist MPEP: comparison with dizocilpine. Psychopharmacology. 2008;200:167–76. doi: 10.1007/s00213-008-1191-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Porter RH, et al. Fenobam: a clinically validated nonbenzodiazepine anxiolytic is a potent, selective, and noncompetitive mGlu5 receptor antagonist with inverse agonist activity. The Journal of pharmacology and experimental therapeutics. 2005a;315:711–21. doi: 10.1124/jpet.105.089839. [DOI] [PubMed] [Google Scholar]
  252. Porter RH, et al. Fenobam: a clinically validated nonbenzodiazepine anxiolytic is a potent, selective, and noncompetitive mGlu5 receptor antagonist with inverse agonist activity. J Pharmacol Exp Ther. 2005b;315:711–21. doi: 10.1124/jpet.105.089839. [DOI] [PubMed] [Google Scholar]
  253. Poslusney MS, et al. Spirocyclic replacements for the isatin in the highly selective, muscarinic M1 PAM ML137: the continued optimization of an MLPCN probe molecule. Bioorg Med Chem Lett. 2013;23:1860–4. doi: 10.1016/j.bmcl.2013.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Prashanth LK, et al. l-Dopa-induced dyskinesia-clinical presentation, genetics, and treatment. International review of neurobiology. 2011;98:31–54. doi: 10.1016/B978-0-12-381328-2.00002-X. [DOI] [PubMed] [Google Scholar]
  255. Przedborski S. Pathogenesis of nigral cell death in Parkinson’s disease. Parkinsonism Relat Disord. 2005;11(Suppl 1):S3–7. doi: 10.1016/j.parkreldis.2004.10.012. [DOI] [PubMed] [Google Scholar]
  256. Raffa RB. The M5 muscarinic receptor as possible target for treatment of drug abuse. Journal of clinical pharmacy and therapeutics. 2009;34:623–9. doi: 10.1111/j.1365-2710.2009.01059.x. [DOI] [PubMed] [Google Scholar]
  257. Reid PR, et al. Discovery and optimization of a novel, selective and brain penetrant M1 positive allosteric modulator (PAM): the development of ML169, an MLPCN probe. Bioorg Med Chem Lett. 2011;21:2697–701. doi: 10.1016/j.bmcl.2010.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Robichaud AJ, et al. Recent progress on the identification of metabotropic glutamate 4 receptor ligands and their potential utility as CNS therapeutics. ACS Chem Neurosci. 2011;2:433–49. doi: 10.1021/cn200043e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Robinson L, et al. Involvement of the cholinergic system in conditioning and perceptual memory. Behavioural brain research. 2011;221:443–65. doi: 10.1016/j.bbr.2011.01.055. [DOI] [PubMed] [Google Scholar]
  260. Rodriguez AL, et al. 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;78:1105–23. doi: 10.1124/mol.110.067207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Rodriguez AL, et al. A close structural analog of 2-methyl-6-(phenylethynyl)-pyridine acts as a neutral allosteric site ligand on metabotropic glutamate receptor subtype 5 and blocks the effects of multiple allosteric modulators. Mol Pharmacol. 2005a;68:1793–802. doi: 10.1124/mol.105.016139. [DOI] [PubMed] [Google Scholar]
  262. Rodriguez AL, et al. A close structural analog of 2-methyl-6-(phenylethynyl)-pyridine acts as a neutral allosteric site ligand on metabotropic glutamate receptor subtype 5 and blocks the effects of multiple allosteric modulators. Molecular pharmacology. 2005b;68:1793–802. doi: 10.1124/mol.105.016139. [DOI] [PubMed] [Google Scholar]
  263. Rook JM, et al. Unique signaling profiles of positive allosteric modulators of metabotropic glutamate receptor subtype 5 determine differences in in vivo activity. Biol Psychiatry. 2013a;73:501–9. doi: 10.1016/j.biopsych.2012.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Rook JM, et al. Unique signaling profiles of positive allosteric modulators of metabotropic glutamate receptor subtype 5 determine differences in in vivo activity. Biological psychiatry. 2013b;73:501–9. doi: 10.1016/j.biopsych.2012.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Rosenbrock H, et al. Functional interaction of metabotropic glutamate receptor 5 and NMDA-receptor by a metabotropic glutamate receptor 5 positive allosteric modulator. Eur J Pharmacol. 2010;639:40–6. doi: 10.1016/j.ejphar.2010.02.057. [DOI] [PubMed] [Google Scholar]
  266. Rowland LM. Subanesthetic ketamine: how it alters physiology and behavior in humans. Aviat Space Environ Med. 2005;76:C52–8. [PubMed] [Google Scholar]
  267. Rylander D, et al. A mGluR5 antagonist under clinical development improves L-DOPA-induced dyskinesia in parkinsonian rats and monkeys. Neurobiol Dis. 2010;39:352–61. doi: 10.1016/j.nbd.2010.05.001. [DOI] [PubMed] [Google Scholar]
  268. Rylander D, et al. Pharmacological modulation of glutamate transmission in a rat model of L-DOPA-induced dyskinesia: effects on motor behavior and striatal nuclear signaling. J Pharmacol Exp Ther. 2009;330:227–35. doi: 10.1124/jpet.108.150425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Sachpatzidis A, et al. Identification of allosteric peptide agonists of CXCR4. J Biol Chem. 2003;278:896–907. doi: 10.1074/jbc.M204667200. [DOI] [PubMed] [Google Scholar]
  270. Salamone JD, et al. Neostriatal muscarinic receptor subtypes involved in the generation of tremulous jaw movements in rodents implications for cholinergic involvement in parkinsonism. Life Sci. 2001;68:2579–84. doi: 10.1016/s0024-3205(01)01055-4. [DOI] [PubMed] [Google Scholar]
  271. Samadi P, et al. mGluR5 metabotropic glutamate receptors and dyskinesias in MPTP monkeys. Neurobiol Aging. 2008;29:1040–51. doi: 10.1016/j.neurobiolaging.2007.02.005. [DOI] [PubMed] [Google Scholar]
  272. Sanacora G, et al. Subtype-specific alterations of gamma-aminobutyric acid and glutamate in patients with major depression. Archives of general psychiatry. 2004;61:705–13. doi: 10.1001/archpsyc.61.7.705. [DOI] [PubMed] [Google Scholar]
  273. Santoro MR, et al. Molecular mechanisms of fragile X syndrome: a twenty-year perspective. Annual review of pathology. 2012;7:219–45. doi: 10.1146/annurev-pathol-011811-132457. [DOI] [PubMed] [Google Scholar]
  274. Schkeryantz JM, et al. Prospects for metabotropic glutamate 1 receptor antagonists in the treatment of neuropathic pain. Journal of medicinal chemistry. 2007;50:2563–8. doi: 10.1021/jm060950g. [DOI] [PubMed] [Google Scholar]
  275. Schlumberger C, et al. Effects of a positive allosteric modulator of mGluR5 ADX47273 on conditioned avoidance response and PCP-induced hyperlocomotion in the rat as models for schizophrenia. Pharmacol Biochem Behav. 2010;95:23–30. doi: 10.1016/j.pbb.2009.12.002. [DOI] [PubMed] [Google Scholar]
  276. Schlumberger C, et al. 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. 2009;623:73–83. doi: 10.1016/j.ejphar.2009.09.006. [DOI] [PubMed] [Google Scholar]
  277. Schoepp DD, Marek GJ. Preclinical pharmacology of mGlu2/3 receptor agonists: novel agents for schizophrenia? Curr Drug Targets CNS Neurol Disord. 2002;1:215–25. doi: 10.2174/1568007024606177. [DOI] [PubMed] [Google Scholar]
  278. Schoepp DD, et al. LY354740, an mGlu2/3 receptor agonist as a novel approach to treat anxiety/stress. Stress. 2003;6:189–97. doi: 10.1080/1025389031000146773. [DOI] [PubMed] [Google Scholar]
  279. Scott SA, et al. Design of isoform-selective phospholipase D inhibitors that modulate cancer cell invasiveness. Nat Chem Biol. 2009;5:108–17. doi: 10.1038/nchembio.140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Sheffler DJ, Conn PJ. Allosteric potentiators of metabotropic glutamate receptor subtype 1a differentially modulate independent signaling pathways in baby hamster kidney cells. Neuropharmacology. 2008;55:419–27. doi: 10.1016/j.neuropharm.2008.06.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  281. Sheffler DJ, et al. Development of a novel, CNS-penetrant, metabotropic glutamate receptor 3 (mGlu3) NAM probe (ML289) derived from a closely related mGlu5 PAM. Bioorganic & medicinal chemistry letters. 2012a;22:3921–5. doi: 10.1016/j.bmcl.2012.04.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Sheffler DJ, et al. Development of a novel, CNS-penetrant, metabotropic glutamate receptor 3 (mGlu3) NAM probe (ML289) derived from a closely related mGlu5 PAM. Bioorg Med Chem Lett. 2012b;22:3921–5. doi: 10.1016/j.bmcl.2012.04.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  283. Shekhar A, et al. Selective muscarinic receptor agonist xanomeline as a novel treatment approach for schizophrenia. Am J Psychiatry. 2008;165:1033–9. doi: 10.1176/appi.ajp.2008.06091591. [DOI] [PubMed] [Google Scholar]
  284. Shigemoto R, et al. Immunohistochemical localization of a metabotropic glutamate receptor, mGluR5, in the rat brain. Neurosci Lett. 1993;163:53–7. doi: 10.1016/0304-3940(93)90227-c. [DOI] [PubMed] [Google Scholar]
  285. Shirey JK, et al. A selective allosteric potentiator of the M1 muscarinic acetylcholine receptor increases activity of medial prefrontal cortical neurons and restores impairments in reversal learning. J Neurosci. 2009;29:14271–86. doi: 10.1523/JNEUROSCI.3930-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Shirey JK, et al. An allosteric potentiator of M4 mAChR modulates hippocampal synaptic transmission. Nat Chem Biol. 2008;4:42–50. doi: 10.1038/nchembio.2007.55. [DOI] [PubMed] [Google Scholar]
  287. Sibille P, et al. Synthesis and biological evaluation of 1-amino-2-phosphonomethylcyclopropanecarboxylic acids, new group III metabotropic glutamate receptor agonists. J Med Chem. 2007;50:3585–95. doi: 10.1021/jm070262c. [DOI] [PubMed] [Google Scholar]
  288. Silverman JL, et al. Negative allosteric modulation of the mGluR5 receptor reduces repetitive behaviors and rescues social deficits in mouse models of autism. Sci Transl Med. 2012;4:131ra51. doi: 10.1126/scitranslmed.3003501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Slawinska A, et al. The antipsychotic-like effects of mGlu4 receptor positive allosteric modulators in rodents. Br J Pharmacol. 2013a doi: 10.1111/bph.12254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  290. Slawinska A, et al. Anxiolytic- but not antidepressant-like activity of Lu AF21934, a novel, selective positive allosteric modulator of the mGlu(4) receptor. Neuropharmacology. 2013b;66:225–35. doi: 10.1016/j.neuropharm.2012.05.001. [DOI] [PubMed] [Google Scholar]
  291. Smith R, et al. Cholinergic neuronal defect without cell loss in Huntington’s disease. Hum Mol Genet. 2006;15:3119–31. doi: 10.1093/hmg/ddl252. [DOI] [PubMed] [Google Scholar]
  292. Sofuoglu M, Mooney M. Cholinergic functioning in stimulant addiction: implications for medications development. CNS Drugs. 2009;23:939–52. doi: 10.2165/11310920-000000000-00000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Spalding TA, et al. Discovery of an ectopic activation site on the M(1) muscarinic receptor. Mol Pharmacol. 2002;61:1297–302. doi: 10.1124/mol.61.6.1297. [DOI] [PubMed] [Google Scholar]
  294. Stefani MR, Moghaddam B. Activation of type 5 metabotropic glutamate receptors attenuates deficits in cognitive flexibility induced by NMDA receptor blockade. European journal of pharmacology. 2010a;639:26–32. doi: 10.1016/j.ejphar.2010.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  295. Stefani MR, Moghaddam B. Activation of type 5 metabotropic glutamate receptors attenuates deficits in cognitive flexibility induced by NMDA receptor blockade. Eur J Pharmacol. 2010b;639:26–32. doi: 10.1016/j.ejphar.2010.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Sur C, et al. N-desmethylclozapine, an allosteric agonist at muscarinic 1 receptor, potentiates N-methyl-D-aspartate receptor activity. Proc Natl Acad Sci U S A. 2003;100:13674–9. doi: 10.1073/pnas.1835612100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Swanson CJ, et al. Metabotropic glutamate receptors as novel targets for anxiety and stress disorders. Nature reviews. Drug discovery. 2005;4:131–44. doi: 10.1038/nrd1630. [DOI] [PubMed] [Google Scholar]
  298. Tabolacci E, et al. The mGluR5 antagonist AFQ056 does not affect methylation and transcription of the mutant FMR1 gene in vitro. BMC medical genetics. 2012;13:13. doi: 10.1186/1471-2350-13-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  299. Tandon R, et al. Muscarinic cholinergic hyperactivity in schizophrenia. Relationship to positive and negative symptoms. Schizophr Res. 1991;4:23–30. doi: 10.1016/0920-9964(91)90006-d. [DOI] [PubMed] [Google Scholar]
  300. Taylor DL, et al. Activation of microglial group III metabotropic glutamate receptors protects neurons against microglial neurotoxicity. J Neurosci. 2003;23:2150–60. doi: 10.1523/JNEUROSCI.23-06-02150.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  301. Thomas AM, et al. Group I metabotropic glutamate receptor antagonists alter select behaviors in a mouse model for fragile X syndrome. Psychopharmacology. 2012;219:47–58. doi: 10.1007/s00213-011-2375-4. [DOI] [PubMed] [Google Scholar]
  302. Thomas NK, et al. (S)-3,4-DCPG, a potent and selective mGlu8a receptor agonist, activates metabotropic glutamate receptors on primary afferent terminals in the neonatal rat spinal cord. Neuropharmacology. 2001;40:311–8. doi: 10.1016/s0028-3908(00)00169-6. [DOI] [PubMed] [Google Scholar]
  303. Thomsen M, et al. Attenuation of cocaine’s reinforcing and discriminative stimulus effects via muscarinic M1 acetylcholine receptor stimulation. J Pharmacol Exp Ther. 2010;332:959–69. doi: 10.1124/jpet.109.162057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  304. Thomsen M, et al. Contribution of both M1 and M4 receptors to muscarinic agonist-mediated attenuation of the cocaine discriminative stimulus in mice. Psychopharmacology (Berl) 2012;220:673–85. doi: 10.1007/s00213-011-2516-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  305. Tsai P, Sahin M. Mechanisms of neurocognitive dysfunction and therapeutic considerations in tuberous sclerosis complex. Current opinion in neurology. 2011;24:106–13. doi: 10.1097/WCO.0b013e32834451c4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  306. Ugolini A, et al. AMN082, an allosteric mGluR7 agonist that inhibits afferent glutamatergic transmission in rat basolateral amygdala. Neuropharmacology. 2008;55:532–6. doi: 10.1016/j.neuropharm.2008.04.020. [DOI] [PubMed] [Google Scholar]
  307. Uslaner JM, et al. The muscarinic M1 receptor positive allosteric modulator PQCA improves cognitive measures in rat, cynomolgus macaque, and rhesus macaque. Psychopharmacology (Berl) 2013;225:21–30. doi: 10.1007/s00213-012-2788-8. [DOI] [PubMed] [Google Scholar]
  308. Utley T, et al. Synthesis and SAR of a novel metabotropic glutamate receptor 4 (mGlu4) antagonist: unexpected ‘molecular switch’ from a closely related mGlu4 positive allosteric modulator. Bioorg Med Chem Lett. 2011;21:6955–9. doi: 10.1016/j.bmcl.2011.09.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Valenti O, et al. Group III metabotropic glutamate-receptor-mediated modulation of excitatory transmission in rodent substantia nigra pars compacta dopamine neurons. J Pharmacol Exp Ther. 2005;313:1296–304. doi: 10.1124/jpet.104.080481. [DOI] [PubMed] [Google Scholar]
  310. Valenti O, et al. Modulation of excitatory transmission onto midbrain dopaminergic neurons of the rat by activation of group III metabotropic glutamate receptors. Ann N Y Acad Sci. 2003a;1003:479–80. doi: 10.1196/annals.1300.058. [DOI] [PubMed] [Google Scholar]
  311. Valenti O, et al. Group III metabotropic glutamate receptor-mediated modulation of the striatopallidal synapse. J Neurosci. 2003b;23:7218–26. doi: 10.1523/JNEUROSCI.23-18-07218.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. van Bon BW, et al. The phenotype of recurrent 10q22q23 deletions and duplications. Eur J Hum Genet. 2011;19:400–8. doi: 10.1038/ejhg.2010.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  313. Vanover KE, et al. Antipsychotic-like behavioral effects and cognitive enhancement by a potent and selective muscarinic M-sub-1 receptor agonist, AC-260584. Behav Neurosci. 2008;122:570–5. doi: 10.1037/0735-7044.122.3.570. [DOI] [PubMed] [Google Scholar]
  314. Vardigan JD, et al. 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;95:223–9. doi: 10.1016/j.pbb.2010.01.010. [DOI] [PubMed] [Google Scholar]
  315. Varty GB, et al. The effects of adenosine A2A receptor antagonists on haloperidol-induced movement disorders in primates. Psychopharmacology. 2008;200:393–401. doi: 10.1007/s00213-008-1214-8. [DOI] [PubMed] [Google Scholar]
  316. Vilaro MT, et al. Localization of m5 muscarinic receptor mRNA in rat brain examined by in situ hybridization histochemistry. Neuroscience letters. 1990;114:154–9. doi: 10.1016/0304-3940(90)90064-g. [DOI] [PubMed] [Google Scholar]
  317. Wang H, et al. Decreased amphetamine-induced locomotion and improved latent inhibition in mice mutant for the M5 muscarinic receptor gene found in the human 15q schizophrenia region. Neuropsychopharmacology. 2004;29:2126–39. doi: 10.1038/sj.npp.1300502. [DOI] [PubMed] [Google Scholar]
  318. Wei H, et al. Independent beta-arrestin 2 and G protein-mediated pathways for angiotensin II activation of extracellular signal-regulated kinases 1 and 2. Proc Natl Acad Sci U S A. 2003;100:10782–7. doi: 10.1073/pnas.1834556100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  319. Weiner DM, et al. Expression of muscarinic acetylcholine and dopamine receptor mRNAs in rat basal ganglia. Proceedings of the National Academy of Sciences of the United States of America. 1990a;87:7050–4. doi: 10.1073/pnas.87.18.7050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  320. Weiner DM, et al. Expression of muscarinic acetylcholine and dopamine receptor mRNAs in rat basal ganglia. Proc Natl Acad Sci U S A. 1990b;87:7050–4. doi: 10.1073/pnas.87.18.7050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. Wichmann J, et al. Alkyl diphenylacetyl, 9H-xanthene- and 9H-thioxanthene-carbonyl carbamates as positive allosteric modulators of mGlu1 receptors. Farmaco. 2002;57:989–92. doi: 10.1016/s0014-827x(02)01283-1. [DOI] [PubMed] [Google Scholar]
  322. Wichmann T, DeLong MR. Functional and pathophysiological models of the basal ganglia. Curr Opin Neurobiol. 1996;6:751–8. doi: 10.1016/s0959-4388(96)80024-9. [DOI] [PubMed] [Google Scholar]
  323. Wieronska JM, et al. The antipsychotic-like effects of the mGlu group III orthosteric agonist, LSP1-2111, involves 5-HT(1)A signalling. Psychopharmacology (Berl) 2013;227:711–25. doi: 10.1007/s00213-013-3005-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  324. Wieronska JM, et al. Opposing efficacy of group III mGlu receptor activators, LSP1-2111 and AMN082, in animal models of positive symptoms of schizophrenia. Psychopharmacology (Berl) 2012;220:481–94. doi: 10.1007/s00213-011-2502-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Williams MJ, Adinoff B. The role of acetylcholine in cocaine addiction. Neuropsychopharmacology. 2008;33:1779–97. doi: 10.1038/sj.npp.1301585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Wood MR, et al. “Molecular switches” on mGluR allosteric ligands that modulate modes of pharmacology. Biochemistry. 2011;50:2403–10. doi: 10.1021/bi200129s. [DOI] [PMC free article] [PubMed] [Google Scholar]
  327. Woolley ML, et al. Attenuation of amphetamine-induced activity by the non-selective muscarinic receptor agonist, xanomeline, is absent in muscarinic M4 receptor knockout mice and attenuated in muscarinic M1 receptor knockout mice. Eur J Pharmacol. 2009;603:147–9. doi: 10.1016/j.ejphar.2008.12.020. [DOI] [PubMed] [Google Scholar]
  328. Wootten D, et al. Emerging paradigms in GPCR allostery: implications for drug discovery. Nat Rev Drug Discov. 2013;12:630–44. doi: 10.1038/nrd4052. [DOI] [PubMed] [Google Scholar]
  329. Yamada M, et al. Cholinergic dilation of cerebral blood vessels is abolished in M(5) muscarinic acetylcholine receptor knockout mice. Proc Natl Acad Sci U S A. 2001;98:14096–101. doi: 10.1073/pnas.251542998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  330. Yang FV, et al. Parallel synthesis of N-biaryl quinolone carboxylic acids as selective M(1) positive allosteric modulators. Bioorg Med Chem Lett. 2010;20:531–6. doi: 10.1016/j.bmcl.2009.11.100. [DOI] [PubMed] [Google Scholar]
  331. Yu F, et al. Metabotropic glutamate receptor I (mGluR1) antagonism impairs cocaine-induced conditioned place preference via inhibition of protein synthesis. Neuropsychopharmacology. 2013;38:1308–21. doi: 10.1038/npp.2013.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  332. Zarate CA, Jr, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63:856–64. doi: 10.1001/archpsyc.63.8.856. [DOI] [PubMed] [Google Scholar]
  333. Zavitsanou K, et al. Investigation of m1/m4 muscarinic receptors in the anterior cingulate cortex in schizophrenia, bipolar disorder, and major depression disorder. Neuropsychopharmacology. 2004;29:619–25. doi: 10.1038/sj.npp.1300367. [DOI] [PubMed] [Google Scholar]
  334. Zerbib F, et al. Efficacy, tolerability and pharmacokinetics of a modified release formulation of ADX10059, a negative allosteric modulator of metabotropic glutamate receptor 5: an esophageal pH-impedance study in healthy subjects. Neurogastroenterol Motil. 2010;22:859–65. e231. doi: 10.1111/j.1365-2982.2010.01484.x. [DOI] [PubMed] [Google Scholar]
  335. Zhang Y, et al. Allosteric potentiators of metabotropic glutamate receptor subtype 5 have differential effects on different signaling pathways in cortical astrocytes. J Pharmacol Exp Ther. 2005;315:1212–9. doi: 10.1124/jpet.105.090308. [DOI] [PubMed] [Google Scholar]

References

  1. Amato RJ, Felts AS, et al. Substituted 1-Phenyl-3-(pyridin-2-yl)urea Negative Allosteric Modulators of mGlu: Discovery of a New Tool Compound VU0463841 with Activity in Rat Models of Cocaine Addiction. ACS Chem Neurosci. 2013 doi: 10.1021/cn400070k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Annoura H, FA, Uesugi M, Tatsuoka T, Horikawa Y. A novel class of antagonists for metabotropic glutamate receptors, 7-(ydroxyimino)cyclopropa[b]chromen-1a-carboxylates. Bioorganic & medicinal chemistry letters. 1996;6(7):763–766. [Google Scholar]
  3. Ayala JE, Chen Y, et al. 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]
  4. Benneyworth MA, Xiang Z, et al. 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]
  5. Betz AJ, McLaughlin PJ, et al. The muscarinic receptor antagonist tropicamide suppresses tremulous jaw movements in a rodent model of parkinsonian tremor: possible role of M4 receptors. Psychopharmacology (Berl) 2007;194(3):347–359. doi: 10.1007/s00213-007-0844-6. [DOI] [PubMed] [Google Scholar]
  6. Brady AE, Jones CK, et al. Centrally active allosteric potentiators of the M4 muscarinic acetylcholine receptor reverse amphetamine-induced hyperlocomotor activity in rats. J Pharmacol Exp Ther. 2008;327(3):941–953. doi: 10.1124/jpet.108.140350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bridges TM, LeBois EP, et al. The antipsychotic potential of muscarinic allosteric modulation. Drug News Perspect. 2010;23(4):229–240. doi: 10.1358/dnp.2010.23.4.1416977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bridges TM, Lewis LM, et al. Probe Reports from the NIH Molecular Libraries Program. Bethesda (MD): 2010. Discovery and development of the a highly selective M1 Positive Allosteric Modulator (PAM) [PubMed] [Google Scholar]
  9. Bridges TM, Marlo JE, et al. Discovery of the first highly M5-preferring muscarinic acetylcholine receptor ligand, an M5 positive allosteric modulator derived from a series of 5-trifluoromethoxy N-benzyl isatins. J Med Chem. 2009;52(11):3445–3448. doi: 10.1021/jm900286j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bridges TM, Niswender CM, et al. Probe Reports from the NIH Molecular Libraries Program. Bethesda (MD): 2010. Discovery of a Highly Selective in vitro and in vivo M4 Positive Allosteric Modulator (PAM) Series with Greatly Improved Human Receptor Activity. [PubMed] [Google Scholar]
  11. Bridges TM, Reid PR, et al. Probe Reports from the NIH Molecular Libraries Program. Bethesda (MD): 2010. Discovery and development of a second highly selective M1 Positive Allosteric Modulator (PAM) [PubMed] [Google Scholar]
  12. Celanire S, Campo B. Recent advances in the drug discovery of metabotropic glutamate receptor 4 (mGluR4) activators for the treatment of CNS and non-CNS disorders. Expert Opin Drug Discov. 2012;7(3):261–280. doi: 10.1517/17460441.2012.660914. [DOI] [PubMed] [Google Scholar]
  13. Chan WY, McKinzie DL, et al. Allosteric modulation of the muscarinic M4 receptor as an approach to treating schizophrenia. Proc Natl Acad Sci U S A. 2008;105(31):10978–10983. doi: 10.1073/pnas.0800567105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cosford ND, Tehrani L, et al. 3-[(2-Methyl-1,3-thiazol-4-yl)ethynyl]-pyridine: a potent and highly selective metabotropic glutamate subtype 5 receptor antagonist with anxiolytic activity. Journal of medicinal chemistry. 2003;46(2):204–206. doi: 10.1021/jm025570j. [DOI] [PubMed] [Google Scholar]
  15. Digby GJ, Noetzel MJ, et al. Novel allosteric agonists of M1 muscarinic acetylcholine receptors induce brain region-specific responses that correspond with behavioral effects in animal models. J Neurosci. 2012;32(25):8532–8544. doi: 10.1523/JNEUROSCI.0337-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Digby GJ, Utley TJ, et al. Chemical modification of the M(1) agonist VU0364572 reveals molecular switches in pharmacology and a bitopic binding mode. ACS Chem Neurosci. 2012;3(12):1025–1036. doi: 10.1021/cn300103e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. East SP, Bamford S, et al. An orally bioavailable positive allosteric modulator of the mGlu4 receptor with efficacy in an animal model of motor dysfunction. Bioorganic & medicinal chemistry letters. 2010;20(16):4901–4905. doi: 10.1016/j.bmcl.2010.06.078. [DOI] [PubMed] [Google Scholar]
  18. Galici R, Jones CK, et al. 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]
  19. Gasparini F, Lingenhohl K, et al. 2-Methyl-6-(phenylethynyl)-pyridine (MPEP), a potent, selective and systemically active mGlu5 receptor antagonist. Neuropharmacology. 1999;38(10):1493–1503. doi: 10.1016/s0028-3908(99)00082-9. [DOI] [PubMed] [Google Scholar]
  20. Gastambide F, Gilmour G, et al. The mGlu(5) positive allosteric modulator LSN2463359 differentially modulates motor, instrumental and cognitive effects of NMDA receptor antagonists in the rat. Neuropharmacology. 2013;64:240–247. doi: 10.1016/j.neuropharm.2012.07.039. [DOI] [PubMed] [Google Scholar]
  21. Gilmour G, Broad LM, et al. In vitro characterisation of the novel positive allosteric modulators of the mGlu(5) receptor, LSN2463359 and LSN2814617, and their effects on sleep architecture and operant responding in the rat. Neuropharmacology. 2013;64:224–239. doi: 10.1016/j.neuropharm.2012.07.030. [DOI] [PubMed] [Google Scholar]
  22. Goeldner C, Ballard TM, et al. Cognitive impairment in major depression and the mGlu2 receptor as a therapeutic target. Neuropharmacology. 2013;64:337–346. doi: 10.1016/j.neuropharm.2012.08.001. [DOI] [PubMed] [Google Scholar]
  23. Hashimoto K, Malchow B, et al. Glutamate modulators as potential therapeutic drugs in schizophrenia and affective disorders. European archives of psychiatry and clinical neuroscience. 2013 doi: 10.1007/s00406-013-0399-y. [DOI] [PubMed] [Google Scholar]
  24. Hemstapat K, Da Costa H, et al. A novel family of potent negative allosteric modulators of group II metabotropic glutamate receptors. The Journal of pharmacology and experimental therapeutics. 2007;322(1):254–264. doi: 10.1124/jpet.106.117093. [DOI] [PubMed] [Google Scholar]
  25. Hemstapat K, de Paulis T, et al. A novel class of positive allosteric modulators of metabotropic glutamate receptor subtype 1 interact with a site distinct from that of negative allosteric modulators. Mol Pharmacol. 2006;70(2):616–626. doi: 10.1124/mol.105.021857. [DOI] [PubMed] [Google Scholar]
  26. Hikichi H, Murai T, et al. Effects of a novel metabotropic glutamate receptor 7 negative allosteric modulator, 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazonolo[4,5-c]pyridin-4(5H)-one (MMPIP), on the central nervous system in rodents. Eur J Pharmacol. 2010;639(1–3):106–114. doi: 10.1016/j.ejphar.2009.08.047. [DOI] [PubMed] [Google Scholar]
  27. Hughes ZA, Neal SJ, et al. Negative allosteric modulation of metabotropic glutamate receptor 5 results in broad spectrum activity relevant to treatment resistant depression. Neuropharmacology. 2013;66:202–214. doi: 10.1016/j.neuropharm.2012.04.007. [DOI] [PubMed] [Google Scholar]
  28. Jacquemont S, Curie A, et al. Epigenetic modification of the FMR1 gene in fragile X syndrome is associated with differential response to the mGluR5 antagonist AFQ056. Sci Transl Med. 2011;3(64):64ra61. doi: 10.1126/scitranslmed.3001708. [DOI] [PubMed] [Google Scholar]
  29. Johnson MP, Baez M, et al. 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. Journal of medicinal chemistry. 2003;46(15):3189–3192. doi: 10.1021/jm034015u. [DOI] [PubMed] [Google Scholar]
  30. Jones CK, Brady AE, et al. Novel selective allosteric activator of the M1 muscarinic acetylcholine receptor regulates amyloid processing and produces antipsychotic-like activity in rats. J Neurosci. 2008;28(41):10422–10433. doi: 10.1523/JNEUROSCI.1850-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Jones CK, Bubser M, et al. The metabotropic glutamate receptor 4-positive allosteric modulator VU0364770 produces efficacy alone and in combination with L-DOPA or an adenosine 2A antagonist in preclinical rodent models of Parkinson’s disease. J Pharmacol Exp Ther. 2012;340(2):404–421. doi: 10.1124/jpet.111.187443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Kalinichev M, Rouillier M, et al. ADX71743, a potent and selective negative allosteric modulator of metabotropic glutamate receptor 7: in vitro and in vivo characterization. The Journal of pharmacology and experimental therapeutics. 2013;344(3):624–636. doi: 10.1124/jpet.112.200915. [DOI] [PubMed] [Google Scholar]
  33. Knoflach F, Mutel V, et al. Positive allosteric modulators of metabotropic glutamate 1 receptor: characterization, mechanism of action, and binding site. Proc Natl Acad Sci U S A. 2001;98(23):13402–13407. doi: 10.1073/pnas.231358298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Langmead CJ, Austin NE, et al. Characterization of a CNS penetrant, selective M1 muscarinic receptor agonist, 77-LH-28-1. Br J Pharmacol. 2008;154(5):1104–1115. doi: 10.1038/bjp.2008.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Lavreysen H, Pereira SN, et al. Metabotropic glutamate 1 receptor distribution and occupancy in the rat brain: a quantitative autoradiographic study using [3H]R214127. Neuropharmacology. 2004;46(5):609–619. doi: 10.1016/j.neuropharm.2003.11.014. [DOI] [PubMed] [Google Scholar]
  36. Lazareno S, Birdsall B, et al. Allosteric effects of four stereoisomers of a fused indole ring system with 3H-N-methylscopolamine and acetylcholine at M1-M4 muscarinic receptors. Life Sci. 1999;64(6–7):519–526. doi: 10.1016/s0024-3205(98)00596-7. [DOI] [PubMed] [Google Scholar]
  37. Lazareno S, Dolezal V, et al. Thiochrome enhances acetylcholine affinity at muscarinic M4 receptors: receptor subtype selectivity via cooperativity rather than affinity. Mol Pharmacol. 2004;65(1):257–266. doi: 10.1124/mol.65.1.257. [DOI] [PubMed] [Google Scholar]
  38. Lebois EP, Bridges TM, et al. Discovery and characterization of novel subtype-selective allosteric agonists for the investigation of M(1) receptor function in the central nervous system. ACS Chem Neurosci. 2010;1(2):104–121. doi: 10.1021/cn900003h. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Lindemann L, Jaeschke G, et al. CTEP: a novel, potent, long-acting, and orally bioavailable metabotropic glutamate receptor 5 inhibitor. The Journal of pharmacology and experimental therapeutics. 2011;339(2):474–486. doi: 10.1124/jpet.111.185660. [DOI] [PubMed] [Google Scholar]
  40. Liu F, Grauer S, et al. ADX47273 [S-(4-fluoro-phenyl)-{3-[3-(4-fluoro-phenyl)-[1,2,4]-oxadiazol-5-yl]-piperidin-1-yl}-methanone]: a novel metabotropic glutamate receptor 5-selective positive allosteric modulator with preclinical antipsychotic-like and procognitive activities. The Journal of pharmacology and experimental therapeutics. 2008;327(3):827–839. doi: 10.1124/jpet.108.136580. [DOI] [PubMed] [Google Scholar]
  41. Maj M, Bruno V, et al. (-)-PHCCC, a positive allosteric modulator of mGluR4: characterization, mechanism of action, and neuroprotection. Neuropharmacology. 2003;45(7):895–906. doi: 10.1016/s0028-3908(03)00271-5. [DOI] [PubMed] [Google Scholar]
  42. Marino MJ, Williams DL, Jr, et al. Allosteric modulation of group III metabotropic glutamate receptor 4: a potential approach to Parkinson’s disease treatment. Proc Natl Acad Sci U S A. 2003;100(23):13668–13673. doi: 10.1073/pnas.1835724100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Niswender CM, Johnson KA, et al. Context-dependent pharmacology exhibited by negative allosteric modulators of metabotropic glutamate receptor 7. Molecular pharmacology. 2010;77(3):459–468. doi: 10.1124/mol.109.058768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Niswender CM, Johnson KA, et al. Discovery, characterization, and antiparkinsonian effect of novel positive allosteric modulators of metabotropic glutamate receptor 4. Mol Pharmacol. 2008;74(5):1345–1358. doi: 10.1124/mol.108.049551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Porter RH, Jaeschke G, et al. Fenobam: a clinically validated nonbenzodiazepine anxiolytic is a potent, selective, and noncompetitive mGlu5 receptor antagonist with inverse agonist activity. The Journal of pharmacology and experimental therapeutics. 2005;315(2):711–721. doi: 10.1124/jpet.105.089839. [DOI] [PubMed] [Google Scholar]
  46. Rodriguez AL, Grier MD, et al. 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]
  47. Rodriguez AL, Nong Y, et al. A close structural analog of 2-methyl-6-(phenylethynyl)-pyridine acts as a neutral allosteric site ligand on metabotropic glutamate receptor subtype 5 and blocks the effects of multiple allosteric modulators. Molecular pharmacology. 2005;68(6):1793–1802. doi: 10.1124/mol.105.016139. [DOI] [PubMed] [Google Scholar]
  48. Sheffler DJ, Wenthur CJ, et al. Development of a novel, CNS-penetrant, metabotropic glutamate receptor 3 (mGlu3) NAM probe (ML289) derived from a closely related mGlu5 PAM. Bioorg Med Chem Lett. 2012;22(12):3921–3925. doi: 10.1016/j.bmcl.2012.04.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Sheffler DJ, Wenthur CJ, et al. Development of a novel, CNS-penetrant, metabotropic glutamate receptor 3 (mGlu3) NAM probe (ML289) derived from a closely related mGlu5 PAM. Bioorganic & medicinal chemistry letters. 2012;22(12):3921–3925. doi: 10.1016/j.bmcl.2012.04.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Shirey JK, Brady AE, et al. A selective allosteric potentiator of the M1 muscarinic acetylcholine receptor increases activity of medial prefrontal cortical neurons and restores impairments in reversal learning. J Neurosci. 2009;29(45):14271–14286. doi: 10.1523/JNEUROSCI.3930-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Slawinska A, Wieronska JM, et al. The antipsychotic-like effects of mGlu4 receptor positive allosteric modulators in rodents. Br J Pharmacol. 2013 doi: 10.1111/bph.12254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Slawinska A, Wieronska JM, et al. Anxiolytic- but not antidepressant-like activity of Lu AF21934, a novel, selective positive allosteric modulator of the mGlu(4) receptor. Neuropharmacology. 2013;66:225–235. doi: 10.1016/j.neuropharm.2012.05.001. [DOI] [PubMed] [Google Scholar]
  53. Spalding TA, Trotter C, et al. Discovery of an ectopic activation site on the M(1) muscarinic receptor. Mol Pharmacol. 2002;61(6):1297–1302. doi: 10.1124/mol.61.6.1297. [DOI] [PubMed] [Google Scholar]
  54. Sur C, Mallorga PJ, et al. N-desmethylclozapine, an allosteric agonist at muscarinic 1 receptor, potentiates N-methyl-D-aspartate receptor activity. Proc Natl Acad Sci U S A. 2003;100(23):13674–13679. doi: 10.1073/pnas.1835612100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Thomas AM, Bui N, et al. Group I metabotropic glutamate receptor antagonists alter select behaviors in a mouse model for fragile X syndrome. Psychopharmacology. 2012;219(1):47–58. doi: 10.1007/s00213-011-2375-4. [DOI] [PubMed] [Google Scholar]
  56. Thomas NK, Wright RA, et al. (S)-3,4-DCPG, a potent and selective mGlu8a receptor agonist, activates metabotropic glutamate receptors on primary afferent terminals in the neonatal rat spinal cord. Neuropharmacology. 2001;40(3):311–318. doi: 10.1016/s0028-3908(00)00169-6. [DOI] [PubMed] [Google Scholar]
  57. Ugolini A, Large CH, et al. AMN082, an allosteric mGluR7 agonist that inhibits afferent glutamatergic transmission in rat basolateral amygdala. Neuropharmacology. 2008;55(4):532–536. doi: 10.1016/j.neuropharm.2008.04.020. [DOI] [PubMed] [Google Scholar]
  58. Uslaner JM, Eddins D, et al. The muscarinic M1 receptor positive allosteric modulator PQCA improves cognitive measures in rat, cynomolgus macaque, and rhesus macaque. Psychopharmacology (Berl) 2013;225(1):21–30. doi: 10.1007/s00213-012-2788-8. [DOI] [PubMed] [Google Scholar]
  59. Wichmann J, Bleicher K, et al. Alkyl diphenylacetyl, 9H-xanthene- and 9H-thioxanthene-carbonyl carbamates as positive allosteric modulators of mGlu1 receptors. Farmaco. 2002;57(12):989–992. doi: 10.1016/s0014-827x(02)01283-1. [DOI] [PubMed] [Google Scholar]

RESOURCES