Abstract
Allosteric modulation has emerged as an innovative pharmacological approach to selectively activate or inhibit a number of Class C GPCRs. Of the Class C GPCRs, metabotropic glutamate (mGlu) receptors represent the most promising candidates for clinical success, and both positive allosteric modulators (PAMs) and negative allosteric modulators (NAMs) of mGluRs have demonstrated therapeutic potential for a range of psychiatric and neurological disorders such as pain, depression, anxiety, cognition, Fragile X syndrome, Parkinson’s disease and schizophrenia.
Keywords: G-protein coupled receptor, positive allosteric modulator (PAM), negative allosteric modulator (NAM), metabotropic glutamate receptor
Introduction
G Protein-coupled receptors (GPCRs) represent the largest class of cell surface receptors, and small molecules targeting ~50 GPCRs constitute >50% of currently marketed drugs [1–5]. Over 300 GPCRs remain to be the targets of novel therapeutics, and, despite intense efforts, viable ligands have failed to be identified [1–7]. Classical approaches afford ligands that interact with the highly conserved orthosteric binding site (OBS), leading to poor selectivity amongst GPCR subtypes, ligands with poor physiochemical properties, and potential adverse events. Therefore, alternative, functional screening approaches have been developed to identify allosteric ligands that bind at a site distinct from the OBS, that modulate (potentiate or inhibit) the binding and/or signaling of the native orthosteric ligand [1–12]. Allosteric ligands afford unprecedented GPCR subtype selectivity, attractive chemotypes and diminished potential for adverse events. This strategy has been highly successful for Class C GPCRs, providing robust in vivo probes and clinical candidates for a number of CNS disorders [1–12].
Class C GPCRs and Metabotropic Glutamate Receptors
GPCRs are commonly divided into three major classes (or families) based on sequence homology and functional roles: Class A (e.g., dopamine D1), Class B (e.g., glucagon-like peptide, GLP-1) and Class C (e.g, metabotropic glutamate receptors, mGlu5) [1–15]. The Class C GPCRs are distinguished by two unique structural features: 1) a large extracellular, venus fly-trap amino-terminal agonist binding site linked to the heptahelical transmembrane (7TM) domain via a cysteine-rich region and 2) the formation of constitutive homo- and heterodimers that engender diverse activation modes (Fig. 1), where the allosteric and orthosteric ligands can occupy the same receptor or a canonical trans-mode (Fig. 1B). Unlike Class A and B GPCRs, the orthosteric binding site for Class C GPCRs is not located within the 7TM domain. Class C GPCRs are comprised of several members: metabotropic glutamate receptors (mGlu receptors), γ-aminobutyric acidB receptors (GABAB receptors), calcium sensing receptors (CaS receptors), taste (sweet and umami) receptors (T1 receptors), fish odorant receptors and a number of orphan receptors. Of these, the mGlu receptors and the GABAB receptors represent exciting molecular targets for the development of central nervous system (CNS) therapeutic agents, with mGlu receptors showing the greatest promise [1–15].
Figure 1.
A) A schematic representation of a Class C GPCR highlighting the large, extracellular bi-lobed (‘Venus fly-trap’) N-terminal agonist binding domain connected to the N-terminal domain via a cysteine-rich region to the 7TM domain. B) mGlu receptors function as homodimers. Unlike Class A and B GPCRs, the orthosteric binding site is not located within the 7TM; in contrast, glutamate, the orthosteric ligand, binds in the N-terminal extracellular bi-lobed domain. Thus far, the 7TM domain contains the binding sites for all known positive, negative and silent allosteric modulators of the mGlu receptors. C) Representative endogenous (glutamate, 1) and synthetic orthosteric agonists 2 and 3, along with the first mGlu allosteric ligands 4 and 5. D) The ‘triple add screening’ paradigm in a kinetic calcium flourescene assay as a surrogate for GPCR activation. Compound is added (1st add), followed 2 minutes later by an EC20 of glutamate (2nd add) and then 1 minute later by an EC80 of glutamate (3rd add). E) An agonist will elicit fluorescence upon compound addition. A PAM will have no effect on receptor activation alone, but will potentiate the EC20. A NAM will block both the EC20 and EC80.
Glutamate, L-glutamic acid, is the major excitatory transmitter in the mammalian central nervous system (CNS), exerting its effects through both ionotropic and metabotropic glutamate receptors [1–15]. Glutamate, the orthosteric agonist, along with the other known agonists and competitive antagonists of the mGlu receptors, bind within the extracellular N-terminal region; in contrast, all of the positive allosteric modulators (PAMs) and negative allosteric modulators (NAMs) described thus far bind within the 7TM domain (Fig. 1). To date, eight mGluRs have been cloned and sequenced, and have been assigned to three groups based on their structure, preferred coupling to effector mechanisms and pharmacology [1–15]. Group I receptors (mGlu1 and mGlu5) are predominantly located post-synaptically and couple to Gαq, resulting in increases in intracellular calcium. Group II (mGlu2 and mGlu3) and Group III (mGlu4, mGlu6, mGlu7 and mGlu8) receptors are coupled through Gi/Go resulting in decreases in cAMP levels. Group II and III mGlu receptors are located presynaptically and generally have inhibitory effects on neurotransmitter release [1–15].
Orthosteric and Allosteric GPCR Pharmacology
Classical orthosteric GPCR ligands modulate signaling by directly stimulating a receptor response (agonist), blocking the binding of the native agonist (competitive antagonist) or blocking constitutive activity (inverse agonist) of the GPCR, and have typically been discovered and optimized using orthosteric radioligand binding methods [1–15]. While some of these agonists and antagonists are group-selective, it has been difficult to identify subgroup-selective ligands, presumably due to evolutionary pressure to conserve the glutamate binding site across the mGlu receptors. For the mGlu receptors, a number of synthetic orthosteric agonists and competitive antagonists, analogs of glutamate 1 [1–15], have been identified and include the clinically validated mGlu2/3 agonist LY354740 (2) [16] and the mGlu4 preferring agonist LSP3-2074-I (3) [17] (Fig. 1). Moreover, poor oral bioavailability, limited CNS exposure and poor physical properties for these amino acid analogs have limited their utility in the study of the therapeutic potential of individual mGlu pharmacology and often require pro-drug formulations to achieve appropriate exposure in vivo [1–18].
The advent of high-throughput functional assays has enabled scientists to screen for compounds capable of modulating the activity of a receptor without regard to the binding site(s), and ideally identify modulators that bind at distinct allosteric sites [1–15]. This functional screening strategy constitutes a major shift from historical ligand discovery efforts, and has successfully identified a number of allosteric modulators (PAMs, NAMs, ago-PAMs, silent allosteric modulators (SAMs), allosteric agonists and partial antagonists) of GPCRs that can modify receptor function without necessarily affecting radioligand binding [1–15]. Key to the success of functional screening was the advent of the ‘triple add’ protocol (compound add, 2 min later EC20 glutamate add, 1 min later EC80 glutamate add), allowing for the identification of agonists, ago-PAMs, PAMs and NAMs in a single kinetic assay (Fig. 1) [2,19]. This works well for group I mGluRs, which natively couple to Gq to release intracellular calcium. For group II and group III mGluRs, cell lines must be generated that co-express the desired group II and III mGlu receptor with a chimeric G protein, such as Gqi5, which links the Gi/o-coupled group II and III mGlu receptor to the phospholipase C/Ca2+ pathway, and enables the ‘triple add’ kinetic assay platform [14]. In the case of the mGlu receptors, the first small molecules that were clearly shown to exert allosteric effects were negative allosteric modulators of mGlu1 (CPCCOEt, 4) and mGlu5 (MPEP, 5) [1–15], and recently, mGlu3 and mGlu4 NAMs [20,21] have been identified as well (Fig. 2). Soon after, selective positive allosteric modulators, small molecules that enhance the agonist response without intrinsic agonist activity, were reported for mGlu1 [22–24], mGlu2 [25], mGlu4 [26] and mGlu5 [27]. The ability to identify sub-type selective allosteric modulators suggests that unlike the case of the agonist binding site, where there is significant evolutionary pressure to conserve residues across mGluRs, the allosteric binding sites are under less evolutionary pressure for their conservation [1–15, 23–28]. In addition to greater receptor sub-type selectivity, allosteric ligands offer numerous advantages: 1) the effects of an allosteric modulator are saturable (ie., a ‘ceiling effect’) 2) a modulator that lacks agonistic activity will only exert its effects when the endogenous agonist is present, resulting in temporal and spatial activity, also known as state dependence, of the endogenous ligand and 3) improved chemical tractability. In addition, ago-PAMs and partial antagonists provide unique modes of pharmacology, while ligand-biased signaling phenomena can be both welcomed and admonished [1–15, 28].
Figure 2.
Representative Group I mGlu allosteric modulator ligands. mGlu1 NAMs 6 and 7, mGlu1 PAMs 8 and 9, mGlu5 NAMs 10–13 and mGlu5 PAMs 14–18.
While many advantages over orthosteric ligands have been realized (Table 1), allosteric modulation is far from a panacea for GPCR drug discovery, and many issues have arisen as the field matures [1–15]. First, the SAR for allosteric modulators can be extremely ‘steep’ (subtle modifications to a ligand leads to complete loss of activity), and coupled with ‘molecular switch’ events (subtle structural changes that modulate modes of pharmacology or subtype selectivity), chemical optimization can be complex; however, this can also provide entry into ligands for other mGlu receptors. Thus, the ‘triple add’ screening technology is critical both for HTS campaigns and for the routine primary assay driving the optimization, along with periodic mGlu selectivity screens. Second, optimization must consider EC50/IC50, fold-shift and efficacy (% Glu Max) as well as standard in vitro and in vivo DMPK parameters. Third, the lack of evolutionary pressure on allosteric sites can lead to significant species differences, which can complicate IND-enabling studies. Finally, the state-dependence of allosteric modulators could be a liability in degenerative diseases, such as Alzheimer’s disease, where there is a progressive loss of endogenous orthosteric cholinergic tone [1–15].
Table 1.
Technology Summary Table.
| Name of technology | Technology 1 | Technology 2 | Technology 3 | Technology 4 |
|---|---|---|---|---|
| Radioligand binding assay | Kinetic Functional Assay | Orthosteric ligands | Allosteric Ligands | |
| Pros | Established approach Know binding site of ligand, typically orthosteric |
High-throughput, identifies allosteric ligands ‘Triple Add’ Identifies PAMs, NAMs, ago-PAMs, SAMs, allosteric agonists in single assay |
Radioligands typically available for occupancy studies High conservation afford little or no species differences |
Excellent subtype selectivity amongst Class C GPCR families Ligands with excellent physiochemical properties and CNS penetration |
| Cons | Only orthosteric agonists and antagonists Radioactive ligands Only provides binding data – no function |
Requires expensive kinetic plate readers and robotic addition arms. No information on site of binding |
Poor subtype selectivity amongst Class C GPCR families Ligands with poor physiochemical properties and poor CNS penetration |
Often no radioligand or PET tracers Unpredictable, shallow SAR ‘Molecular Switches’ Do not know if ligands bind to same or different allosteric sites Ligand biased signaling Potential species differences |
| references | [1–15] | [1–15] | [1–15] | [1–15] |
Therapeutic potential for CNS disorders
The therapeutic potential of mGlu receptor allosteric modulation is quite broad, and dysfunction in glutamatergic systems has been implicated in a number of CNS pathologies including pain, anxiety, depression, Parkinson’s disease, cognition, addiction, Fragile X syndrome and schizophrenia [1–15,22–28]. A diverse array of allosteric ligands have been reported for mGlu1, mGlu2, mGlu3, mGlu4, and mGlu5; however, selective allosteric ligands are lacking for mGlu6 and only few examples exist for mGlu7 and mGlu8 (Figs. 2–4) [1–15,22–31]. Here we will summarize key data and therapeutic potential for the known allosteric mGlu receptor ligands.
Figure 4.
Representative Group III mGlu allosteric modulator ligands. mGlu4 PAMs 26–30, mGlu4 NAM 31, mGlu7 allosteric agonist 32, mGlu7 NAMs 33 and 34 and mGlu8 PAM, 35.
Group I mGlu Receptors (mGlu1 and mGlu5)
The mGlu1 receptor controls the post-synaptic release of the key neurotransmitters GABA and glutamate, and also interacts with the NMDA receptor [21–24]. mGlu1 knock-out mice display motor dysfunction and diminished learning and memory capabilities [24]. Recent genetic data indicates loss of function mutation in the GRM1 gene, which encodes mGlu1, in patients suffering from schizophrenia and bipolar disorder [32,33], and impaired mGlu1 signaling has also been shown in cocaine treated mice [34]. Thus, mGlu1 PAMs may represent novel treatment approaches for both schizophrenia and addiction; however, few mGlu1 PAMs have been described, and the known ligands represent two distinct scaffolds (Fig. 2). In contrast, a large number of mGlu1 NAMs with tremendous structural diversity have been reported, with robust in vivo efficacy in preclinical models of pain, stroke, epilepsy, drug addiction, Huntington’s disease, anxiety and neurodegeneration by increasing neuronal excitability and modulation of multiple ion channels (Fig. 2) [22–24,32–34].
Allosteric modulation of mGlu5 affords opportunities for the treatment of a wide range of CNS disorders [1–15]. With respect to mGlu5 PAMs, the major therapeutic interests are focused on schizophrenia and cognition, via potentiation of the N-methyl-D-aspartate (NMDA) receptor and the NMDA hypofunction hypothesis of schizophrenia [1–15,35]. mGlu5 physically and functionally interacts with NMDA receptors, and multiple mGlu5 PAMs (Fig. 2) have been shown to potentiate NMDA currents, and afford robust efficacy in preclinical models predictive of antipsychotic activity. Moreover, several series of mGlu5 PAMs have demonstrated pro-cognitive profiles in a variety of preclinical cognition assays and have also shown potential to treat absence epilepsy [1–15,27,35]. However, no clinical data has yet to be reported. For mGlu5 PAMs, ligands have been developed that bind at one of three distinct allosteric sites, further adding complexity, as the MPEP binding site is the only characterized allosteric site on mGlu5. It is not yet clear if the unique conformations stabilized by ligands at the other allosteric sites offer advantages or disadvantages over modulation via the MPEP site. Regardless, pure mGlu5 PAMs are essential, as ago-PAMs, may suffer the same adverse events as orthosteric Group I agonists [1–15,35,36].
The most advanced mGlu allosteric ligands, in terms of clinical trials in humans, are the mGlu5 NAMs [1–15,28]. At present, nine chemically distinct mGlu5 NAMs are in clinical testing for a wide range of indications including anxiety, Fragile X syndrome, migraine, chromic neuropathic pain, treatment resistant depression, L-DOPA-induced diskinesia (PD-LID) and gastroesophageal reflux disease (GERD); moreover, positive results have been reported [2]. Preclinically, a wide range of mGlu5 NAM chemotypes have demonstrated efficacy in rodent models of the same CNS disorders listed above [1–15]. Interestingly, mGlu5 partial antagonists, allosteric ligands that fully occupy the allosteric site but only partially block signaling, have been discovered, and represent a new mode of pharmacology that may limit potential adverse events associated with ablation of receptor signaling with NAMs (also referred to as non-competitive antagonists) [37]. Only recently have non-MPEP (non-acetylene containing) mGlu5 NAM chemotypes been reported, which display improved ancillary pharmacology and DMPK profiles [1–15].
Group II mGlu Receptors (mGlu2 and mGlu3)
Group II mGlu receptors, mGlu2 and mGlu3, inhibit further release of glutamate once levels accumulate in the perisynaptic extracellular region, and thus modulate glutamatergic neurotransmission [1–15,25]. Recent preclinical rodent and human clinical data provide strong evidence that the mGlu2/3 agonist (3) is effective in the treatment of the positive symptoms of schizophrenia. Data from knock-out mice, coupled with the fact that the efficacy of 3 has been recapitulated preclinically with mGlu2 PAMs, has resulted in an enormous efforts by multiple pharmaceutical companies to develop mGlu2 PAMs (Fig. 3). Beyond schizophrenia, mGlu2 PAMs have also displayed efficacy in anxiety assays, drug dependence (cocaine) and sleep-wake architecture (normalizing REM states characteristic of depression) [1–15,25]. As of this writing, no selective mGlu3 PAMs have been reported; therefore the therapeutic potential of selective mGlu3 activation remains unclear.
Figure 3.
Representative Group II mGlu allosteric modulator ligands. mGlu2 PAMs 19–22, mGlu2/3 NAMs 23 and 24 and an mGlu3NAM 25.
Until recently, the only known Group II NAMs were dual mGlu2/3 NAMs, representing only two chemotypes (Fig. 3). In 2012, a selective mGlu3 NAM (24) was developed starting from a potent mGlu5 PAM, highlighting the value of ‘molecular switches’ [20,38]. Therapeutically, Group II NAMs have shown efficacy in reversing hyperlocomotion and in improving memory; however, it is not yet clear if this is mGlu2 or mGlu3 driven, and additional small molecule tools are required to dissect the contribution of the two receptors to the observed efficacy [1–15,25].
Group III mGlu Receptors (mGlu4, mGlu6, mGlu7 and mGlu8)
Of the Group III mGlu receptors, mGlu4 is the most advanced, with a diverse array of PAMs and ago-PAMs (Fig. 4) affording robust efficacy in multiple preclinical models of Parkinson’s disease (PD), both for symptomatic motor disturbances and for neuroprotection of dopaminergic neurons [26]. Recent studies have demonstrated that mGlu4 PAMs also synergize with A2A antagonists (in haloperidol-induced catalepsy) and with L-DOPA (in 6-OHDA rats), offering additional co-treatment options for PD patients and reducing the necessary dosage of L-DOPA [39]. In parallel, new roles for mGlu4 PAMs in the treatment of pain, anxiety and mood disorders are being developed. Thus far, only one report of an mGlu4 NAM has been disclosed [21], but the therapeutic potential of inhibiting mGlu4 is not clear.
Of the remaining Group III mGlu receptors, mGlu6 is restricted to the retina, and therefore not relevant to CNS disorders; moreover, no selective ligands for mGlu6 have been disclosed [1–15]. Selective activation of mGlu7 (Fig. 4), with the lone mGlu7 allosteric agonist AMN082 (32) has been shown to reduce the rewarding effects of cocaine and ethanol and facilitate the extinction of conditioned fear; thus, mGlu7 has potential in addiction, anxiety and post-traumatic stress disorder (PTSD) [29]. In addition, two selective mGlu7 antagonists/NAMs, 33 and 34, has also been described [2,30]. Similarly, there is only one selective mGlu8 PAM, AZ12216052 (35), reported thus far, and data from animal models indicate activation of mGlu8 may have anxiolytic effects (Fig. 4) [31]. Additional data suggests that mGlu7 and mGlu8, either alone or in combination, may have efficacy in neurodegeneration, mood disorders, pain, addiction and epilepsy [29–31].
Conclusion
The advent of kinetic, functional assay paradigms has led to a revolution in drug discovery aimed at Class C GPCRs, providing new opportunities to modulate receptor function through novel, allosteric mechanisms. These novel modes of pharmacology lead to small molecules that can function as PAMs, ago-PAMs, NAMs, silent allosteric modulators, allosteric agonists and partial allosteric antagonists, highlight the unprecedented potential for fine tuning cellular signaling processes in either a positive or negative manner. Emerging from this new paradigm is Cinacalcet™, a PAM of the CaS receptor, and the first class C GPCR to become a marketed therapeutic [40]. A number of mGluR allosteric modulators are now in clinical development for a range of CNS disorders, and many are showing robust efficacy in a number of CNS pathologies. However, despite the positive attributes of allosteric modulation, it is not a panacea, and lead optimization of mGlu receptor allosteric clinical candidates is burdened with many caveats and challenges. Application of technology, both within pharmacology and medicinal chemistry are critical for the successful optimization of class C allosteric ligands. Functional ‘triple add’ assays allow allosteric ligands to be identified as well as capturing mode switching, ie., PAM to NAM. In contrast, classical radioligand binding assays, employing orthosteric radioligands, will only provide traditional orthosteric ligands, and require additional functional assays to determine if potent binders have any functional activity. Allosteric ligands are best optimized through iterative parallel synthesis, as opposed to classical single compound synthesis, due to the ‘steep’ SAR and propensity for ‘molecular switches’ to modulate modes of pharmacology and subtype selectivity. Yet, it remains to be definitively proven if class C allosteric ligands will prove more successful in the clinic than the second generation of orthosteric ligands; however, subtype selectivity, temporal control and improved physiochemical properties bode well for allosteric ligands to be a driving force in class C GPCR drug discovery for decades to come.
Glossary
- Orthosteric Site
The binding site of the receptor’s endogenous agonist
- Allosteric Site
A ligand binding site topographically distinct from the orthosteric binding site
- Positive Allosteric Modulator (PAM)
A ligand that can increase the action of an orthosteric ligand by binding at an allosteric site. The PAM may enhance the affinity and/or efficacy of the orthosteric ligand without any intrinsic activity. The PAM may also enhance coupling to the G-Protein
- Negative Allosteric Modulator (NAM)
A ligand that can decrease the action of an orthosteric ligand by binding at an allosteric site. The NAM may also inhibit coupling to the G-protein. Also known as a non-competitive antagonist
- Silent Allosteric Modulator (SAM)
A ligand that fully occupies an allosteric site, but exerts no pharmacological function on its own, but can block the allosteric activity of both PAMs and NAMs. Also referred to as neutral or pharmacologically silent ligands
- Partial Antagonist (PA)
A NAM that fully occupies the allosteric binding site, but induces only a partial blockade of receptor signaling
- Allosteric Agonist (AA)
A ligand capable of receptor activation alone, in the absence of the orthosteric ligand, by binding at an allosteric site and engendering an active conformation of the receptor
- Ago-Potentiator (ago-PAM)
An allosteric ligand that functions as both a PAM and an allosteric against; however, the agonism is usually observed at concentrations higher than that for the PAM activity
Footnotes
Conflict of Interest
Dr. Lindsley and Dr. Engers are actively developing a wide range of Class C allosteric ligands for numerous CNS indications with corporate partners, and receive research funding from Johnson & Johnson and Seaside Therapeutics.
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.Conn PJ, et al. Allosteric Modulators of GPCRs as a Novel Approach to Treatment of CNS Disorders. Nat Rev Drug Discov. 2009;8:41–54. doi: 10.1038/nrd2760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Melancon BJ, et al. Allosteric Modulation of 7 Transmembrane Spanning Receptors: Theory, Practice and Opportunities for CNS Drug Discovery. J Med Chem. 2012;55:1445–1464. doi: 10.1021/jm201139r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bridges TM, Lindsley CW. G-protein coupled receptors: From classical modes of modulation to allosteric mechanisms. ACS Chem Biol. 2008;3:530–542. doi: 10.1021/cb800116f. [DOI] [PubMed] [Google Scholar]
- 4.Langerstrom MC, Schioth HB. Structural diversity of G protein-coupled receptors and significance for drug discovery. Nat Rev Drug Discov. 2008;7:339–357. doi: 10.1038/nrd2518. [DOI] [PubMed] [Google Scholar]
- 5.Chun L, et al. Structure and ligand recognition of class C GPCRs. Acta Pharmacologica Sinica. 2012;33:312–323. doi: 10.1038/aps.2011.186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Christopoulos A, Kenakin T. G protein-coupled receptors allosterism and complexing. Pharmacol Rev. 2002;54:323–374. doi: 10.1124/pr.54.2.323. [DOI] [PubMed] [Google Scholar]
- 7.Christopoulos A. Allosteric binding sites on cell surface receptors: novel targets for drug discovery. Nat Rev Drug Discov. 2002;1:198–210. doi: 10.1038/nrd746. [DOI] [PubMed] [Google Scholar]
- 8.Kenakin TP. 7TM receptor allostery: putting numbers to shapeshifting proteins. Trends Pharmacol Sci. 2009;30:460–469. doi: 10.1016/j.tips.2009.06.007. [DOI] [PubMed] [Google Scholar]
- 9.Langmead CJ, Christopoulos A. Allosteric agonists of 7TM receptors: expanding the pharmacological toolbox. Trends Pharmacol Sci. 2006;27:475–481. doi: 10.1016/j.tips.2006.07.009. [DOI] [PubMed] [Google Scholar]
- 10.May LT, et al. Allosteric modulation of G protein-coupled receptors. Annu Rev Pharmacol Toxicol. 2007;47:1–51. doi: 10.1146/annurev.pharmtox.47.120505.105159. [DOI] [PubMed] [Google Scholar]
- 11.Lewis JA, et al. Allosteric modulators of kinases and GPCRs. Design principles and structural diversity. Curr Opin Chem Biol. 2008;12:269–279. doi: 10.1016/j.cbpa.2008.02.014. [DOI] [PubMed] [Google Scholar]
- 12.Urwyler S. Allosteric modulation of Family C G protein-coupled receptors: from molecular insights to therapeutic perspectives. Pharmacol Rev. 2011;63:59–126. doi: 10.1124/pr.109.002501. [DOI] [PubMed] [Google Scholar]
- 13.Rocheville M, Garland SL. An industrial perspective on positive allosteric modulation as a means to discover safe and selective drugs. Drug Discov Today Technol. 2010;7:e87–e94. doi: 10.1016/j.ddtec.2010.06.004. [DOI] [PubMed] [Google Scholar]
- 14.Niswender CM, Conn PJ. Metabotropic glutamate receptors: physiology, pharmacology and disease. Annu Rev Pharmaol Tox. 2010;50:295–322. doi: 10.1146/annurev.pharmtox.011008.145533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Conn PJ, et al. Activation of metabotropic glutamate receptors as a novel approach for the treatment of schizophrenia. Trends Pharm Sci. 2009;30:25–31. doi: 10.1016/j.tips.2008.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Schoepp DD, et al. LY354740 is a potent and highly selective group II metabotropic glutamate receptor agonist in cells expressing human glutamate receptors. Neuropharmacology. 1997;36:1–11. doi: 10.1016/s0028-3908(96)00160-8. [DOI] [PubMed] [Google Scholar]
- 17.Goudet C, et al. A novel selective metabotropic glutamate receptor 4 agonist reveals new possibilities for developing subtype selective ligands with therapeutic potential. FASEB J. 2012;26:1682–1693. doi: 10.1096/fj.11-195941. [DOI] [PubMed] [Google Scholar]
- 18.Rorick-Kehn LM. Improved bioavailability of the mGlu2/3 receptor agonist LY354740 using a prodrug strategy: In vivo pharmacology of LY544344. J Pharmacol Exp Ther. 2006;316:905–913. doi: 10.1124/jpet.105.091926. [DOI] [PubMed] [Google Scholar]
- 19.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 Pharm. 2010;78:1105–1123. doi: 10.1124/mol.110.067207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.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. 2012;22:3921–3925. doi: 10.1016/j.bmcl.2012.04.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Utley T, et al. Synthesis and SAR of a novel metabotropic glutamate receptor 4 (mGlu4) antagonist: Unexpected ‘molecular switch’ from a closely related mGlu4 PAM scaffold. Bioorg Med, Chem Lett. 2011;21:6955–6959. doi: 10.1016/j.bmcl.2011.09.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Knoflach F, et al. Positive allosteric modulators of metabotropic glutamate 1 receptor: characterization, mechanism of action and binding site. Proc Natl Acad Sci USA. 2001;98:13402–13407. doi: 10.1073/pnas.231358298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.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;70:616–626. doi: 10.1124/mol.105.021857. [DOI] [PubMed] [Google Scholar]
- 24.Owen DR. Recent advances in the medicinal chemistry of the metabotropic glutamate receptor 1 (mGlu1) ACS Chem Neurosci. 2011;2:394–401. doi: 10.1021/cn2000124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Sheffler DJ, et al. Recent progress in the synthesis and characterization of group II metabotropic glutamate receptor allosteric modulators. ACS Chem Neurosci. 2011;2:382–393. doi: 10.1021/cn200008d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.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;2:433–449. doi: 10.1021/cn200043e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Stauffer SR, et al. Progress toward positive allosteric modulators of the metabotropic glutamate receptor subtype 5 (mGlu5) ACS Chem Neurosci. 2011;2:450–470. doi: 10.1021/cn2000519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Emmitte KA, et al. Recent progress in the design and development of negative allosteric modulators of mGlu5. ACS Chem Neurosci. 2011;2:411–432. doi: 10.1021/cn2000266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ugolini A, et al. AMN082, an allosteric mGluR7 agonist that inhibits afferent glutamatergic transmission in rat basolateral amygdala. Neuropharmacology. 2008;55:532–536. doi: 10.1016/j.neuropharm.2008.04.020. [DOI] [PubMed] [Google Scholar]
- 30.Suzuki G, et al. In vitro pharmacological characterization of novel isoxazolopyridone derivatives as allosteric metabotropic glutamate receptor 7 antagonists. J Pharmacol Exp Ther. 2007;323:147–153. doi: 10.1124/jpet.107.124701. [DOI] [PubMed] [Google Scholar]
- 31.Duvoisin RM, et al. Opposing roles of mGluR8 in measures of anxiety involving non-social and social challenges. Behav Brian Res. 2011;221:50–54. doi: 10.1016/j.bbr.2011.02.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Frank RA, et al. Clustered coding variants in the glutamate receptor complexes of individuals with schizophrenia and bipolar disorder. PLoS One. 2011;6:e19011. doi: 10.1371/journal.pone.0019011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ayoub MA, et al. Deleterious GRM1 mutations in schizophrenia. PLoS One. 2012;7:e32849. doi: 10.1371/journal.pone.0032849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bellone C, et al. In utero exposure to cocaine delays postnatal synaptic maturation of glutamatergic transmission in the VTA. Nat Neurosci. 2011;14:1439–1447. doi: 10.1038/nn.2930. [DOI] [PubMed] [Google Scholar]
- 35.Lindsley CW, et al. Progress towards validating the NMDA receptor hypofunction hypothesis of schizophrenia. Curr Topics in Med Chem. 2006;8:771–784. doi: 10.2174/156802606777057599. [DOI] [PubMed] [Google Scholar]
- 36.Williams DL, Jr, Lindsley CW. Positive allosteric modulators of metabotropic glutamate receptor subtype 5 (mGluR5) Curr Topics in Med Chem. 2005;5:825–838. doi: 10.2174/1568026054750290. [DOI] [PubMed] [Google Scholar]
- 37.Sharma S, et al. Synthesis and SAR of a mGluR5 allosteric partial antagonist lead: Unexpected modulation of pharmacology with slight structural modifications to a 5-(phenylethynyl)pyrimidine scaffold. Bioorg Med Chem Lett. 2008;18:4098–4101. doi: 10.1016/j.bmcl.2008.05.091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wood MR, et al. Molecular switches’ on allosteric ligands that modulate modes of pharmacology’. Biochemistry. 2011;50:2403–2410. doi: 10.1021/bi200129s. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Jones CK, et al. The mGlu4 positive allosteric modulator VU0364770 produces efficacy alone and in combination with L-DOPA or an adenosine A2A antagonist in preclinical rodent models of Parkinson’s disease’. J Pharmacol & Exp Ther. 2012;340:404–421. doi: 10.1124/jpet.111.187443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Nemeth EF, et al. Pharmacodynamics of the type II calcimimetic compound cinacalcet HCl. J Pharmacol Exp Ther. 2004;308:627–635. doi: 10.1124/jpet.103.057273. [DOI] [PubMed] [Google Scholar]




