51
|
Turlington M, Malosh C, Jacobs J, Manka JT, Noetzel MJ, Vinson PN, Jadhav S, Herman EJ, Lavreysen H, Mackie C, Bartolomé-Nebreda JM, Conde-Ceide S, Martín-Martín ML, Tong HM, López S, MacDonald GJ, Steckler T, Daniels JS, Weaver CD, Niswender CM, Jones CK, Conn PJ, Lindsley CW, Stauffer SR. Tetrahydronaphthyridine and dihydronaphthyridinone ethers as positive allosteric modulators of the metabotropic glutamate receptor 5 (mGlu₅). J Med Chem 2014; 57:5620-37. [PMID: 24914612 PMCID: PMC4096224 DOI: 10.1021/jm500259z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022]
Abstract
Positive allosteric modulators (PAMs) of metabotropic glutamate receptor 5 (mGlu5) represent a promising therapeutic strategy for the treatment of schizophrenia. Starting from an acetylene-based lead from high throughput screening, an evolved bicyclic dihydronaphthyridinone was identified. We describe further refinements leading to both dihydronaphthyridinone and tetrahydronaphthyridine mGlu5 PAMs containing an alkoxy-based linkage as an acetylene replacement. Exploration of several structural features including western pyridine ring isomers, positional amides, linker connectivity/position, and combinations thereof, reveal that these bicyclic modulators generally exhibit steep SAR and within specific subseries display a propensity for pharmacological mode switching at mGlu5 as well as antagonist activity at mGlu3. Structure-activity relationships within a dihydronaphthyridinone subseries uncovered 12c (VU0405372), a selective mGlu5 PAM with good in vitro potency, low glutamate fold-shift, acceptable DMPK properties, and in vivo efficacy in an amphetamine-based model of psychosis.
Collapse
Affiliation(s)
- Mark Turlington
- Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Pomierny-Chamioło L, Rup K, Pomierny B, Niedzielska E, Kalivas PW, Filip M. Metabotropic glutamatergic receptors and their ligands in drug addiction. Pharmacol Ther 2014; 142:281-305. [DOI: 10.1016/j.pharmthera.2013.12.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/13/2013] [Accepted: 12/02/2013] [Indexed: 02/07/2023]
|
53
|
Yin S, Niswender CM. Progress toward advanced understanding of metabotropic glutamate receptors: structure, signaling and therapeutic indications. Cell Signal 2014; 26:2284-97. [PMID: 24793301 DOI: 10.1016/j.cellsig.2014.04.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/08/2014] [Accepted: 04/27/2014] [Indexed: 12/24/2022]
Abstract
The metabotropic glutamate (mGlu) receptors are a group of Class C seven-transmembrane spanning/G protein-coupled receptors (7TMRs/GPCRs). These receptors are activated by glutamate, one of the standard amino acids and the major excitatory neurotransmitter. By activating G protein-dependent and non-G protein-dependent signaling pathways, mGlus modulate glutamatergic transmission both in the periphery and throughout the central nervous system. Since the discovery of the first mGlu receptor, and especially during the last decade, a great deal of progress has been made in understanding the signaling, structure, pharmacological manipulation and therapeutic indications of the 8 mGlu members.
Collapse
Affiliation(s)
- Shen Yin
- Department of Pharmacology, Vanderbilt University Medical School, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical School, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University Medical School, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical School, Nashville, TN 37232, USA.
| |
Collapse
|
54
|
Wu H, Wang C, Gregory KJ, Han GW, Cho HP, Xia Y, Niswender CM, Katritch V, Meiler J, Cherezov V, Conn PJ, Stevens RC. Structure of a class C GPCR metabotropic glutamate receptor 1 bound to an allosteric modulator. Science 2014; 344:58-64. [PMID: 24603153 DOI: 10.1126/science.1249489] [Citation(s) in RCA: 403] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/01/2023]
Abstract
The excitatory neurotransmitter glutamate induces modulatory actions via the metabotropic glutamate receptors (mGlus), which are class C G protein-coupled receptors (GPCRs). We determined the structure of the human mGlu1 receptor seven-transmembrane (7TM) domain bound to a negative allosteric modulator, FITM, at a resolution of 2.8 angstroms. The modulator binding site partially overlaps with the orthosteric binding sites of class A GPCRs but is more restricted than most other GPCRs. We observed a parallel 7TM dimer mediated by cholesterols, which suggests that signaling initiated by glutamate's interaction with the extracellular domain might be mediated via 7TM interactions within the full-length receptor dimer. A combination of crystallography, structure-activity relationships, mutagenesis, and full-length dimer modeling provides insights about the allosteric modulation and activation mechanism of class C GPCRs.
Collapse
Affiliation(s)
- Huixian Wu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Mølck C, Harpsøe K, Gloriam DE, Mathiesen JM, Nielsen SM, Bräuner-Osborne H. mGluR5: Exploration of Orthosteric and Allosteric Ligand Binding Pockets and Their Applications to Drug Discovery. Neurochem Res 2014; 39:1862-75. [DOI: 10.1007/s11064-014-1248-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/06/2013] [Revised: 01/14/2014] [Accepted: 01/21/2014] [Indexed: 10/25/2022]
|
56
|
Cacabelos R, Cacabelos P, Torrellas C, Tellado I, Carril JC. Pharmacogenomics of Alzheimer's disease: novel therapeutic strategies for drug development. Methods Mol Biol 2014; 1175:323-556. [PMID: 25150875 DOI: 10.1007/978-1-4939-0956-8_13] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a major problem of health and disability, with a relevant economic impact on our society. Despite important advances in pathogenesis, diagnosis, and treatment, its primary causes still remain elusive, accurate biomarkers are not well characterized, and the available pharmacological treatments are not cost-effective. As a complex disorder, AD is a polygenic and multifactorial clinical entity in which hundreds of defective genes distributed across the human genome may contribute to its pathogenesis. Diverse environmental factors, cerebrovascular dysfunction, and epigenetic phenomena, together with structural and functional genomic dysfunctions, lead to amyloid deposition, neurofibrillary tangle formation, and premature neuronal death, the major neuropathological hallmarks of AD. Future perspectives for the global management of AD predict that genomics and proteomics may help in the search for reliable biomarkers. In practical terms, the therapeutic response to conventional drugs (cholinesterase inhibitors, multifactorial strategies) is genotype-specific. Genomic factors potentially involved in AD pharmacogenomics include at least five categories of gene clusters: (1) genes associated with disease pathogenesis; (2) genes associated with the mechanism of action of drugs; (3) genes associated with drug metabolism (phase I and II reactions); (4) genes associated with drug transporters; and (5) pleiotropic genes involved in multifaceted cascades and metabolic reactions. The implementation of pharmacogenomic strategies will contribute to optimize drug development and therapeutics in AD and related disorders.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Chair of Genomic Medicine, Camilo José Cela University, 28692, Villanueva de la Cañada, Madrid, Spain,
| | | | | | | | | |
Collapse
|
57
|
Nickols HH, Conn PJ. Development of allosteric modulators of GPCRs for treatment of CNS disorders. Neurobiol Dis 2014; 61:55-71. [PMID: 24076101 PMCID: PMC3875303 DOI: 10.1016/j.nbd.2013.09.013] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/25/2013] [Revised: 09/13/2013] [Accepted: 09/17/2013] [Indexed: 12/14/2022] Open
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 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.
Collapse
Key Words
- (+)-6-(2,4-dimethylphenyl)-2-ethyl-6,7-dihydrobenzo[d]oxazol-4(5H)-one
- (1-(4-cyano-4-(pyridine-2-yl)piperidine-1-yl)methyl-4-oxo-4H-quinolizine-3-carboxylic acid)
- (1S,2S)-N(1)-(3,4-dichlorophenyl)cyclohexane-1,2-dicarboxamide
- (1S,3R,4S)-1-aminocyclo-pentane-1,3,4-tricarboxylic acid
- (3,4-dihydro-2H-pyrano[2,3]b quinolin-7-yl)(cis-4-methoxycyclohexyl) methanone
- (3aS,5S,7aR)-methyl 5-hydroxy-5-(m-tolylethynyl)octahydro-1H-indole-1-carboxylate
- 1-(1′-(2-methylbenzyl)-1,4′-bipiperidin-4-yl)-1H-benzo[d]imidazol-2(3H)-one
- 1-[3-(4-butyl-1-piperidinyl)propyl]-3,4-dihydro-2(1H)-quinolinone
- 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- 2-(2-(3-methoxyphenyl)ethynyl)-5-methylpyridine
- 2-chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-1Himidazol-4-yl)ethynyl)pyridine
- 2-methyl-6-(2-phenylethenyl)pyridine
- 2-methyl-6-(phenylethynyl)-pyridine
- 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide
- 3-cyclohexyl-5-fluoro-6-methyl-7-(2-morpholin-4-ylethoxy)-4H-chromen-4-one
- 3[(2-methyl-1,3-thiazol-4-yl)ethylnyl]pyridine
- 4-((E)-styryl)-pyrimidin-2-ylamine
- 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
- 4-n-butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]-piperidine
- 5-methyl-6-(phenylethynyl)-pyridine
- 5MPEP
- 6-(4-methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo[4,5-c]pyridin-4(5H)-one
- 6-OHDA
- 6-hydroxydopamine
- 6-methyl-2-(phenylazo)-3-pyridinol
- 77-LH-28-1
- 7TMR
- AC-42
- ACPT-1
- AChE
- AD
- ADX71743
- AFQ056
- APP
- Allosteric modulator
- Alzheimer's disease
- BINA
- BQCA
- CDPPB
- CFMMC
- CNS
- CPPHA
- CTEP
- DA
- DFB
- DHPG
- Drug discovery
- ERK1/2
- FMRP
- FTIDC
- FXS
- Fragile X syndrome
- GABA
- GPCR
- JNJ16259685
- L-AP4
- L-DOPA
- Lu AF21934
- Lu AF32615
- M-5MPEP
- MMPIP
- MPEP
- MPTP
- MTEP
- Metabotropic glutamate receptor
- Muscarinic acetylcholine receptor
- N-[4-chloro-2[(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)methyl]phenyl]-2-hydrobenzamide
- N-methyl-d-aspartate
- N-phenyl-7-(hydroxylimino)cyclopropa[b]chromen-1a-carboxamide
- NAM
- NMDA
- PAM
- PCP
- PD
- PD-LID
- PET
- PHCCC
- PQCA
- Parkinson's disease
- Parkinson's disease levodopa-induced dyskinesia
- SAM
- SIB-1757
- SIB-1893
- TBPB
- [(3-fluorophenyl)methylene]hydrazone-3-fluorobenzaldehyde
- acetylcholinesterase
- amyloid precursor protein
- benzylquinolone carboxylic acid
- central nervous system
- dihydroxyphenylglycine
- dopamine
- extracellular signal-regulated kinase 1/2
- fragile X mental retardation protein
- l-(+)-2-amino-4-phosphonobutyric acid
- l-3,4-dihydroxyphenylalanine
- mGlu
- metabotropic glutamate receptor
- negative allosteric modulator
- phencyclidine
- positive allosteric modulator
- positron emission tomography
- potassium 30-([(2-cyclopentyl-6-7-dimethyl-1-oxo-2,3-dihydro-1H-inden-5yl)oxy]methyl)biphenyl l-4-carboxylate
- seven transmembrane receptor
- silent allosteric modulator
- γ-aminobutyric acid
Collapse
Affiliation(s)
- Hilary Highfield Nickols
- Division of Neuropathology, Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, 37232, USA
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
58
|
Shineman DW, Carman AJ, Dacks PA, Lane RF, Fillit HM. Progress in novel cognitive enhancers for cognitive aging and Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2013; 5:45. [PMID: 24083622 PMCID: PMC3979029 DOI: 10.1186/alzrt209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Academic Contribution Register] [Indexed: 11/23/2022]
Abstract
Increased knowledge of the biology of synaptic function has led to the development of novel cognitive-enhancing therapeutic strategies with the potential for increased efficacy and safety. This editorial highlights a diverse array of approaches currently being explored to target cognitive dysfunction due to aging and/or Alzheimer’s disease.
Collapse
Affiliation(s)
- Diana W Shineman
- Alzheimer's Drug Discovery Foundation, 57 West 57th Street, Suite 904, New York, NY 10019, USA
| | - Aaron J Carman
- Alzheimer's Drug Discovery Foundation, 57 West 57th Street, Suite 904, New York, NY 10019, USA
| | - Penny A Dacks
- Alzheimer's Drug Discovery Foundation, 57 West 57th Street, Suite 904, New York, NY 10019, USA
| | - Rachel F Lane
- Alzheimer's Drug Discovery Foundation, 57 West 57th Street, Suite 904, New York, NY 10019, USA
| | - Howard M Fillit
- Alzheimer's Drug Discovery Foundation, 57 West 57th Street, Suite 904, New York, NY 10019, USA
| |
Collapse
|
59
|
Abstract
The presence of druggable, topographically distinct allosteric sites on a wide range of receptor families has offered new paradigms for small molecules to modulate receptor function. Moreover, ligands that target allosteric sites offer significant advantages over the corresponding orthosteric ligands in terms of selectivity, including subtype selectivity within receptor families, and can also impart improved physicochemical properties. However, allosteric ligands are not a panacea. Many chemical issues (e.g., flat structure-activity relationships) and pharmacological issues (e.g., ligand-biased signaling) that are allosteric centric have emerged. Notably, the fact that allosteric sites are less evolutionarily conserved leads to improved selectivity; however, this can also lead to species differences that can hinder safety assessment. Many allosteric ligands possess molecular switches, wherein a small structural change (chemical or metabolic) can modulate the mode of pharmacology or receptor subtype selectivity. As the field has matured, as described here, key principles and strategies have emerged for the design of ligands/drugs for allosteric sites.
Collapse
Affiliation(s)
- Cody J Wenthur
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600;
| | | | | | | |
Collapse
|
60
|
Metabotropic glutamate receptor 5 in schizophrenia: emerging evidence for the development of antipsychotic drugs. Future Med Chem 2013; 5:1471-4. [DOI: 10.4155/fmc.13.137] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/17/2022] Open
|
61
|
Gregory KJ, Herman EJ, Ramsey AJ, Hammond AS, Byun NE, Stauffer SR, Manka JT, Jadhav S, Bridges TM, Weaver CD, Niswender CM, Steckler T, Drinkenburg WH, Ahnaou A, Lavreysen H, Macdonald GJ, Bartolomé JM, Mackie C, Hrupka BJ, Caron MG, Daigle TL, Lindsley CW, Conn PJ, Jones CK. N-aryl piperazine metabotropic glutamate receptor 5 positive allosteric modulators possess efficacy in preclinical models of NMDA hypofunction and cognitive enhancement. J Pharmacol Exp Ther 2013; 347:438-57. [PMID: 23965381 DOI: 10.1124/jpet.113.206623] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022] Open
Abstract
Impaired transmission through glutamatergic circuits has been postulated to play a role in the underlying pathophysiology of schizophrenia. Furthermore, inhibition of the N-methyl-d-aspartate (NMDA) subtype of ionotropic glutamate receptors (NMDAR) induces a syndrome that recapitulates many of the symptoms observed in patients with schizophrenia. Selective activation of metabotropic glutamate receptor subtype 5 (mGlu5) may provide a novel therapeutic approach for treatment of symptoms associated with schizophrenia through facilitation of transmission through central glutamatergic circuits. Here, we describe the characterization of two novel N-aryl piperazine mGlu5 positive allosteric modulators (PAMs): 2-(4-(2-(benzyloxy)acetyl)piperazin-1-yl)benzonitrile (VU0364289) and 1-(4-(2,4-difluorophenyl)piperazin-1-yl)-2-((4-fluorobenzyl)oxy)ethanone (DPFE). VU0364289 and DPFE induced robust leftward shifts in the glutamate concentration-response curves for Ca(2+) mobilization and extracellular signal-regulated kinases 1 and 2 phosphorylation. Both PAMs displayed micromolar affinity for the common mGlu5 allosteric binding site and high selectivity for mGlu5. VU0364289 and DPFE possessed suitable pharmacokinetic properties for dosing in vivo and produced robust dose-related effects in reversing amphetamine-induced hyperlocomotion, a preclinical model predictive of antipsychotic-like activity. In addition, DPFE enhanced acquisition of contextual fear conditioning in rats and reversed behavioral deficits in a mouse model of NMDAR hypofunction. In contrast, DPFE had no effect on reversing apomorphine-induced disruptions of prepulse inhibition of the acoustic startle reflex. These mGlu5 PAMs also increased monoamine levels in the prefrontal cortex, enhanced performance in a hippocampal-mediated memory task, and elicited changes in electroencephalogram dynamics commensurate with procognitive effects. Collectively, these data support and extend the role for the development of novel mGlu5 PAMs for the treatment of psychosis and cognitive deficits observed in individuals with schizophrenia.
Collapse
Affiliation(s)
- K J Gregory
- Department of Pharmacology and Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee (K.J.G., E.J.H., A.S.H., N.E.B., S.R.S., J.T.M., S.J., T.M.B., C.D.W., C.M.N., C.W.L., P.J.C., C.K.J.); Drug Discovery Biology, MIPS, Monash University, Parkville, Victoria, Australia (K.J.G.); Department of Pharmacology and Toxicology, University of Toronto, Ontario, Canada (A.J.R.); Institute of Imaging and Science, Vanderbilt University (N.E.B.); Janssen Research & Development, Beerse, Belgium (T.S., W.H.D., A.A., H.L., G.J.M., C.M., B.J.H.); Janssen Research & Development, Toledo, Spain (J.M.B.); Department of Cell Biology, Duke University, Durham, North Carolina (M.G.C., T.L.D.); Department of Chemistry, Vanderbilt University Medical Center, Nashville, Tennessee (C.W.L.); and U.S. Department of Veterans Affairs, Nashville, Tennessee (C.K.J.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Cioffi CL. Modulation of NMDA receptor function as a treatment for schizophrenia. Bioorg Med Chem Lett 2013; 23:5034-44. [PMID: 23916256 DOI: 10.1016/j.bmcl.2013.07.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/07/2013] [Revised: 07/03/2013] [Accepted: 07/13/2013] [Indexed: 11/30/2022]
Abstract
Schizophrenia is a devastating mental illness that afflicts nearly 1% of the world's population. Currently available antipsychotics treat positive symptoms, but are largely ineffective at addressing negative symptoms and cognitive dysfunction. Thus, improved pharmacotherapies that treat all aspects of the disease remain a critical unmet need. There is mounting evidence that links NMDA receptor hypofunction and the expression of schizophrenia, and numerous drug discovery programs have developed agents that directly or indirectly potentiate NMDA receptor-mediated neurotransmission. Several compounds have emerged that show promise for treating all symptom sub-domains in both preclinical models and clinical studies, and we will review recent developments in many of these areas.
Collapse
|