1
|
Frouni I, Kim E, Shaqfah J, Bédard D, Kwan C, Belliveau S, Huot P. [ 3H]-NFPS binding to the glycine transporter 1 in the hemi-parkinsonian rat brain. Exp Brain Res 2024; 242:1203-1214. [PMID: 38526743 PMCID: PMC11078860 DOI: 10.1007/s00221-024-06815-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 02/23/2024] [Indexed: 03/27/2024]
Abstract
L-3,4-dihydroxyphenylalanine (L-DOPA) is the main treatment for Parkinson's disease (PD) but with long term administration, motor complications such as dyskinesia are induced. Glycine transporter 1 (GlyT1) inhibition was shown to produce an anti-dyskinetic effect in parkinsonian rats and primates, coupled with an improvement in the anti-parkinsonian action of L-DOPA. The expression of GlyT1 in the brain in the dyskinetic state remains to be investigated. Here, we quantified the levels of GlyT1 across different brain regions using [3H]-NFPS in the presence of Org-25,935. Brain sections were chosen from sham-lesioned rats, L-DOPA-naïve 6-hydroxydopamine (6-OHDA)-lesioned rats and 6-OHDA-lesioned rats exhibiting mild or severe abnormal involuntary movements (AIMs). [3H]-NFPS binding decreased in the ipsilateral and contralateral thalamus, by 28% and 41%, in 6-OHDA-lesioned rats with severe AIMs compared to sham-lesioned animals (P < 0.01 and 0.001). [3H]-NFPS binding increased by 21% in the ipsilateral substantia nigra of 6-OHDA-lesioned rats with severe AIMs compared to 6-OHDA-lesioned rats with mild AIMs (P < 0.05). [3H]-NFPS binding was lower by 19% in the contralateral primary motor cortex and by 20% in the contralateral subthalamic nucleus of 6-OHDA-lesioned rats with mild AIMs animals compared to rats with severe AIMs (both P < 0.05). The severity of AIMs scores positively correlated with [3H]-NFPS binding in the ipsilateral substantia nigra (P < 0.05), ipsilateral entopeduncular nucleus (P < 0.05) and contralateral primary motor cortex (P < 0.05). These data provide an anatomical basis to explain the efficacy of GlyT1 inhibitors in dyskinesia in PD.
Collapse
Affiliation(s)
- Imane Frouni
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), 3801 University St, Montreal, QC, H3A 2B4, Canada
| | - Esther Kim
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), 3801 University St, Montreal, QC, H3A 2B4, Canada
| | - Judy Shaqfah
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), 3801 University St, Montreal, QC, H3A 2B4, Canada
| | - Dominique Bédard
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), 3801 University St, Montreal, QC, H3A 2B4, Canada
| | - Cynthia Kwan
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), 3801 University St, Montreal, QC, H3A 2B4, Canada
| | - Sébastien Belliveau
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), 3801 University St, Montreal, QC, H3A 2B4, Canada
| | - Philippe Huot
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada.
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), 3801 University St, Montreal, QC, H3A 2B4, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- Movement Disorder Clinic, Division of Neurology, Department of Neurosciences, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
2
|
Affiliation(s)
- Christopher L. Cioffi
- Departments of Basic and Clinical Sciences and Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences , Albany, NY, USA
| |
Collapse
|
3
|
Talpos JC. Symptomatic thinking: the current state of Phase III and IV clinical trials for cognition in schizophrenia. Drug Discov Today 2017; 22:1017-1026. [PMID: 28461223 DOI: 10.1016/j.drudis.2017.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 04/19/2017] [Accepted: 04/21/2017] [Indexed: 12/20/2022]
Abstract
Research indicates that relieving the cognitive and negative symptoms of schizophrenia is crucial for improving patient quality of life. However effective pharmacotherapies for cognitive and negative symptoms do not currently exist. A review of ongoing Phase III clinical trials indicates that, despite numerous compounds being investigated for cognition in schizophrenia, few are actually novel and most are not backed by empirically driven preclinical research efforts. Based on these trials, and a general disinvestment in development of novel therapies for schizophrenia, the likelihood of a major advancement in treating cognitive differences in schizophrenia does not look promising. Possible ways in which the remaining resources for development of novel treatment for schizophrenia can best be leveraged are discussed.
Collapse
Affiliation(s)
- John C Talpos
- National Center for Toxicological Research, 3900 NCTR Rd, Jefferson, AR 72079, USA.
| |
Collapse
|
4
|
Thomas R, Baker G, Dursun S, Todd K, Dhami K, Chue J, Chue P. Glycine Reuptake Inhibitors in the Treatment of Negative Symptoms of Schizophrenia. ACTA ACUST UNITED AC 2016. [DOI: 10.5455/bcp.20140918102748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Reji Thomas
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Glen Baker
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Serdar Dursun
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Kathryn Todd
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Kamaldeep Dhami
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - James Chue
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Pierre Chue
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Cioffi CL, Liu S, Wolf MA, Guzzo PR, Sadalapure K, Parthasarathy V, Loong DTJ, Maeng JH, Carulli E, Fang X, Karunakaran K, Matta L, Choo SH, Panduga S, Buckle RN, Davis RN, Sakwa SA, Gupta P, Sargent BJ, Moore NA, Luche MM, Carr GJ, Khmelnitsky YL, Ismail J, Chung M, Bai M, Leong WY, Sachdev N, Swaminathan S, Mhyre AJ. Synthesis and Biological Evaluation of N-((1-(4-(Sulfonyl)piperazin-1-yl)cycloalkyl)methyl)benzamide Inhibitors of Glycine Transporter-1. J Med Chem 2016; 59:8473-94. [PMID: 27559615 DOI: 10.1021/acs.jmedchem.6b00914] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We previously disclosed the discovery of rationally designed N-((1-(4-(propylsulfonyl)piperazin-1-yl)cycloalkyl)methyl)benzamide inhibitors of glycine transporter-1 (GlyT-1), represented by analogues 10 and 11. We describe herein further structure-activity relationship exploration of this series via an optimization strategy that primarily focused on the sulfonamide and benzamide appendages of the scaffold. These efforts led to the identification of advanced leads possessing a desirable balance of excellent in vitro GlyT-1 potency and selectivity, favorable ADME and in vitro pharmacological profiles, and suitable pharmacokinetic and safety characteristics. Representative analogue (+)-67 exhibited robust in vivo activity in the cerebral spinal fluid glycine biomarker model in both rodents and nonhuman primates. Furthermore, rodent microdialysis experiments also demonstrated that oral administration of (+)-67 significantly elevated extracellular glycine levels within the medial prefrontal cortex (mPFC).
Collapse
Affiliation(s)
- Christopher L Cioffi
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Shuang Liu
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Mark A Wolf
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Peter R Guzzo
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Kashinath Sadalapure
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Visweswaran Parthasarathy
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - David T J Loong
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Jun-Ho Maeng
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Edmund Carulli
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Xiao Fang
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Kalesh Karunakaran
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Lakshman Matta
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Sok Hui Choo
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Shailijia Panduga
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Ronald N Buckle
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Randall N Davis
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Samuel A Sakwa
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Priya Gupta
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Bruce J Sargent
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Nicholas A Moore
- Department of Medicinal Chemistry, AMRI , East Campus, 3 University Place, Rensselaer, New York 12144, United States
| | - Michele M Luche
- Bothell Research Center, AMRI , 22215 26th Ave SE, Bothell, Washington 98021-4425, United States
| | - Grant J Carr
- Bothell Research Center, AMRI , 22215 26th Ave SE, Bothell, Washington 98021-4425, United States
| | - Yuri L Khmelnitsky
- Drug Metabolism and Pharmacokinetics, AMRI , East Campus, 17 University Place, Rensselaer, New York 12144, United States
| | - Jiffry Ismail
- Drug Metabolism and Pharmacokinetics, AMRI , East Campus, 17 University Place, Rensselaer, New York 12144, United States
| | - Mark Chung
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Mei Bai
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Wei Yee Leong
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Nidhi Sachdev
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Srividya Swaminathan
- Discovery Research and Development Chemistry, Singapore Research Center, AMRI , 61 Science Park Road, Science Park III, 117525, Singapore
| | - Andrew J Mhyre
- Bothell Research Center, AMRI , 22215 26th Ave SE, Bothell, Washington 98021-4425, United States
| |
Collapse
|
6
|
Jin H, Zhang X, Yue X, Liu H, Li J, Yang H, Flores H, Su Y, Parsons SM, Perlmutter JS, Tu Z. Kinetics modeling and occupancy studies of a novel C-11 PET tracer for VAChT in nonhuman primates. Nucl Med Biol 2015; 43:131-9. [PMID: 26872437 DOI: 10.1016/j.nucmedbio.2015.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 10/29/2015] [Accepted: 11/05/2015] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Deficits in cholinergic function have been found in the aged brain and in neurodegenerative diseases including Alzheimer's disease (AD) and Parkinson's disease (PD). The vesicular acetylcholine transporter (VAChT) is a reliable biomarker for the cholinergic system. We previously reported the initial in vitro and ex vivo characterization of (-)-[(11)C]TZ659 as a VAChT specific ligand. Here, we report the in vivo specificity, tracer kinetics, and dose-occupancy studies in the nonhuman primate brain. METHODS MicroPET brain imaging of (-)-[(11)C]TZ659 was performed under baseline conditions in two male macaques. Tracer kinetic modeling was carried out using a two-tissue compartment model (2TCM) and Logan plot with arterial blood input function and using a simplified reference tissue model (SRTM) and Logan plot (LoganREF) without blood input. Specificity for VAChT was demonstrated by pretreatment with (+)-pentazocine, (-)-vesamicol, or S-(-)-eticlopride. Target occupancy (Occ) was calculated following pretreatment with escalating doses of (-)-vesamicol. RESULTS Baseline PET imaging revealed selective retention in the striatum with rapid clearance from the cerebellar hemispheres as a reference region. Total volume of distribution (VT) values derived from both 2TCM and Logan analysis with blood input revealed ~3-fold higher levels of (-)-[(11)C]TZ659 in the striatum than the cerebellar hemispheres. Injection of (-)-vesamicol either as a blocking or displacing agent significantly reduced striatal uptake of (-)-[(11)C]TZ659. In contrast, pretreatment with the sigma-1 ligand (+)-pentazocine had no impact. Pretreatment with the S-(-)-eticlopride, a dopamine D2-like receptor antagonist, increased striatal uptake of (-)-[(11)C]TZ659. Striatal binding potential (BPND, range of 0.33-1.6 with cerebellar hemispheres as the reference region) showed good correlation (r(2)=0.97) between SRTM and LoganREF. Occupancy studies found that ~0.0057 mg/kg of (-)-vesamicol produced 50% VAChT occupancy in the striatum. CONCLUSION (-)-[(11)C]TZ659 demonstrated specific and reversible VAChT binding and favorable pharmacokinetic properties for assessing the density of VAChT in the living brain.
Collapse
Affiliation(s)
- Hongjun Jin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiang Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xuyi Yue
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hui Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Junfeng Li
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hao Yang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hubert Flores
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yi Su
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Stanley M Parsons
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
7
|
Assmus F, Seelig A, Gobbi L, Borroni E, Glaentzlin P, Fischer H. Label-free assay for the assessment of nonspecific binding of positron emission tomography tracer candidates. Eur J Pharm Sci 2015; 79:27-35. [DOI: 10.1016/j.ejps.2015.08.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/02/2015] [Accepted: 08/25/2015] [Indexed: 10/23/2022]
|
8
|
Liu H, Jin H, Yue X, Zhang X, Yang H, Li J, Flores H, Su Y, Perlmutter JS, Tu Z. Preclinical evaluation of a promising C-11 labeled PET tracer for imaging phosphodiesterase 10A in the brain of living subject. Neuroimage 2015. [PMID: 26216275 DOI: 10.1016/j.neuroimage.2015.07.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Phosphodiesterase 10A (PDE10A) plays a key role in the regulation of brain striatal signaling. A PET tracer for PDE10A may serve as a tool to evaluate PDE10A expression in vivo in central nervous system disorders with striatal pathology. Here, we further characterized the binding properties of a previously reported radioligand we developed for PDE10A, [(11)C]TZ1964B, in rodents and nonhuman primates (NHPs). The tritiated counterpart [(3)H]TZ1964B was used for in vitro binding characterizations in rat striatum homogenates and in vitro autoradiographic studies in rat brain slices. The carbon-11 labeled [(11)C]TZ1964B was utilized in the ex vivo autoradiography studies for the brain of rats and microPET imaging studies for the brain of NHPs. MicroPET scans of [(11)C]TZ1964B in NHPs were conducted at baseline, as well as with using a selective PDE10A inhibitor MP-10 for either pretreatment or displacement. The in vivo regional target occupancy (Occ) was obtained by pretreating with different doses of MP-10 (0.05-2.00 mg/kg). Both in vitro binding assays and in vitro autoradiographic studies revealed a nanomolar binding affinity of [(3)H]TZ1964B to the rat striatum. The striatal binding of [(3)H]TZ1964B and [(11)C]TZ1964B was either displaced or blocked by MP-10 in rats and NHPs. Autoradiography and microPET imaging confirmed that the specific binding of the radioligand was found in the striatum but not in the cerebellum. Blocking studies also confirmed the suitability of the cerebellum as an appropriate reference region. The binding potentials (BPND) of [(11)C]TZ1964B in the NHP striatum that were calculated using either the Logan reference model (LoganREF, 3.96 ± 0.17) or the simplified reference tissue model (SRTM, 4.64 ± 0.47), with the cerebellum as the reference region, was high and had good reproducibility. The occupancy studies indicated a MP-10 dose of 0.31 ± 0.09 mg/kg (LoganREF)/0.45 ± 0.17mg/kg (SRTM) occupies 50% striatal PDE10A binding sites. Studies in rats and NHPs demonstrated radiolabeled TZ1964B has a high binding affinity and good specificity for PDE10A, as well as favorable in vivo pharmacokinetic properties and binding profiles. Our data suggests that [(11)C]TZ1964B is a promising radioligand for in vivo imaging PDE10A in the brain of living subject.
Collapse
Affiliation(s)
- Hui Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hongjun Jin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xuyi Yue
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiang Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hao Yang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Junfeng Li
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hubert Flores
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yi Su
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
9
|
Castner SA, Murthy NV, Ridler K, Herdon H, Roberts BM, Weinzimmer DP, Huang Y, Zheng MQ, Rabiner EA, Gunn RN, Carson RE, Williams GV, Laruelle M. Relationship between glycine transporter 1 inhibition as measured with positron emission tomography and changes in cognitive performances in nonhuman primates. Neuropsychopharmacology 2014; 39:2742-9. [PMID: 24487737 PMCID: PMC4200505 DOI: 10.1038/npp.2014.4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 11/04/2013] [Accepted: 12/13/2013] [Indexed: 01/22/2023]
Abstract
Several lines of evidence suggest that schizophrenia is associated with deficits in glutamatergic transmission at the N-methyl-d-aspartate (NMDA) receptors. Glycine is a NMDA receptor co-agonist, and extracellular levels of glycine are regulated in the forebrain by the glycine type-1 transporters (GlyT-1). GlyT-1 inhibitors elevate extracellular glycine and thus potentiate NMDA transmission. This mechanism represents a promising new avenue for the treatment of schizophrenia. Here, the recently introduced positron emission tomography radiotracer [11C]GSK931145 was used to quantify the relationship between occupancy of GlyT-1 by a GlyT-1 inhibitor, Org 25935, and its impact on spatial working memory performances in rhesus monkeys. The effect of Org 25935 on working memory was assessed both in control conditions and during a state of relative NMDA hypofunction induced by ketamine administration, at a dose selected for each animal to reduce task performance by about 50%. Under control conditions, Org 25935 had no effect on working memory at GlyT-1 occupancies lower than 75% and significantly impaired working memory at occupancies higher than 75%. Under ketamine conditions, Org 25935 reversed the deficit in working memory induced by ketamine and did so optimally in the 40-70% GlyT-1 occupancy range. The results confirm the efficacy of this mechanism to correct working memory deficits associated with NMDA hypofunction. These data also suggest the existence of an inverted-U dose-response curve in the potential therapeutic effect of this class of compounds.
Collapse
Affiliation(s)
- S A Castner
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - N V Murthy
- Neurosciences Centre for Excellence in Drug Discovery, GlaxoSmithKline, Harlow, UK
| | - K Ridler
- Clinical Imaging Centre, GlaxoSmithKline, Hammersmith Hospital–Imperial College, London, UK
| | - H Herdon
- Neurosciences Centre for Excellence in Drug Discovery, GlaxoSmithKline, Harlow, UK
| | - B M Roberts
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - D P Weinzimmer
- Department of Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Y Huang
- Department of Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - M Q Zheng
- Department of Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - E A Rabiner
- Clinical Imaging Centre, GlaxoSmithKline, Hammersmith Hospital–Imperial College, London, UK
| | - R N Gunn
- Clinical Imaging Centre, GlaxoSmithKline, Hammersmith Hospital–Imperial College, London, UK
| | - R E Carson
- Department of Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - G V Williams
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - M Laruelle
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA,Neurosciences Centre for Excellence in Drug Discovery, GlaxoSmithKline, Harlow, UK,Department of Radiology, Yale University School of Medicine, New Haven, CT, USA,UCB Pharma, Braine-l'Alleud, Brussels, Belgium,UCB Pharma, Chemin du Foriest, Braine-l'Alleud 1420, Belgium, Tel: +1 914 316 0923, Fax: +322 386 2550, E-mail:
| |
Collapse
|
10
|
Glycine transporters as novel therapeutic targets in schizophrenia, alcohol dependence and pain. Nat Rev Drug Discov 2014; 12:866-85. [PMID: 24172334 DOI: 10.1038/nrd3893] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glycine transporters are endogenous regulators of the dual functions of glycine, which acts as a classical inhibitory neurotransmitter at glycinergic synapses and as a modulator of neuronal excitation mediated by NMDA (N-methyl-D-aspartate) receptors at glutamatergic synapses. The two major subtypes of glycine transporters, GlyT1 and GlyT2, have been linked to the pathogenesis and/or treatment of central and peripheral nervous system disorders, including schizophrenia and related affective and cognitive disturbances, alcohol dependence, pain, epilepsy, breathing disorders and startle disease (also known as hyperekplexia). This Review examines the rationale for the therapeutic potential of GlyT1 and GlyT2 inhibition, and surveys the latest advances in the biology of glycine reuptake and transport as well as the drug discovery and clinical development of compounds that block glycine transporters.
Collapse
|
11
|
GlyT-1 Inhibitors: From Hits to Clinical Candidates. SMALL MOLECULE THERAPEUTICS FOR SCHIZOPHRENIA 2014. [DOI: 10.1007/7355_2014_53] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
12
|
Glycine transporter-1 inhibition promotes striatal axon sprouting via NMDA receptors in dopamine neurons. J Neurosci 2013; 33:16778-89. [PMID: 24133278 DOI: 10.1523/jneurosci.3041-12.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
NMDA receptor activity is involved in shaping synaptic connections throughout development and adulthood. We recently reported that brief activation of NMDA receptors on cultured ventral midbrain dopamine neurons enhanced their axon growth rate and induced axonal branching. To test whether this mechanism was relevant to axon regrowth in adult animals, we examined the reinnervation of dorsal striatum following nigral dopamine neuron loss induced by unilateral intrastriatal injections of the toxin 6-hydroxydopamine. We used a pharmacological approach to enhance NMDA receptor-dependent signaling by treatment with an inhibitor of glycine transporter-1 that elevates levels of extracellular glycine, a coagonist required for NMDA receptor activation. All mice displayed sprouting of dopaminergic axons from spared fibers in the ventral striatum to the denervated dorsal striatum at 7 weeks post-lesion, but the reinnervation in mice treated for 4 weeks with glycine uptake inhibitor was approximately twice as dense as in untreated mice. The treated mice also displayed higher levels of striatal dopamine and a complete recovery from lateralization in a test of sensorimotor behavior. We confirmed that the actions of glycine uptake inhibition on reinnervation and behavioral recovery required NMDA receptors in dopamine neurons using targeted deletion of the NR1 NMDA receptor subunit in dopamine neurons. Glycine transport inhibitors promote functionally relevant sprouting of surviving dopamine axons and could provide clinical treatment for disorders such as Parkinson's disease.
Collapse
|
13
|
Glycine transporter type 1 occupancy by bitopertin: a positron emission tomography study in healthy volunteers. Neuropsychopharmacology 2013; 38:504-12. [PMID: 23132267 PMCID: PMC3547202 DOI: 10.1038/npp.2012.212] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Deficient N-methyl-D-aspartate (NMDA) receptor transmission is thought to underlie schizophrenia. An approach for normalizing glutamate neurotransmission by enhancing NMDA receptor transmission is to increase glycine availability by inhibiting the glycine transporter type 1 (GlyT1). This study investigated the relationship between the plasma concentration of the glycine reuptake inhibitor bitopertin (RG1678) and brain GlyT1 occupancy. Healthy male volunteers received up to 175 mg bitopertin once daily, for 10-12 days. Three positron emission tomography scans, preceded by a single intravenous infusion of ∼30 mCi [(11)C]RO5013853, were performed: at baseline, on the last day of bitopertin treatment, and 2 days after drug discontinuation. Eighteen subjects were enrolled. At baseline, regional volume of distribution (V(T)) values were highest in the pons, thalamus, and cerebellum (1.7-2.7 ml/cm(3)) and lowest in cortical areas (∼0.8 ml/cm(3)). V(T) values were reduced to a homogeneous level following administration of 175 mg bitopertin. Occupancy values derived by a two-tissue five-parameter (2T5P) model, a simplified reference tissue model (SRTM), and a pseudoreference tissue model (PRTM) were overall comparable. At steady state, the relationship between bitopertin plasma concentration and GlyT1 occupancy derived by the 2T5P model, SRTM, and PRTM exhibited an EC(50) of ∼190, ∼200, and ∼130 ng/ml, respectively. E(max) was ∼92% independently of the model used. Bitopertin plasma concentration was a reliable predictor of occupancy because the concentration-occupancy relationship was superimposable at steady state and 2 days after drug discontinuation. These data allow understanding of the concentration-occupancy-efficacy relationship of bitopertin and support dose selection of future molecules.
Collapse
|
14
|
Beyoğlu D, Idle JR. The glycine deportation system and its pharmacological consequences. Pharmacol Ther 2012; 135:151-67. [PMID: 22584143 PMCID: PMC3665358 DOI: 10.1016/j.pharmthera.2012.05.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 04/27/2012] [Indexed: 12/13/2022]
Abstract
The glycine deportation system is an essential component of glycine catabolism in man whereby 400 to 800mg glycine per day are deported into urine as hippuric acid. The molecular escort for this deportation is benzoic acid, which derives from the diet and from gut microbiota metabolism of dietary precursors. Three components of this system, involving hepatic and renal metabolism, and renal active tubular secretion help regulate systemic and central nervous system levels of glycine. When glycine levels are pathologically high, as in congenital nonketotic hyperglycinemia, the glycine deportation system can be upregulated with pharmacological doses of benzoic acid to assist in normalization of glycine homeostasis. In congenital urea cycle enzymopathies, similar activation of the glycine deportation system with benzoic acid is useful for the excretion of excess nitrogen in the form of glycine. Drugs which can substitute for benzoic acid as substrates for the glycine deportation system have adverse reactions that may involve perturbations of glycine homeostasis. The cancer chemotherapeutic agent ifosfamide has an unacceptably high incidence of encephalopathy. This would appear to arise as a result of the production of toxic aldehyde metabolites which deplete ATP production and sequester NADH in the mitochondrial matrix, thereby inhibiting the glycine deportation system and causing de novo glycine synthesis by the glycine cleavage system. We hypothesize that this would result in hyperglycinemia and encephalopathy. This understanding may lead to novel prophylactic strategies for ifosfamide encephalopathy. Thus, the glycine deportation system plays multiple key roles in physiological and neurotoxicological processes involving glycine.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Hepatology Research Group, Department of Clinical Research, University of Bern, 3010 Bern, Switzerland
| | - Jeffrey R. Idle
- Hepatology Research Group, Department of Clinical Research, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
15
|
Wong DF, Ostrowitzki S, Zhou Y, Raymont V, Hofmann C, Borroni E, Kumar A, Parkar N, Brašić JR, Hilton J, Dannals RF, Martin-Facklam M. Characterization of [11C]RO5013853, a novel PET tracer for the glycine transporter type 1 (GlyT1) in humans. Neuroimage 2011; 75:282-290. [PMID: 22155032 DOI: 10.1016/j.neuroimage.2011.11.052] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 11/08/2011] [Accepted: 11/14/2011] [Indexed: 11/26/2022] Open
Abstract
We characterize a novel radioligand for the glycine transporter type 1 (GlyT1), [(11)C]RO5013853, in humans. Ten healthy male volunteers, 23-60 years of age, were enrolled in this PET study; seven subjects participated in the evaluation of test-retest reliability and three subjects in whole body dosimetry. Subjects were administered intravenous bolus injections of approximately 1100 MBq (30 mCi) [(11)C]RO5013853 with a high specific activity of about 481 GBq (13 Ci)/μmol. Standard compartmental model analysis with arterial plasma input function, and an alternative noninvasive analysis method which was evaluated and validated by occupancy studies in both baboons and humans, were performed. Mean parameter estimates of the volumes of distribution (VT) obtained by a 2-tissue 5-parameter model were higher in the cerebellum, pons, and thalamus (1.99 to 2.59 mL/mL), and lower in the putamen, caudate, and cortical areas (0.86 to 1.13 mL/mL), with estimates showing less than 10% difference between test and retest scans. Tracer retention was effectively blocked by the specific glycine reuptake inhibitor (GRI), bitopertin (RG1678). [(11)C]RO5013853 was safe and well tolerated. Human dosimetry studies showed that the effective dose was approximately 0.0033 mSv/MBq, with the liver receiving the highest absorbed dose. In conclusion, quantitative dynamic PET of the human brain after intravenous injection of [(11)C]RO5013853 attains reliable measurements of GlyT1 binding in accordance with the expected transporter distribution in the human brain. [(11)C]RO5013853 is a radioligand suitable for further clinical PET studies. Full characterization of a novel radiotracer for GlyT1 in humans is provided. The tracer has subsequently been used to assess receptor occupancy in healthy volunteers and to estimate occupancy at doses associated with best efficacy in a clinical trial with schizophrenic patients with predominantly negative symptoms.
Collapse
Affiliation(s)
- Dean F Wong
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, Room 3245, Johns Hopkins Outpatient Center, Baltimore, MD 21287 -0807, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287-0807, USA; Department of Environmental Health Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287-0807, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287 -0807, USA; Honorary Professor of Neuroscience and Pharmacology, University of Copenhagen, Denmark.
| | - Susanne Ostrowitzki
- F. Hoffmann-La Roche Ltd., Pharmaceutical Division, Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Yun Zhou
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287 -0807, USA
| | - Vanessa Raymont
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287 -0807, USA
| | - Carsten Hofmann
- F. Hoffmann-La Roche Ltd., Pharmaceutical Division, Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Edilio Borroni
- F. Hoffmann-La Roche Ltd., Pharmaceutical Division, Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Anil Kumar
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287 -0807, USA
| | - Nikhat Parkar
- F. Hoffmann-La Roche Ltd., Pharmaceutical Division, Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - James R Brašić
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287 -0807, USA
| | - John Hilton
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287 -0807, USA
| | - Robert F Dannals
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287 -0807, USA
| | - Meret Martin-Facklam
- F. Hoffmann-La Roche Ltd., Pharmaceutical Division, Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| |
Collapse
|