1
|
Howes O, Marcinkowska J, Turkheimer FE, Carr R. Synaptic changes in psychiatric and neurological disorders: state-of-the art of in vivo imaging. Neuropsychopharmacology 2024; 50:164-183. [PMID: 39134769 PMCID: PMC11525650 DOI: 10.1038/s41386-024-01943-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/03/2024] [Accepted: 07/19/2024] [Indexed: 11/01/2024]
Abstract
Synapses are implicated in many neuropsychiatric illnesses. Here, we provide an overview of in vivo techniques to index synaptic markers in patients. Several positron emission tomography (PET) tracers for synaptic vesicle glycoprotein 2 A (SV2A) show good reliability and selectivity. We review over 50 clinical studies including over 1700 participants, and compare findings in healthy ageing and across disorders, including addiction, schizophrenia, depression, posttraumatic stress disorder, and neurodegenerative disorders, including tauopathies, Huntington's disease and α-synucleinopathies. These show lower SV2A measures in cortical brain regions across most of these disorders relative to healthy volunteers, with the most well-replicated findings in tauopathies, whilst changes in Huntington's chorea, Parkinson's disease, corticobasal degeneration and progressive supranuclear palsy are predominantly subcortical. SV2A PET measures are correlated with functional connectivity across brain networks, and a number of other measures of brain function, including glucose metabolism. However, the majority of studies found no relationship between grey matter volume measured with magnetic resonance imaging and SV2A PET measures. Cognitive dysfunction, in domains including working memory and executive function, show replicated inverse relationships with SV2A measures across diagnoses, and initial findings also suggest transdiagnostic relationships with mood and anxiety symptoms. This suggests that synaptic abnormalities could be a common pathophysiological substrate underlying cognitive and, potentially, affective symptoms. We consider limitations of evidence and future directions; highlighting the need to develop postsynaptic imaging markers and for longitudinal studies to test causal mechanisms.
Collapse
Affiliation(s)
- Oliver Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England.
- South London & the Maudsley NHS Trust, London, England.
- London Institute of Medical Sciences, London, England.
| | - Julia Marcinkowska
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
| | - Richard Carr
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
- South London & the Maudsley NHS Trust, London, England
- London Institute of Medical Sciences, London, England
| |
Collapse
|
2
|
Koush Y, Rothman DL, Behar KL, de Graaf RA, Hyder F. Human brain functional MRS reveals interplay of metabolites implicated in neurotransmission and neuroenergetics. J Cereb Blood Flow Metab 2022; 42:911-934. [PMID: 35078383 PMCID: PMC9125492 DOI: 10.1177/0271678x221076570] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/26/2021] [Accepted: 01/05/2022] [Indexed: 01/28/2023]
Abstract
While functional MRI (fMRI) localizes brain activation and deactivation, functional MRS (fMRS) provides insights into the underlying metabolic conditions. There is much interest in measuring task-induced and resting levels of metabolites implicated in neuroenergetics (e.g., lactate, glucose, or β-hydroxybutyrate (BHB)) and neurotransmission (e.g., γ-aminobutyric acid (GABA) or pooled glutamate and glutamine (Glx)). Ultra-high magnetic field (e.g., 7T) has boosted the fMRS quantification precision, reliability, and stability of spectroscopic observations using short echo-time (TE) 1H-MRS techniques. While short TE 1H-MRS lacks sensitivity and specificity for fMRS at lower magnetic fields (e.g., 3T or 4T), most of these metabolites can also be detected by J-difference editing (JDE) 1H-MRS with longer TE to filter overlapping resonances. The 1H-MRS studies show that JDE can detect GABA, Glx, lactate, and BHB at 3T, 4T and 7T. Most recently, it has also been demonstrated that JDE 1H-MRS is capable of reliable detection of metabolic changes in different brain areas at various magnetic fields. Combining fMRS measurements with fMRI is important for understanding normal brain function, but also clinically relevant for mechanisms and/or biomarkers of neurological and neuropsychiatric disorders. We provide an up-to-date overview of fMRS research in the last three decades, both in terms of applications and technological advances. Overall the emerging fMRS techniques can be expected to contribute substantially to our understanding of metabolism for brain function and dysfunction.
Collapse
Affiliation(s)
- Yury Koush
- Magnetic Resonance Research Center, Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Douglas L Rothman
- Magnetic Resonance Research Center, Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Kevin L Behar
- Magnetic Resonance Research Center, Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Robin A de Graaf
- Magnetic Resonance Research Center, Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Fahmeed Hyder
- Magnetic Resonance Research Center, Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
3
|
Bartlett EA, Ogden RT, Mann JJ, Zanderigo F. Source-to-Target Automatic Rotating Estimation (STARE) - a publicly-available, blood-free quantification approach for PET tracers with irreversible kinetics: Theoretical framework and validation for [ 18F]FDG. Neuroimage 2022; 249:118901. [PMID: 35026425 PMCID: PMC8969778 DOI: 10.1016/j.neuroimage.2022.118901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/16/2021] [Accepted: 01/09/2022] [Indexed: 10/30/2022] Open
Abstract
INTRODUCTION Full quantification of positron emission tomography (PET) data requires an input function. This generally means arterial blood sampling, which is invasive, labor-intensive and burdensome. There is no current, standardized method to fully quantify PET radiotracers with irreversible kinetics in the absence of blood data. Here, we present Source-to-Target Automatic Rotating Estimation (STARE), a novel, data-driven approach to quantify the net influx rate (Ki) of irreversible PET radiotracers, that requires only individual-level PET data and no blood data. We validate STARE with human [18F]FDG PET scans and assess its performance using simulations. METHODS STARE builds upon a source-to-target tissue model, where the tracer time activity curves (TACs) in multiple "target" regions are expressed at once as a function of a "source" region, based on the two-tissue irreversible compartment model, and separates target region Ki from source Ki by fitting the source-to-target model across all target regions simultaneously. To ensure identifiability, data-driven, subject-specific anchoring is used in the STARE minimization, which takes advantage of the PET signal in a vasculature cluster in the field of view (FOV) that is automatically extracted and partial volume-corrected. To avoid the need for any a priori determination of a single source region, each of the considered regions acts in turn as the source, and a final Ki is estimated in each region by averaging the estimates obtained in each source rotation. RESULTS In a large dataset of human [18F]FDG scans (N=69), STARE Ki estimates were correlated with corresponding arterial blood-based Ki estimates (r=0.80), with an overall regression slope of 0.88, and were precisely estimated, as assessed by comparing STARE Ki estimates across several runs of the algorithm (coefficient of variation across runs=6.74 ± 2.48%). In simulations, STARE Ki estimates were largely robust to factors that influence the individualized anchoring used within its algorithm. CONCLUSION Through simulations and application to [18F]FDG PET data, feasibility is demonstrated for STARE blood-free, data-driven quantification of Ki. Future work will include applying STARE to PET data obtained with a portable PET camera and to other irreversible radiotracers.
Collapse
Affiliation(s)
- Elizabeth A Bartlett
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Medical Center, New York, USA.
| | - R Todd Ogden
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Medical Center, New York, USA; Department of Biostatistics, Mailman School of Public Health, Columbia University Medical Center, New York, USA
| | - J John Mann
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Medical Center, New York, USA; Department of Radiology, Columbia University Medical Center, New York, USA
| | - Francesca Zanderigo
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, USA; Department of Psychiatry, Columbia University Medical Center, New York, USA
| |
Collapse
|
4
|
Schubert J, Tonietto M, Turkheimer F, Zanotti-Fregonara P, Veronese M. Supervised clustering for TSPO PET imaging. Eur J Nucl Med Mol Imaging 2021; 49:257-268. [PMID: 33779770 PMCID: PMC8712290 DOI: 10.1007/s00259-021-05309-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE This technical note seeks to act as a practical guide for implementing a supervised clustering algorithm (SVCA) reference region approach and to explain the main strengths and limitations of the technique in the context of 18-kilodalton translocator protein (TSPO) positron emission tomography (PET) studies in experimental medicine. BACKGROUND TSPO PET is the most widely used imaging technique for studying neuroinflammation in vivo in humans. Quantifying neuroinflammation with PET can be a challenging and invasive procedure, especially in frail patients, because it often requires blood sampling from an arterial catheter. A widely used alternative to arterial sampling is SVCA, which identifies the voxels with minimal specific binding in the PET images, thus extracting a pseudo-reference region for non-invasive quantification. Unlike other reference region approaches, SVCA does not require specification of an anatomical reference region a priori, which alleviates the limitation of TSPO contamination in anatomically-defined reference regions in individuals with underlying inflammatory processes. Furthermore, SVCA can be applied to any TSPO PET tracer across different neurological and neuropsychiatric conditions, providing noninvasivequantification of TSPO expression. METHODS We provide an overview of the development of SVCA as well as step-by-step instructions for implementing SVCA with suggestions for specific settings. We review the literature on SVCAapplications using first- and second- generation TSPO PET tracers and discuss potential clinically relevant limitations and applications. CONCLUSIONS The correct implementation of SVCA can provide robust and reproducible estimates of brain TSPO expression. This review encourages the standardisation of SVCA methodology in TSPO PET analysis, ultimately aiming to improve replicability and comparability across study sites.
Collapse
Affiliation(s)
- Julia Schubert
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Matteo Tonietto
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Paolo Zanotti-Fregonara
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
5
|
Frouni I, Belliveau S, Maddaford S, Nuara SG, Gourdon JC, Huot P. Effect of the glycine transporter 1 inhibitor ALX-5407 on dyskinesia, psychosis-like behaviours and parkinsonism in the MPTP-lesioned marmoset. Eur J Pharmacol 2021; 910:174452. [PMID: 34480885 DOI: 10.1016/j.ejphar.2021.174452] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 11/26/2022]
Abstract
Dyskinesia and psychosis are complications encountered in advanced Parkinson's disease (PD) following long-term therapy with L-3,4-dihydroxyphenylalanine (L-DOPA). Disturbances in the glutamatergic system have been associated with both dyskinesia and psychosis, making glutamatergic modulation a potential therapeutic approach for these. Treatments thus far have sought to dampen glutamatergic transmission, for example through blockade of N-methyl-D-aspartate (NMDA) receptors or modulation of metabotropic glutamate receptors 5. In contrast, activation of the glycine-binding site on NMDA receptors is required for their physiological response. Here, we investigated whether indirectly enhancing glutamatergic transmission through inhibition of glycine re-uptake would be efficacious in diminishing both dyskinesia and psychosis-like behaviours (PLBs) in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned common marmoset. Six marmosets were rendered parkinsonian by MPTP injection. Following repeated administration of L-DOPA to induce dyskinesia and PLBs, they underwent acute challenges of the glycine transporter 1 (GlyT1) inhibitor ALX-5407 (0.01, 0.1 and 1 mg/kg) or vehicle, in combination with L-DOPA, after which the severity of dyskinesia, PLBs and parkinsonian disability was evaluated. In combination with L-DOPA, ALX-5407 0.1 and 1 mg/kg significantly reduced the severity of dyskinesia, by 51% and 41% (both P < 0.001), when compared to vehicle. ALX-5407 0.01, 0.1 and 1 mg/kg also decreased the severity of global PLBs, by 25%, 51% and 38% (all P < 0.001), when compared to vehicle. The benefits on dyskinesia and PLBs were achieved without compromising the therapeutic effect of L-DOPA on parkinsonism. Our results suggest that GlyT1 inhibition may be a novel strategy to attenuate dyskinesia and PLBs in PD, without interfering with L-DOPA anti-parkinsonian action.
Collapse
Affiliation(s)
- Imane Frouni
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada; Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Sébastien Belliveau
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | | | - Stephen G Nuara
- Comparative Medicine & Animal Resource Centre, McGill University, Montreal, QC, Canada
| | - Jim C Gourdon
- Comparative Medicine & Animal Resource Centre, McGill University, Montreal, QC, Canada
| | - Philippe Huot
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada; Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada; Movement Disorder Clinic, Division of Neurology, Department of Neuroscience, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
6
|
Gobbi L, Mercier J, Bang-Andersen B, Nicolas JM, Reilly J, Wagner B, Whitehead D, Briard E, Maguire RP, Borroni E, Auberson YP. A Comparative Study of in vitro Assays for Predicting the Nonspecific Binding of PET Imaging Agents in vivo. ChemMedChem 2019; 15:585-592. [PMID: 31797561 DOI: 10.1002/cmdc.201900608] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/27/2019] [Indexed: 01/23/2023]
Abstract
Nonspecific binding (NSB) is a key parameter in optimizing PET imaging tracers. We compared the ability to predict NSB of three available methods: LIMBA, rat fu,brain , and CHI(IAM). Even though NSB is often associated with lipophilicity, we observed that logD does not correlate with any of these assays, clearly indicating that lipophilicity, while influencing NSB, is insufficient to predict it. A cross-comparison of the methods showed that all three correlate and are useful predictors of NSB. The three assays, however, rank the molecules slightly differently, illustrating the challenge of comparing molecules within a narrow chemical space. We also noted that CHI(IAM) values more effectively predict VNS , a measure of in vivo NSB in the human brain. CHI(IAM) measurements might be a closer model of the actual physicochemical interaction between PET tracer candidates and cell membranes, and seems to be the method of choice for the optimization of in vivo NSB.
Collapse
Affiliation(s)
- Luca Gobbi
- Pharma Research and Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd., 4070, Basel, Switzerland
| | - Joël Mercier
- UCB Early Solutions, UCB Biopharma sprl, 1420, Braine-l'Alleud, Belgium
| | - Benny Bang-Andersen
- Molecular Discovery and Innovation, H. Lundbeck A/S, 9 Ottiliavej, 2500, Valby, Denmark
| | | | - John Reilly
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Fabrikstrasse 2, 4056, Basel, Switzerland
| | - Björn Wagner
- Pharma Research and Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd., 4070, Basel, Switzerland
| | - David Whitehead
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Fabrikstrasse 2, 4056, Basel, Switzerland
| | - Emmanuelle Briard
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Fabrikstrasse 2, 4056, Basel, Switzerland
| | - R Paul Maguire
- UCB Early Solutions, UCB Biopharma sprl, 1420, Braine-l'Alleud, Belgium
| | - Edilio Borroni
- Pharma Research and Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd., 4070, Basel, Switzerland
| | - Yves P Auberson
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Fabrikstrasse 2, 4056, Basel, Switzerland
| |
Collapse
|
7
|
Feng D, Baumgartner R, Svetnik V. Bayesian Estimation of Agreement of Biomarkers in Early Drug Development. Stat Biopharm Res 2019. [DOI: 10.1080/19466315.2018.1537887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Dai Feng
- Biometrics Research, Merck & Co., Inc., Rahway, NJ
| | | | | |
Collapse
|
8
|
Sandiego CM, Barret O, Lee H, Alagille D, Amenta A, Fowles K, Holden D, Seibyl JP, Tamagnan G. Imaging histamine H3 receptors with [ 18 F]FMH3: Test-retest and occupancy studies in the non-human primate. Synapse 2019; 73:e22096. [PMID: 30835877 DOI: 10.1002/syn.22096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/01/2019] [Indexed: 01/03/2023]
Abstract
A positron emission tomography (PET) radioligand, [18 F]FMH3, has been developed to interrogate histamine receptor subtype 3 (H3R), where dysfunction at this site is linked with obesity, sleep abnormality, and cognitive disorders. [18 F]FMH3 was evaluated for imaging central H3R sites in non-human primates through test-retest (TRT) and dose-receptor occupancy studies with two selective H3R antagonists in order to support clinical investigations. Two adult female baboons underwent [18 F]FMH3 PET brain scans in the HR+, at repeated baseline (n = 7) and following administration of escalating doses of ABT-239 (0.003-0.1m/kg, n = 4) and ciproxifan (0.5-2.1 mg/kg, n = 7). Volume of distribution (VT ) in brain regions was estimated using the 2-tissue compartment model. TRT variability of VT across repeated baseline scans was reported as % coefficient of variation (COV). ABT-239 and ciproxifan occupancy at H3R was estimated using the occupancy plot, and the relationship of occupancy with dose and plasma levels was determined. In baboons, distribution of [18 F]FMH3 was high in the striatum, intermediate in cortical regions, and low in the brain stem. COV of baseline VT was 7.0 ± 3.5%, averaged across regions and animals. Dose-dependent effects of ABT-239 and ciproxifan measured the brain. ED50 and EC50, respectively, were 0.011 mg/kg and 0.942 ng/ml for ABT-239 and 0.73 mg/kg and 208.3 ng/ml for ciproxifan. [18 F]FMH3 demonstrated high TRT reliability and can be used to measure occupancy of H3R-targeted drugs. Validation in non-human primates support [18 F]FMH3 PET studies toward clinical investigations of H3R.
Collapse
Affiliation(s)
| | - Olivier Barret
- Invicro, a Konica Minolta Company, New Haven, Connecticut
| | - Hsiaoju Lee
- Invicro, a Konica Minolta Company, New Haven, Connecticut
| | - David Alagille
- Invicro, a Konica Minolta Company, New Haven, Connecticut
| | - Amy Amenta
- Invicro, a Konica Minolta Company, New Haven, Connecticut
| | - Krista Fowles
- Department of Diagnostic Radiology, Yale University, New Haven, Connecticut
| | - Daniel Holden
- Department of Diagnostic Radiology, Yale University, New Haven, Connecticut
| | - John P Seibyl
- Invicro, a Konica Minolta Company, New Haven, Connecticut
| | | |
Collapse
|
9
|
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
|
10
|
Rabiner EA. Imaging Synaptic Density: A Different Look at Neurologic Diseases. J Nucl Med 2017; 59:380-381. [PMID: 29123011 DOI: 10.2967/jnumed.117.198317] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/30/2017] [Indexed: 11/16/2022] Open
Affiliation(s)
- Eugenii A Rabiner
- Imanova Ltd. and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology, and Neuroscience, King's College, London, United Kingdom
| |
Collapse
|
11
|
Abstract
As the field of PET has expanded and an ever-increasing number and variety of compounds have been radiolabeled as potential in vivo tracers of biochemistry, transporters have become important primary targets or facilitators of radiotracer uptake and distribution. A transporter can be the primary target through the development of a specific high-affinity radioligand: examples are the multiple high-affinity radioligands for the neuronal membrane neurotransmitter or vesicular transporters, used to image nerve terminals in the brain. The goal of a radiotracer might be to study the function of a transporter through the use of a radiolabeled substrate, such as the application of 3-O-[11C]methyl]glucose to measure rates of glucose transport through the blood-brain barrier. In many cases, transporters are required for radiotracer distributions, but the targeted biochemistries might be unrelated: an example is the use of 2-deoxy-2-[18F]FDG for imaging glucose metabolism, where initial passage of the radiotracer through cell membranes requires the action of specific glucose transporters. Finally, there are transporters such as p-glycoprotein that function to extrude small molecules from tissues, and can effectively work against successful uptake of radiotracers. The diversity of structures and functions of transporters, their importance in human health and disease, and their role in therapeutic drug disposition suggest that in vivo imaging of transporter location and function will continue to be a point of emphasis in PET radiopharmaceutical development. In this review, the variety of transporters and their importance for in vivo PET radiotracer development and application are discussed. Transporters have thus joined the other major protein targets such as G-protein coupled receptors, ligand-gated ion channels, enzymes, and aggregated proteins as of high interest for understanding human health and disease.
Collapse
Affiliation(s)
- Michael R Kilbourn
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI.
| |
Collapse
|
12
|
Zanderigo F, Mann JJ, Ogden RT. A hybrid deconvolution approach for estimation of in vivo non-displaceable binding for brain PET targets without a reference region. PLoS One 2017; 12:e0176636. [PMID: 28459878 PMCID: PMC5411064 DOI: 10.1371/journal.pone.0176636] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/13/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND AIM Estimation of a PET tracer's non-displaceable distribution volume (VND) is required for quantification of specific binding to its target of interest. VND is generally assumed to be comparable brain-wide and is determined either from a reference region devoid of the target, often not available for many tracers and targets, or by imaging each subject before and after blocking the target with another molecule that has high affinity for the target, which is cumbersome and involves additional radiation exposure. Here we propose, and validate for the tracers [11C]DASB and [11C]CUMI-101, a new data-driven hybrid deconvolution approach (HYDECA) that determines VND at the individual level without requiring either a reference region or a blocking study. METHODS HYDECA requires the tracer metabolite-corrected concentration curve in blood plasma and uses a singular value decomposition to estimate the impulse response function across several brain regions from measured time activity curves. HYDECA decomposes each region's impulse response function into the sum of a parametric non-displaceable component, which is a function of VND, assumed common across regions, and a nonparametric specific component. These two components differentially contribute to each impulse response function. Different regions show different contributions of the two components, and HYDECA examines data across regions to find a suitable common VND. HYDECA implementation requires determination of two tuning parameters, and we propose two strategies for objectively selecting these parameters for a given tracer: using data from blocking studies, and realistic simulations of the tracer. Using available test-retest data, we compare HYDECA estimates of VND and binding potentials to those obtained based on VND estimated using a purported reference region. RESULTS For [11C]DASB and [11C]CUMI-101, we find that regardless of the strategy used to optimize the tuning parameters, HYDECA provides considerably less biased estimates of VND than those obtained, as is commonly done, using a non-ideal reference region. HYDECA test-retest reproducibility is comparable to that obtained using a VND determined from a non-ideal reference region, when considering the binding potentials BPP and BPND. CONCLUSIONS HYDECA can provide subject-specific estimates of VND without requiring a blocking study for tracers and targets for which a valid reference region does not exist.
Collapse
Affiliation(s)
- Francesca Zanderigo
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York, United States of America
- Department of Psychiatry, Columbia University, New York, New York, United States of America
| | - J. John Mann
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York, United States of America
- Department of Psychiatry, Columbia University, New York, New York, United States of America
- Department of Radiology, Columbia University, New York, New York, United States of America
| | - R. Todd Ogden
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York, United States of America
- Department of Psychiatry, Columbia University, New York, New York, United States of America
- Department of Biostatistics, Columbia University, Mailman School of Public Health, New York, New York, United States of America
| |
Collapse
|
13
|
Schain M, Zanderigo F, Mann J, Ogden R. Estimation of the binding potential BPND without a reference region or blood samples for brain PET studies. Neuroimage 2017; 146:121-131. [PMID: 27856316 DOI: 10.1016/j.neuroimage.2016.11.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/13/2016] [Indexed: 02/02/2023] Open
|
14
|
Suhara T, Chaki S, Kimura H, Furusawa M, Matsumoto M, Ogura H, Negishi T, Saijo T, Higuchi M, Omura T, Watanabe R, Miyoshi S, Nakatani N, Yamamoto N, Liou SY, Takado Y, Maeda J, Okamoto Y, Okubo Y, Yamada M, Ito H, Walton NM, Yamawaki S. Strategies for Utilizing Neuroimaging Biomarkers in CNS Drug Discovery and Development: CINP/JSNP Working Group Report. Int J Neuropsychopharmacol 2016; 20:285-294. [PMID: 28031269 PMCID: PMC5604546 DOI: 10.1093/ijnp/pyw111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/15/2016] [Indexed: 01/07/2023] Open
Abstract
Despite large unmet medical needs in the field for several decades, CNS drug discovery and development has been largely unsuccessful. Biomarkers, particularly those utilizing neuroimaging, have played important roles in aiding CNS drug development, including dosing determination of investigational new drugs (INDs). A recent working group was organized jointly by CINP and Japanese Society of Neuropsychopharmacology (JSNP) to discuss the utility of biomarkers as tools to overcome issues of CNS drug development.The consensus statement from the working group aimed at creating more nuanced criteria for employing biomarkers as tools to overcome issues surrounding CNS drug development. To accomplish this, a reverse engineering approach was adopted, in which criteria for the utilization of biomarkers were created in response to current challenges in the processes of drug discovery and development for CNS disorders. Based on this analysis, we propose a new paradigm containing 5 distinct tiers to further clarify the use of biomarkers and establish new strategies for decision-making in the context of CNS drug development. Specifically, we discuss more rational ways to incorporate biomarker data to determine optimal dosing for INDs with novel mechanisms and targets, and propose additional categorization criteria to further the use of biomarkers in patient stratification and clinical efficacy prediction. Finally, we propose validation and development of new neuroimaging biomarkers through public-private partnerships to further facilitate drug discovery and development for CNS disorders.
Collapse
Affiliation(s)
- Tetsuya Suhara
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Shigeyuki Chaki
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Haruhide Kimura
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Makoto Furusawa
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Mitsuyuki Matsumoto
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Hiroo Ogura
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Takaaki Negishi
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Takeaki Saijo
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Makoto Higuchi
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Tomohiro Omura
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Rira Watanabe
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Sosuke Miyoshi
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Noriaki Nakatani
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Noboru Yamamoto
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Shyh-Yuh Liou
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Yuhei Takado
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Jun Maeda
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Yasumasa Okamoto
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Yoshiaki Okubo
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Makiko Yamada
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Hiroshi Ito
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Noah M. Walton
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Shigeto Yamawaki
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| |
Collapse
|
15
|
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
|
16
|
Role of 18F-fluorodeoxyglucose Positron Emission Tomography in the Monitoring of Inflammatory Activity in Crohn's Disease. Inflamm Bowel Dis 2016; 22:2619-2629. [PMID: 27753695 DOI: 10.1097/mib.0000000000000924] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Fluorine-fluorodeoxyglucose positron emission tomography (F-FDG PET) has recently attracted interest for the measurement of disease activity in Crohn's disease (CD). The aim of this study was to assess the utility of FDG-PET as a marker of progression of inflammatory activity and its response to treatment in patients with CD. METHODS Twenty-two patients with active CD were recruited prospectively to undergo FDG-PET scanning at 2 time points. All 22 index scans were used to assess sensitivity and specificity against a reference standard magnetic resonance imaging measure. Correlations with clinicopathological markers of severity (Harvey-Bradshaw Index, C-reactive protein, and calprotectin) were also performed. Of note, 17/22 patients participated in the longitudinal component and underwent scanning before and 12 weeks after the initiation of anti-tumor necrosis factor alpha therapy. Patients were subcategorized on the basis of a clinically significant response, and responsiveness of the PET measures was assessed using previously described indices. Of note, 5/22 patients took part in the test-retest component of the study and underwent scanning twice within a target interval of 1 week, to assess the reproducibility of the PET measures. RESULTS The sensitivity and specificity of F-FDG PET were 88% and 70%, respectively. Standardized uptake value (SUV)-related PET measures correlated significantly both with C-reactive protein and Harvey-Bradshaw Index in cross-sectional and longitudinal analyses. (G)SUVMAX and (G)SUVMEAN demonstrated favorable responsiveness and reliability characteristics (responsiveness ratio of Guyatt >0.80 and % variability <20%) compared with volume-dependent FDG-PET measures. A proportion of the FDG signal (10%-30%) was found to originate from the lumen of diseased segments. CONCLUSIONS F-FDG PET may be useful for longitudinal monitoring of inflammatory activity in CD.
Collapse
|
17
|
Pitcher MR, Quevedo J. Tools for studying drug transport and metabolism in the brain. Expert Opin Drug Metab Toxicol 2016; 12:161-8. [DOI: 10.1517/17425255.2016.1132307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Meagan R. Pitcher
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - João Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Neuroscience Graduate Program, Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| |
Collapse
|
18
|
Zheng MQ, Lin SF, Holden D, Naganawa M, Ropchan JR, Najafzaden S, Kapinos M, Tabriz M, Carson RE, Hamill TG, Huang Y. Comparative evaluation of two glycine transporter 1 radiotracers [11C]GSK931145 and [18F]MK-6577 in baboons. Synapse 2015; 70:112-20. [PMID: 26671330 DOI: 10.1002/syn.21879] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 12/02/2015] [Accepted: 12/11/2015] [Indexed: 01/24/2023]
Abstract
Glycine transporter type-1 (GlyT1) has been proposed as a target for drug development for schizophrenia. PET imaging with a GlyT1 specific radiotracer will allow for the measurement of target occupancy of GlyT1 inhibitors, and for in vivo investigation of GlyT1 alterations in schizophrenia. We conducted a comparative evaluation of two GlyT1 radiotracers, [(11) C]GSK931145, and [(18) F]MK-6577, in baboons. Two baboons were imaged with [(11) C]GSK931145 and [(18) F]MK-6577. Blocking studies with GSK931145 (0.3 or 0.2 mg/kg) were conducted to determine the level of tracer specific binding. [(11) C]GSK931145 and [(18) F]MK-6577 were synthesized in good yield and high specific activity. Moderately fast metabolism was observed for both tracers, with ∼ 30% of parent at 30 min post-injection. In the brain, both radiotracers showed good uptake and distribution profiles consistent with regional GlyT1 densities. [(18) F]MK-6577 displayed higher uptake and faster kinetics than [(11) C]GSK931145. Time activity curves were well described by the two-tissue compartment model. Regional volume of distribution (VT ) values were higher for [(18) F]MK-6577 than [(11) C]GSK931145. Pretreatment with GSK931145 reduced tracer uptake to a homogeneous level throughout the brain, indicating in vivo binding specificity and lack of a reference region for both radiotracers. Linear regression analysis of VT estimates between tracers indicated higher specific binding for [(18) F]MK-6577 than [(11) C]GSK931145, consistent with higher regional binding potential (BPND ) values of [(18) F]MK-6577 calculated using VT from the baseline scans and non-displaceable distribution volume (VND ) derived from blocking studies. [(18) F]MK-6577 appears to be a superior radiotracer with higher brain uptake, faster kinetics, and higher specific binding signals than [(11) C]GSK931145.
Collapse
Affiliation(s)
- Ming-Qiang Zheng
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, PET Center, New Haven, Connecticut
| | - Shu-Fei Lin
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, PET Center, New Haven, Connecticut
| | - Daniel Holden
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, PET Center, New Haven, Connecticut
| | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, PET Center, New Haven, Connecticut
| | - Jim R Ropchan
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, PET Center, New Haven, Connecticut
| | - Soheila Najafzaden
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, PET Center, New Haven, Connecticut
| | - Michael Kapinos
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, PET Center, New Haven, Connecticut
| | - Mike Tabriz
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, PET Center, New Haven, Connecticut
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, PET Center, New Haven, Connecticut
| | - Terence G Hamill
- Discovery Imaging, Merck Research Laboratories, West Point, Pennsylvania
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, PET Center, New Haven, Connecticut
| |
Collapse
|
19
|
Measurement of Bmax and Kd with the glycine transporter 1 radiotracer ¹⁸F-MK6577 using a novel multi-infusion paradigm. J Cereb Blood Flow Metab 2015. [PMID: 26198176 PMCID: PMC4671121 DOI: 10.1038/jcbfm.2015.163] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glycine is a co-agonist of glutamate at the NMDA receptor. Glycine transporter 1 (GlyT1) inhibitors are reported to be potential therapeutic agents for schizophrenia. (18)F-MK6577 is a new positron emission tomography (PET) radiotracer useful for imaging brain GlyT1 and its occupancy in humans. We devised a novel multi-infusion paradigm of radiolabeled and unlabeled compound and an iterative linear/nonlinear alternating fitting method to allow for the determination of in vivo affinity (Kd) and target concentration (Bmax) images, constraining Kd to be uniform across the brain. This paradigm was tested with (18)F-MK6577 in baboons. Voxel-based analysis produced high quality Bmax images and reliable Kd estimates, and also suggested that the nondisplaceable distribution volume (VND) is not uniform throughout the brain. In vivo GlyT1 Kd was estimated to be 1.87 nmol/L for (18)F-MK6577, and the rank order of GlyT1 distribution measured in the baboon brain was: high in the brainstem (133 nmol/L), medium in the cerebellum (83 nmol/L), and low in the cortex (30 nmol/L). These in vivo Kd and Bmax values agreed well with those determined in vitro, thus validating our novel multi-infusion approach.
Collapse
|
20
|
Gunn RN, Slifstein M, Searle GE, Price JC. Quantitative imaging of protein targets in the human brain with PET. Phys Med Biol 2015; 60:R363-411. [DOI: 10.1088/0031-9155/60/22/r363] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
21
|
Wey HY, Wang C, Schroeder FA, Logan J, Price JC, Hooker JM. Kinetic Analysis and Quantification of [¹¹C]Martinostat for in Vivo HDAC Imaging of the Brain. ACS Chem Neurosci 2015; 6:708-15. [PMID: 25768025 DOI: 10.1021/acschemneuro.5b00066] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Epigenetic mechanisms mediated by histone deacetylases (HDACs) have been implicated in a wide-range of CNS disorders and may offer new therapeutic opportunities. In vivo evaluation of HDAC density and drug occupancy has become possible with [(11)C]Martinostat, which exhibits selectivity for a subset of class I/IIb HDAC enzymes. In this study, we characterize the kinetic properties of [(11)C]Martinostat in the nonhuman primate (NHP) brain in preparation for human neuroimaging studies. The goal of this work was to determine whether classic compartmental analysis techniques were appropriate and to further determine if arterial plasma is required for future NHP studies. Using an arterial plasma input function, several analysis approaches were evaluated for robust outcome measurements. [(11)C]Martinostat showed high baseline distribution volume (VT) ranging from 29.9 to 54.4 mL/cm(3) in the brain and large changes in occupancy (up to 99%) with a blocking dose approaching full enzyme saturation. An averaged nondisplaceable tissue uptake (VND) of 8.6 ± 3.7 mL/cm(3) suggests high specific binding of [(11)C]Martinostat. From a two-tissue compartment model, [(11)C]Martinostat exhibits a high K1 (averaged K1 of 0.65 mL/cm(3)/min) and a small k4 (average of 0.0085 min(-1)). Our study supports that [(11)C]Martinostat can be used to detect changes in HDAC density and occupancy in vivo and that simplified analysis not using arterial blood could be appropriate.
Collapse
Affiliation(s)
- Hsiao-Ying Wey
- Athinoula
A Martinos Center for Biomedical Imaging, Department of Radiology,
Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Changning Wang
- Athinoula
A Martinos Center for Biomedical Imaging, Department of Radiology,
Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Frederick A. Schroeder
- Athinoula
A Martinos Center for Biomedical Imaging, Department of Radiology,
Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Jean Logan
- Center
for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York 10016, United States
| | - Julie C. Price
- Department
of Radiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Jacob M. Hooker
- Athinoula
A Martinos Center for Biomedical Imaging, Department of Radiology,
Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
22
|
The simplified reference tissue model: model assumption violations and their impact on binding potential. J Cereb Blood Flow Metab 2015; 35:304-11. [PMID: 25425078 PMCID: PMC4426748 DOI: 10.1038/jcbfm.2014.202] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/17/2014] [Accepted: 10/21/2014] [Indexed: 11/09/2022]
Abstract
Reference tissue models have gained significant traction over the last two decades as the methods of choice for the quantification of brain positron emission tomography data because they balance quantitative accuracy with less invasive procedures. The principal advantage is the elimination of the need to perform arterial cannulation of the subject to measure blood and metabolite concentrations for input function generation. In particular, the simplified reference tissue model (SRTM) has been widely adopted as it uses a simplified model configuration with only three parameters that typically produces good fits to the kinetic data and a stable parameter estimation process. However, the model's simplicity and its ability to generate good fits to the data, even when the model assumptions are not met, can lead to misplaced confidence in binding potential (BPND) estimates. Computer simulation were used to study the bias introduced in BPND estimates as a consequence of violating each of the four core SRTM model assumptions. Violation of each model assumption led to bias in BPND (both over and underestimation). Careful assessment of the bias in SRTM BPND should be performed for new tracers and applications so that an appropriate decision about its applicability can be made.
Collapse
|
23
|
Naganawa M, Mishina M, Sakata M, Oda K, Hiura M, Ishii K, Ishiwata K. Test-retest variability of adenosine A2A binding in the human brain with (11)C-TMSX and PET. EJNMMI Res 2014; 4:76. [PMID: 25621197 PMCID: PMC4293456 DOI: 10.1186/s13550-014-0076-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 12/10/2014] [Indexed: 11/23/2022] Open
Abstract
Background The goal of the present study was to evaluate the reproducibility of cerebral adenosine A2A receptor (A2AR) quantification using 11C-TMSX and PET in a test-retest study. Methods Five healthy volunteers were studied twice. The test-retest variability was assessed for distribution volume (VT) and binding potential relative to non-displaceable uptake (BPND) based on either metabolite-corrected arterial blood sampling or a reference region. The cerebral cortex and centrum semiovale were used as candidate reference regions. Results Test-retest variability of VT was good in all regions (6% to 13%). In the putamen, BPND using the centrum semiovale displayed a lower test-retest variability (3%) than that of BPND using the cerebral cortex as a reference region (5%). The noninvasive method showed a higher or similar level of test-retest reproducibility compared to the invasive method. Conclusions Binding reproducibility is sufficient to use 11C-TMSX as a tool to measure the change in A2AR in the human brain. Electronic supplementary material The online version of this article (doi:10.1186/s13550-014-0076-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mika Naganawa
- PET Center, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT 06520-8048 USA ; Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015 Japan
| | - Masahiro Mishina
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015 Japan ; Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-0022 Japan
| | - Muneyuki Sakata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015 Japan
| | - Keiichi Oda
- Department of Radiological Technology, Faculty of Health Sciences, Hokkaido University of Science, Hokkaido, 006-8585 Japan
| | - Mikio Hiura
- Faculty of Sports and Health Studies, Hosei University, Tokyo, 194-0298 Japan
| | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015 Japan
| | - Kiichi Ishiwata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015 Japan
| |
Collapse
|
24
|
Kinetic modeling of (11)C-LY2795050, a novel antagonist radiotracer for PET imaging of the kappa opioid receptor in humans. J Cereb Blood Flow Metab 2014; 34:1818-25. [PMID: 25182664 PMCID: PMC4269759 DOI: 10.1038/jcbfm.2014.150] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/15/2014] [Accepted: 07/31/2014] [Indexed: 11/08/2022]
Abstract
(11)C-LY2795050 is a novel kappa opioid receptor (KOR) antagonist tracer for positron emission tomography (PET) imaging. The purpose of this first-in-human study was to determine the optimal kinetic model for analysis of (11)C-LY2795050 imaging data. Sixteen subjects underwent baseline scans and blocking scans after oral naltrexone. Compartmental modeling and multilinear analysis-1 (MA1) were applied using the arterial input functions. Two-tissue compartment model and MA1 were found to be the best models to provide reliable measures of binding parameters. The rank order of (11)C-LY2795050 distribution volume (VT) matched the known regional KOR densities in the human brain. Blocking scans with naltrexone indicated no ideal reference region for (11)C-LY2795050. Three methods for calculation of the nondisplaceable distribution volume (VND) were assessed: (1) individual VND estimated from naltrexone occupancy plots, (2) mean VND across subjects, and (3) a fixed fraction of cerebellum VT. Approach (3) produced the lowest intersubject variability in the calculation of binding potentials (BPND, BPF, and BPP). Therefore, binding potentials of (11)C-LY2795050 can be determined if the specific binding fraction in the cerebellum is presumed to be unchanged by diseases and experimental conditions. In conclusion, results from the present study show the suitability of (11)C-LY2795050 to image and quantify KOR in humans.
Collapse
|
25
|
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
|
26
|
Joshi AD, Sanabria-Bohórquez SM, Bormans G, Koole M, De Hoon J, Van Hecken A, Depre M, De Lepeleire I, Van Laere K, Sur C, Hamill TG. Characterization of the novel GlyT1 PET tracer [18F]MK-6577 in humans. Synapse 2014; 69:33-40. [PMID: 25196464 DOI: 10.1002/syn.21782] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/06/2014] [Accepted: 07/26/2014] [Indexed: 01/05/2023]
Abstract
Decreased glutamatergic neurotransmission is hypothesized to be involved in the pathophysiology of schizophrenia. Inhibition of glycine transporter Type-1 (GlyT1) reuptake is expected to increase the glutamatergic neurotransmission and may serve as treatment for cognitive and negative symptoms of schizophrenia. In this article, we present human data from a novel GlyT1 PET tracer, [(18) F]MK-6577. In the process of developing a GlyT1 inhibitor therapeutic, a PET tracer can assist in determining the dose with a high probability of sufficiently testing the mechanism of action. This article reports the human PET studies with [(18) F]MK-6577 for measuring GlyT1 receptor availability at baseline in normal human subjects and occupancy with a GlyT1 inhibitor, MK-2637. Studies were also performed to measure radiation burden and the baseline test-retest (T-RT) variability of the tracer. The effective dose from sequential whole-body dosimetry scans in three male subjects was estimated to be 24.5 ± 2.9 µSV/MBq (mean ± SD). The time-activity curves from T-RT scans modeled satisfactorily using a two tissue compartmental model. The tracer uptake was highest in the pons (VT = 6.7 ± 0.9, BPND = 4.1 ± 0.43) and lowest in the cortex (VT = 2.1 ± 0.5, BPND = 0.60 ± 0.23). VT T-RT variability measured in three subjects was <12% on average. The occupancy scans performed in a cohort of 15 subjects indicated absence of a reference region. The in vivo potency (Occ50 ) of MK-2637 was determined using two methods: A: Lassen plot with a population input function (Occ50 = 106 nM, SE = 20 nM) and B: pseudo reference tissue model using cortex as the pseudo reference region (Occ50 = 141 nM, SE = 21 nM).
Collapse
|
27
|
Barret O, Hannestad J, Alagille D, Vala C, Tavares A, Papin C, Morley T, Fowles K, Lee H, Seibyl J, Tytgat D, Laruelle M, Tamagnan G. Adenosine 2A receptor occupancy by tozadenant and preladenant in rhesus monkeys. J Nucl Med 2014; 55:1712-8. [PMID: 25082853 DOI: 10.2967/jnumed.114.142067] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Motor symptoms in Parkinson disease (PD) are caused by a loss of dopamine input from the substantia nigra to the striatum. Blockade of adenosine 2A (A(2A)) receptors facilitates dopamine D(2) receptor function. In phase 2 clinical trials, A(2A) antagonists (istradefylline, preladenant, and tozadenant) improved motor function in PD. We developed a new A(2A) PET radiotracer, (18)F-MNI-444, and used it to investigate the relationship between plasma levels and A(2A) occupancy by preladenant and tozadenant in nonhuman primates (NHP). METHODS A series of 20 PET experiments was conducted in 5 adult rhesus macaques. PET data were analyzed with both plasma-input (Logan graphical analysis) and reference-region-based (simplified reference tissue model and noninvasive Logan graphical analysis) methods. Whole-body PET images were acquired for radiation dosimetry estimates. Human pharmacokinetic parameters for tozadenant and preladenant were used to predict A(2A) occupancy in humans, based on median effective concentration (EC(50)) values estimated from the NHP PET measurements. RESULTS (18)F-MNI-444 regional uptake was consistent with A(2A) receptor distribution in the brain. Selectivity was demonstrated by dose-dependent blocking by tozadenant and preladenant. The specific-to-nonspecific ratio was superior to that of other A(2A) PET radiotracers. Pharmacokinetic modeling predicted that tozadenant and preladenant may have different profiles of A(2A) receptor occupancy in humans. CONCLUSION (18)F-MNI-444 appears to be a better PET radiotracer for A(2A) imaging than currently available radiotracers. Assuming that EC(50) in humans is similar to that in NHP, it appears that tozadenant will provide a more sustained A(2A) receptor occupancy than preladenant in humans at clinically tested doses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hsiaoju Lee
- Molecular NeuroImaging, LLC, New Haven, Connecticut
| | - John Seibyl
- Molecular NeuroImaging, LLC, New Haven, Connecticut
| | | | | | | |
Collapse
|
28
|
Barret O, Thomae D, Tavares A, Alagille D, Papin C, Waterhouse R, McCarthy T, Jennings D, Marek K, Russell D, Seibyl J, Tamagnan G. In Vivo Assessment and Dosimetry of 2 Novel PDE10A PET Radiotracers in Humans: 18F-MNI-659 and 18F-MNI-654. J Nucl Med 2014; 55:1297-304. [DOI: 10.2967/jnumed.113.122895] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 03/27/2014] [Indexed: 11/16/2022] Open
|
29
|
Hwang DR, Hu E, Rumfelt S, Easwaramoorthy B, Castrillon J, Davis C, Allen JR, Chen H, Treanor J, Abi-Dargham A, Slifstein M. Initial characterization of a PDE10A selective positron emission tomography tracer [11C]AMG 7980 in non-human primates. Nucl Med Biol 2014; 41:343-9. [PMID: 24607437 DOI: 10.1016/j.nucmedbio.2014.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 12/19/2013] [Accepted: 01/07/2014] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Phosphodiesterase 10A (PDE10A) is an intracellular enzyme responsible for the breakdown of cyclic nucleotides which are important secondary messengers in the central nervous system. Inhibition of PDE10A has been identified as a potential therapeutic target for treatment of various neuropsychiatric disorders. To assist the drug development program, we have identified a selective PDE10A PET tracer, [(11)C]AMG 7980, for imaging PDE10A distribution using positron emission tomography. METHODS [(11)C]AMG 7980 was prepared in a one-pot, two-step reaction. Dynamic PET scans were performed in non-human primates following a bolus or bolus plus constant infusion tracer injection paradigm. Regions-of-interest were defined on individuals' MRIs and transferred to the co-registered PET images. Data were analyzed using Logan graphical analysis with metabolite-corrected input function, the simplified reference tissue model (SRTM) method and occupancy plots. A benchmark PDE10A inhibitor was used to demonstrate PDE10A-specific binding. RESULTS [(11)C]AMG 7980 was prepared with a mean specific activity of 99 ± 74 GBq/μmol (n=10) and a synthesis time of 45 min. Specific binding of the tracer was localized to the striatum and globus pallidus (GP) and low in other brain regions. Thalamus was used as the reference tissue to derive binding potentials (BPND). The BPND for caudate, putamen, and GP were 0.23, 0.65, 0.51, respectively by the graphical method, and 0.42, 0.76, and 0.75 from the SRTM method. A dose dependent decrease of BPND was observed with the pre-treatment of a PDE10A inhibitor. A bolus plus infusion injection paradigm yielded similar results. CONCLUSION [(11)C]AMG 7980 has been successfully used for imaging PDE10A in non-human primate brain. Despite the fast brain kinetics it can be used to measure target occupancy of PDE10A inhibitors in non-human primates and potentially applicable to humans.
Collapse
Affiliation(s)
- Dah-Ren Hwang
- Department of Medical Sciences, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States.
| | - Essa Hu
- Department of Small Molecule Chemistry, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Shannon Rumfelt
- Department of Small Molecule Chemistry, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Balu Easwaramoorthy
- Department of Psychiatry, Columbia University, New York, NY, USA; New York State Psychiatric Institute, NY, USA
| | | | - Carl Davis
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Jennifer R Allen
- Department of Small Molecule Chemistry, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Hang Chen
- Department of Neuroscience, Amgen Inc., South San Francisco, CA
| | - James Treanor
- Department of Neuroscience, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Anissa Abi-Dargham
- Department of Psychiatry, Columbia University, New York, NY, USA; Department of Radiology, Columbia University, New York, NY, USA; New York State Psychiatric Institute, NY, USA
| | - Mark Slifstein
- Department of Psychiatry, Columbia University, New York, NY, USA; New York State Psychiatric Institute, NY, USA
| |
Collapse
|
30
|
Huang C, Ackerman JL, Petibon Y, Normandin MD, Brady TJ, El Fakhri G, Ouyang J. Motion compensation for brain PET imaging using wireless MR active markers in simultaneous PET-MR: phantom and non-human primate studies. Neuroimage 2014; 91:129-37. [PMID: 24418501 DOI: 10.1016/j.neuroimage.2013.12.061] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 12/16/2013] [Accepted: 12/30/2013] [Indexed: 11/19/2022] Open
Abstract
Brain PET scanning plays an important role in the diagnosis, prognostication and monitoring of many brain diseases. Motion artifacts from head motion are one of the major hurdles in brain PET. In this work, we propose to use wireless MR active markers to track head motion in real time during a simultaneous PET-MR brain scan and incorporate the motion measured by the markers in the listmode PET reconstruction. Several wireless MR active markers and a dedicated fast MR tracking pulse sequence module were built. Data were acquired on an ACR Flangeless PET phantom with multiple spheres and a non-human primate with and without motion. Motions of the phantom and monkey's head were measured with the wireless markers using a dedicated MR tracking sequence module. The motion PET data were reconstructed using list-mode reconstruction with and without motion correction. Static reference was used as gold standard for quantitative analysis. The motion artifacts, which were prominent on the images without motion correction, were eliminated by the wireless marker based motion correction in both the phantom and monkey experiments. Quantitative analysis was performed on the phantom motion data from 24 independent noise realizations. The reduction of bias of sphere-to-background PET contrast by active marker based motion correction ranges from 26% to 64% and 17% to 25% for hot (i.e., radioactive) and cold (i.e., non-radioactive) spheres, respectively. The motion correction improved the channelized Hotelling observer signal-to-noise ratio of the spheres by 1.2 to 6.9 depending on their locations and sizes. The proposed wireless MR active marker based motion correction technique removes the motion artifacts in the reconstructed PET images and yields accurate quantitative values.
Collapse
Affiliation(s)
- Chuan Huang
- Center for Advanced Medical Imaging Sciences, Division of Nuclear Medicine and Molecular Imaging, Department of Imaging, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Radiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Jerome L Ackerman
- Department of Radiology, Harvard Medical School, Boston, MA 02115, USA; Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129, USA.
| | - Yoann Petibon
- Center for Advanced Medical Imaging Sciences, Division of Nuclear Medicine and Molecular Imaging, Department of Imaging, Massachusetts General Hospital, Boston, MA 02114, USA; Laboratoire d'imagerie fonctionnelle (LIF), UMRS-678, INSERM, Université Pierre et Marie Curie, CHU Pitié-Salpêtrière, Paris, France.
| | - Marc D Normandin
- Center for Advanced Medical Imaging Sciences, Division of Nuclear Medicine and Molecular Imaging, Department of Imaging, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Radiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Thomas J Brady
- Center for Advanced Medical Imaging Sciences, Division of Nuclear Medicine and Molecular Imaging, Department of Imaging, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Radiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Georges El Fakhri
- Center for Advanced Medical Imaging Sciences, Division of Nuclear Medicine and Molecular Imaging, Department of Imaging, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Radiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Jinsong Ouyang
- Center for Advanced Medical Imaging Sciences, Division of Nuclear Medicine and Molecular Imaging, Department of Imaging, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Radiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
31
|
Poels EMP, Kegeles LS, Kantrowitz JT, Slifstein M, Javitt DC, Lieberman JA, Abi-Dargham A, Girgis RR. Imaging glutamate in schizophrenia: review of findings and implications for drug discovery. Mol Psychiatry 2014; 19:20-9. [PMID: 24166406 DOI: 10.1038/mp.2013.136] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 08/25/2013] [Accepted: 09/09/2013] [Indexed: 12/11/2022]
Abstract
Currently, all treatments for schizophrenia (SCZ) function primarily by blocking D(2)-type dopamine receptors. Given the limitations of these medications, substantial efforts have been made to identify alternative neurochemical targets for treatment development in SCZ. One such target is brain glutamate. The objective of this article is to review and synthesize the proton magnetic resonance spectroscopy ((1)H MRS) and positron emission tomography (PET)/single-photon emission computed tomography (SPECT) investigations that have examined glutamatergic indices in SCZ, including those of modulatory compounds such as glutathione (GSH) and glycine, as well as data from ketamine challenge studies. The reviewed (1)H MRS and PET/SPECT studies support the theory of hypofunction of the N-methyl-D-aspartate receptor (NMDAR) in SCZ, as well as the convergence between the dopamine and glutamate models of SCZ. We also review several advances in MRS and PET technologies that have opened the door for new opportunities to investigate the glutamate system in SCZ and discuss some ways in which these imaging tools can be used to facilitate a greater understanding of the glutamate system in SCZ and the successful and efficient development of new glutamate-based treatments for SCZ.
Collapse
Affiliation(s)
- E M P Poels
- 1] Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY, USA [2] New York State Psychiatric Institute, New York, NY, USA
| | - L S Kegeles
- 1] Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY, USA [2] New York State Psychiatric Institute, New York, NY, USA
| | - J T Kantrowitz
- 1] Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY, USA [2] New York State Psychiatric Institute, New York, NY, USA
| | - M Slifstein
- 1] Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY, USA [2] New York State Psychiatric Institute, New York, NY, USA
| | - D C Javitt
- 1] Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY, USA [2] New York State Psychiatric Institute, New York, NY, USA
| | - J A Lieberman
- 1] Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY, USA [2] New York State Psychiatric Institute, New York, NY, USA
| | - A Abi-Dargham
- 1] Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY, USA [2] New York State Psychiatric Institute, New York, NY, USA [3] Department of Radiology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - R R Girgis
- 1] Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY, USA [2] New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
32
|
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]
|
33
|
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] [Scholar 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
|
34
|
Rizzo SJS, Edgerton JR, Hughes ZA, Brandon NJ. Future Viable Models of Psychiatry Drug Discovery in Pharma. ACTA ACUST UNITED AC 2013; 18:509-21. [DOI: 10.1177/1087057113475871] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The unmet need for the treatment of disorders of the nervous system is growing, and as highlighted in the media and elsewhere, the results of an aging population will ensure this continues with an upward trajectory. Incredibly, the efforts within industry to identify new drugs to treat these conditions have seemingly disappeared despite the growing need. There has been a run of extraordinary failure in the later stages of the drug discovery process for neurological and psychiatric disorders, which has many causes. We believe, though, that we have to confront this dire situation, both by using learnings from the post hoc analysis of our historical failure, as well as harnessing the bewildering array of new technologies and data now available to us, to ensure we are making the right decisions along the very complicated path of drug discovery to registration.
Collapse
Affiliation(s)
| | - Jeremy R. Edgerton
- Neuroscience Research Unit, Pfizer Research and Development, Cambridge, MA, USA
| | - Zoë A. Hughes
- Neuroscience Research Unit, Pfizer Research and Development, Cambridge, MA, USA
| | | |
Collapse
|
35
|
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
|
36
|
Tavares AADS, Barret O, Batis J, Alagille D, Koren A, Papin C, Kudej G, Nice K, Kordower JH, Cosgrove KP, Kloczynski T, Brenner E, Seibyl J, Tamagnan G. Kinetic modeling and occupancy measures of the norepinephrine transporters in baboons using single photon emission computed tomography with (123)I-INER. Synapse 2012; 67:30-41. [PMID: 23042643 DOI: 10.1002/syn.21613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 10/01/2012] [Indexed: 11/11/2022]
Abstract
This study aims to investigate the pharmacokinetics of a recently developed radiotracer for imaging of the norepinephrine transporter (NET) in baboon brain, (123)I-INER, using single photon emission computed tomography (SPECT). In addition, it also aims to determine NET occupancy by atomoxetine and reboxetine, two selective norepinephrine reuptake inhibitors, using (123)I-INER in baboons. Baseline and preblocking studies with a high dose of atomoxetine (0.85 mg/kg) were conducted in three baboons using SPECT with (123)I-INER administered as a bolus. Kinetic modeling analysis was investigated for different models, namely invasive and reference tissue models. Bolus plus constant infusion experiments with displacement at equilibrium using six different doses of atomoxetine (0.03-0.85 mg/kg) and four different doses of reboxetine (0.5-3.0 mg/kg) were carried out in several baboons to obtain occupancy measurements as a function of dose for the two NET selective drugs. Results showed that reference tissue models can be used to estimate binding potential values and occupancy measures of (123)I-INER in different brain regions. In addition, the apparent volume of distribution was estimated by dividing concentration in tissue by the concentration in blood at 3 hours postinjection. After administration of atomoxetine or reboxetine, a dose-dependent occupancy was observed in brain regions known to contain high densities of NET. In conclusion, pharmacokinetic properties of (123) I-INER were successfully described, and obtained results may be used to simplify future data acquisition and image processing. Dose-dependent NET occupancy for two selective norepinephrine reuptake inhibitors was successfully measured in vivo in baboon brain using SPECT and (123) I-INER.
Collapse
|
37
|
Fuchigami T, Takano A, Gulyás B, Jia Z, Finnema SJ, Andersson JD, Nakao R, Magata Y, Haratake M, Nakayama M, Halldin C. Synthesis and evaluation of 2-chloro N-[(S)-{(S)-1-[11 C]methylpiperidin-2-yl} (phenyl)methyl]3-trifluoromethyl-benzamide ([11 C]N-methyl-SSR504734) as a PET radioligand for glycine transporter 1. EJNMMI Res 2012; 2:37. [PMID: 22776065 PMCID: PMC3531252 DOI: 10.1186/2191-219x-2-37] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/01/2012] [Indexed: 11/10/2022] Open
Abstract
UNLABELLED BACKGROUND Dysfunction of the glycine transporter 1 (GlyT1) has been suggested to be involved in psychiatric disorders such as schizophrenia. GlyT1 inhibitors have therefore been considered to have antipsychotic therapeutic potential. Positron emission tomography (PET) imaging probes for GlyT1 are, consequently, expected to be useful for investigating the mechanism of such disease conditions and for measuring occupancy of GlyT1 inhibitors in vivo. The aim of this study was to assess the potential of 2-chloro N-[(S)-{(S)-1-[11 C]methylpiperidin-2-yl} (phenyl)methyl] 3-trifluoromethyl-benzamide ([11 C]N-methyl-SSR504734) as a PET imaging agent for GlyT1. METHODS [11 C]N-methyl-SSR504734 was synthesized by N-[11 C]methylation of SSR504734 via [11 C]CH3OTf. In vitro brain distribution of [11 C]N-methyl-SSR504734 was tested in whole-hemisphere autoradiography (ARG) on human brain slices. Initial PET studies were performed using a cynomolgus monkey at baseline and after pretreatment with 0.1 to 1.5 mg/kg of SSR504734. Then, PET studies using rhesus monkeys were performed with arterial blood sampling at baseline and after pretreatment with 1.5 to 4.5 mg/kg SSR504734. Distribution volumes (VT) were calculated with a two-tissue compartment model, and GlyT1 occupancy by SSR504734 was estimated using a Lassen plot approach. RESULTS [11 C]N-methyl-SSR504734 was successfully synthesized in moderate radiochemical yield and high specific radioactivity. In the ARG experiments, [11 C]N-methyl-SSR504734 showed specific binding in the white matter and pons. In the initial PET experiments in a cynomolgus monkey, [11 C]N-methyl-SSR504734 showed high brain uptake and consistent distribution with previously reported GlyT1 expression in vivo (thalamus, brainstem > cerebellum > cortical regions). However, the brain uptake increased after pretreatment with SSR504734. Further PET studies in rhesus monkeys showed a similar increase of brain uptake after pretreatment with SSR504734. However, the VT of [11 C]N-methyl-SSR504734 was found to decrease after pretreatment of SSR504734 in a dose-dependent manner. GlyT1 occupancy was calculated to be 45% and 73% at 1.5 and 4.5 mg/kg of SSR504734, respectively. CONCLUSIONS [11 C]N-methyl-SSR504734 is demonstrated to be a promising PET radioligand for GlyT1 in nonhuman primates. The present results warrant further PET studies in human subjects.
Collapse
Affiliation(s)
- Takeshi Fuchigami
- Karolinska Institutet, Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska University Hospital, R5:02, Stockholm 171 76, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Borroni E, Zhou Y, Ostrowitzki S, Alberati D, Kumar A, Hainzl D, Hartung T, Hilton J, Dannals RF, Wong DF. Pre-clinical characterization of [11C]R05013853 as a novel radiotracer for imaging of the glycine transporter type 1 by positron emission tomography. Neuroimage 2011; 75:291-300. [PMID: 22178811 DOI: 10.1016/j.neuroimage.2011.11.090] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 11/24/2011] [Accepted: 11/30/2011] [Indexed: 11/19/2022] Open
Abstract
A specific positron emission tomography (PET) radiotracer for the glycine transporter type 1 (GlyT1) would constitute an imaging biomarker to investigate the distribution of GlyT1 in normal individuals and those with neuropsychiatric disorders. In addition it could demonstrate the ability of a novel drug to reach its target in the brain and enable receptor occupancy studies, thus facilitating drug development. In this article we describe the evaluation in non-human primates of two candidate PET radiotracers ([(11)C]RO5013852 and [(11)C]RO5013853) previously characterized in the rat. Both radiotracers showed acceptable uptake in the baboon brain and heterogeneous distribution consistent with that reported for GlyT1. In vivo blockade studies with two specific glycine reuptake inhibitors (GRIs), RO5013853 and bitopertin (RG1678, reduced uptake of both tracers to homogenous levels across brain regions and demonstrated specificity of the signal. [(11)C]RO5013853 showed a larger specific signal and slightly higher brain uptake and was therefore selected for further characterization. Quantitative compartmental analysis of PET data showed that the 2-tissue compartment model with 5 parameters was the most appropriate to describe the kinetics of [(11)C]RO5013853. Two additional methods were used: a) the Logan graphical analysis using plasma input and, b) a linear parametric imaging approach with the 2-tissue compartmental model. These produced VT estimates of comparable magnitude, namely, pons, thalamus and cerebellum>caudate, putamen and cortical regions. High resolution autoradiography with tritiated RO5013853 was used to confirm the binding pattern observed by PET. In vivo metabolism studies in the baboon demonstrated the formation of a single, radiolabeled metabolite more polar than the parent compound. Finally, [(11)C]RO5013853 was used to quantify the degree of cerebral GlyT1 occupancy observed in the baboon following oral administration of bitopertin, a selective GRI presently in Phase III clinical trial. Plasma concentrations of approximately 150-300 ng/mL were estimated to produce 50% GlyT1 occupancy in the thalamus, the cerebellum and the pons. [(11)C]RO5013853 is a promising radiotracer for in vivo imaging of the GlyT1. It can be easily radiolabeled, exhibits moderate metabolism, displays a good specific signal, and is suitable for receptor occupancy studies of therapeutic compounds that target the GlyT1. The successful characterization of [(11)C]RO5013853 in healthy volunteers is presented in this NeuroImage issue (Wong et al., 2013).
Collapse
Affiliation(s)
- Edilio Borroni
- Neuroscience Department, Pharmaceutical Division, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland.
| | - Yun Zhou
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287-0807, USA
| | - Susanne Ostrowitzki
- Neuroscience Department, Pharmaceutical Division, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Daniela Alberati
- Neuroscience Department, Pharmaceutical Division, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Anil Kumar
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287-0807, USA
| | - Dominik Hainzl
- Nonclinical Safety Department, Pharmaceutical Division, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Thomas Hartung
- Process Research & Synthesis Department, Pharmaceutical Division, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - John Hilton
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287-0807, USA
| | - Robert F Dannals
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287-0807, USA
| | - Dean F Wong
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287-0807, USA; Department of Psychiatry, The Johns Hopkins University School of Medicine, 601 N. Caroline St., JHOC, Baltimore, MD 21287-0807, USA; Department of Neuroscience, The Johns Hopkins University School of Medicine, 601 N. Caroline St., JHOC, Baltimore, MD 21287-0807, USA; Department of Environmental Health Sciences, The Johns Hopkins University School of Medicine, 601 N. Caroline St., JHOC, Baltimore, MD 21287-0807, USA; Honorary Professor of Neuroscience and Pharmacology, University of Copenhagen, Denmark
| |
Collapse
|
39
|
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
|
40
|
Gunn RN, Guo Q, Salinas CA, Tziortzi AC, Searle GE. Advances in biomathematical modeling for PET neuroreceptor imaging. DRUG DISCOVERY TODAY. TECHNOLOGIES 2011; 8:e45-e51. [PMID: 24990262 DOI: 10.1016/j.ddtec.2012.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The quantitative application of PET neuroreceptor imaging to study pathophysiology, diagnostics and drug development has continued to benefit from associated advances in biomathematical imaging methodology. We review some of these advances with particular focus on multi-modal image processing, tracer kinetic modeling, occupancy studies and discovery and development of novel radioligands.:
Collapse
Affiliation(s)
- Roger N Gunn
- Department of Medicine, Imperial College London, London, UK.
| | - Qi Guo
- Department of Medicine, Imperial College London, London, UK
| | - Cristian A Salinas
- Imanova Limited, Burlington Danes Building, Imperial College London, Hammersmith Hospital, London, Du Cane Road, W12 0NN, UK
| | - Andri C Tziortzi
- FMRIB Centre, Department of Clinical Neurology, University of Oxford, Oxford, UK
| | - Graham E Searle
- Imanova Limited, Burlington Danes Building, Imperial College London, Hammersmith Hospital, London, Du Cane Road, W12 0NN, UK
| |
Collapse
|