1
|
Maccioni L, Michelle CM, Brusaferri L, Silvestri E, Bertoldo A, Schubert JJ, Nettis MA, Mondelli V, Howes O, Turkheimer FE, Bottlaender M, Bodini B, Stankoff B, Loggia ML, Veronese M. A blood-free modeling approach for the quantification of the blood-to-brain tracer exchange in TSPO PET imaging. Front Neurosci 2024; 18:1395769. [PMID: 39104610 PMCID: PMC11299498 DOI: 10.3389/fnins.2024.1395769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Introduction Recent evidence suggests the blood-to-brain influx rate (K1 ) in TSPO PET imaging as a promising biomarker of blood-brain barrier (BBB) permeability alterations commonly associated with peripheral inflammation and heightened immune activity in the brain. However, standard compartmental modeling quantification is limited by the requirement of invasive and laborious procedures for extracting an arterial blood input function. In this study, we validate a simplified blood-free methodologic framework for K1 estimation by fitting the early phase tracer dynamics using a single irreversible compartment model and an image-derived input function (1T1K-IDIF). Methods The method is tested on a multi-site dataset containing 177 PET studies from two TSPO tracers ([11C]PBR28 and [18F]DPA714). Firstly, 1T1K-IDIF K1 estimates were compared in terms of both bias and correlation with standard kinetic methodology. Then, the method was tested on an independent sample of [11C]PBR28 scans before and after inflammatory interferon-α challenge, and on test-retest dataset of [18F]DPA714 scans. Results Comparison with standard kinetic methodology showed good-to-excellent intra-subject correlation for regional 1T1K-IDIF-K1 (ρintra = 0.93 ± 0.08), although the bias was variable depending on IDIF ability to approximate blood input functions (0.03-0.39 mL/cm3/min). 1T1K-IDIF-K1 unveiled a significant reduction of BBB permeability after inflammatory interferon-α challenge, replicating results from standard quantification. High intra-subject correlation (ρ = 0.97 ± 0.01) was reported between K1 estimates of test and retest scans. Discussion This evidence supports 1T1K-IDIF as blood-free alternative to assess TSPO tracers' unidirectional blood brain clearance. K1 investigation could complement more traditional measures in TSPO studies, and even allow further mechanistic insight in the interpretation of TSPO signal.
Collapse
Affiliation(s)
- Lucia Maccioni
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Carranza Mellana Michelle
- Department of Information Engineering, University of Padova, Padova, Italy
- Paris Brain Institute, ICM, CNRS, Inserm, Sorbonne Université, Paris, France
| | - Ludovica Brusaferri
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
- Computer Science and Informatics, School of Engineering, London South Bank University, London, United Kingdom
| | - Erica Silvestri
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Alessandra Bertoldo
- Department of Information Engineering, University of Padova, Padova, Italy
- Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Julia J. Schubert
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Maria A. Nettis
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Valeria Mondelli
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Oliver Howes
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Federico E. Turkheimer
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Michel Bottlaender
- BioMaps, Service Hospitalier Frédéric Joliot CEA, CNRS Inserm, Université Paris-Saclay, Orsay, France
| | - Benedetta Bodini
- Paris Brain Institute, ICM, CNRS, Inserm, Sorbonne Université, Paris, France
| | - Bruno Stankoff
- Paris Brain Institute, ICM, CNRS, Inserm, Sorbonne Université, Paris, France
| | - Marco L. Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Mattia Veronese
- Department of Information Engineering, University of Padova, Padova, Italy
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| |
Collapse
|
2
|
Lopresti BJ, Stehouwer J, Reese AC, Mason NS, Royse SK, Narendran R, Laymon CM, Lopez OL, Cohen AD, Mathis CA, Villemagne VL. Kinetic modeling of the monoamine oxidase-B radioligand [ 18F]SMBT-1 in human brain with positron emission tomography. J Cereb Blood Flow Metab 2024:271678X241254679. [PMID: 38735059 DOI: 10.1177/0271678x241254679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
This paper describes pharmacokinetic analyses of the monoamine-oxidase-B (MAO-B) radiotracer [18F](S)-(2-methylpyrid-5-yl)-6-[(3-fluoro-2-hydroxy)propoxy]quinoline ([18F]SMBT-1) for positron emission tomography (PET) brain imaging. Brain MAO-B expression is widespread, predominantly within astrocytes. Reactive astrogliosis in response to neurodegenerative disease pathology is associated with MAO-B overexpression. Fourteen elderly subjects (8 control, 5 mild cognitive impairment, 1 Alzheimer's disease) with amyloid ([11C]PiB) and tau ([18F]flortaucipir) imaging assessments underwent dynamic [18F]SMBT-1 PET imaging with arterial input function determination. [18F]SMBT-1 showed high brain uptake and a retention pattern consistent with the known MAO-B distribution. A two-tissue compartment (2TC) model where the K1/k2 ratio was fixed to a whole brain value best described [18F]SMBT-1 kinetics. The 2TC total volume of distribution (VT) was well identified and highly correlated (r2∼0.8) with post-mortem MAO-B indices. Cerebellar grey matter (CGM) showed the lowest mean VT of any region and is considered the optimal pseudo-reference region. Simplified analysis methods including reference tissue models, non-compartmental models, and standard uptake value ratios (SUVR) agreed with 2TC outcomes (r2 > 0.9) but with varying bias. We found the CGM-normalized 70-90 min SUVR to be highly correlated (r2 = 0.93) with the 2TC distribution volume ratio (DVR) with acceptable bias (∼10%), representing a practical alternative for [18F]SMBT-1 analyses.
Collapse
Affiliation(s)
- Brian J Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeffrey Stehouwer
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexandria C Reese
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neale S Mason
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sarah K Royse
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Charles M Laymon
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Dept. of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oscar L Lopez
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Clinical and Translational Sciences Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Gu F, Wu Q. Quantitation of dynamic total-body PET imaging: recent developments and future perspectives. Eur J Nucl Med Mol Imaging 2023; 50:3538-3557. [PMID: 37460750 PMCID: PMC10547641 DOI: 10.1007/s00259-023-06299-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 06/05/2023] [Indexed: 10/04/2023]
Abstract
BACKGROUND Positron emission tomography (PET) scanning is an important diagnostic imaging technique used in disease diagnosis, therapy planning, treatment monitoring, and medical research. The standardized uptake value (SUV) obtained at a single time frame has been widely employed in clinical practice. Well beyond this simple static measure, more detailed metabolic information can be recovered from dynamic PET scans, followed by the recovery of arterial input function and application of appropriate tracer kinetic models. Many efforts have been devoted to the development of quantitative techniques over the last couple of decades. CHALLENGES The advent of new-generation total-body PET scanners characterized by ultra-high sensitivity and long axial field of view, i.e., uEXPLORER (United Imaging Healthcare), PennPET Explorer (University of Pennsylvania), and Biograph Vision Quadra (Siemens Healthineers), further stimulates valuable inspiration to derive kinetics for multiple organs simultaneously. But some emerging issues also need to be addressed, e.g., the large-scale data size and organ-specific physiology. The direct implementation of classical methods for total-body PET imaging without proper validation may lead to less accurate results. CONCLUSIONS In this contribution, the published dynamic total-body PET datasets are outlined, and several challenges/opportunities for quantitation of such types of studies are presented. An overview of the basic equation, calculation of input function (based on blood sampling, image, population or mathematical model), and kinetic analysis encompassing parametric (compartmental model, graphical plot and spectral analysis) and non-parametric (B-spline and piece-wise basis elements) approaches is provided. The discussion mainly focuses on the feasibilities, recent developments, and future perspectives of these methodologies for a diverse-tissue environment.
Collapse
Affiliation(s)
- Fengyun Gu
- School of Mathematics and Physics, North China Electric Power University, 102206, Beijing, China.
- School of Mathematical Sciences, University College Cork, T12XF62, Cork, Ireland.
| | - Qi Wu
- School of Mathematical Sciences, University College Cork, T12XF62, Cork, Ireland
| |
Collapse
|
4
|
Galfalvy H, Shea E, de Vegvar J, Pantazatos S, Huang YY, Burke AK, Sublette ME, Oquendo MA, Zanderigo F, Miller JM, Mann JJ. Brain serotonin 1A receptor binding: relationship to peripheral blood DNA methylation, recent life stress and childhood adversity in unmedicated major depression. Br J Psychiatry 2023; 223:415-421. [PMID: 37395098 PMCID: PMC10514224 DOI: 10.1192/bjp.2023.13] [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: 07/21/2022] [Revised: 12/19/2022] [Accepted: 01/12/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Childhood and lifetime adversity may reduce brain serotonergic (5-HT) neurotransmission by epigenetic mechanisms. AIMS We tested the relationships of childhood adversity and recent stress to serotonin 1A (5-HT1A) receptor genotype, DNA methylation of this gene in peripheral blood monocytes and in vivo 5-HT1A receptor binding potential (BPF) determined by positron emission tomography (PET) in 13 a priori brain regions, in participants with major depressive disorder (MDD) and healthy volunteers (controls). METHOD Medication-free participants with MDD (n = 192: 110 female, 81 male, 1 other) and controls (n = 88: 48 female, 40 male) were interviewed about childhood adversity and recent stressors and genotyped for rs6295. DNA methylation was assayed at three upstream promoter sites (-1019, -1007, -681) of the 5-HT1A receptor gene. A subgroup (n = 119) had regional brain 5-HT1A receptor BPF quantified by PET. Multi-predictor models were used to test associations between diagnosis, recent stress, childhood adversity, genotype, methylation and BPF. RESULTS Recent stress correlated positively with blood monocyte methylation at the -681 CpG site, adjusted for diagnosis, and had positive and region-specific correlations with 5-HT1A BPF in participants with MDD, but not in controls. In participants with MDD, but not in controls, methylation at the -1007 CpG site had positive and region-specific correlations with binding potential. Childhood adversity was not associated with methylation or BPF in participants with MDD. CONCLUSIONS These findings support a model in which recent stress increases 5-HT1A receptor binding, via methylation of promoter sites, thus affecting MDD psychopathology.
Collapse
MESH Headings
- Humans
- Male
- Female
- Depressive Disorder, Major/diagnostic imaging
- Depressive Disorder, Major/genetics
- Depressive Disorder, Major/drug therapy
- Receptor, Serotonin, 5-HT1A/genetics
- Receptor, Serotonin, 5-HT1A/metabolism
- Receptor, Serotonin, 5-HT1A/therapeutic use
- DNA Methylation
- Serotonin/metabolism
- Serotonin/therapeutic use
- Depression
- Brain/pathology
- Positron-Emission Tomography/methods
- Stress, Psychological/genetics
Collapse
Affiliation(s)
- Hanga Galfalvy
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Department of Biostatistics, Columbia University Mailman School of Public Health, New York, New York, USA
| | | | - Jacqueline de Vegvar
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Spiro Pantazatos
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - Yung-yu Huang
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - Ainsley K. Burke
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - M. Elizabeth Sublette
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - Maria A. Oquendo
- Psychiatry Department, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Francesca Zanderigo
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - Jeffrey M. Miller
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - J. John Mann
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA; and Department of Radiology, Columbia University, New York, New York, USA
| |
Collapse
|
5
|
Murgaš M, Unterholzner J, Stöhrmann P, Philippe C, Godbersen GM, Nics L, Reed MB, Vraka C, Vanicek T, Wadsak W, Kranz GS, Hahn A, Mitterhauser M, Hacker M, Kasper S, Lanzenberger R, Baldinger-Melich P. Effects of bilateral sequential theta-burst stimulation on 5-HT 1A receptors in the dorsolateral prefrontal cortex in treatment-resistant depression: a proof-of-concept trial. Transl Psychiatry 2023; 13:33. [PMID: 36725835 PMCID: PMC9892572 DOI: 10.1038/s41398-023-02319-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 02/03/2023] Open
Abstract
Theta-burst stimulation (TBS) represents a brain stimulation technique effective for treatment-resistant depression (TRD) as underlined by meta-analyses. While the methodology undergoes constant refinement, bilateral stimulation of the dorsolateral prefrontal cortex (DLPFC) appears promising to restore left DLPFC hypoactivity and right hyperactivity found in depression. The post-synaptic inhibitory serotonin-1A (5-HT1A) receptor, also occurring in the DLPFC, might be involved in this mechanism of action. To test this hypothesis, we performed PET-imaging using the tracer [carbonyl-11C]WAY-100635 including arterial blood sampling before and after a three-week treatment with TBS in 11 TRD patients compared to sham stimulation (n = 8 and n = 3, respectively). Treatment groups were randomly assigned, and TBS protocol consisted of excitatory intermittent TBS to the left and inhibitory continuous TBS to the right DLPFC. A linear mixed model including group, hemisphere, time, and Hamilton Rating Scale for Depression (HAMD) score revealed a 3-way interaction effect of group, time, and HAMD on specific distribution volume (VS) of 5-HT1A receptor. While post-hoc comparisons showed no significant changes of 5-HT1A receptor VS in either group, higher 5-HT1A receptor VS after treatment correlated with greater difference in HAMD (r = -0.62). The results of this proof-of-concept trial hint towards potential effects of TBS on the distribution of the 5-HT1A receptor. Due to the small sample size, all results must, however, be regarded with caution.
Collapse
Affiliation(s)
- Matej Murgaš
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Jakob Unterholzner
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Peter Stöhrmann
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Cécile Philippe
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Godber M Godbersen
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Murray B Reed
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Chrysoula Vraka
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas Vanicek
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Wadsak
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Georg S Kranz
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Andreas Hahn
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Markus Mitterhauser
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
- Department of Chemistry, Institute of Inorganic Chemistry, University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria.
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria.
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria.
| | - Pia Baldinger-Melich
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Xiu Z, Muzi M, Huang J, Wolsztynski E. Patient-Adaptive Population-Based Modeling of Arterial Input Functions. IEEE TRANSACTIONS ON MEDICAL IMAGING 2023; 42:132-147. [PMID: 36094987 PMCID: PMC10008518 DOI: 10.1109/tmi.2022.3205940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Kinetic modeling of dynamic PET data requires knowledge of tracer concentration in blood plasma, described by the arterial input function (AIF). Arterial blood sampling is the gold standard for AIF measurement, but is invasive and labour intensive. A number of methods have been proposed to accurately estimate the AIF directly from blood sampling and/or imaging data. Here we consider fitting a patient-adaptive mixture of historical population time course profiles to estimate individual AIFs. Travel time of a tracer atom from the injection site to the right ventricle of the heart is modeled as a realization from a Gamma distribution, and the time this atom spends in circulation before being sampled is represented by a subject-specific linear mixture of population profiles. These functions are estimated from independent population data. Individual AIFs are obtained by projection onto this basis of population profile components. The model incorporates knowledge of injection duration into the fit, allowing for varying injection protocols. Analyses of arterial sampling data from 18F-FDG, 15O-H2O and 18F-FLT clinical studies show that the proposed model can outperform reference techniques. The statistically significant gain achieved by using population data to train the basis components, instead of fitting these from the single individual sampling data, is measured on the FDG cohort. Kinetic analyses of simulated data demonstrate the reliability and potential benefit of this approach in estimating physiological parameters. These results are further supported by numerical simulations that demonstrate convergence and stability of the proposed technique under varying training population sizes and noise levels.
Collapse
|
7
|
Matheson GJ, Ogden RT. Simultaneous multifactor Bayesian analysis (SiMBA) of PET time activity curve data. Neuroimage 2022; 256:119195. [PMID: 35452807 PMCID: PMC9470242 DOI: 10.1016/j.neuroimage.2022.119195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/24/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022] Open
Abstract
Positron emission tomography (PET) is an in vivo imaging method essential for studying the neurochemical pathophysiology of psychiatric and neurological disease. However, its high cost and exposure of participants to radiation make it unfeasible to employ large sample sizes. The major shortcoming of PET imaging is therefore its lack of power for studying clinically-relevant research questions. Here, we introduce a new method for performing PET quantification and analysis called SiMBA, which helps to alleviate these issues by improving the efficiency of PET analysis by exploiting similarities between both individuals and regions within individuals. In simulated [11C]WAY100635 data, SiMBA greatly improves both statistical power and the consistency of effect size estimation without affecting the false positive rate. This approach makes use of hierarchical, multifactor, multivariate Bayesian modelling to effectively borrow strength across the whole dataset to improve stability and robustness to measurement error. In so doing, parameter identifiability and estimation are improved, without sacrificing model interpretability. This comes at the cost of increased computational overhead, however this is practically negligible relative to the time taken to collect PET data. This method has the potential to make it possible to test clinically-relevant hypotheses which could never be studied before given the practical constraints. Furthermore, because this method does not require any additional information over and above that required for traditional analysis, it makes it possible to re-examine data which has already previously been collected at great expense. In the absence of dramatic advancements in PET image data quality, radiotracer development, or data sharing, PET imaging has been fundamentally limited in the scope of research hypotheses which could be studied. This method, especially combined with the recent steps taken by the PET imaging community to embrace data sharing, will make it possible to greatly improve the research possibilities and clinical relevance of PET neuroimaging.
Collapse
Affiliation(s)
- Granville J Matheson
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY 10032, USA.
| | - R Todd Ogden
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| |
Collapse
|
8
|
Bartlett EA, Zanderigo F, Shieh D, Miller J, Hurley P, Rubin-Falcone H, Oquendo MA, Sublette ME, Ogden RT, Mann JJ. Serotonin transporter binding in major depressive disorder: impact of serotonin system anatomy. Mol Psychiatry 2022; 27:3417-3424. [PMID: 35487966 PMCID: PMC9616969 DOI: 10.1038/s41380-022-01578-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/09/2022]
Abstract
Serotonin transporter (5-HTT) binding deficits are reported in major depressive disorder (MDD). However, most studies have not considered serotonin system anatomy when parcellating brain regions of interest (ROIs). We now investigate 5-HTT binding in MDD in two novel ways: (1) use of a 5-HTT tract-based analysis examining binding along serotonergic axons; and (2) using the Copenhagen University Hospital Neurobiology Research Unit (NRU) 5-HT Atlas, based on brain-wide binding patterns of multiple serotonin receptor types. [11C]DASB 5-HTT PET scans were obtained in 60 unmedicated participants with MDD in a current depressive episode and 31 healthy volunteers (HVs). Binding potential (BPP) was quantified with empirical Bayesian estimation in graphical analysis (EBEGA). Within the [11C]DASB tract, the MDD group showed significantly lower BPP compared with HVs (p = 0.02). This BPP diagnosis difference also significantly varied by tract location (p = 0.02), with the strongest MDD binding deficit most proximal to brainstem raphe nuclei. NRU 5-HT Atlas ROIs showed a BPP diagnosis difference that varied by region (p < 0.001). BPP was lower in MDD in 3/10 regions (p-values < 0.05). Neither [11C]DASB tract or NRU 5-HT Atlas BPP correlated with depression severity, suicidal ideation, suicide attempt history, or antidepressant medication exposure. Future studies are needed to determine the causes of this deficit in 5-HTT binding being more pronounced in proximal axon segments and in only a subset of ROIs for the pathogenesis of MDD. Such regional specificity may have implications for targeting antidepressant treatment, and may extend to other serotonin-related disorders.
Collapse
Affiliation(s)
- Elizabeth A Bartlett
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA.
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA.
| | - Francesca Zanderigo
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Denise Shieh
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
- Department of Biostatistics, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | - Jeffrey Miller
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Patrick Hurley
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Harry Rubin-Falcone
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Maria A Oquendo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M Elizabeth Sublette
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - R Todd Ogden
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
- Department of Biostatistics, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | - J John Mann
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
9
|
Lan MJ, Zanderigo F, Pantazatos SP, Sublette ME, Miller J, Ogden RT, Mann JJ. Serotonin 1A Receptor Binding of [11C]CUMI-101 in Bipolar Depression Quantified Using Positron Emission Tomography: Relationship to Psychopathology and Antidepressant Response. Int J Neuropsychopharmacol 2022; 25:534-544. [PMID: 34996114 PMCID: PMC9352178 DOI: 10.1093/ijnp/pyac001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/15/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The pathophysiology of bipolar disorder (BD) remains largely unknown despite it causing significant disability and suicide risk. Serotonin signaling may play a role in the pathophysiology, but direct evidence for this is lacking. Treatment of the depressed phase of the disorder is limited. Previous studies have indicated that positron emission tomography (PET) imaging of the serotonin 1A receptor (5HT1AR) may predict antidepressant response. METHODS A total of 20 participants with BD in a current major depressive episode and 16 healthy volunteers had PET imaging with [11C]CUMI-101, employing a metabolite-corrected input function for quantification of binding potential to the 5HT1AR. Bipolar participants then received an open-labeled, 6-week clinical trial with a selective serotonin reuptake inhibitor (SSRI) in addition to their mood stabilizer. Clinical ratings were obtained at baseline and during SSRI treatment. RESULTS Pretreatment binding potential (BPF) of [11C]CUMI-101 was associated with a number of pretreatment clinical variables within BD participants. Within the raphe nucleus, it was inversely associated with the baseline Montgomery Åsberg Rating Scale (P = .026), the Beck Depression Inventory score (P = .0023), and the Buss Durkee Hostility Index (P = .0058), a measure of lifetime aggression. A secondary analysis found [11C]CUMI-101 BPF was higher in bipolar participants compared with healthy volunteers (P = .00275). [11C]CUMI-101 BPF did not differ between SSRI responders and non-responders (P = .907) to treatment and did not predict antidepressant response (P = .580). Voxel-wise analyses confirmed the results obtained in regions of interest analyses. CONCLUSIONS A disturbance of serotonin system function is associated with both the diagnosis of BD and its severity of depression. Pretreatment 5HT1AR binding did not predict SSRI antidepressant outcome.The study was listed on clinicaltrials.gov with identifier NCT02473250.
Collapse
Affiliation(s)
- Martin J Lan
- Correspondence: Martin Lan, MD, PhD, 1051 Riverside Dr., Unit 42, New York, NY 10032, USA ()
| | - Francesca Zanderigo
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Spiro P Pantazatos
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - M Elizabeth Sublette
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Jeffrey Miller
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - R Todd Ogden
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - J John Mann
- Department of Psychiatry, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,Department of Radiology, Vagelos College of Physicians and Surgeons at Columbia University, New York, NY, USA
| |
Collapse
|
10
|
Melhem NM, Zhong Y, Miller JM, Zanderigo F, Ogden RT, Sublette ME, Newell M, Burke A, Keilp JG, Lesanpezeshki M, Bartlett E, Brent DA, Mann JJ. Brain 5-HT1A Receptor PET Binding, Cortisol Responses to Stress, and the Familial Transmission of Suicidal Behavior. Int J Neuropsychopharmacol 2021; 25:36-45. [PMID: 34555145 PMCID: PMC8756092 DOI: 10.1093/ijnp/pyab060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/22/2021] [Accepted: 10/04/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The serotonin 1A (5-HT1A) receptor has been implicated in depression and suicidal behavior. Lower resting cortisol levels are associated with higher 5-HT1A receptor binding, and both differentiate suicide attempters with depression. However, it is not clear whether 5-HT1A receptor binding and cortisol responses to stress are related to familial risk and resilience for suicidal behavior. METHODS [11C]CUMI-101 positron emission tomography imaging to quantify regional brain 5-HT1A receptor binding was conducted in individuals considered to be at high risk for mood disorder or suicidal behavior on the basis of having a first- or second-degree relative(s) with an early onset mood disorder and history of suicidal behavior. These high-risk individuals were subdivided into the following groups: high risk resilient having no mood disorder or suicidal behavior (n = 29); high risk with mood disorder and no suicidal behavior history (n = 31); and high risk with mood disorder and suicidal behavior (n = 25). Groups were compared with healthy volunteers without a family history of mood disorder or suicidal behavior (n = 34). Participants underwent the Trier Social Stress Task (TSST). All participants were free from psychotropic medications at the time of the TSST and PET scanning. RESULTS We observed no group differences in 5-HT1A receptor binding considering all regions simultaneously, nor did we observe heterogeneity of the effect of group across regions. These results were similar across outcome measures (BPND for all participants and BPp in a subset of the sample) and definitions of regions of interest (ROIs; standard or serotonin system-specific ROIs). We also found no group differences on TSST outcomes. Within the high risk with mood disorder and suicidal behavior group, lower BPp binding (β = -0.084, SE = 0.038, P = .048) and higher cortisol reactivity to stress (β = 9.25, 95% CI [3.27,15.23], P = .004) were associated with higher lethality attempts. There were no significant relationships between 5-HT1A binding and cortisol outcomes. CONCLUSIONS 5-HT1A receptor binding in ROIs was not linked to familial risk or resilience protecting against suicidal behavior or mood disorder although it may be related to lethality of suicide attempt. Future studies are needed to better understand the biological mechanisms implicated in familial risk for suicidal behavior and how hypothalamic-pituitary-adrenal axis function influences such risk.
Collapse
Affiliation(s)
- Nadine M Melhem
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Correspondence: Nadine Melhem, PhD, 3811 O’Hara Street, Pittsburgh, PA 15213, USA ()
| | - Yongqi Zhong
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Graduate School of Public Health, University of Pittsburgh
| | - Jeffrey M Miller
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - Francesca Zanderigo
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - R Todd Ogden
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York, United States
| | - M Elizabeth Sublette
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - Madison Newell
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
| | - Ainsley Burke
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - John G Keilp
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - Mohammad Lesanpezeshki
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - Elizabeth Bartlett
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - David A Brent
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - J John Mann
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| |
Collapse
|
11
|
Salinas C, Lohith TG, Purohit A, Struyk A, Sur C, Bennacef I, Beaver J, Martarello L. Test-retest characteristic of [ 18F]MK-6240 quantitative outcomes in cognitively normal adults and subjects with Alzheimer's disease. J Cereb Blood Flow Metab 2020; 40:2179-2187. [PMID: 31711342 PMCID: PMC7585918 DOI: 10.1177/0271678x19887781] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
[18F]MK-6240 is a selective, high-affinity PET radiotracer for imaging neurofibrillary tangles (NFT) in Alzheimer's disease (AD). Herein, we report test-retest (T-RT) reproducibility of [18F]MK-6240 in AD and healthy volunteers (HV). Twelve subjects with AD and three cognitively normal HV were enrolled in the study and dynamically scanned for 150 min with [18F]MK-6240 under a T-RT protocol. Two radioactivity doses were investigated: 165 ± 3 MBq (n = 6) and 300 ± 40 MBq (n = 9). Serial arterial blood samples were taken for each scan to obtain metabolite-corrected input functions. Following intravenous administration of [18F]MK-6240, the tracer rapidly partitioned into the brain and its heterogenous distribution pattern was consistent with known NFT pathology in AD. In contrast, uptake in HV was low and uniform across the brain parenchyma. Across all subjects, average T-RT variabilities in NFT-rich regions were ∼21%, ∼14% and ∼6% for various quantitative metrics: total distribution volume (VT), binding potential (BPND), and standardized uptake ratio (SUVR90-120), respectively. No significant differences in SUVR T-RT variability were observed between the high and low injected radioactivity groups (5.6% and 6.1%, respectively). This work suggests [18F]MK-6240 has adequate SUVR T-RT characteristics supporting the use of this outcome in future studies.
Collapse
Affiliation(s)
| | - Talakad G Lohith
- Translational Biomarkers, Merck & Co., Inc., West Point, PA, USA
| | | | - Arie Struyk
- Translational Pharmacology, Merck & Co., Inc., North Wales, PA, USA
| | - Cyrille Sur
- Translational Biomarkers, Merck & Co., Inc., West Point, PA, USA
| | - Idriss Bennacef
- Translational Biomarkers, Merck & Co., Inc., West Point, PA, USA
| | | | | |
Collapse
|
12
|
Cselényi Z, Jucaite A, Kristensson C, Stenkrona P, Ewing P, Varrone A, Johnström P, Schou M, Vazquez-Romero A, Moein MM, Bolin M, Siikanen J, Grybäck P, Larsson B, Halldin C, Grime K, Eriksson UG, Farde L. Quantification and reliability of [ 11C]VC - 002 binding to muscarinic acetylcholine receptors in the human lung - a test-retest PET study in control subjects. EJNMMI Res 2020; 10:59. [PMID: 32495011 PMCID: PMC7270393 DOI: 10.1186/s13550-020-00634-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/22/2020] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The radioligand [11C]VC-002 was introduced in a small initial study long ago for imaging of muscarinic acetylcholine receptors (mAChRs) in human lungs using positron emission tomography (PET). The objectives of the present study in control subjects were to advance the methodology for quantification of [11C]VC-002 binding in lung and to examine the reliability using a test-retest paradigm. This work constituted a self-standing preparatory step in a larger clinical trial aiming at estimating mAChR occupancy in the human lungs following inhalation of mAChR antagonists. METHODS PET measurements using [11C]VC-002 and the GE Discovery 710 PET/CT system were performed in seven control subjects at two separate occasions, 2-19 days apart. One subject discontinued the study after the first measurement. Radioligand binding to mAChRs in lung was quantified using an image-derived arterial input function. The total distribution volume (VT) values were obtained on a regional and voxel-by-voxel basis. Kinetic one-tissue and two-tissue compartment models (1TCM, 2TCM), analysis based on linearization of the compartment models (multilinear Logan) and image analysis by data-driven estimation of parametric images based on compartmental theory (DEPICT) were applied. The test-retest repeatability of VT estimates was evaluated by absolute variability (VAR) and intraclass correlation coefficients (ICCs). RESULTS The 1TCM was the statistically preferred model for description of [11C]VC-002 binding in the lungs. Low VAR (< 10%) across analysis methods indicated good reliability of the PET measurements. The VT estimates were stable after 60 min. CONCLUSIONS The kinetic behaviour and good repeatability of [11C]VC-002 as well as the novel lung image analysis methodology support its application in applied studies on drug-induced mAChR receptor occupancy and the pathophysiology of pulmonary disorders. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT03097380, registered: 31 March 2017.
Collapse
Affiliation(s)
- Zsolt Cselényi
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden.
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.
| | - Aurelija Jucaite
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | | | - Per Stenkrona
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Pär Ewing
- BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden
| | - Andrea Varrone
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Peter Johnström
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Magnus Schou
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Ana Vazquez-Romero
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Mohammad Mahdi Moein
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Martin Bolin
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jonathan Siikanen
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Pär Grybäck
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Bengt Larsson
- BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Ken Grime
- BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden
| | | | - Lars Farde
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
13
|
Kim MJ, Lee JH, Juarez Anaya F, Hong J, Miller W, Telu S, Singh P, Cortes MY, Henry K, Tye GL, Frankland MP, Montero Santamaria JA, Liow JS, Zoghbi SS, Fujita M, Pike VW, Innis RB. First-in-human evaluation of [ 11C]PS13, a novel PET radioligand, to quantify cyclooxygenase-1 in the brain. Eur J Nucl Med Mol Imaging 2020; 47:3143-3151. [PMID: 32399622 DOI: 10.1007/s00259-020-04855-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 05/04/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE This study assessed whether the newly developed PET radioligand [11C]PS13, which has shown excellent in vivo selectivity in previous animal studies, could be used to quantify constitutive levels of cyclooxygenase-1 (COX-1) in healthy human brain. METHODS Brain test-retest scans with concurrent arterial blood samples were obtained in 10 healthy individuals. The one- and unconstrained two-tissue compartment models, as well as the Logan graphical analysis were compared, and test-retest reliability and time-stability of total distribution volume (VT) were assessed. Correlation analyses were conducted between brain regional VT and COX-1 transcript levels provided in the Allen Human Brain Atlas. RESULTS In the brain, [11C]PS13 showed highest uptake in the hippocampus and occipital cortex. The pericentral cortex also showed relatively higher uptake compared with adjacent neocortices. The two-tissue compartment model showed the best fit in all the brain regions, and the results from the Logan graphical analysis were consistent with those from the two-tissue compartment model. VT values showed excellent test-retest variability (range 6.0-8.5%) and good reliability (intraclass correlation coefficient range 0.74-0.87). VT values also showed excellent time-stability in all brain regions, confirming that there was no radiometabolite accumulation and that shorter scans were still able to reliably measure VT. Significant correlation was observed between VT and COX-1 transcript levels (r = 0.82, P = 0.007), indicating that [11C]PS13 binding reflects actual COX-1 density in the human brain. CONCLUSIONS These results from the first-in-human evaluation of the ability of [11C]PS13 to image COX-1 in the brain justifies extending the study to disease populations with neuroinflammation. CLINICAL TRIAL REGISTRATION NCT03324646 at https://clinicaltrials.gov/ . Registered October 30, 2017. Retrospectively registered.
Collapse
Affiliation(s)
- Min-Jeong Kim
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA.
| | - Jae-Hoon Lee
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA.,Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Fernanda Juarez Anaya
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Jinsoo Hong
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - William Miller
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Sanjay Telu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Prachi Singh
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Michelle Y Cortes
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Katharine Henry
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - George L Tye
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Michael P Frankland
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Jose A Montero Santamaria
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Masahiro Fujita
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bldg. 10, Rm B1D43, Bethesda, MD, 20892-1026, USA
| |
Collapse
|
14
|
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: 1.6] [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
|
15
|
Paredes S, Cantillo S, Candido KD, Knezevic NN. An Association of Serotonin with Pain Disorders and Its Modulation by Estrogens. Int J Mol Sci 2019; 20:E5729. [PMID: 31731606 PMCID: PMC6888666 DOI: 10.3390/ijms20225729] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
Ovarian hormones play an important role in pain perception, and are responsible, at least in part, for the pain threshold differences between the sexes. Modulation of pain and its perception are mediated by neurochemical changes in several pathways, affecting both the central and peripheral nervous systems. One of the most studied neurotransmitters related to pain disorders is serotonin. Estrogen can modify serotonin synthesis and metabolism, promoting a general increase in its tonic effects. Studies evaluating the relationship between serotonin and disorders such as irritable bowel syndrome, fibromyalgia, migraine, and other types of headache suggest a clear impact of this neurotransmitter, thereby increasing the interest in serotonin as a possible future therapeutic target. This literature review describes the importance of substances such as serotonin and ovarian hormones in pain perception and illustrates the relationship between those two, and their direct influence on the presentation of the aforementioned pain-related conditions. Additionally, we review the pathways and receptors implicated in each disorder. Finally, the objective was to stimulate future pharmacological research to experimentally evaluate the potential of serotonin modulators and ovarian hormones as therapeutic agents to regulate pain in specific subpopulations.
Collapse
Affiliation(s)
- Stephania Paredes
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, 836 W. Wellington Ave. Suite 4815, Chicago, IL 60657, USA; (S.P.); (S.C.); (K.D.C.)
| | - Santiago Cantillo
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, 836 W. Wellington Ave. Suite 4815, Chicago, IL 60657, USA; (S.P.); (S.C.); (K.D.C.)
| | - Kenneth D. Candido
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, 836 W. Wellington Ave. Suite 4815, Chicago, IL 60657, USA; (S.P.); (S.C.); (K.D.C.)
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
- Department of Surgery, University of Illinois, Chicago, IL 60612, USA
| | - Nebojsa Nick Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, 836 W. Wellington Ave. Suite 4815, Chicago, IL 60657, USA; (S.P.); (S.C.); (K.D.C.)
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
- Department of Surgery, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
16
|
Gopaldas M, Zanderigo F, Zhan S, Ogden RT, Miller JM, Rubin-Falcone H, Cooper TB, Oquendo MA, Sullivan G, Mann JJ, Sublette ME. Brain serotonin transporter binding, plasma arachidonic acid and depression severity: A positron emission tomography study of major depression. J Affect Disord 2019; 257:495-503. [PMID: 31319341 PMCID: PMC6886679 DOI: 10.1016/j.jad.2019.07.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/11/2019] [Accepted: 07/04/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Serotonin transporter (5-HTT) binding and polyunsaturated fatty acids (PUFAs) are implicated in major depressive disorder (MDD). Links between the two systems in animal models have not been investigated in humans. METHODS Using positron emission tomography (PET) and [11C]DASB, we studied relationships between 5-HTT binding potential and plasma levels of PUFAs docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and arachidonic acid (AA) in medication-free MDD patients (n = 21). PUFAs were quantified using transesterification and gas chromatography. Binding potential BPP, and alternative outcome measures BPF and BPND, were determined for [11C]DASB in six a priori brain regions of interest (ROIs) using likelihood estimation in graphical analysis (LEGA) to calculate radioligand total distribution volume (VT), and a validated hybrid deconvolution approach (HYDECA) that estimates radioligand non-displaceable distribution volume (VND) without a reference region. Linear mixed models used PUFA levels as predictors and binding potential measures as outcomes across the specified ROIs; age and sex as fixed effects; and subject as random effect to account for across-region binding correlations. As nonlinear relationships were observed, a quadratic term was added to final models. RESULTS AA predicted both 5-HTT BPP and depression severity nonlinearly, described by an inverted U-shaped curve. 5-HTT binding potential mediated the relationship between AA and depression severity. LIMITATIONS Given the small sample and multiple comparisons, results require replication. CONCLUSIONS Our findings suggest that AA status may impact depression pathophysiology through effects on serotonin transport. Future studies should examine whether these relationships explain therapeutic effects of PUFAs in the treatment of MDD.
Collapse
Affiliation(s)
- Manesh Gopaldas
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,Department of Psychiatry & Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Francesca Zanderigo
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Serena Zhan
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY
| | - R. Todd Ogden
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY
| | - Jeffrey M. Miller
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Harry Rubin-Falcone
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Thomas B. Cooper
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Maria A. Oquendo
- Psychiatry Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - J. John Mann
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,Department of Radiology, Columbia University, New York, NY, USA
| | - M. Elizabeth Sublette
- Department of Psychiatry, Columbia University, New York, NY, USA,Molecular Imaging & Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA,To whom correspondence should be addressed: New York State Psychiatric Institute, 1051 Riverside Drive, Unit 42, New York, NY 10032, Tel: 646 774-7514, Fax: 646 774-7589,
| |
Collapse
|
17
|
Jain S, Nataraja NP. The Effect of Fatigue on Working Memory and Auditory Perceptual Abilities in Trained Musicians. Am J Audiol 2019; 28:483-494. [PMID: 31461329 DOI: 10.1044/2019_aja-ind50-18-0102] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Purpose Many perceptual processing processes, such as cognition, vision, sensation, and audition, are affected due to fatigue. Musicians are prone to fatigue because of long working hours and demanding profession. The research on assessing working memory abilities in musicians is heterogeneous, as none of the studies has controlled the effect of fatigue. This study was thus designed to assess the effect of fatigue on working memory and auditory perception in trained instrumental musicians. Method Twenty-six trained instrumental musicians and 25 nonmusicians participated in this study. The effect of fatigue was assessed by administering the working memory and auditory perception tests in prefatigue and postfatigue conditions. The fatigue was assessed using a 100-point visual analog scale. The working memory was assessed using Operation Span, Reading Span, and Digit Span (Forward and Backward) tests (Knutson et al., 1991). Auditory perception was assessed using speech-perception-in-noise and temporal integration tests. Results Working memory and speech perception were affected due to fatigue. Musicians, despite having better working memory abilities than nonmusicians, are equally prone to fatigue-related distress. Conclusion The advantage of music training for the working memory abilities may be reduced due to fatigue; hence, measures to reduce fatigue should be employed by the musicians. Fatigue should also be treated as a variable in all future research related to assess cognitive processing in musicians.
Collapse
Affiliation(s)
- Saransh Jain
- Department of Speech and Hearing, Jagadguru Sri Shivarathreeshwara Institute of Speech and Hearing, JSS Research Foundation, Mysuru, Karnataka, India
| | - Nuggehalli Puttareviyah Nataraja
- Department of Speech and Hearing, Jagadguru Sri Shivarathreeshwara Institute of Speech and Hearing, JSS Research Foundation, Mysuru, Karnataka, India
| |
Collapse
|
18
|
Pillai RL, Zhang M, Yang J, Mann JJ, Oquendo MA, Parsey RV, DeLorenzo C. Molecular connectivity disruptions in males with major depressive disorder. J Cereb Blood Flow Metab 2019; 39. [PMID: 29519187 PMCID: PMC6681531 DOI: 10.1177/0271678x18764053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In most positron emission tomography (PET) molecular brain imaging studies, regions of interest have been defined anatomically and examined in isolation. However, by defining regions based on physiology and examining relationships between them, we may derive more sensitive measures of receptor abnormalities in conditions such as major depressive disorder (MDD). Using an average of 52 normalized binding potential maps, acquired using radiotracer [11C]-WAY100635 and full arterial input analysis, we identified two molecular volumes of interest (VOIs) with contiguously high serotonin 1A receptor (5-HT1A) binding sites: the olfactory sulcus (OLFS) and a band of tissue including piriform, olfactory, and entorhinal cortex (PRF). We applied these VOIs to a separate cohort of 25 healthy control males and 16 males with MDD who received [11C]-WAY100635 imaging. Patients with MDD had significantly higher binding than controls in both VOIs, (p < 0.01). To identify potential homeostatic disruptions in MDD, we examined molecular connectivity, i.e. the correlation between binding of raphe nucleus (RN) 5-HT1A autoreceptors and post-synaptic receptors in molecular VOIs. Molecular connectivity was significant in healthy controls (p < 0.01), but not in patients with MDD. This disruption in molecular connectivity allowed identification of MDD cases with high sensitivity (81%) and specificity (88%).
Collapse
Affiliation(s)
| | - Mengru Zhang
- 2 Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - Jie Yang
- 3 Department of Family, Population, & Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - J John Mann
- 4 Department of Psychiatry, Molecular Imaging and Neuropathology Division, Columbia University, New York, NY, USA
| | - Maria A Oquendo
- 5 Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Ramin V Parsey
- 1 Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| | - Christine DeLorenzo
- 1 Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA.,4 Department of Psychiatry, Molecular Imaging and Neuropathology Division, Columbia University, New York, NY, USA
| |
Collapse
|
19
|
Nørgaard M, Ganz M, Svarer C, Frokjaer VG, Greve DN, Strother SC, Knudsen GM. Optimization of preprocessing strategies in Positron Emission Tomography (PET) neuroimaging: A [ 11C]DASB PET study. Neuroimage 2019; 199:466-479. [PMID: 31158479 DOI: 10.1016/j.neuroimage.2019.05.055] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 03/21/2019] [Accepted: 05/21/2019] [Indexed: 11/26/2022] Open
Abstract
Positron Emission Tomography (PET) is an important neuroimaging tool to quantify the distribution of specific molecules in the brain. The quantification is based on a series of individually designed data preprocessing steps (pipeline) and an optimal preprocessing strategy is per definition associated with less noise and improved statistical power, potentially allowing for more valid neurobiological interpretations. In spite of this, it is currently unclear how to design the best preprocessing pipeline and to what extent the choice of each preprocessing step in the pipeline minimizes subject-specific errors. To evaluate the impact of various preprocessing strategies, we systematically examined 384 different pipeline strategies in data from 30 healthy participants scanned twice with the serotonin transporter (5-HTT) radioligand [11C]DASB. Five commonly used preprocessing steps with two to four options were investigated: (1) motion correction (MC) (2) co-registration (3) delineation of volumes of interest (VOI's) (4) partial volume correction (PVC), and (5) kinetic modeling. To quantitatively compare and evaluate the impact of various preprocessing strategies, we used the performance metrics: test-retest bias, within- and between-subject variability, the intraclass-correlation coefficient, and global signal-to-noise ratio. We also performed a power analysis to estimate the required sample size to detect either a 5% or 10% difference in 5-HTT binding as a function of preprocessing pipeline. The results showed a complex downstream dependency between the various preprocessing steps on the performance metrics. The choice of MC had the most profound effect on 5-HTT binding, prior to the effects caused by PVC and kinetic modeling, and the effects differed across VOI's. Notably, we observed a negative bias in 5-HTT binding across test and retest in 98% of pipelines, ranging from 0 to 6% depending on the pipeline. Optimization of the performance metrics revealed a trade-off in within- and between-subject variability at the group-level with opposite effects (i.e. minimization of within-subject variability increased between-subject variability and vice versa). The sample size required to detect a given effect size was also compromised by the preprocessing strategy, resulting in up to 80% increases in sample size needed to detect a 5% difference in 5-HTT binding. This is the first study to systematically investigate and demonstrate the effect of choosing different preprocessing strategies on the outcome of dynamic PET studies. We provide a framework to show how optimal and maximally powered neuroimaging results can be obtained by choosing appropriate preprocessing strategies and we provide recommendations depending on the study design. In addition, the results contribute to a better understanding of methodological uncertainty and variability in preprocessing decisions for future group- and/or longitudinal PET studies.
Collapse
Affiliation(s)
- Martin Nørgaard
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Melanie Ganz
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Computer Science, University of Copenhagen, Copenhagen, Denmark
| | - Claus Svarer
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Vibe G Frokjaer
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Douglas N Greve
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen C Strother
- Rotman Research Institute, Baycrest, Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Gitte M Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
20
|
Steinberg LJ, Rubin-Falcone H, Galfalvy HC, Kaufman J, Miller JM, Sublette ME, Cooper TB, Min E, Keilp JG, Stanley BH, Oquendo MA, Ogden RT, Mann JJ. Cortisol Stress Response and in Vivo PET Imaging of Human Brain Serotonin 1A Receptor Binding. Int J Neuropsychopharmacol 2019; 22:329-338. [PMID: 30927011 PMCID: PMC6499240 DOI: 10.1093/ijnp/pyz009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/07/2018] [Accepted: 02/15/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Abnormalities in the hypothalamic-pituitary-adrenal axis, serotonergic system, and stress response have been linked to the pathogenesis of major depressive disorder. State-dependent hyper-reactivity of the hypothalamic-pituitary-adrenal axis is seen in major depressive disorder, and higher binding to the serotonin 1A receptor is observed as a trait in both currently depressed and remitted untreated major depressive disorder. Here, we sought to examine whether a relationship exists between cortisol secretion in response to a stressor and serotonin 1A receptor binding throughout the brain, both in healthy controls and participants with major depressive disorder. METHODS Research participants included 42 medication-free, depressed subjects and 31 healthy volunteers. Participants were exposed to either an acute, physical stressor (radial artery catheter insertion) or a psychological stressor (Trier Social Stress Test). Levels of serotonin 1A receptor binding on positron emission tomography with [11C]WAY-100635 were also obtained from all participants. The relationship between [11C]WAY-100635 binding and cortisol was examined using mixed linear effects models with group (major depressive disorder vs control), cortisol, brain region, and their interactions as fixed effects and subject as a random effect. RESULTS We found a positive correlation between post-stress cortisol measures and serotonin 1A receptor ligand binding levels across multiple cortical and subcortical regions, independent of diagnosis and with both types of stress. The relationship between [11C]WAY-100635 binding and cortisol was homogenous across all a priori brain regions. In contrast, resting cortisol levels were negatively correlated with serotonin 1A receptor ligand binding levels independently of diagnosis, except in the RN. There was no significant difference in cortisol between major depressive disorder participants and healthy volunteers with either stressor. Similarly, there was no correlation between cortisol and depression severity in either stressor group. CONCLUSIONS This study suggests that there may be a common underlying mechanism that links abnormalities in the serotonin system and hypothalamic-pituitary-adrenal axis hyper-reactivity to stress. Future studies need to determine how hypothalamic-pituitary-adrenal axis dysfunction affects mood to increase the risk of suicide in major depression.
Collapse
Affiliation(s)
- Louisa J Steinberg
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY,Correspondence: Louisa J. Steinberg, MD, PhD, 1051 Riverside Drive, New York, NY 10032 ()
| | - Harry Rubin-Falcone
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - Hanga C Galfalvy
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY
| | - Joshua Kaufman
- Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - Jeffrey M Miller
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - M Elizabeth Sublette
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - Thomas B Cooper
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY,Nathan S. Kline Institute for Psychiatric Research, New York, NY
| | - Eli Min
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - John G Keilp
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - Barbara H Stanley
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY
| | - Maria A Oquendo
- Psychiatry Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - R Todd Ogden
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY,Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY
| | - J John Mann
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY,Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York, NY,Department of Radiology, Columbia University, New York, NY
| |
Collapse
|
21
|
Metts AV, Rubin-Falcone H, Ogden RT, Lin X, Wilner DE, Burke AK, Sublette ME, Oquendo MA, Miller JM, Mann JJ. Antidepressant medication exposure and 5-HT 1A autoreceptor binding in major depressive disorder. Synapse 2019; 73:e22089. [PMID: 30693567 DOI: 10.1002/syn.22089] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/13/2019] [Accepted: 01/23/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVE We have previously reported higher brain serotonin 1A (5-HT1A ) autoreceptor binding in antidepressant-naïve patients with Major Depressive Disorder (MDD) compared with healthy volunteers, and a decrease in binding in MDD after selective serotonin reuptake inhibitor (SSRI) treatment. This SSRI effect is also present in rodents administered SSRIs chronically. We therefore sought to determine the duration of antidepressant medication effects on 5-HT1A receptor binding after medication discontinuation. METHODS Positron emission tomography (PET) imaging with the 5-HT1A receptor radioligand [11 C]WAY-100635 was performed in 66 individuals with current DSM-IV MDD to examine relationships between 5-HT1A binding and time since most recent antidepressant treatment. All subjects were medication-free for at least 2 weeks prior to scanning. Thirty-two additional MDD comparison subjects were antidepressant naïve. RESULTS No differences in [11 C]WAY-100635 binding were observed between antidepressant naïve and antidepressant exposed MDD groups in 13 a priori cortical and subcortical regions of interest, including raphe autoreceptors, assessed simultaneously in linear mixed effects models. Furthermore, [11 C]WAY-100635 binding did not correlate with time off antidepressants in the antidepressant exposed patients considering these ROIs. The same results were observed when effects of treatment discontinuation of any psychotropic medication used to treat their depression was examined. CONCLUSION These results indicate that any antidepressant-associated downregulation of 5-HT1A autoreceptor binding reverses within 2 weeks of medication discontinuation. Since this effect is hypothesized to mediate the antidepressant action of SSRIs, and perhaps other antidepressants, it suggests that patients who need ongoing treatment may relapse rapidly when medication is discontinued. Moreover, 2 weeks appears to be a sufficiently long washout of antidepressant medications for a reliable measure of illness-related binding levels.
Collapse
Affiliation(s)
- A V Metts
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - H Rubin-Falcone
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - R T Ogden
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - X Lin
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - D E Wilner
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - A K Burke
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - M E Sublette
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - M A Oquendo
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - J M Miller
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - J J Mann
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| |
Collapse
|
22
|
Cortes-Altamirano JL, Olmos-Hernandez A, Jaime HB, Carrillo-Mora P, Bandala C, Reyes-Long S, Alfaro-Rodríguez A. Review: 5-HT1, 5-HT2, 5-HT3 and 5-HT7 Receptors and their Role in the Modulation of Pain Response in the Central Nervous System. Curr Neuropharmacol 2018; 16:210-221. [PMID: 28901281 PMCID: PMC5883380 DOI: 10.2174/1570159x15666170911121027] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 08/24/2017] [Accepted: 09/01/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The aim of this review was to identify the mechanisms by which serotonin receptors involved at the central level are able to modulate the nociceptive response. Pain is a defense mechanism of the body that entails physiological, anatomical, neurochemical, and psychological changes, and is defined as an unpleasant sensory and emotional experience with potential risk of tissue damage, comprising the leading cause of appointments with Physicians worldwide. Treatment for this symptom has generated several neuropharmacological lines of research, due to the different types of pain and the various drugs employed to treat this condition. Serotonin [5- HydroxyTryptamine (5-HT)] is a neurotransmitter with seven families (5-HT1-5-HT7) and approximately 15 receptor subtypes. Serotonin modulates neuronal activity; however, this neurotransmitter is related with a number of physiological processes, such as cardiovascular function, gastric motility, renal function, etc. On the other hand, several researches reported that serotonin modulates nociceptive response through 5-HT1, 5-HT2, 5-HT3, and 5-HT7 receptors in the Central Nervous System (CNS). METHOD In this review, a search was conducted on PubMed, ProQuest, EBSCO, and the Science Citation Index for studies evaluating the effects of 5-HT1, 5-HT2, 5-HT3, and 5-HT7 receptors in the CNS on the modulation of different types of pain. CONCLUSION We concluded that 5-HT1, 5-HT2, 5-HT3, and 5-HT7 receptors in the CNS modulate the pain, but this depends on the distribution of the receptors, dose of agonists or antagonists, administration route, pain type and duration in order to inhibit, excite, or even maintain the nociceptive response.
Collapse
Affiliation(s)
- Jose Luis Cortes-Altamirano
- PhD Program in Biological and Health Sciences, Universidad Autonoma Metropolitana Iztapalapa-Xochimilco- Cuajimalpa, Mexico, Calzada del Hueso 1100, Col. Villa Quietud, Mexico, D.F. 04960, Mexico
| | - Adriana Olmos-Hernandez
- Departament of Neurosciences, Instituto Nacional de Rehabilitacion, "Luis Guillermo Ibarra Ibarra", Secretaria de Salud, Col. Arenal de Guadalupe, Mexico, D.F. 14389, Mexico
| | - Herlinda Bonilla Jaime
- Departament of Reproductive Biology, Universidad Autonoma Metropolitana Campus Iztapalapa, Mexico, D.F. 09340, Mexico
| | - Paul Carrillo-Mora
- Departament of Neurosciences, Instituto Nacional de Rehabilitacion, "Luis Guillermo Ibarra Ibarra", Secretaria de Salud, Col. Arenal de Guadalupe, Mexico, D.F. 14389, Mexico
| | - Cindy Bandala
- Departament of Neurosciences, Instituto Nacional de Rehabilitacion, "Luis Guillermo Ibarra Ibarra", Secretaria de Salud, Col. Arenal de Guadalupe, Mexico, D.F. 14389, Mexico
| | - Samuel Reyes-Long
- Departament of Neurosciences, Instituto Nacional de Rehabilitacion, "Luis Guillermo Ibarra Ibarra", Secretaria de Salud, Col. Arenal de Guadalupe, Mexico, D.F. 14389, Mexico
| | - Alfonso Alfaro-Rodríguez
- Departament of Neurosciences, Instituto Nacional de Rehabilitacion, "Luis Guillermo Ibarra Ibarra", Secretaria de Salud, Col. Arenal de Guadalupe, Mexico, D.F. 14389, Mexico
| |
Collapse
|
23
|
Abstract
OBJECTIVE The current state-of-the-art for compartment modeling of dynamic PET data can be described as a two-stage approach. In Stage 1, individual estimates of kinetic parameters are obtained by fitting models using standard techniques, such as nonlinear least squares, to each individual's data one subject at a time. Population-level effects, such as the difference between diagnostic groups, are analyzed in Stage 2 using standard statistical methods by treating the individual estimates as if they were observed data. While this approach is generally valid, it is possible to increase efficiency and precision of the analysis, allow more complex models to be fitted, and also to permit parameter-specific investigation by fitting data across subjects simultaneously. We explore the application of nonlinear mixed-effects (NLME) models for estimation and inference in this setting. METHODS In the NLME framework, subjects are modeled simultaneously through the inclusion of random effects of subjects for each kinetic parameter; meanwhile, population parameters are estimated directly in a joint model. RESULTS Simulation results indicate that NLME outperforms the two-stage approach in estimating group-level effects and also has improved power to detect differences across groups. We applied our NLME approach to clinical PET data and found effects not detected by the two-stage approach. CONCLUSION The proposed NLME approach is more accurate and correspondingly more powerful than the two-stage approach in compartment modeling of PET data. SIGNIFICANCE The NLME method can broaden the methodological scope of PET modeling because of its efficiency and stability.
Collapse
|
24
|
Milak MS, Pantazatos S, Rashid R, Zanderigo F, DeLorenzo C, Hesselgrave N, Ogden RT, Oquendo MA, Mulhern ST, Miller JM, Burke AK, Parsey RV, Mann JJ. Higher 5-HT 1A autoreceptor binding as an endophenotype for major depressive disorder identified in high risk offspring - A pilot study. Psychiatry Res Neuroimaging 2018; 276:15-23. [PMID: 29702461 PMCID: PMC5959803 DOI: 10.1016/j.pscychresns.2018.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/10/2018] [Accepted: 04/12/2018] [Indexed: 01/10/2023]
Abstract
Higher serotonin-1A (5-HT1A) receptor binding potential (BPF) has been found in major depressive disorder (MDD) during and between major depressive episodes. We investigated whether higher 5-HT1A binding is a biologic trait transmitted to healthy high risk (HR) offspring of MDD probands. Data were collected contemporaneously from: nine HR, 30 depressed not-recently medicated (NRM) MDD, 18 remitted NRM MDD, 51 healthy volunteer (HV) subjects. Subjects underwent positron emission tomography (PET) using [11C]WAY100635 to quantify 5-HT1A BPF, estimated using metabolite, free fraction-corrected arterial input function and cerebellar white matter as reference region. Multivoxel pattern analyses (MVPA) of PET data evaluated group status classification of individuals. When tested across 13 regions of interest, an effect of diagnosis is found on BPF which remains significant after correction for sex, age, injected mass and dose: HR have higher BPF than HV (84.3% higher in midbrain raphe, 40.8% higher in hippocampus, mean BPF across all 13 brain regions is 49.9% ± 11.8% higher). Voxel-level BPF maps distinguish HR vs. HV. Elevated 5-HT1A BPF appears to be a familially transmitted trait abnormality. Future studies are needed to replicate this finding in a larger cohort and demonstrate the link to the familial transmission of mood disorders.
Collapse
Affiliation(s)
- Matthew S Milak
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States.
| | - Spiro Pantazatos
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | - Rain Rashid
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | - Francesca Zanderigo
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | | | - Natalie Hesselgrave
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | - R Todd Ogden
- Department of Biostatistics, Columbia University, Mailman School of Public Health, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | - Maria A Oquendo
- Department of Psychiatry, Perelman School of Medicine, United States
| | - Stephanie T Mulhern
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | - Jeffrey M Miller
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | - Ainsley K Burke
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| | - Ramin V Parsey
- Department of Psychiatry, Stony Brook Medicine, Stony Brook, New York, United States
| | - J John Mann
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, NY, United States; Department of Radiology, Columbia University, College of Physicians and Surgeons, New York, NY, United States; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, United States
| |
Collapse
|
25
|
Schain M, Zanderigo F, Todd Ogden R. Likelihood estimation of drug occupancy for brain PET studies. Neuroimage 2018; 178:255-265. [PMID: 29753104 DOI: 10.1016/j.neuroimage.2018.05.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/27/2018] [Accepted: 05/05/2018] [Indexed: 11/16/2022] Open
Abstract
Neuroimaging with PET is unique in its capability to measure in vivo the occupancy of a drug. The occupancy is typically obtained by conducting PET measurements before and after administration of the drug. For radioligands for which no reference region exists, however, the only established procedure to estimate the occupancy from these data is via linear regression analysis, forming the basis for the so-called Lassen plot. There are several reasons why simple linear regression analysis is not ideal for analyzing these data, including regression attenuation and correlated errors. Here, we propose the use of Likelihood Estimation of Occupancy (LEO) in such a situation. Similar to the Lassen plot, LEO uses the total distribution volume estimates at baseline and at block condition as input, but estimates the non-displaceable distribution volume (VND) and fractional occupancy (Δ) via direct maximum likelihood estimation (MLE). This study outlines the rationale for using MLE to estimate Δ and VND from PET data, and evaluates its performance in relation to the Lassen Plot via two separate simulation experiments. Finally, LEO and Lassen plot are applied to a PET dataset acquired with [11C]WAY-100635. LEO can exploit the covariance structure of the data to improve the accuracy and precision of the estimates of Δ and VND. Theoretically, the covariance matrix can be extracted from a test-retest dataset for the radioligand at hand. Several procedures to estimate the covariance matrix were considered as part of the simulation experiments, and the effect of the test-retest sample size was also assessed. The results are conclusive in that MLE can be used to estimate Δ and VND from PET data, avoiding the limitations associated with linear regression. The performance of LEO was, naturally, dependent on the procedure used to estimate the covariance matrix, and the test-retest sample size. Given a test-retest sample size of at least 5, but preferably 10 individuals, LEO provides higher accuracy and precision than Lassen plot in the estimation of Δ and VND. We conclude that LEO is valuable in drug occupancy studies.
Collapse
Affiliation(s)
- Martin Schain
- Department of Psychiatry, Columbia University, New York, NY, USA.
| | - Francesca Zanderigo
- Department of Psychiatry, Columbia University, New York, NY, USA; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, USA
| | - R Todd Ogden
- Department of Psychiatry, Columbia University, New York, NY, USA; Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, USA; Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
26
|
Pillai RLI, Zhang M, Yang J, Boldrini M, Mann JJ, Oquendo MA, Parsey RV, DeLorenzo C. Will imaging individual raphe nuclei in males with major depressive disorder enhance diagnostic sensitivity and specificity? Depress Anxiety 2018; 35:411-420. [PMID: 29365217 PMCID: PMC5934332 DOI: 10.1002/da.22721] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/01/2017] [Accepted: 01/05/2018] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Positron emission tomography (PET) studies in major depressive disorder (MDD) have reported higher serotonin 1A (5-HT1A ) autoreceptor binding in the raphe. In males, the difference is so large that it can potentially be used as the first biological marker for MDD. However, the raphe includes several nuclei, which project to different regions of the brain and spinal cord and may be differentially involved in disease. We aimed to identify 5-HT1A differences in individual raphe nuclei using PET in order to determine whether use of subnuclei would provide greater sensitivity and specificity of diagnosing MDD. METHODS We identified individual nuclei using a hybrid set-level technique on an average [11 C]-WAY100635 PET image derived from 52 healthy volunteers (HV). We delineated three nuclei: dorsal raphe nucleus (DRN), median raphe nucleus (MRN), and raphe magnus (RMg). An atlas image of these nuclei was created and nonlinearly warped to each subject (through an associated MRI) in a separate sample of 41 males (25 HV, 16 MDD) who underwent [11 C]-WAY100635 PET. RESULTS 5-HT1A binding was elevated in DRN in MDD (P < .01), and was not different in the RMg and MRN between groups. Receiver operating characteristic (ROC) curves showed that combining DRN and MRN produces highest sensitivity (94%) and specificity (84%) to identify MDD. CONCLUSION In agreement with postmortem studies, we found higher 5-HT1A autoreceptor binding in MDD selectively in the DRN. 5-HT1A autoreceptor binding in the combined DRN and MRN is a better biomarker for MDD than in the raphe as a whole.
Collapse
Affiliation(s)
| | - Mengru Zhang
- Department of Applied Mathematics and Statistics, Columbia University, 630 W 168 St, New York, NY 10032
| | - Jie Yang
- Department of Family, Population, & Preventive Medicine, Columbia University, 630 W 168 St, New York, NY 10032
| | - Maura Boldrini
- Department of Psychiatry, Molecular Imaging and Neuropathology Division, University of Pennsylvania, 3525 Market Street, Philadelphia, PA 19104
| | - J. John Mann
- Department of Psychiatry, Molecular Imaging and Neuropathology Division, University of Pennsylvania, 3525 Market Street, Philadelphia, PA 19104
| | - Maria A. Oquendo
- Department of Psychiatry, University of Pennsylvania, 3525 Market Street, Philadelphia, PA 19104
| | - Ramin V. Parsey
- Department of Psychiatry, Stony Brook University, 101 Nicolls Rd, Stony Brook NY 11794
| | - Christine DeLorenzo
- Department of Psychiatry, Stony Brook University, 101 Nicolls Rd, Stony Brook NY 11794,Department of Psychiatry, Molecular Imaging and Neuropathology Division, University of Pennsylvania, 3525 Market Street, Philadelphia, PA 19104
| |
Collapse
|
27
|
Dai Y, Chen X, Yin J, Wang G, Wang B, Zhan Y, Nie Y, Wu K, Liang J. Investigation of the influence of sampling schemes on quantitative dynamic fluorescence imaging. BIOMEDICAL OPTICS EXPRESS 2018; 9:1859-1870. [PMID: 29675325 PMCID: PMC5905930 DOI: 10.1364/boe.9.001859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/22/2018] [Accepted: 03/18/2018] [Indexed: 05/08/2023]
Abstract
Dynamic optical data from a series of sampling intervals can be used for quantitative analysis to obtain meaningful kinetic parameters of probe in vivo. The sampling schemes may affect the quantification results of dynamic fluorescence imaging. Here, we investigate the influence of different sampling schemes on the quantification of binding potential (BP) with theoretically simulated and experimentally measured data. Three groups of sampling schemes are investigated including the sampling starting point, sampling sparsity, and sampling uniformity. In the investigation of the influence of the sampling starting point, we further summarize two cases by considering the missing timing sequence between the probe injection and sampling starting time. Results show that the mean value of BP exhibits an obvious growth trend with an increase in the delay of the sampling starting point, and has a strong correlation with the sampling sparsity. The growth trend is much more obvious if throwing the missing timing sequence. The standard deviation of BP is inversely related to the sampling sparsity, and independent of the sampling uniformity and the delay of sampling starting time. Moreover, the mean value of BP obtained by uniform sampling is significantly higher than that by using the non-uniform sampling. Our results collectively suggest that a suitable sampling scheme can help compartmental modeling of dynamic fluorescence imaging provide more accurate results and simpler operations.
Collapse
Affiliation(s)
- Yunpeng Dai
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education & School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, China
- These authors contributed equally to this work
| | - Xueli Chen
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education & School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, China
- These authors contributed equally to this work
| | - Jipeng Yin
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
- These authors contributed equally to this work
| | - Guodong Wang
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Bo Wang
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education & School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, China
| | - Yonghua Zhan
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education & School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jimin Liang
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education & School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, China
| |
Collapse
|
28
|
Zanderigo F, Pantazatos S, Rubin-Falcone H, Ogden RT, Chhetry BT, Sullivan G, Oquendo M, Miller JM, Mann JJ. In vivo relationship between serotonin 1A receptor binding and gray matter volume in the healthy brain and in major depressive disorder. Brain Struct Funct 2018; 223:2609-2625. [PMID: 29550938 DOI: 10.1007/s00429-018-1649-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 03/09/2018] [Indexed: 12/17/2022]
Abstract
Serotonin 1A (5-HT1A) receptors mediate serotonin trophic role in brain neurogenesis. Gray matter volume (GMV) loss and 5-HT1A receptor binding alterations have been identified in major depressive disorder (MDD). Here we investigated the relationship between 5-HT1A receptor binding and GMV in 40 healthy controls (HCs) and, for the first time, 47 antidepressant-free MDD patients using Voxel-Based Morphometry and [11C]WAY100635 Positron Emission Tomography. Values of GMV and 5-HT1A binding (expressed as BPF, one of the types of binding potentials that refer to displaceable or specific binding that can be quantified in vivo with PET) were obtained in 13 regions of interest, including raphe, and at the voxel level. We used regression analysis within each group to predict GMV from BPF, while covarying for age, sex, total gray matter volume and medication status. In the HCs group, we found overall a positive correlation between terminal field 5-HT1A receptor binding and GMV, which reached statistical significance in regions such as hippocampus, insula, orbital prefrontal cortex, and parietal lobe. We observed a trend towards inverse correlation between raphe 5-HT1A autoreceptor binding and anterior cingulate GMV in both groups, and a statistically significant positive correlation between raphe 5-HT1A binding and temporal GMV in MDD. Analysis of covariance at the voxel-level revealed a trend towards interaction between diagnosis and raphe 5-HT1A binding in predicting GMV in cerebellum and supramarginal gyrus (higher correlation in HCs compared with MDD). Our results replicated previous findings in the normative brain, but did not extend them to the brain in MDD, and indicated a trend towards dissociation between MDD and HCs in the relationship of raphe 5-HT1A binding with postsynaptic GMV. These results suggest that 5-HT1A receptors contribute to altered neuroplasticity in MDD, possibly via effects predating depression onset.
Collapse
Affiliation(s)
- Francesca Zanderigo
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA. .,Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY, 10032, USA.
| | - Spiro Pantazatos
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA.,Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Harry Rubin-Falcone
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA
| | - R Todd Ogden
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA.,Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY, 10032, USA.,Department of Biostatistics, Columbia University, Mailman School of Public Health, 722 W 168th Street, New York, NY, 10032, USA
| | - Binod Thapa Chhetry
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Gregory Sullivan
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Maria Oquendo
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA.,Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Jeffrey M Miller
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA.,Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY, 10032, USA
| | - J John Mann
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA.,Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY, 10032, USA.,Department of Radiology, Columbia University, 622 W 168th Street, New York, NY, 10032, USA
| |
Collapse
|
29
|
Zeng F, Nye JA, Voll RJ, Howell L, Goodman MM. Synthesis and Evaluation of Pyridyloxypyridyl Indole Carboxamides as Potential PET Imaging Agents for 5-HT 2C Receptors. ACS Med Chem Lett 2018. [PMID: 29541358 DOI: 10.1021/acsmedchemlett.7b00443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Nine pyridyloxypyridyl indole carboxamides were synthesized and displayed high affinities for 5-HT2C receptors and high selectivity over 5-HT2A and 5-HT2B. Among them, 6-methyl-N-[6-[(2-methyl-3-pyridinyl)oxy]-3-pyridinyl]1H-indole-3-carboxamide (8) exhibits the highest 5-HT2C binding affinity (Ki = 1.3 nM) and high selectivity over 5-HT2A (∼1000 times) and 5-HT2B (∼140 times). [11C]8 was synthesized by palladium-catalyzed coupling reaction between pinacolboranate 16 and [11C]CH3I with an average radiochemical yield of 27 ± 4% (n = 8, decay-corrected from end of [11C]CH3I synthesis). MicroPET imaging studies in rhesus monkeys showed regional uptake of [11C]8 in the choroid plexus, whereas the bindings in all other brain regions were low. The specific binding in the choroid plexus was confirmed by administration of a blocking dose of 0.1 mg/kg of the 5-HT2C antagonist SB-242084.
Collapse
|
30
|
Baumgartner R, Joshi A, Feng D, Zanderigo F, Ogden RT. Statistical evaluation of test-retest studies in PET brain imaging. EJNMMI Res 2018; 8:13. [PMID: 29435678 PMCID: PMC5809632 DOI: 10.1186/s13550-018-0366-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/30/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
| | - Aniket Joshi
- Novartis Institutes for Biomedical Research, Cambridge, USA
| | - Dai Feng
- Merck and Co., Inc., Kenilworth, NJ, USA
| | - Francesca Zanderigo
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA.,Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, USA
| | - R Todd Ogden
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, USA.,Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
31
|
Zanderigo F, D’Agostino AE, Joshi N, Schain M, Kumar D, Parsey RV, DeLorenzo C, Mann JJ. [11C]Harmine Binding to Brain Monoamine Oxidase A: Test-Retest Properties and Noninvasive Quantification. Mol Imaging Biol 2018; 20:667-681. [DOI: 10.1007/s11307-018-1165-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
32
|
Avendaño-Estrada A, Ávila-Rodríguez MA. Reference tissue models in the assessment of 11 C-DTBZ binding to the VMAT2 in rat striatum: A test-retest reproducibility study. Synapse 2018; 72:e22029. [PMID: 29381820 DOI: 10.1002/syn.22029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/12/2022]
Abstract
Dopaminergic PET imaging is a useful tool to assess the dopaminergic integrity and to follow-up longitudinal studies. The aim of this study was to evaluate the reliability and reproducibility of different reference tissue-based methods to determine the non-displaceable binding potential (BPND ) as a quantitative measure of 11 C-DTBZ binding to the VMAT2 in rat striatum using cerebellum as reference region. Eight healthy Wistar rats underwent two microPET scans at the age of 12 (test) and 20 weeks (retest). BPND was determined using the simplified reference tissue model, Logan reference tissue model, and multilinear reference tissue models (MRTMo and MRTM2). Additionally, a striatal-to-cerebellar-ratio (SCR) analysis was performed. The reproducibility between the two scans was assessed using the interclass correlation coefficients (ICC) and the variability index. Repeatability indices showed acceptable ICC = 0.66 (SCR) to excellent ICC = 0.98 (MRTM2) reliability for this study and a variability ranging from 12.26% (SCR) to 3.28% (MRTM2). To the best of our knowledge, this is the first report on longitudinal studies for 11 C-DTBZ in rats using reference tissue methods. Excellent intersubject and intrasubject reproducibility was obtained with the multilinear reference MRTM2, suggesting this as the best method to compare longitudinal studies, whereas the SCR method had poor reliability. Logan method, however, is a method simple to compute that shows accurate reproducibility with a reasonable level of inter- and intra-subject variability allowing crossover studies to follow-up the uptake of 11 C-DTBZ in rat striatum.
Collapse
Affiliation(s)
- Arturo Avendaño-Estrada
- División de Investigación, Unidad Radiofarmacia-Ciclotrón, Universidad Nacional Autónoma de México, Mexico City, México
| | - Miguel Angel Ávila-Rodríguez
- División de Investigación, Unidad Radiofarmacia-Ciclotrón, Universidad Nacional Autónoma de México, Mexico City, México
| |
Collapse
|
33
|
Bartlett E, DeLorenzo C, Parsey R, Huang C. Noise contamination from PET blood sampling pump: Effects on structural MRI image quality in simultaneous PET/MR studies. Med Phys 2017; 45:678-686. [PMID: 29210075 DOI: 10.1002/mp.12715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/19/2017] [Accepted: 11/26/2017] [Indexed: 11/11/2022] Open
Abstract
PURPOSE To fully quantify PET imaging outcome measures, a blood sampling pump is often used during the PET acquisition. With simultaneous PET/MR studies, a structural magnetization-prepared rapid gradient-echo (MP-RAGE) may also be acquired while the pump is generating electromagnetic noise. This study investigated whether this noise contamination would be detrimental to the quantification of volume and cortical thickness measures obtained from automated segmentation of the MP-RAGE image. METHODS MP-RAGE T1w structural images were acquired for a phantom and 10 healthy volunteers (five female, 27.2 ± 5.1 y old) with the blood sampling pump and without. The white matter signal-to-noise ratio (SNR) was computed for all images. Region-wise cortical thickness and volume were extracted with Freesurfer 5.3.0. RESULTS The phantom SNR and the white matter human subject SNR was degraded in the MP-RAGE images acquired with the pump (P = 0.005; white matter SNR: 43.9 and 50.8 with the pump and without). Intrasession, region-wise volume and cortical thickness estimates were significantly overestimated with the pump (percent difference: 1.14 ± 2.67% for volume (P = 0.0003) and 0.34 ± 1.59% (P = 0.02) for cortical thickness). Regions with percent differences greater than 5% between pump conditions were those close to tissue-air interfaces: entorhinal, frontal pole, parsorbitalis, temporal pole, and medial orbitofrontal. Synthetically adding Gaussian noise to the without pump MP-RAGE images yielded similar, significant detriments to cortical morphometry compared to without the pump. CONCLUSIONS This study provides evidence that the use of PET blood sampling pumps may generate unstructured, Gaussian-distributed noise in MP-RAGE images that significantly alters the accuracy of Freesurfer-derived volume and cortical thickness estimates. While many cortical regions showed a percent difference of less than 1% with the pump, regions close to tissue-air interfaces, subject to larger susceptibility artifacts, were significantly affected. This potential for decreased accuracy should be considered in PET/MR research studies utilizing blood sampling pumps, as well as any MRI study utilizing radiofrequency noise producing devices such as functional MRI task equipment and physiologic monitoring devices.
Collapse
Affiliation(s)
- Elizabeth Bartlett
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Christine DeLorenzo
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA.,Department of Psychiatry, Stony Brook Medicine, Stony Brook, NY, 11794, USA
| | - Ramin Parsey
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA.,Department of Psychiatry, Stony Brook Medicine, Stony Brook, NY, 11794, USA.,Department of Radiology, Stony Brook Medicine, Stony Brook, NY, 11794, USA
| | - Chuan Huang
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA.,Department of Psychiatry, Stony Brook Medicine, Stony Brook, NY, 11794, USA.,Department of Radiology, Stony Brook Medicine, Stony Brook, NY, 11794, USA
| |
Collapse
|
34
|
Vraka C, Mijailovic S, Fröhlich V, Zeilinger M, Klebermass EM, Wadsak W, Wagner KH, Hacker M, Mitterhauser M. Expanding LogP: Present possibilities. Nucl Med Biol 2017; 58:20-32. [PMID: 29309919 DOI: 10.1016/j.nucmedbio.2017.11.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/16/2017] [Accepted: 11/22/2017] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Due to the high candidate exclusion rate during a drug development process, an early prediction of the pharmacokinetic behavior would be needed. Accordingly, high performance bioaffinity chromatography (HPBAC) approaches are growing in popularity, however, there is a lack of knowledge and no consensus about the relation between HPBAC measurements, in vivo distribution and blood brain barrier (BBB) penetration behavior. With respect to radiotracers, there is almost no reference data available for plasma protein binding (PPB), permeability (Pm) and the membrane coefficient (KIAM). Thus, this study was aimed at exploring the relevance of measuring PPB, Pm and KIAM for the prediction of BBB penetration. METHODS Measurements of %PPB, Pm and KIAM were performed using HPBAC. In total, 113 compounds were tested, 43 with brain uptake, 30 not showing brain uptake and 40 with known interactions with efflux transporters. Additionally, ClogP and HPLC logPowpH7.4 data were collected. RESULTS %PPB, KIAM, Pm and ClogP values were in the same range for each of the three groups. A significant difference was observed for the HPLC logPowpH7.4 between CNS penetrating drug group (CNSpos) and the non-penetrating drug group (CNSneg), as well as for the CNSneg towards the drug group interacting with efflux transporters (DRUGefflux). However, as the other experimental data, also the HPLC logPowpH7.4 showed a broad overlapping of the single values between the groupings. CONCLUSION Experimental reference values (logP, Pm, KIAM & PPB) of commonly used PET tracers and drugs showing different BBB penetration behavior are provided. The influence of the logP on brain uptake depends strongly on the selected method. However, using a single parameter (experimental or calculated) to predict BBB penetration or for the classification of drug groups is inexpedient.
Collapse
Affiliation(s)
- Chrysoula Vraka
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria; Department for Nutritional Science, University of Vienna, Vienna, Austria
| | - Sanja Mijailovic
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Vanessa Fröhlich
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Zeilinger
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria; Faculty of Engineering, University of Applied Sciences Wiener Neustadt, Wiener Neustadt, Austria
| | - Eva-Maria Klebermass
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Wadsak
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria; Department of Inorganic Chemistry, University of Vienna, Vienna, Austria; CBmed, Graz, Austria
| | - Karl-Heinz Wagner
- Department for Nutritional Science, University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Mitterhauser
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria.
| |
Collapse
|
35
|
Pillai RLI, Malhotra A, Rupert DD, Weschler B, Williams JC, Zhang M, Yang J, Mann JJ, Oquendo MA, Parsey RV, DeLorenzo C. Relations between cortical thickness, serotonin 1A receptor binding, and structural connectivity: A multimodal imaging study. Hum Brain Mapp 2017; 39:1043-1055. [PMID: 29323797 DOI: 10.1002/hbm.23903] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 11/19/2017] [Accepted: 11/22/2017] [Indexed: 01/03/2023] Open
Abstract
Serotonin 1A (5-HT1A ) receptors play a direct role in neuronal development, cell proliferation, and dendritic branching. We hypothesized that variability in 5-HT1A binding can affect cortical thickness, and may account for a subtype of major depressive disorder (MDD) in which both are altered. To evaluate this, we measured cortical thickness from structural magnetic resonance imaging (MRI) and 5-HT1A binding by positron emission tomography (PET) in an exploratory study. To examine a range of 5-HT1A binding and cortical thickness values, we recruited 25 healthy controls and 19 patients with MDD. We hypothesized increased 5-HT1A binding in the raphe nucleus (RN) would be negatively associated with cortical thickness due to reduced serotonergic transmission. Contrary to our hypothesis, raphe 5-HT1A binding was positively correlated with cortical thickness in right posterior cingulate cortex (PCC), a region implicated in the default mode network. Cortical thickness was also positively correlated with 5-HT1A in each cortical region. We further hypothesized that the strength of 5-HT1A -cortical thickness correlation depends on the number of axons between the raphe nucleus and each region. To explore this we related 5-HT1A -cortical thickness correlation coefficients to the number of tracts connecting that region and the raphe, as measured by diffusion tensor imaging (DTI) in an independent sample. The 5-HT1A -cortical thickness association correlated significantly with the number of tracts to each region, supporting our hypothesis. We posit a defect in the raphe may affect the PCC within the default mode network in MDD through serotonergic fibers, resulting in increased ruminative processing.
Collapse
Affiliation(s)
- Rajapillai L I Pillai
- Stony Brook University SOM, Stony Brook, New York.,Department of Psychiatry, Stony Brook University, Stony Brook, New York.,Center for Understanding Biology using Imaging Technology, Stony Brook University, Stony Brook, New York
| | - Ashwin Malhotra
- Department of Neurology, New York-Presbyterian Weill Cornell Medical Center, New York, New York
| | | | | | | | - Mengru Zhang
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York
| | - Jie Yang
- Department of Family, Population, and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | - J John Mann
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Maria A Oquendo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philidelphia, Pennsylvania
| | - Ramin V Parsey
- Department of Psychiatry, Stony Brook University, Stony Brook, New York.,Center for Understanding Biology using Imaging Technology, Stony Brook University, Stony Brook, New York
| | - Christine DeLorenzo
- Department of Psychiatry, Stony Brook University, Stony Brook, New York.,Center for Understanding Biology using Imaging Technology, Stony Brook University, Stony Brook, New York.,Department of Biomedical Engineering, Columbia University, New York, New York
| |
Collapse
|
36
|
Regional Differences in Serotonin Transporter Occupancy by Escitalopram: An [ 11C]DASB PK-PD Study. Clin Pharmacokinet 2017; 56:371-381. [PMID: 27557550 DOI: 10.1007/s40262-016-0444-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND OBJECTIVE Escitalopram is one of the most commonly prescribed selective serotonin reuptake inhibitors (SSRIs). It is thought to act by blocking the serotonin transporter (SERT). However, its dose-SERT occupancy relationship is not well known, so it is not clear what level of SERT blockade is achieved by currently approved doses. METHODS To determine the dose-occupancy relationship, we measured serial SERT occupancy using [11C]DASB [3-amino-4-(2-dimethylaminomethylphenylsulfanyl)-benzonitrile] positron emission tomography (PET) and plasma drug concentrations after the administration of escitalopram in 12 healthy volunteers. We then built a pharmacokinetic-pharmacodynamic model to characterize the dose-occupancy relationship in the putamen and the dorsal raphe nucleus. RESULTS Escitalopram at approved doses occupied less SERT than expected and the SERT occupancy showed regional effects [occupancy was higher in the dorsal raphe nucleus than in the putamen (p < 0.001)]. The drug concentration when 50 % of receptors are occupied (EC50) value and Hill coefficient were significantly different between the putamen (EC50 4.30, Hill coefficient 0.459) and the dorsal raphe nucleus (EC50 2.89, Hill coefficient 0.817). CONCLUSIONS Higher doses of escitalopram than 20 mg are needed to achieve 80 % or greater SERT occupancy. Higher occupancy by escitalopram in the dorsal raphe nucleus relative to the striatum may explain the delayed onset of action of SSRIs by modulating autoreceptor function. The prevention of the 5-HT1A autoreceptor-mediated negative feedback could be a strategy for accelerating the clinical antidepressant effects.
Collapse
|
37
|
Iscan Z, Rakesh G, Rossano S, Yang J, Zhang M, Miller J, Sullivan GM, Sharma P, McClure M, Oquendo MA, Mann JJ, Parsey RV, DeLorenzo C. A positron emission tomography study of the serotonergic system in relation to anxiety in depression. Eur Neuropsychopharmacol 2017; 27:1011-1021. [PMID: 28811068 PMCID: PMC5623123 DOI: 10.1016/j.euroneuro.2017.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 06/02/2017] [Accepted: 07/29/2017] [Indexed: 12/17/2022]
Abstract
Symptoms of anxiety are highly comorbid with major depressive disorder (MDD) and are known to alter the course of the disease. To help elucidate the biological underpinnings of these prevalent disorders, we previously examined the relationship between components of anxiety (somatic, psychic and motoric) and serotonin 1A receptor (5-HT1A) binding in MDD and found that higher psychic and lower somatic anxiety was associated with greater 5-HT1A binding. In this work, we sought to examine the correlation between these anxiety symptom dimensions and 5-HTT binding. Positron emission tomography with [11C]-3-amino-4-(3-dimethylamino-methylphenylsulfanyl)-benzonitrile ([11C]DASB) and a metabolite-corrected arterial input function were used to estimate regional 5-HTT binding in 55 subjects with MDD and anxiety symptoms. Somatic anxiety was negatively correlated with 5-HTT binding in the thalamus (β=-.33, p=.025), amygdala (β=-.31, p=.007) and midbrain (β=-.72, p<.001). Psychic anxiety was positively correlated with 5-HTT binding in midbrain only (β=.46, p=.0025). To relate to our previous study, correlation between 5-HT1A and 5-HTT binding was examined, and none was found. We also examined how much of the variance in anxiety symptom dimensions could be explained by both 5-HTT and 5-HT1A binding. The developed model was able to explain 68% (p<.001), 38% (p=.012) and 32% (p=.038) of the total variance in somatic, psychic, and motoric anxiety, respectively. Results indicate the tight coupling between the serotonergic system and anxiety components, which may be confounded when using aggregate anxiety measures. Uncovering serotonin's role in anxiety and depression in this way may give way to a new generation of therapeutics and treatment strategies.
Collapse
Affiliation(s)
- Zafer Iscan
- Centre for Cognition and Decision Making, National Research University, Higher School of Economics, Russian Federation; Cognitive Neuroimaging Unit, CEA DRF/Joliot Institute, INSERM, Université Paris-Sud, Université Paris-Saclay, NeuroSpin Center, 91191 Gif-sur-Yvette, France.
| | | | - Samantha Rossano
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| | - Jie Yang
- Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Mengru Zhang
- Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - Jeffrey Miller
- New York State Psychiatric Institute and Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Gregory M Sullivan
- Tonix Pharmaceuticals, Inc., 509 Madison Avenue Suite 306, New York, NY, USA
| | - Priya Sharma
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| | - Matthew McClure
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| | - Maria A Oquendo
- New York State Psychiatric Institute and Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - J John Mann
- New York State Psychiatric Institute and Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Ramin V Parsey
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA; Radiology, Stony Brook University, Stony Brook, NY, USA
| | - Christine DeLorenzo
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA; New York State Psychiatric Institute and Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
38
|
Adhikarla V, Dunlop BW, Jarkas N, Goodman MM, Mayberg H, Owens MJ, Nye JA. Test-Retest Reliability of the SERT Imaging Agent 11C-HOMADAM in Healthy Humans. J Nucl Med 2017; 59:315-319. [PMID: 28935840 DOI: 10.2967/jnumed.117.196915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/18/2017] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to measure the test-retest reliability of 11C-N,N-dimethyl-2-(2'-amino-4'-hydroxymethylphenylthio)benzylamine (11C-HOMADAM) imaging of serotonin transporter (SERT) density in healthy control subjects. Methods: Two female and 2 male volunteers participated in the study, with each undergoing three 90-min 11C-HOMADAM PET scans. Time-activity curves were derived from SERT-rich structures and fit to 2 models: a simplified reference tissue model and a multilinear graphical model. Binding potential, the ratio of specifically bound uptake to nondisplaceable uptake at equilibrium, was calculated from the model parameter estimates. Ninety-five percent confidence intervals and the intraclass correlation coefficient (ICC) were calculated and adjusted for repeated measures. Results: The ICC values ranged from -0.13 in the dorsal raphe to 0.88 in the caudate nucleus. The highest average ICC values were in the striatum, but other regions were sensitive to measurement outliers. Conclusion: Good-to-excellent test-retest reliability was observed for SERT binding in the striatum. The dorsal raphe ICC value was sensitive to a measurement outlier. 11C-HOMADAM binding potential calculated from the simplified reference tissue model and the multilinear graphical model were robust and in good agreement.
Collapse
Affiliation(s)
- Vikram Adhikarla
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia; and
| | - Boadie W Dunlop
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia
| | - Nashwa Jarkas
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia; and
| | - Mark M Goodman
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia; and.,Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia
| | - Helen Mayberg
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia
| | - Michael J Owens
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia
| | - Jonathon A Nye
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia; and
| |
Collapse
|
39
|
DeLorenzo C, Gallezot JD, Gardus J, Yang J, Planeta B, Nabulsi N, Ogden RT, Labaree DC, Huang YH, Mann JJ, Gasparini F, Lin X, Javitch JA, Parsey RV, Carson RE, Esterlis I. In vivo variation in same-day estimates of metabotropic glutamate receptor subtype 5 binding using [ 11C]ABP688 and [ 18F]FPEB. J Cereb Blood Flow Metab 2017; 37:2716-2727. [PMID: 27742888 PMCID: PMC5536783 DOI: 10.1177/0271678x16673646] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/02/2016] [Accepted: 09/12/2016] [Indexed: 01/11/2023]
Abstract
Positron emission tomography tracers [11C]ABP688 and [18F]FPEB target the metabotropic glutamate receptor subtype 5 providing quantification of the brain glutamatergic system in vivo. Previous [11C]ABP688 positron emission tomography human test-retest studies indicate that, when performed on the same day, significant binding increases are observed; however, little deviation is reported when scans are >7 days apart. Due to the small cohorts examined previously (eight and five males, respectively), we aimed to replicate the same-day test-retest studies in a larger cohort including both males and females. Results confirmed large within-subject binding differences (ranging from -23% to 108%), suggesting that measurements are greatly affected by study design. We further investigated whether this phenomenon was specific to [11C]ABP688. Using [18F]FPEB and methodology that accounts for residual radioactivity from the test scan, four subjects were scanned twice on the same day. In these subjects, binding estimates increased between 5% and 39% between scans. Consistent with [11C]ABP688, mean absolute test-retest variability was previously reported as <12% when scans were >21 days apart. This replication study and pilot extension to [18F]FPEB suggest that observed within-day binding variation may be due to characteristics of mGluR5; for example, diurnal variation in mGluR5 may affect measurement of this receptor.
Collapse
Affiliation(s)
- Christine DeLorenzo
- Department of Psychiatry, Stony Brook University, Stony Brook, USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, USA
- Department of Psychiatry, Columbia University, New York, USA
| | | | - John Gardus
- Department of Psychiatry, Stony Brook University, Stony Brook, USA
| | - Jie Yang
- Department of Preventive Medicine, Stony Brook University, Stony Brook, USA
| | - Beata Planeta
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, USA
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, USA
| | - R Todd Ogden
- Department of Psychiatry, Columbia University, New York, USA
| | - David C Labaree
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, USA
| | - Yiyun H Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, USA
| | - J John Mann
- Department of Psychiatry, Columbia University, New York, USA
| | | | - Xin Lin
- Department of Psychiatry, Columbia University, New York, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, USA
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University, New York, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, USA
- Department of Pharmacology, Columbia University, New York, USA
| | - Ramin V Parsey
- Department of Psychiatry, Stony Brook University, Stony Brook, USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, USA
- Department of Radiology, Stony Brook University, Stony Brook, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, USA
- Department of Biomedical Engineering, Yale University, New Haven, USA
| | - Irina Esterlis
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, USA
- Department of Psychiatry, Yale University, New Haven, USA
| |
Collapse
|
40
|
Griesenauer RH, Weis JA, Arlinghaus LR, Meszoely IM, Miga MI. Breast tissue stiffness estimation for surgical guidance using gravity-induced excitation. Phys Med Biol 2017; 62:4756-4776. [PMID: 28520556 DOI: 10.1088/1361-6560/aa700a] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Tissue stiffness interrogation is fundamental in breast cancer diagnosis and treatment. Furthermore, biomechanical models for predicting breast deformations have been created for several breast cancer applications. Within these applications, constitutive mechanical properties must be defined and the accuracy of this estimation directly impacts the overall performance of the model. In this study, we present an image-derived computational framework to obtain quantitative, patient specific stiffness properties for application in image-guided breast cancer surgery and interventions. The method uses two MR acquisitions of the breast in different supine gravity-loaded configurations to fit mechanical properties to a biomechanical breast model. A reproducibility assessment of the method was performed in a test-retest study using healthy volunteers and was further characterized in simulation. In five human data sets, the within subject coefficient of variation ranged from 10.7% to 27% and the intraclass correlation coefficient ranged from 0.91-0.944 for assessment of fibroglandular and adipose tissue stiffness. In simulation, fibroglandular content and deformation magnitude were shown to have significant effects on the shape and convexity of the objective function defined by image similarity. These observations provide an important step forward in characterizing the use of nonrigid image registration methodologies in conjunction with biomechanical models to estimate tissue stiffness. In addition, the results suggest that stiffness estimation methods using gravity-induced excitation can reliably and feasibly be implemented in breast cancer surgery/intervention workflows.
Collapse
Affiliation(s)
- Rebekah H Griesenauer
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, United States of America. Vanderbilt Institute in Surgery and Engineering (VISE), Nashville, TN, United States of America
| | | | | | | | | |
Collapse
|
41
|
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: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/13/2016] [Indexed: 02/02/2023] Open
|
42
|
Yang KC, Stepanov V, Amini N, Martinsson S, Takano A, Nielsen J, Bundgaard C, Bang-Andersen B, Grimwood S, Halldin C, Farde L, Finnema SJ. Characterization of [ 11C]Lu AE92686 as a PET radioligand for phosphodiesterase 10A in the nonhuman primate brain. Eur J Nucl Med Mol Imaging 2016; 44:308-320. [PMID: 27817159 PMCID: PMC5215309 DOI: 10.1007/s00259-016-3544-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/03/2016] [Indexed: 11/28/2022]
Abstract
Purpose [11C]Lu AE92686 is a positron emission tomography (PET) radioligand that has recently been validated for examining phosphodiesterase 10A (PDE10A) in the human striatum. [11C]Lu AE92686 has high affinity for PDE10A (IC50 = 0.39 nM) and may also be suitable for examination of the substantia nigra, a region with low density of PDE10A. Here, we report characterization of regional [11C]Lu AE92686 binding to PDE10A in the nonhuman primate (NHP) brain. Methods A total of 11 PET measurements, seven baseline and four following pretreatment with unlabeled Lu AE92686 or the structurally unrelated PDE10A inhibitor MP-10, were performed in five NHPs using a high resolution research tomograph (HRRT). [11C]Lu AE92686 binding was quantified using a radiometabolite-corrected arterial input function and compartmental and graphical modeling approaches. Results Regional time-activity curves were best described with the two-tissue compartment model (2TCM). However, the distribution volume (VT) values for all regions were obtained by the Logan plot analysis, as reliable cerebellar VT values could not be derived by the 2TCM. For cerebellum, a proposed reference region, VT values increased by ∼30 % with increasing PET measurement duration from 63 to 123 min, while VT values in target regions remained stable. Both pretreatment drugs significantly decreased [11C]Lu AE92686 binding in target regions, while no significant effect on cerebellum was observed. Binding potential (BPND) values, derived with the simplified reference tissue model (SRTM), were 13–17 in putamen and 3–5 in substantia nigra and correlated well to values from the Logan plot analysis. Conclusions The method proposed for quantification of [11C]Lu AE92686 binding in applied studies in NHP is based on 63 min PET data and SRTM with cerebellum as a reference region. The study supports that [11C]Lu AE92686 can be used for PET examinations of PDE10A binding also in substantia nigra. Electronic supplementary material The online version of this article (doi:10.1007/s00259-016-3544-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kai-Chun Yang
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Vladimir Stepanov
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Nahid Amini
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Stefan Martinsson
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Akihiro Takano
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jacob Nielsen
- Synaptic Transmission, H. Lundbeck A/S, Valby, Denmark
| | | | | | - Sarah Grimwood
- Neuroscience and Pain Research Unit, Pfizer Inc., Cambridge, MA, USA
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Farde
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Personalized Health Care and Biomarkers, AstraZeneca PET Science Center at Karolinska Institutet, Stockholm, Sweden
| | - Sjoerd J Finnema
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| |
Collapse
|
43
|
Oquendo MA, Galfalvy H, Sullivan GM, Miller JM, Milak MM, Sublette ME, Cisneros-Trujillo S, Burke AK, Parsey RV, Mann JJ. Positron Emission Tomographic Imaging of the Serotonergic System and Prediction of Risk and Lethality of Future Suicidal Behavior. JAMA Psychiatry 2016; 73:1048-1055. [PMID: 27463606 PMCID: PMC6552665 DOI: 10.1001/jamapsychiatry.2016.1478] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
IMPORTANCE Biomarkers that predict suicidal behavior, especially highly lethal behavior, are urgently needed. In cross-sectional studies, individuals with depression who attempt suicide have lower midbrain serotonin transporter binding potential compared with those who do not attempt suicide, and higher serotonin1A binding potential in the raphe nuclei (RN) is associated with greater lethality of past suicide attempts and suicidal intent and ideation. OBJECTIVES To determine whether serotonin transporter binding potential in the lower midbrain predicts future suicide attempts and whether higher RN serotonin1A binding potential predicts future suicidal ideation and intent and lethality of future suicide attempts. DESIGN, SETTING, AND PARTICIPANTS In this prospective 2-year observational study, a well-characterized cohort of 100 patients presenting for treatment of a major depressive episode of at least moderate severity underwent positron emission tomography using carbon 11-labeled N-(2-(1-(4-(2-methoxyphenyl)-1-piperazinyl)ethyl))-N-(2-pyridyl)-cyclohexanecarboxamide ([11C]WAY-100635), a serotonin1A antagonist; a subset of 50 patients also underwent imaging with carbon 11-labeled 3-amino-4-(2-dimethylaminomethyl-phenylsulfanyl)- benzonitrile ([11C]DASB), a serotonin transporter radioligand. Imaging was performed at Columbia University Medical Center from May 3, 1999, to March 11, 2008. Follow-up was completed on May 28, 2010, and data were analyzed from August 1, 2013, to March 1, 2016. EXPOSURES Patients were treated naturalistically in the community and followed up for 2 years with documentation of suicidal behavior, its lethality, and suicidal ideation and intent. MAIN OUTCOMES AND MEASURES Suicide attempt or suicide. RESULTS Of the 100 patients undergoing follow-up for more than 2 years (39 men; 61 women; mean [SD] age, 40.2 [11.2] years), 15 made suicide attempts, including 2 who died by suicide. Higher RN serotonin1A binding potential predicted more suicidal ideation at 3 (b = 0.02; t = 3.45; P = .001) and 12 (b = 0.02; t = 3.63; P = .001) months and greater lethality of subsequent suicidal behavior (b = 0.08; t = 2.89; P = .01). Exploratory analyses suggest that the serotonin1A binding potential of the insula (t = 2.41; P = .04), anterior cingulate (t = 2.27; P = .04), and dorsolateral prefrontal cortex (t = 2.44; P = .03) were also predictive of lethality. Contrary to our hypotheses, suicidal intent was not predicted by serotonin1A binding potential in any brain region (F1,10 = 0.83; P = .38), and midbrain serotonin transporter binding potential did not predict future attempts (log-rank χ21 = 0.4; P = .54), possibly owing to low power. CONCLUSIONS AND RELEVANCE Greater RN serotonin1A binding potential predicted higher suicidal ideation and more lethal suicidal behavior during a 2-year period. This effect may be mediated through less serotonin neuron firing and release, which affects mood and suicidal ideation and thereby decision making.
Collapse
Affiliation(s)
- Maria A. Oquendo
- New York State Psychiatric Institute, New York,Department of Psychiatry, Columbia University, New York, New York
| | - Hanga Galfalvy
- New York State Psychiatric Institute, New York,Department of Psychiatry, Columbia University, New York, New York
| | | | - Jeffrey M. Miller
- New York State Psychiatric Institute, New York,Department of Psychiatry, Columbia University, New York, New York
| | - Matthew M. Milak
- New York State Psychiatric Institute, New York,Department of Psychiatry, Columbia University, New York, New York
| | - M. Elizabeth Sublette
- New York State Psychiatric Institute, New York,Department of Psychiatry, Columbia University, New York, New York
| | - Sebastian Cisneros-Trujillo
- New York State Psychiatric Institute, New York,Department of Psychiatry, Columbia University, New York, New York
| | - Ainsley K. Burke
- New York State Psychiatric Institute, New York,Department of Psychiatry, Columbia University, New York, New York
| | - Ramin V. Parsey
- Department of Psychiatry and Behavioral Science, Stony Brook University School of Medicine, Stony Brook, New York
| | - J. John Mann
- New York State Psychiatric Institute, New York,Department of Radiology, Columbia University, New York, New York
| |
Collapse
|
44
|
Strupp-Levitsky M, Miller JM, Rubin-Falcone H, Zanderigo F, Milak MS, Sullivan G, Ogden RT, Oquendo MA, DeLorenzo C, Simpson N, Parsey RV, Mann JJ. Lack of association between the serotonin transporter and serotonin 1A receptor: an in vivo PET imaging study in healthy adults. Psychiatry Res 2016; 255:81-86. [PMID: 27567324 PMCID: PMC5175477 DOI: 10.1016/j.pscychresns.2016.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/11/2016] [Accepted: 08/06/2016] [Indexed: 01/12/2023]
Abstract
The serotonin neurotransmitter system is modulated in part by the uptake of synaptically released serotonin (5-HT) by the serotonin transporter (5-HTT), and by specific serotonin autoreceptors such as the somatodendritic 5-HT1A receptor, which can limit serotonin neuron depolarization. However, little is known about how 5-HTT and 5-HT1A are related in vivo. To study this question, we reanalyzed positron emission tomography (PET) data obtained earlier in 40 healthy participants (21 females) using [(11)C]WAY-100635 for quantification of 5-HT1A binding and [(11)C](+)-McN-5652 for quantification of 5-HTT binding. We hypothesized negative correlations between 5-HT1A binding in the raphe nuclei (RN) and 5-HTT binding in RN terminal field regions. Controlling for sex, no significant correlations were found (all p>0.05). Similarly, an exploratory analysis correlating whole-brain voxel-wise 5-HTT binding with 5-HT1A binding in RN identified no significant clusters meeting our a priori statistical threshold. The lack of correlation between 5-HT1A and 5-HTT binding observed in the current study may be due to the different temporal responsiveness of regulatory processes controlling the somatodendritic 5-HT1A receptor and 5-HTT in response to changing availability of intrasynaptic serotonin.
Collapse
Affiliation(s)
- Michael Strupp-Levitsky
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive #42, New York, NY 10032, USA
| | - Jeffrey M Miller
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive #42, New York, NY 10032, USA; Department of Psychiatry, Columbia University, 1051 Riverside Drive #42, New York, NY 10032, USA.
| | - Harry Rubin-Falcone
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive #42, New York, NY 10032, USA; Department of Psychiatry, Columbia University, 1051 Riverside Drive #42, New York, NY 10032, USA
| | - Francesca Zanderigo
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive #42, New York, NY 10032, USA; Department of Psychiatry, Columbia University, 1051 Riverside Drive #42, New York, NY 10032, USA
| | - Matthew S Milak
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive #42, New York, NY 10032, USA; Department of Psychiatry, Columbia University, 1051 Riverside Drive #42, New York, NY 10032, USA
| | - Gregory Sullivan
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive #42, New York, NY 10032, USA; Department of Psychiatry, Columbia University, 1051 Riverside Drive #42, New York, NY 10032, USA
| | - R Todd Ogden
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive #42, New York, NY 10032, USA; Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Maria A Oquendo
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive #42, New York, NY 10032, USA; Department of Psychiatry, Columbia University, 1051 Riverside Drive #42, New York, NY 10032, USA
| | - Christine DeLorenzo
- Now at Department of Psychiatry, Department of Radiology, Stony Brook Medicine, Stony Brook, NY, USA
| | - Norman Simpson
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive #42, New York, NY 10032, USA; Department of Psychiatry, Columbia University, 1051 Riverside Drive #42, New York, NY 10032, USA
| | - Ramin V Parsey
- Now at Department of Psychiatry, Department of Radiology, Stony Brook Medicine, Stony Brook, NY, USA
| | - J John Mann
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, 1051 Riverside Drive #42, New York, NY 10032, USA; Department of Psychiatry, Columbia University, 1051 Riverside Drive #42, New York, NY 10032, USA
| |
Collapse
|
45
|
Tonietto M, Rizzo G, Veronese M, Bertoldo A. Modelling arterial input functions in positron emission tomography dynamic studies. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2016; 2015:2247-50. [PMID: 26736739 DOI: 10.1109/embc.2015.7318839] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The quantification of dynamic positron emission tomography (PET) images often requires the invasive measures of the arterial plasma tracer concentration to be used as arterial input function (AIF). In several situations, a mathematical model is fit to the hematic data to obtain a continuous and noise-free description of the AIF. In common practice, the tri-exponential and Feng's models are generally adopted. Despite their general applicability, often these approximations of blood tracer activity do not properly describe the complex behavior of the AIF (e.g. different clearance rates of the tracers) as well as they do not account for the length of the radiotracer injection. Here we propose two models able to include the injection duration as additional information in the AIF modeling and we compare their performances in eight different datasets acquired from different PET facilities.
Collapse
|
46
|
Kumar JSD, Walker M, Packiarajan M, Jubian V, Prabhakaran J, Chandrasena G, Pratap M, Parsey RV, Mann JJ. Radiosynthesis and in Vivo Evaluation of Neuropeptide Y5 Receptor (NPY5R) PET Tracers. ACS Chem Neurosci 2016; 7:540-5. [PMID: 26886507 DOI: 10.1021/acschemneuro.5b00315] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Neuropeptide Y receptor type 5 (NPY5R) is a G-protein coupled receptor (GPCR) that belongs to the subfamily of neuropeptide receptors (NPYR) that mediate the action of endogenous neuropeptide Y (NPY). Animal models and preclinical studies indicate a role for NPY5R in the pathophysiology of depression, anxiety, and obesity and as a target of potential therapeutic drugs. To better understand the pathophysiological involvement of NPY5R, and to measure target occupancy by potential therapeutic drugs, it would be advantageous to measure NPY5R binding in vivo by positron emission tomography (PET). Four potent and selective NPY5R antagonists were radiolabeled via nucleophilic aromatic substitution reactions with [(18)F]fluoride. Of the four radioligands investigated, PET studies in anesthetized baboons showed that [(18)F]LuAE00654 ([(18)F]N-[trans-4-({[4-(2-fluoropyridin-3-yl)thiazol-2-yl]amino}methyl)cyclohexyl]propane-2-sulfonamide) penetrates blood brain barrier (BBB) and a small amount is retained in the brain. Slow metabolism of [(18)F]LuAE00654 was observed in baboon plasma. Blocking studies with a specific NPY5R antagonist demonstrated up to 60% displacement of radioactivity in striatum, the brain region with highest NPY5R binding. Our studies suggest that [(18)F]LuAE00654 can be a potential PET radiotracer for the quantification and occupancy studies of NPY5R drug candidates.
Collapse
Affiliation(s)
- J. S. Dileep Kumar
- Division
of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York 10032, United States
- Department
of Psychiatry and Behavioral Medicine, Stony Brook University, New York, New York 11794, United States
| | - Mary Walker
- Chemical & Pharmacokinetic Sciences and Synaptic Transmission, Disease Biological Unit, Lundbeck Research USA, Paramus, New Jersey 07652, United States
| | - Mathivanan Packiarajan
- Chemical & Pharmacokinetic Sciences and Synaptic Transmission, Disease Biological Unit, Lundbeck Research USA, Paramus, New Jersey 07652, United States
| | - Vrej Jubian
- Chemical & Pharmacokinetic Sciences and Synaptic Transmission, Disease Biological Unit, Lundbeck Research USA, Paramus, New Jersey 07652, United States
| | - Jaya Prabhakaran
- Department
of Psychiatry, Columbia University Medical Center, New York, New York 10032, United States
| | - Gamini Chandrasena
- Chemical & Pharmacokinetic Sciences and Synaptic Transmission, Disease Biological Unit, Lundbeck Research USA, Paramus, New Jersey 07652, United States
| | - Mali Pratap
- Division
of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Ramin V. Parsey
- Department
of Psychiatry and Behavioral Medicine, Stony Brook University, New York, New York 11794, United States
| | - J. John Mann
- Division
of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York 10032, United States
- Department
of Psychiatry, Columbia University Medical Center, New York, New York 10032, United States
| |
Collapse
|
47
|
Mukherjee J, Bajwa AK, Wooten DW, Hillmer AT, Pan ML, Pandey SK, Saigal N, Christian BT. Comparative assessment of (18) F-Mefway as a serotonin 5-HT1A receptor PET imaging agent across species: Rodents, nonhuman primates, and humans. J Comp Neurol 2016; 524:1457-71. [PMID: 26509362 PMCID: PMC4783179 DOI: 10.1002/cne.23919] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 08/22/2015] [Accepted: 10/23/2015] [Indexed: 02/02/2023]
Abstract
We have developed (18) F-trans-Mefway ((18) F-Mefway) for positron emission tomography (PET) imaging studies of serotonin 5-HT1A receptors which are implicated in various brain functions. Translation of imaging the 5-HT1A receptor in animal models to humans will facilitate an understanding of the role of the receptor in human brain disorders. We report comparative brain distribution of (18) F-Mefway in normal mice, rats, monkeys, and healthy human volunteers. Mefway was found to be very selective, with subnanomolar affinity for the 5-HT1A receptor. Affinities of >55 nM were found for all other human-cloned receptor subtypes tested. Mefway was found to be a poor substrate (>30 μM) for the multidrug resistance 1 protein, suggesting low likelihood of brain uptake being affected by P-glycoprotein. Cerebellum was used as a reference region in all imaging studies across all species due to the low levels of (18) F-Mefway binding. Consistent binding of (18) F-Mefway in cortical regions, hippocampus, and raphe was observed across all species. (18) F-Mefway in the human brain regions correlated with the known postmortem distribution of 5-HT1A receptors. Quantitation of raphe was affected by the resolution of the PET scanners in rodents, whereas monkeys and humans showed a raphe to cerebellum ratio of approximately 3. (18) F-Mefway appears to be an effective 5-HT1A receptor imaging agent in all models, including humans. (18) F-Mefway therefore may be used to quantify 5-HT1A receptor distribution in brain regions for the study of various CNS disorders. J. Comp. Neurol. 524:1457-1471, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California, Irvine, California, 92697-5000
| | - Alisha K Bajwa
- Preclinical Imaging, Department of Radiological Sciences, University of California, Irvine, California, 92697-5000
| | - Dustin W Wooten
- Department of Medical Physics and Waisman Center, University of Wisconsin, Madison, Wisconsin, 53705
| | - Ansel T Hillmer
- Department of Medical Physics and Waisman Center, University of Wisconsin, Madison, Wisconsin, 53705
| | - Min-Liang Pan
- Preclinical Imaging, Department of Radiological Sciences, University of California, Irvine, California, 92697-5000
| | - Suresh K Pandey
- Preclinical Imaging, Department of Radiological Sciences, University of California, Irvine, California, 92697-5000
| | - Neil Saigal
- Preclinical Imaging, Department of Radiological Sciences, University of California, Irvine, California, 92697-5000
| | - Bradley T Christian
- Department of Medical Physics and Waisman Center, University of Wisconsin, Madison, Wisconsin, 53705
| |
Collapse
|
48
|
Miller JM, Everett BA, Oquendo MA, Ogden RT, Mann JJ, Parsey RV. Positron emission tomography quantification of serotonin transporter binding in medication-free bipolar disorder. Synapse 2016; 70:24-32. [PMID: 26426356 PMCID: PMC4654655 DOI: 10.1002/syn.21868] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 09/26/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Bipolar disorder (BD) is associated with abnormalities in the serotonin transporter (5-HTT), but specific in vivo findings have been discrepant. Using positron emission tomography (PET) and [(11)C]DASB, we compared 5-HTT binding between unmedicated depressed BD subjects and healthy volunteers (HVs). EXPERIMENTAL DESIGN 5-HTT binding in six brain regions was compared between 17 depressed, unmedicated BD subjects and 31 HVs, using the outcome measure of VT/fP (proportional to the total number of available transporters). Alternative outcome measures were examined as well. 47% of BD were BP I; and 65% reported a prior suicide attempt. PRINCIPAL OBSERVATIONS 5-HTT binding (VT/fP ) did not differ between BD and HV groups considering six brain regions of interest simultaneously (P = 0.24). In contrast, alternative outcome measures (BPF*, BPP*, and BPND*) indicated lower binding in BD compared with HV across these six regions of interest (BPF*: P = 0.047; BPP*: P = 0.032; BPND*: P = 0.031). 5-HTT binding was unrelated to suicide attempt history, depression severity, bipolar subtype, or history of past substance use disorder. CONCLUSIONS Choice of outcome measure strongly affects comparisons of serotonin transporter binding using PET with [(11)C]DASB. We do not find evidence of abnormal 5-HTT binding in bipolar depression using our primary outcome measure, VT /fP . However, we did observe lower 5-HTT binding in BD with alternative outcome measures that are frequently used with [(11)C]DASB. Relative merits and assumptions of different outcome measures are discussed. Evaluation in larger samples and during different mood states, including remission, is warranted.
Collapse
Affiliation(s)
- Jeffrey M. Miller
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY
- Department of Psychiatry, Columbia University, New York, NY
| | - Benjamin A. Everett
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY
| | - Maria A. Oquendo
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY
- Department of Psychiatry, Columbia University, New York, NY
| | - R. Todd Ogden
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY
| | - J. John Mann
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY
- Department of Psychiatry, Columbia University, New York, NY
| | | |
Collapse
|
49
|
Zanderigo F, Parsey RV, Ogden RT. Model-free quantification of dynamic PET data using nonparametric deconvolution. J Cereb Blood Flow Metab 2015; 35:1368-79. [PMID: 25873427 PMCID: PMC4528013 DOI: 10.1038/jcbfm.2015.65] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 02/25/2015] [Accepted: 03/18/2015] [Indexed: 11/09/2022]
Abstract
Dynamic positron emission tomography (PET) data are usually quantified using compartment models (CMs) or derived graphical approaches. Often, however, CMs either do not properly describe the tracer kinetics, or are not identifiable, leading to nonphysiologic estimates of the tracer binding. The PET data are modeled as the convolution of the metabolite-corrected input function and the tracer impulse response function (IRF) in the tissue. Using nonparametric deconvolution methods, it is possible to obtain model-free estimates of the IRF, from which functionals related to tracer volume of distribution and binding may be computed, but this approach has rarely been applied in PET. Here, we apply nonparametric deconvolution using singular value decomposition to simulated and test-retest clinical PET data with four reversible tracers well characterized by CMs ([(11)C]CUMI-101, [(11)C]DASB, [(11)C]PE2I, and [(11)C]WAY-100635), and systematically compare reproducibility, reliability, and identifiability of various IRF-derived functionals with that of traditional CMs outcomes. Results show that nonparametric deconvolution, completely free of any model assumptions, allows for estimates of tracer volume of distribution and binding that are very close to the estimates obtained with CMs and, in some cases, show better test-retest performance than CMs outcomes.
Collapse
Affiliation(s)
- Francesca Zanderigo
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute and Columbia University, New York, New York, USA
| | - Ramin V Parsey
- Department of Psychiatry and Radiology, Stony Brook University, Stony Brook, New York, USA
| | - R Todd Ogden
- 1] Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York, USA [2] Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, New York, USA [3] Department of Biostatistics, Columbia University, Mailman School of Public Health, New York, New York, USA
| |
Collapse
|
50
|
Choi JY, Lyoo CH, Kim JS, Kim KM, Kang JH, Choi SH, Kim JJ, Ryu YH. 18F-Mefway PET imaging of serotonin 1A receptors in humans: a comparison with 18F-FCWAY. PLoS One 2015; 10:e0121342. [PMID: 25830772 PMCID: PMC4382022 DOI: 10.1371/journal.pone.0121342] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 01/30/2015] [Indexed: 11/19/2022] Open
Abstract
Introduction The purpose of this research is to evaluate the prospects for the use of 4-(trans-18F-fluoranylmethyl)-N-[2-[4-(2-methoxyphenyl)piperazin-1-yl]ethyl]-N-pyridin-2-ylcyclohexane-1-carboxamide (18F-Mefway) in comparison to 18F-trans-4-fluoro-N-2-[4-(2-methoxyphenyl)piperazin-1-yl]ethyl]-N-(2-pyridyl)cyclohexanecarboxamide (18F-FCWAY) for the quantification of 5-HT1A receptors in human subjects. Method Five healthy male controls were included for two positron emission tomography (PET) studies: 18F-FCWAY PET after the pretreatment with 500 mg of disulfiram and two months later, 18F-Mefway PET without disulfiram. Regional time-activity curves (TACs) were extracted from nine cortical and subcortical regions in dynamic PET images. Using cerebellar cortex without vermis as reference tissue, in vivo kinetics for both radioligands were compared based on the distribution volume ratio (DVR) calculated by non-invasive Logan graphical analysis and area under the curve ratio of the TACs (AUC ratio). Result Although the pattern of regional uptakes in the 18F-Mefway PET was similar to that of the 18F-FCWAY PET (highest in the hippocampus and lowest in the cerebellar cortex), the amount of regional uptake in 18F-Mefway PET was almost half of that in 18F-FCWAY PET. The skull uptake in 18F-Mefway PET was only 25% of that in 18F-FCWAY PET with disulfiram pretreatment. The regional DVR values and AUC ratio values for 18F-Mefway were 17—40% lower than those of 18F-FCWAY. In contrast to a small overestimation of DVR values by AUC ratio values (< 10%) in 18F-FCWAY PET, the overestimation bias of AUC ratio values was much higher (up to 21%) in 18F-Mefway PET. Conclusion As 18F-Mefway showed lower DVR values and greater overestimation bias of AUC ratio values, 18F-Mefway may appear less favorable than 18F-FCWAY. However, in contrast to 18F-FCWAY, the resistance to in vivo defluorination of 18F-Mefway obviates the need for the use of a defluorination inhibitor. Thus, 18F-Mefway may be a good candidate PET radioligand for 5-HT1A receptor imaging in human.
Collapse
Affiliation(s)
- Jae Yong Choi
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Su Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Kyeong Min Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Jee Hae Kang
- Department of Chemistry and Biochemistry, Swarthmore College, 500 College Avenue Swarthmore, PA, United States of America
| | - Soo-Hee Choi
- Department of Psychiatry and Institute of Human Behavioral Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Jae-Jin Kim
- Department of Psychiatry and Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|