51
|
Nerella SG, Singh P, Sanam T, Digwal CS. PET Molecular Imaging in Drug Development: The Imaging and Chemistry Perspective. Front Med (Lausanne) 2022; 9:812270. [PMID: 35295604 PMCID: PMC8919964 DOI: 10.3389/fmed.2022.812270] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022] Open
Abstract
Positron emission tomography with selective radioligands advances the drug discovery and development process by revealing information about target engagement, proof of mechanism, pharmacokinetic and pharmacodynamic profiles. Positron emission tomography (PET) is an essential and highly significant tool to study therapeutic drug development, dose regimen, and the drug plasma concentrations of new drug candidates. Selective radioligands bring up target-specific information in several disease states including cancer, cardiovascular, and neurological conditions by quantifying various rates of biological processes with PET, which are associated with its physiological changes in living subjects, thus it reveals disease progression and also advances the clinical investigation. This study explores the major roles, applications, and advances of PET molecular imaging in drug discovery and development process with a wide range of radiochemistry as well as clinical outcomes of positron-emitting carbon-11 and fluorine-18 radiotracers.
Collapse
Affiliation(s)
- Sridhar Goud Nerella
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Priti Singh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Tulja Sanam
- Department of Microbiology and Applied Sciences, University of Agricultural Sciences, Bangalore, India
| | - Chander Singh Digwal
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| |
Collapse
|
52
|
Kinetic modeling and non-invasive approach for translocator protein quantification with 11C-DPA-713. Nucl Med Biol 2022; 108-109:76-84. [DOI: 10.1016/j.nucmedbio.2022.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 11/20/2022]
|
53
|
Wu J, Boutagy NE, Cai Z, Lin SF, Zheng MQ, Feher A, Stendahl JC, Kapinos M, Gallezot JD, Liu H, Mulnix T, Zhang W, Lindemann M, Teng JK, Miller EJ, Huang Y, Carson RE, Sinusas AJ, Liu C. Feasibility study of PET dynamic imaging of [ 18F]DHMT for quantification of reactive oxygen species in the myocardium of large animals. J Nucl Cardiol 2022; 29:216-225. [PMID: 32415628 PMCID: PMC7666654 DOI: 10.1007/s12350-020-02184-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/27/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVES We aimed to develop a dynamic imaging technique for a novel PET superoxide tracer, [18F]DHMT, to allow for absolute quantification of myocardial reactive oxygen species (ROS) production in a large animal model. METHODS Six beagle dogs underwent a single baseline dynamic [18F]DHMT PET study, whereas one animal underwent three serial dynamic studies over the course of chronic doxorubicin administration (1 mg·kg-1·week-1 for 15 weeks). During the scans, sequential arterial blood samples were obtained for plasma metabolite correction. The optimal compartment model and graphical analysis method were identified for kinetic modeling. Values for the left ventricular (LV) net influx rate, Ki, were reported for all the studies and compared with the LV standard uptake values (SUVs) and the LV-to-blood pool SUV ratios from the 60 to 90 minute static images. Parametric images were also generated. RESULTS [18F]DHMT followed irreversible kinetics once oxidized within the myocardium in the presence of superoxide, as evidenced by the fitting generated by the irreversible two-tissue (2Ti) compartment model and the linearity of Patlak analysis. Myocardial Ki values showed a weak correlation with LV SUV (R2 = 0.27), but a strong correlation with LV-to-blood pool SUV ratio (R2 = 0.92). Generation of high-quality parametric images showed superior myocardial to blood contrast compared to static images. CONCLUSIONS A dynamic PET imaging technique for [18F]DHMT was developed with full and simplified kinetic modeling for absolute quantification of myocardial superoxide production in a large animal model.
Collapse
Affiliation(s)
- Jing Wu
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Nabil E Boutagy
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale School of Medicine, New Haven, CT, USA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Shu-Fei Lin
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Ming-Qiang Zheng
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Attila Feher
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale School of Medicine, New Haven, CT, USA
| | - John C Stendahl
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale School of Medicine, New Haven, CT, USA
| | - Michael Kapinos
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Jean-Dominique Gallezot
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Hui Liu
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Tim Mulnix
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Wenjie Zhang
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Marcel Lindemann
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Jo-Ku Teng
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Edward J Miller
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale School of Medicine, New Haven, CT, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
| | - Albert J Sinusas
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA
- Section of Cardiovascular Medicine, Department of Medicine, Yale Translational Research Imaging Center, Yale School of Medicine, New Haven, CT, USA
| | - Chi Liu
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, PO Box 208048, New Haven, CT, 06520-8048, USA.
| |
Collapse
|
54
|
Sari H, Mingels C, Alberts I, Hu J, Buesser D, Shah V, Schepers R, Caluori P, Panin V, Conti M, Afshar-Oromieh A, Shi K, Eriksson L, Rominger A, Cumming P. First results on kinetic modelling and parametric imaging of dynamic 18F-FDG datasets from a long axial FOV PET scanner in oncological patients. Eur J Nucl Med Mol Imaging 2022; 49:1997-2009. [PMID: 34981164 DOI: 10.1007/s00259-021-05623-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/15/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE To investigate the kinetics of 18F-fluorodeoxyglucose (18F-FDG) by positron emission tomography (PET) in multiple organs and test the feasibility of total-body parametric imaging using an image-derived input function (IDIF). METHODS Twenty-four oncological patients underwent dynamic 18F-FDG scans lasting 65 min using a long axial FOV (LAFOV) PET/CT system. Time activity curves (TAC) were extracted from semi-automated segmentations of multiple organs, cerebral grey and white matter, and from vascular structures. The tissue and tumor lesion TACs were fitted using an irreversible two-tissue compartment (2TC) and a Patlak model. Parametric images were also generated using direct and indirect Patlak methods and their performances were evaluated. RESULTS We report estimates of kinetic parameters and metabolic rate of glucose consumption (MRFDG) for different organs and tumor lesions. In some organs, there were significant differences between MRFDG values estimated using 2TC and Patlak models. No statistically significant difference was seen between MRFDG values estimated using 2TC and Patlak methods in tumor lesions (paired t-test, P = 0.65). Parametric imaging showed that net influx (Ki) images generated using direct and indirect Patlak methods had superior tumor-to-background ratio (TBR) to standard uptake value (SUV) images (3.1- and 3.0-fold mean increases in TBRmean, respectively). Influx images generated using the direct Patlak method had twofold higher contrast-to-noise ratio in tumor lesions compared to images generated using the indirect Patlak method. CONCLUSION We performed pharmacokinetic modelling of multiple organs using linear and non-linear models using dynamic total-body 18F-FDG images. Although parametric images did not reveal more tumors than SUV images, the results confirmed that parametric imaging furnishes improved tumor contrast. We thus demonstrate the feasibility of total-body kinetic modelling and parametric imaging in basic research and oncological studies. LAFOV PET can enhance dynamic imaging capabilities by providing high sensitivity parametric images and allowing total-body pharmacokinetic analysis.
Collapse
Affiliation(s)
- Hasan Sari
- Advanced Clinical Imaging Technology, Siemens Healthcare AG, Lausanne, Switzerland.
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland.
| | - Clemens Mingels
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Ian Alberts
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Jicun Hu
- Siemens Medical Solutions, USA Inc., Knoxville, TN, USA
| | - Dorothee Buesser
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Vijay Shah
- Siemens Medical Solutions, USA Inc., Knoxville, TN, USA
| | - Robin Schepers
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Patrik Caluori
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | | | | | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Lars Eriksson
- Siemens Medical Solutions, USA Inc., Knoxville, TN, USA
- Department of Oncology and Pathology, Medical Radiation Physics, Karolinska Institutet, Stockholm, Sweden
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
55
|
Knoll P, Tsapaki V, Varga J, Šámal M. Parametric imaging used in nuclear medicine. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
56
|
Wang Y, Li E, Cherry SR, Wang G. Total-Body PET Kinetic Modeling and Potential Opportunities Using Deep Learning. PET Clin 2021; 16:613-625. [PMID: 34353745 PMCID: PMC8453049 DOI: 10.1016/j.cpet.2021.06.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The uEXPLORER total-body PET/CT system provides a very high level of detection sensitivity and simultaneous coverage of the entire body for dynamic imaging for quantification of tracer kinetics. This article describes the fundamentals and potential benefits of total-body kinetic modeling and parametric imaging focusing on the noninvasive derivation of blood input function, multiparametric imaging, and high-temporal resolution kinetic modeling. Along with its attractive properties, total-body kinetic modeling also brings significant challenges, such as the large scale of total-body dynamic PET data, the need for organ and tissue appropriate input functions and kinetic models, and total-body motion correction. These challenges, and the opportunities using deep learning, are discussed.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Biomedical Engineering, University of California, 451 E. Health Sciences Drive, Davis, CA 95616, USA; Department of Radiology, University of California Davis Medical Center, Ambulatory Care Center, Building Suite 3100, 4860 Y Street, Sacramento, CA 95817, USA
| | - Elizabeth Li
- Department of Biomedical Engineering, University of California, 451 E. Health Sciences Drive, Davis, CA 95616, USA
| | - Simon R Cherry
- Department of Biomedical Engineering, University of California, 451 E. Health Sciences Drive, Davis, CA 95616, USA; Department of Radiology, University of California Davis Medical Center, Ambulatory Care Center, Building Suite 3100, 4860 Y Street, Sacramento, CA 95817, USA
| | - Guobao Wang
- Department of Radiology, University of California Davis Medical Center, Ambulatory Care Center, Building Suite 3100, 4860 Y Street, Sacramento, CA 95817, USA.
| |
Collapse
|
57
|
Veronese M, Rizzo G, Belzunce M, Schubert J, Searle G, Whittington A, Mansur A, Dunn J, Reader A, Gunn RN. Reproducibility of findings in modern PET neuroimaging: insight from the NRM2018 grand challenge. J Cereb Blood Flow Metab 2021; 41:2778-2796. [PMID: 33993794 PMCID: PMC8504414 DOI: 10.1177/0271678x211015101] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/10/2021] [Accepted: 04/03/2021] [Indexed: 11/16/2022]
Abstract
The reproducibility of findings is a compelling methodological problem that the neuroimaging community is facing these days. The lack of standardized pipelines for image processing, quantification and statistics plays a major role in the variability and interpretation of results, even when the same data are analysed. This problem is well-known in MRI studies, where the indisputable value of the method has been complicated by a number of studies that produce discrepant results. However, any research domain with complex data and flexible analytical procedures can experience a similar lack of reproducibility. In this paper we investigate this issue for brain PET imaging. During the 2018 NeuroReceptor Mapping conference, the brain PET community was challenged with a computational contest involving a simulated neurotransmitter release experiment. Fourteen international teams analysed the same imaging dataset, for which the ground-truth was known. Despite a plurality of methods, the solutions were consistent across participants, although not identical. These results should create awareness that the increased sharing of PET data alone will only be one component of enhancing confidence in neuroimaging results and that it will be important to complement this with full details of the analysis pipelines and procedures that have been used to quantify data.
Collapse
Affiliation(s)
- Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | | | - Martin Belzunce
- School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital, King’s College London, London, UK
| | - Julia Schubert
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | | | | | - Ayla Mansur
- Invicro LLC, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Joel Dunn
- School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital, King’s College London, London, UK
- King's College London & Guy's and St. Thomas' PET Centre, London, UK
| | - Andrew Reader
- School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital, King’s College London, London, UK
| | - Roger N Gunn
- Invicro LLC, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - and the Grand Challenge Participants#
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- Invicro LLC, London, UK
- School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital, King’s College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
- King's College London & Guy's and St. Thomas' PET Centre, London, UK
| |
Collapse
|
58
|
Neumaier F, Zlatopolskiy BD, Neumaier B. Drug Penetration into the Central Nervous System: Pharmacokinetic Concepts and In Vitro Model Systems. Pharmaceutics 2021; 13:1542. [PMID: 34683835 PMCID: PMC8538549 DOI: 10.3390/pharmaceutics13101542] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Delivery of most drugs into the central nervous system (CNS) is restricted by the blood-brain barrier (BBB), which remains a significant bottleneck for development of novel CNS-targeted therapeutics or molecular tracers for neuroimaging. Consistent failure to reliably predict drug efficiency based on single measures for the rate or extent of brain penetration has led to the emergence of a more holistic framework that integrates data from various in vivo, in situ and in vitro assays to obtain a comprehensive description of drug delivery to and distribution within the brain. Coupled with ongoing development of suitable in vitro BBB models, this integrated approach promises to reduce the incidence of costly late-stage failures in CNS drug development, and could help to overcome some of the technical, economic and ethical issues associated with in vivo studies in animal models. Here, we provide an overview of BBB structure and function in vivo, and a summary of the pharmacokinetic parameters that can be used to determine and predict the rate and extent of drug penetration into the brain. We also review different in vitro models with regard to their inherent shortcomings and potential usefulness for development of fast-acting drugs or neurotracers labeled with short-lived radionuclides. In this regard, a special focus has been set on those systems that are sufficiently well established to be used in laboratories without significant bioengineering expertise.
Collapse
Affiliation(s)
- Felix Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (B.D.Z.); (B.N.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428 Jülich, Germany
| | - Boris D. Zlatopolskiy
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (B.D.Z.); (B.N.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428 Jülich, Germany
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (B.D.Z.); (B.N.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428 Jülich, Germany
| |
Collapse
|
59
|
Mathematical Models for FDG Kinetics in Cancer: A Review. Metabolites 2021; 11:metabo11080519. [PMID: 34436460 PMCID: PMC8398381 DOI: 10.3390/metabo11080519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 11/21/2022] Open
Abstract
Compartmental analysis is the mathematical framework for the modelling of tracer kinetics in dynamical Positron Emission Tomography. This paper provides a review of how compartmental models are constructed and numerically optimized. Specific focus is given on the identifiability and sensitivity issues and on the impact of complex physiological conditions on the mathematical properties of the models.
Collapse
|
60
|
Veronese M, Tuosto M, Marques TR, Howes O, Pascual B, Yu M, Masdeu JC, Turkheimer F, Bertoldo A, Zanotti-Fregonara P. Parametric Mapping for TSPO PET Imaging with Spectral Analysis Impulsive Response Function. Mol Imaging Biol 2021; 23:560-571. [PMID: 33475944 PMCID: PMC8277653 DOI: 10.1007/s11307-020-01575-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/27/2020] [Accepted: 12/21/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of this study was to investigate the use of spectral analysis (SA) for voxel-wise analysis of TSPO PET imaging studies. TSPO PET quantification is methodologically complicated by the heterogeneity of TSPO expression and its cell-dependent modulation during neuroinflammatory response. Compartmental models to account for this complexity exist, but they are unreliable at the high noise typical of voxel data. On the contrary, SA is noise-robust for parametric mapping and provides useful information about tracer kinetics with a free compartmental structure. PROCEDURES SA impulse response function (IRF) calculated at 90 min after tracer injection was used as main parameter of interest in 3 independent PET imaging studies to investigate its sensitivity to (1) a TSPO genetic polymorphism (rs6971) known to affect tracer binding in a cross-sectional analysis of healthy controls scanned with [11C]PBR28 PET; (2) TSPO density with [11C]PBR28 in a competitive blocking study with a TSPO blocker, XBD173; and (3) the higher affinity of a second radiotracer for TSPO, by using data from a head-to-head comparison between [11C]PBR28 and [11C]ER176 scans. RESULTS SA-IRF produced parametric maps of visually good quality. These were sensitive to TSPO genotype (mean relative difference between high- and mixed-affinity binders = 25 %) and TSPO availability (mean signal displacement after 90 mg oral administration of XBD173 = 39 %). Regional averages of voxel-wise IRF estimates were strongly associated with regional total distribution volume (VT) estimated with a 2-tissue compartmental model with vascular compartment (Pearson's r = 0.86 ± 0.11) but less strongly with standard 2TCM-VT (Pearson's r = 0.76 ± 0.32). Finally, SA-IRF estimates for [11C]ER176 were significantly higher than [11C]PBR28 ones, consistent with the higher amount of specific binding of the former tracer. CONCLUSIONS SA-IRF can be used for voxel-wise quantification of TSPO PET data because it generates high-quality parametric maps, it is sensitive to TSPO availability and genotype, and it accounts for the complexity of TSPO tracer kinetics with no additional assumptions.
Collapse
Affiliation(s)
- Mattia Veronese
- Department of Neuroimaging, IoPPN, King's College London, London, UK.
| | - Marcello Tuosto
- Department of Information Engineering, Padova University, Padova, Italy
| | - Tiago Reis Marques
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
| | - Oliver Howes
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
- MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Belen Pascual
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Meixiang Yu
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | | | - Alessandra Bertoldo
- Department of Information Engineering, Padova University, Padova, Italy
- Padova Neuroscience Centre, Padova University, Padova, Italy
| | - Paolo Zanotti-Fregonara
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| |
Collapse
|
61
|
Abdo RA, Lamare F, Fernandez P, Bentourkia M. Quantification of Hypoxia in Human Glioblastoma using PET with 18F-FMISO. Nucl Med Mol Imaging 2021; 55:107-115. [PMID: 34109007 DOI: 10.1007/s13139-021-00693-8] [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: 09/02/2020] [Revised: 02/16/2021] [Accepted: 03/12/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose This study aimed to investigate the results of compartmental modeling (CM) and spectral analysis (SA) generated with dynamic 18F-FMISO tumor images. Besides, the regular tissue-to-blood ratio (TBR) images were derived and compared with the dynamic models. Methods Nine subjects with glioblastoma underwent PET/CT imaging with the 18F-FMISO tracer. The protocol for PET imaging began with 15 min in dynamic mode and two 10-min duration static images at 120 min and 180 min post-injection. We used the two-tissue compartmental model for CM at the voxel basis, and we conducted SA to estimate the 18F-FMISO accumulation within each voxel. We also investigated the usual tumor-to-blood ratio (TBR) for comparison. Results The images of the tumor showed different patterns of hypoxia and necrosis as a function of PET scanning times, while CM and SA methods based on dynamic PET imaging equally located tumor hypoxia. The mean correlation of Ki images of all subjects between CM and SA was 0.63 ± 0.19 (0.24-0.86). CM produced less noisy K i images than SA, and, in the contrary, SA produced accumulation component images more clear than with CM. CM-K i and SA-K i images were correlated with TBR images (r = 0.72 ± 0.20 and 0.56 ± 0.26, respectively). In the only subject having a continuously increasing tumor time-activity curve, the k 3 image showed a high uptake in the necrosis region which was not apparent in TBR or K i images. Conclusion Based on these results, the combination of CM and SA approaches was found more appropriate in generating voxel-based hypoxia images.
Collapse
Affiliation(s)
- Redha-Alla Abdo
- Department of Nuclear Medicine and Radiobiology, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC J1H 5N4 Canada
| | - Frédéric Lamare
- Service de Médecine Nucléaire, Université de Bordeaux-II, EPHE, Avenue du Haut-Lévêque, 33604 Pessac cedex, Bordeaux, France
| | - Philippe Fernandez
- Service de Médecine Nucléaire, Université de Bordeaux-II, EPHE, Avenue du Haut-Lévêque, 33604 Pessac cedex, Bordeaux, France
| | - M'hamed Bentourkia
- Department of Nuclear Medicine and Radiobiology, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC J1H 5N4 Canada
| |
Collapse
|
62
|
Tuncel H, Boellaard R, Coomans EM, de Vries EFJ, Glaudemans AWJM, Feltes PK, García DV, Verfaillie SCJ, Wolters EE, Sweeney SP, Ryan JM, Ivarsson M, Lynch BA, Schober P, Scheltens P, Schuit RC, Windhorst AD, De Deyn PP, van Berckel BNM, Golla SSV. Kinetics and 28-day test-retest repeatability and reproducibility of [ 11C]UCB-J PET brain imaging. J Cereb Blood Flow Metab 2021; 41:1338-1350. [PMID: 34013797 PMCID: PMC8138337 DOI: 10.1177/0271678x20964248] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 08/19/2020] [Accepted: 09/27/2020] [Indexed: 11/18/2022]
Abstract
[11C]UCB-J is a novel radioligand that binds to synaptic vesicle glycoprotein 2A (SV2A). The main objective of this study was to determine the 28-day test-retest repeatability (TRT) of quantitative [11C]UCB-J brain positron emission tomography (PET) imaging in Alzheimer's disease (AD) patients and healthy controls (HCs). Nine HCs and eight AD patients underwent two 60 min dynamic [11C]UCB-J PET scans with arterial sampling with an interval of 28 days. The optimal tracer kinetic model was assessed using the Akaike criteria (AIC). Micro-/macro-parameters such as tracer delivery (K1) and volume of distribution (VT) were estimated using the optimal model. Data were also analysed for simplified reference tissue model (SRTM) with centrum semi-ovale (white matter) as reference region. Based on AIC, both 1T2k_VB and 2T4k_VB described the [11C]UCB-J kinetics equally well. Analysis showed that whole-brain grey matter TRT for VT, DVR and SRTM BPND were -2.2% ± 8.5, 0.4% ± 12.0 and -8.0% ± 10.2, averaged over all subjects. [11C]UCB-J kinetics can be well described by a 1T2k_VB model, and a 60 min scan duration was sufficient to obtain reliable estimates for both plasma input and reference tissue models. TRT for VT, DVR and BPND was <15% (1SD) averaged over all subjects and indicates adequate quantitative repeatability of [11C]UCB-J PET.
Collapse
Affiliation(s)
- Hayel Tuncel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Emma M Coomans
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Erik FJ de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center, University of Groningen, Groningen, The Netherlands
| | - Andor WJM Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center, University of Groningen, Groningen, The Netherlands
| | - Paula Kopschina Feltes
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center, University of Groningen, Groningen, The Netherlands
| | - David V García
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center, University of Groningen, Groningen, The Netherlands
| | - Sander CJ Verfaillie
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Emma E Wolters
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | | | | | | | | | - Patrick Schober
- Department of Anaesthesiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Philip Scheltens
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Robert C Schuit
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Peter P De Deyn
- Department of Neurology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Alzheimer Research Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart NM van Berckel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sandeep SV Golla
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
63
|
van de Stadt EA, Yaqub M, Lammertsma AA, Poot AJ, Schuit RC, Remmelzwaal S, Schwarte LA, Smit EF, Hendrikse H, Bahce I. Identifying advanced stage NSCLC patients who benefit from afatinib therapy using 18F-afatinib PET/CT imaging. Lung Cancer 2021; 155:156-162. [PMID: 33836373 DOI: 10.1016/j.lungcan.2021.03.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/09/2021] [Accepted: 03/22/2021] [Indexed: 12/09/2022]
Abstract
OBJECTIVES Non-small cell lung cancer (NSCLC) tumors harboring common (exon19del, L858R) and uncommon (e.g. G719X, L861Q) activating epidermal growth factor receptor (EGFR) mutations are best treated with EGFR tyrosine kinase inhibitors (TKI) such as the first-generation EGFR TKI erlotinib, second-generation afatinib or third-generation osimertinib. However, identifying these patients through biopsy is not always possible. Therefore, our aim was to evaluate whether 18F-afatinib PET/CT could identify patients with common and uncommon EGFR mutations. Furthermore, we evaluated the relation between tumor 18F-afatinib uptake and response to afatinib therapy. MATERIALS AND METHODS 18F-afatinib PET/CT was performed in 12 patients: 6 EGFR wild type (WT), 3 EGFR common and 3 EGFR uncommon mutations. Tumor uptake of 18F-afatinib was quantified using TBR_WB60-90 (tumor-to-whole blood activity ratio 60-90 min post-injection) for each tumor. Response was quantified per lesion using percentage of change (PC): [(response measurement (RM)-baseline measurement (BM))/BM]×100. Statistical analyses were performed using t-tests, correlation plots and sensitivity/specificity analysis. RESULTS Twenty-one tumors were identified. Injected dose was 348 ± 31 MBq. Group differences were significant between WT versus EGFR (common and uncommon) activating mutations (p = 0.03). There was no significant difference between EGFR common versus uncommon mutations (p = 0.94). A TBR_WB60-90 cut-off value of 6 showed the best relationship with response with a sensitivity of 70 %, a specificity of 100 % and a positive predictive value of 100 %. CONCLUSION 18F-afatinib uptake was higher in tumors with EGFR mutations (common and uncommon) compared to WT. Furthermore, a TBR_WB60-90 cut-off of 6 was found to best predict response to therapy. 18F-afatinib PET/CT could provide a means to identify EGFR mutation positive patients who benefit from afatinib therapy.
Collapse
Affiliation(s)
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Alex J Poot
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Robert C Schuit
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Sharon Remmelzwaal
- Department of Epidemiology and Data Science, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Lothar A Schwarte
- Department of Anesthesiology, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Egbert F Smit
- Department of Pulmonology, Amsterdam UMC location VUmc, Amsterdam, the Netherlands; Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Harry Hendrikse
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUmc, Amsterdam, the Netherlands; Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Idris Bahce
- Department of Pulmonology, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| |
Collapse
|
64
|
Molotkov A, Carberry P, Dolan MA, Joseph S, Idumonyi S, Oya S, Castrillon J, Konofagou EE, Doubrovin M, Lesser GJ, Zanderigo F, Mintz A. Real-Time Positron Emission Tomography Evaluation of Topotecan Brain Kinetics after Ultrasound-Mediated Blood-Brain Barrier Permeability. Pharmaceutics 2021; 13:405. [PMID: 33803856 PMCID: PMC8003157 DOI: 10.3390/pharmaceutics13030405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 01/13/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary adult brain malignancy with an extremely poor prognosis and a median survival of fewer than two years. A key reason for this high mortality is that the blood-brain barrier (BBB) significantly restricts systemically delivered therapeutics to brain tumors. High-intensity focused ultrasound (HIFU) with microbubbles is a methodology being used in clinical trials to noninvasively permeabilize the BBB for systemic therapeutic delivery to GBM. Topotecan is a topoisomerase inhibitor used as a chemotherapeutic agent to treat ovarian and small cell lung cancer. Studies have suggested that topotecan can cross the BBB and can be used to treat brain metastases. However, pharmacokinetic data demonstrated that topotecan peak concentration in the brain extracellular fluid after systemic injection was ten times lower than in the blood, suggesting less than optimal BBB penetration by topotecan. We hypothesize that HIFU with microbubbles treatment can open the BBB and significantly increase topotecan concentration in the brain. We radiolabeled topotecan with 11C and acquired static and dynamic positron emission tomography (PET) scans to quantify [11C] topotecan uptake in the brains of normal mice and mice after HIFU treatment. We found that HIFU treatments significantly increased [11C] topotecan brain uptake. Moreover, kinetic analysis of the [11C] topotecan dynamic PET data demonstrated a substantial increase in [11C] topotecan volume of distribution in the brain. Furthermore, we found a decrease in [11C] topotecan brain clearance, confirming the potential of HIFU to aid in the delivery of topotecan through the BBB. This opens the potential clinical application of [11C] topotecan as a tool to predict topotecan loco-regional brain concentration in patients with GBMs undergoing experimental HIFU treatments.
Collapse
Affiliation(s)
- Andrei Molotkov
- Department of Radiology, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, USA; (A.M.); (P.C.); (M.A.D.); (S.J.); (S.I.); (S.O.); (J.C.); (M.D.)
| | - Patrick Carberry
- Department of Radiology, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, USA; (A.M.); (P.C.); (M.A.D.); (S.J.); (S.I.); (S.O.); (J.C.); (M.D.)
| | - Martin A. Dolan
- Department of Radiology, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, USA; (A.M.); (P.C.); (M.A.D.); (S.J.); (S.I.); (S.O.); (J.C.); (M.D.)
| | - Simon Joseph
- Department of Radiology, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, USA; (A.M.); (P.C.); (M.A.D.); (S.J.); (S.I.); (S.O.); (J.C.); (M.D.)
| | - Sidney Idumonyi
- Department of Radiology, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, USA; (A.M.); (P.C.); (M.A.D.); (S.J.); (S.I.); (S.O.); (J.C.); (M.D.)
| | - Shunichi Oya
- Department of Radiology, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, USA; (A.M.); (P.C.); (M.A.D.); (S.J.); (S.I.); (S.O.); (J.C.); (M.D.)
| | - John Castrillon
- Department of Radiology, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, USA; (A.M.); (P.C.); (M.A.D.); (S.J.); (S.I.); (S.O.); (J.C.); (M.D.)
| | - Elisa E. Konofagou
- Department of Biomedical Engineering, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, USA;
| | - Mikhail Doubrovin
- Department of Radiology, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, USA; (A.M.); (P.C.); (M.A.D.); (S.J.); (S.I.); (S.O.); (J.C.); (M.D.)
| | - Glenn J. Lesser
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA;
| | - Francesca Zanderigo
- Department of Psychiatry, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, USA;
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032, USA; (A.M.); (P.C.); (M.A.D.); (S.J.); (S.I.); (S.O.); (J.C.); (M.D.)
| |
Collapse
|
65
|
Peretti DE, Renken RJ, Reesink FE, de Jong BM, De Deyn PP, Dierckx RAJO, Doorduin J, Boellaard R, Vállez García D. Feasibility of pharmacokinetic parametric PET images in scaled subprofile modelling using principal component analysis. NEUROIMAGE-CLINICAL 2021; 30:102625. [PMID: 33756179 PMCID: PMC8020472 DOI: 10.1016/j.nicl.2021.102625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/05/2021] [Indexed: 11/30/2022]
Abstract
Scaled subprofile model using principal component analysis (SSM/PCA) is a multivariate analysis technique used, mainly in [18F]-2-fluoro-2-deoxy-d-glucose (FDG) PET studies, for the generation of disease-specific metabolic patterns (DP) that may aid with the classification of subjects with neurological disorders, like Alzheimer’s disease (AD). The aim of this study was to explore the feasibility of using quantitative parametric images for this type of analysis, with dynamic [11C]-labelled Pittsburgh Compound B (PIB) PET data as an example. Therefore, 15 AD patients and 15 healthy control subjects were included in an SSM/PCA analysis to generate four AD-DPs using relative cerebral blood flow (R1), binding potential (BPND) and SUVR images derived from dynamic PIB and static FDG-PET studies. Furthermore, 49 new subjects with a variety of neurodegenerative cognitive disorders were tested against these DPs. The AD-DP was characterized by a reduction in the frontal, parietal, and temporal lobes voxel values for R1 and SUVR-FDG DPs; and by a general increase of values in cortical areas for BPND and SUVR-PIB DPs. In conclusion, the results suggest that the combination of parametric images derived from a single dynamic scan might be a good alternative for subject classification instead of using 2 independent PET studies.
Collapse
Affiliation(s)
- Débora E Peretti
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, The Netherlands.
| | - Remco J Renken
- University of Groningen, University Medical Center Groningen, Cognitive Neuroscience Centre, Department of Biomedical Sciences of Cell & Systems, The Netherlands
| | - Fransje E Reesink
- University of Groningen, University Medical Center Groningen, Department of Neurology, Alzheimer Research Centre, The Netherlands
| | - Bauke M de Jong
- University of Groningen, University Medical Center Groningen, Department of Neurology, Alzheimer Research Centre, The Netherlands
| | - Peter P De Deyn
- University of Groningen, University Medical Center Groningen, Department of Neurology, Alzheimer Research Centre, The Netherlands; University of Antwerp, Institute Born-Bunge, Laboratory of Neurochemistry and Behaviour, Belgium
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, The Netherlands
| | - Janine Doorduin
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, The Netherlands
| | - Ronald Boellaard
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, The Netherlands
| | - David Vállez García
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, The Netherlands
| |
Collapse
|
66
|
Abstract
Positron emission tomography (PET) is a non-invasive imaging technology employed to describe metabolic, physiological, and biochemical processes in vivo. These include receptor availability, metabolic changes, neurotransmitter release, and alterations of gene expression in the brain. Since the introduction of dedicated small-animal PET systems along with the development of many novel PET imaging probes, the number of PET studies using rats and mice in basic biomedical research tremendously increased over the last decade. This article reviews challenges and advances of quantitative rodent brain imaging to make the readers aware of its physical limitations, as well as to inspire them for its potential applications in preclinical research. In the first section, we briefly discuss the limitations of small-animal PET systems in terms of spatial resolution and sensitivity and point to possible improvements in detector development. In addition, different acquisition and post-processing methods used in rodent PET studies are summarized. We further discuss factors influencing the test-retest variability in small-animal PET studies, e.g., different receptor quantification methodologies which have been mainly translated from human to rodent receptor studies to determine the binding potential and changes of receptor availability and radioligand affinity. We further review different kinetic modeling approaches to obtain quantitative binding data in rodents and PET studies focusing on the quantification of endogenous neurotransmitter release using pharmacological interventions. While several studies have focused on the dopamine system due to the availability of several PET tracers which are sensitive to dopamine release, other neurotransmitter systems have become more and more into focus and are described in this review, as well. We further provide an overview of latest genome engineering technologies, including the CRISPR/Cas9 and DREADD systems that may advance our understanding of brain disorders and function and how imaging has been successfully applied to animal models of human brain disorders. Finally, we review the strengths and opportunities of simultaneous PET/magnetic resonance imaging systems to study drug-receptor interactions and challenges for the translation of PET results from bench to bedside.
Collapse
|
67
|
Molecular and Functional Imaging in Central Nervous System Drug Development. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00084-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
68
|
Thomsen MB, Ferreira SA, Schacht AC, Jacobsen J, Simonsen M, Betzer C, Jensen PH, Brooks DJ, Landau AM, Romero-Ramos M. PET imaging reveals early and progressive dopaminergic deficits after intra-striatal injection of preformed alpha-synuclein fibrils in rats. Neurobiol Dis 2020; 149:105229. [PMID: 33352233 DOI: 10.1016/j.nbd.2020.105229] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/24/2022] Open
Abstract
Alpha-synuclein (a-syn) can aggregate and form toxic oligomers and insoluble fibrils which are the main component of Lewy bodies. Intra-neuronal Lewy bodies are a major pathological characteristic of Parkinson's disease (PD). These fibrillar structures can act as seeds and accelerate the aggregation of monomeric a-syn. Indeed, recent studies show that injection of preformed a-syn fibrils (PFF) into the rodent brain can induce aggregation of the endogenous monomeric a-syn resulting in neuronal dysfunction and eventual cell death. We injected 8 μg of murine a-syn PFF, or soluble monomeric a-syn into the right striatum of rats. The animals were monitored behaviourally using the cylinder test, which measures paw asymmetry, and the corridor task that measures lateralized sensorimotor response to sugar treats. In vivo PET imaging was performed after 6, 13 and 22 weeks using [11C]DTBZ, a marker of the vesicular monoamine 2 transporter (VMAT2), and after 15 and 22 weeks using [11C]UCB-J, a marker of synaptic SV2A protein in nerve terminals. Histology was performed at the three time points using antibodies against dopaminergic markers, aggregated a-syn, and MHCII to evaluate the immune response. While the a-syn PFF injection caused only mild behavioural changes, [11C]DTBZ PET showed a significant and progressive decrease of VMAT2 binding in the ipsilateral striatum. This was accompanied by a small progressive decrease in [11C]UCB-J binding in the same area. In addition, our histological analysis revealed a gradual spread of misfolded a-syn pathology in areas anatomically connected to striatum that became bilateral with time. The striatal a-syn PFF injection resulted in a progressive unilateral degeneration of dopamine terminals, and an early and sustained presence of MHCII positive ramified microglia in the ipsilateral striatum and substantia nigra. Our study shows that striatal injections of a-syn fibrils induce progressive pathological synaptic dysfunction prior to cell death that can be detected in vivo with PET. We confirm that intrastriatal injection of a-syn PFFs provides a model of progressive a-syn pathology with loss of dopaminergic and synaptic function accompanied by neuroinflammation, as found in human PD.
Collapse
Affiliation(s)
- Majken B Thomsen
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Sara A Ferreira
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Anna C Schacht
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Jan Jacobsen
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Mette Simonsen
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Cristine Betzer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Poul H Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - David J Brooks
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark; Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Anne M Landau
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark; Translational Neuropsychiatry Unit, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
69
|
Helo Y, Searle GE, Borghese F, Abraham S, Saleem A. Specificity of translocator protein-targeted positron emission tomography in inflammatory joint disease. EJNMMI Res 2020; 10:147. [PMID: 33284369 PMCID: PMC7721924 DOI: 10.1186/s13550-020-00736-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/23/2020] [Indexed: 12/02/2022] Open
Abstract
Objective Expression of the translocator protein (TSPO) on inflammatory cells has facilitated imaging of synovitis with TSPO-targeted positron emission tomography (PET). We aimed to quantitatively assess the specificity of the second-generation TSPO PET radioligand, [11C]PBR28, and to generate simplified PET protocols in patients with inflammatory joint disease (IJD) in this pilot study. Methods Three IJD patients (two rheumatoid arthritis and one osteoarthritis) with knee involvement underwent dynamic [11C]PBR28-PET scans before and after administration of 90 mg of oral emapunil (XBD-173), a TSPO ligand the same day. Radial arterial blood sampling was performed throughout the scan, and total radioactivity and radioactive metabolites were obtained. A semi-automated method was used to generate regions of interest. Standardized uptake value (SUV) and SUV ratio corrected for activity in bone and blood between 50 and 70 min (SUVr50–70 bone, SUVr50–70 blood, respectively) and PET volume of distribution (VT) of the radioligand were calculated. Results A mean [11C]PBR28 radioactivity of 378 (range 362–389) MBq was administered. A significant decrease (p < 0.05) in VT, SUVr50–70 bone and SUVr50–70 blood observed after oral emapunil confirmed the TSPO specificity of [11C]PBR28. A decrease in SUV was not observed in the post-block scan. Conclusion [11C]PBR28 is TSPO-specific radioligand in IJD patients. Simplified PET protocols with static PET acquisition can be used in the management and evaluation of novel therapeutics that target TSPO overexpressing cells.
Collapse
Affiliation(s)
- Yusuf Helo
- Invicro, A Konica Minolta Company, Burlington Danes Building, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Graham E Searle
- Invicro, A Konica Minolta Company, Burlington Danes Building, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Federica Borghese
- Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Sonya Abraham
- Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Azeem Saleem
- Invicro, A Konica Minolta Company, Burlington Danes Building, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK. .,Hull York Medical School, Allam Medical Building, University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| |
Collapse
|
70
|
Papanastasiou G, Rodrigues MA, Wang C, Heurling K, Lucatelli C, Salman RAS, Wardlaw JM, van Beek EJR, Thompson G. Pharmacokinetic modelling for the simultaneous assessment of perfusion and 18F-flutemetamol uptake in cerebral amyloid angiopathy using a reduced PET-MR acquisition time: Proof of concept. Neuroimage 2020; 225:117482. [PMID: 33157265 DOI: 10.1016/j.neuroimage.2020.117482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/24/2020] [Accepted: 10/19/2020] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Cerebral amyloid angiopathy (CAA) is a cerebral small vessel disease associated with perivascular β-amyloid deposition. CAA is also associated with strokes due to lobar intracerebral haemorrhage (ICH). 18F-flutemetamol amyloid ligand PET may improve the early detection of CAA. We performed pharmacokinetic modelling using both full (0-30, 90-120 min) and reduced (30 min) 18F-flutemetamol PET-MR acquisitions, to investigate regional cerebral perfusion and amyloid deposition in ICH patients. METHODS Dynamic18F-flutemetamol PET-MR was performed in a pilot cohort of sixteen ICH participants; eight lobar ICH cases with probable CAA and eight deep ICH patients. A model-based input function (mIF) method was developed for compartmental modelling. mIF 1-tissue (1-TC) and 2-tissue (2-TC) compartmental modelling, reference tissue models and standardized uptake value ratios were assessed in the setting of probable CAA detection. RESULTS The mIF 1-TC model detected perfusion deficits and 18F-flutemetamol uptake in cases with probable CAA versus deep ICH patients, in both full and reduced PET acquisition time (all P < 0.05). In the reduced PET acquisition, mIF 1-TC modelling reached the highest sensitivity and specificity in detecting perfusion deficits (0.87, 0.77) and 18F-flutemetamol uptake (0.83, 0.71) in cases with probable CAA. Overall, 52 and 48 out of the 64 brain areas with 18F-flutemetamol-determined amyloid deposition showed reduced perfusion for 1-TC and 2-TC models, respectively. CONCLUSION Pharmacokinetic (1-TC) modelling using a 30 min PET-MR time frame detected impaired haemodynamics and increased amyloid load in probable CAA. Perfusion deficits and amyloid burden co-existed within cases with CAA, demonstrating a distinct imaging pattern which may have merit in elucidating the pathophysiological process of CAA.
Collapse
Affiliation(s)
- Giorgos Papanastasiou
- Edinburgh Imaging Facility, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | - Mark A Rodrigues
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Chengjia Wang
- Edinburgh Imaging Facility, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | | | - Christophe Lucatelli
- Edinburgh Imaging Facility, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | | | - Joanna M Wardlaw
- Edinburgh Imaging Facility, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Edwin J R van Beek
- Edinburgh Imaging Facility, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Gerard Thompson
- Edinburgh Imaging Facility, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
71
|
Hu J, Panin V, Smith AM, Spottiswoode B, Shah V, CA von Gall C, Baker M, Howe W, Kehren F, Casey M, Bendriem B. Design and Implementation of Automated Clinical Whole Body Parametric PET With Continuous Bed Motion. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2020. [DOI: 10.1109/trpms.2020.2994316] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
72
|
Wang B, Ruan D, Liu H. Noninvasive Estimation of Macro-Parameters by Deep Learning. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2020. [DOI: 10.1109/trpms.2020.2979017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
73
|
Holmes SE, Gallezot JD, Davis MT, DellaGioia N, Matuskey D, Nabulsi N, Krystal JH, Javitch JA, DeLorenzo C, Carson RE, Esterlis I. Measuring the effects of ketamine on mGluR5 using [ 18F]FPEB and PET. J Cereb Blood Flow Metab 2020; 40:2254-2264. [PMID: 31744389 PMCID: PMC7585925 DOI: 10.1177/0271678x19886316] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/23/2019] [Accepted: 10/03/2019] [Indexed: 01/21/2023]
Abstract
The metabotropic glutamate receptor 5 (mGluR5) is a promising treatment target for psychiatric disorders due to its modulatory effects on glutamate transmission. Using [11C]ABP688, we previously showed that the rapidly acting antidepressant ketamine decreases mGluR5 availability. The mGluR5 radioligand [18F]FPEB offers key advantages over [11C]ABP688; however, its suitability for drug challenge studies is unknown. We evaluated whether [18F]FPEB can be used to capture ketamine-induced effects on mGluR5. Seven healthy subjects participated in three [18F]FPEB scans: a baseline, a same-day post-ketamine, and a 24-h post-ketamine scan. The outcome measure was VT/fP, obtained using a two-tissue compartment model and a metabolite-corrected arterial input function. Dissociative symptoms, heart rate and blood pressure increased following ketamine infusion. [18F]FPEB VT/fP decreased by 9% across the cortex after ketamine infusion, with minimal difference between baseline and 24-h scans. Compared to our previous work using [11C]ABP688, the magnitude of the ketamine-induced change in mGluR5 was smaller using [18F]FPEB; however, effect sizes were similar for the same-day post-ketamine vs. baseline scan (Cohen's d = 0.75 for [18F]FPEB and 0.88 for [11C]ABP688). [18F]FPEB is therefore able to capture some of the effects of ketamine on mGluR5, but [11C]ABP688 appears to be more suitable in drug challenge paradigms designed to probe glutamate transmission.
Collapse
Affiliation(s)
- Sophie E Holmes
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | | | - Margaret T Davis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Nicole DellaGioia
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - David Matuskey
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Nabeel Nabulsi
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - John H Krystal
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- U.S. Department of Veteran Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Jonathan A Javitch
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
- Departments of Psychiatry and Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Christine DeLorenzo
- Department of Psychiatry and Behavioral Health, Stony Brook University, New York, NY, USA
| | - Richard E Carson
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
74
|
Wang G, Rahmim A, Gunn RN. PET Parametric Imaging: Past, Present, and Future. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2020; 4:663-675. [PMID: 33763624 PMCID: PMC7983029 DOI: 10.1109/trpms.2020.3025086] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Positron emission tomography (PET) is actively used in a diverse range of applications in oncology, cardiology, and neurology. The use of PET in the clinical setting focuses on static (single time frame) imaging at a specific time-point post radiotracer injection and is typically considered as semi-quantitative; e.g. standardized uptake value (SUV) measures. In contrast, dynamic PET imaging requires increased acquisition times but has the advantage that it measures the full spatiotemporal distribution of a radiotracer and, in combination with tracer kinetic modeling, enables the generation of multiparametric images that more directly quantify underlying biological parameters of interest, such as blood flow, glucose metabolism, and receptor binding. Parametric images have the potential for improved detection and for more accurate and earlier therapeutic response assessment. Parametric imaging with dynamic PET has witnessed extensive research in the past four decades. In this paper, we provide an overview of past and present activities and discuss emerging opportunities in the field of parametric imaging for the future.
Collapse
Affiliation(s)
- Guobao Wang
- Department of Radiology, University of California Davis Health, Sacramento, CA 95817, USA
| | - Arman Rahmim
- University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
75
|
Heeman F, Hendriks J, Lopes Alves I, Ossenkoppele R, Tolboom N, van Berckel BNM, Lammertsma AA, Yaqub M. [ 11C]PIB amyloid quantification: effect of reference region selection. EJNMMI Res 2020; 10:123. [PMID: 33074395 PMCID: PMC7572969 DOI: 10.1186/s13550-020-00714-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/24/2020] [Indexed: 11/24/2022] Open
Abstract
Background The standard reference region (RR) for amyloid-beta (Aβ) PET studies is the cerebellar grey matter (GMCB), while alternative RRs have mostly been utilized without prior validation against the gold standard. This study compared five commonly used RRs to gold standard plasma input-based quantification using the GMCB. Methods Thirteen subjects from a test–retest (TRT) study and 30 from a longitudinal study were retrospectively included (total: 17 Alzheimer’s disease, 13 mild cognitive impairment, 13 controls). Dynamic [11C]PiB PET (90 min) and T1-weighted MR scans were co-registered and time–activity curves were extracted for cortical target regions and the following RRs: GMCB, whole cerebellum (WCB), white matter brainstem/pons (WMBS), whole brainstem (WBS) and eroded subcortical white matter (WMES). A two-tissue reversible plasma input model (2T4k_Vb) with GMCB as RR, reference Logan and the simplified reference tissue model were used to derive distribution volume ratios (DVRs), and standardized uptake value (SUV) ratios were calculated for 40–60 min and 60–90 min intervals. Parameter variability was evaluated using TRT scans, and correlations and agreements with the gold standard (DVR from 2T4k_Vb with GMCB RR) were also assessed. Next, longitudinal changes in SUVs (both intervals) were assessed for each RR. Finally, the ability to discriminate between visually Aβ positive and Aβ negative scans was assessed. Results All RRs yielded stable TRT performance (max 5.1% variability), with WCB consistently showing lower variability. All approaches were able to discriminate between Aβ positive and Aβ negative scans, with highest effect sizes obtained for GMCB (range − 0.9 to − 0.7), followed by WCB (range − 0.8 to − 0.6). Furthermore, all approaches provided good correlations with the gold standard (r ≥ 0.78), while the highest bias (as assessed by the regression slope) was observed using WMES (range slope 0.52–0.67), followed by WBS (range slope 0.58–0.92) and WMBS (range slope 0.62–0.91). Finally, RR SUVs were stable across a period of 2.6 years for all except WBS and WMBS RRs (60–90 min interval). Conclusions GMCB and WCB are considered the best RRs for quantifying amyloid burden using [11C]PiB PET.
Collapse
Affiliation(s)
- Fiona Heeman
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Janine Hendriks
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Isadora Lopes Alves
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Rik Ossenkoppele
- Neurology and Alzheimer Center, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.,Clinical Memory Research Unit, Lund University, Malmö, Sweden
| | - Nelleke Tolboom
- Imaging Division, Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | | |
Collapse
|
76
|
van de Stadt EA, Yaqub M, Lammertsma AA, Poot AJ, Schober PR, Schuit RC, Smit EF, Bahce I, Hendrikse NH. Quantification of [ 18F]afatinib using PET/CT in NSCLC patients: a feasibility study. EJNMMI Res 2020; 10:97. [PMID: 32804306 PMCID: PMC7431492 DOI: 10.1186/s13550-020-00684-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/31/2020] [Indexed: 12/09/2022] Open
Abstract
Introduction Only a subgroup of non-small cell lung cancer (NSCLC) patients benefit from treatment using epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI) such as afatinib. Tumour uptake of [18F]afatinib using positron emission tomography (PET) may identify those patients that respond to afatinib therapy. Therefore, the aim of this study was to find the optimal tracer kinetic model for quantification of [18F]afatinib uptake in NSCLC tumours. Methods [18F]Afatinib PET scans were performed in 10 NSCLC patients. The first patient was scanned for the purpose of dosimetry. Subsequent patients underwent a 20-min dynamic [15O]H2O PET scan (370 MBq) followed by a dynamic [18F]afatinib PET scan (342 ± 24 MBq) of 60 or 90 min. Using the Akaike information criterion (AIC), three pharmacokinetic plasma input models were evaluated with both metabolite-corrected sampler-based input and image-derived (IDIF) input functions in combination with discrete blood samples. Correlation analysis of arterial on-line sampling versus IDIF was performed. In addition, perfusion dependency and simplified measures were assessed. Results Ten patients were included. The injected activity of [18F]afatinib was 341 ± 37 MBq. Fifteen tumours could be identified in the field of view of the scanner. Based on AIC, tumour kinetics were best described using an irreversible two-tissue compartment model and a metabolite-corrected sampler-based input function (Akaike 50%). Correlation of plasma-based input functions with metabolite-corrected IDIF was very strong (r2 = 0.93). The preferred simplified uptake parameter was the tumour-to-blood ratio over the 60- to 90-min time interval (TBR60–90). Tumour uptake of [18F]afatinib was independent of perfusion. Conclusion The preferred pharmacokinetic model for quantifying [18F]afatinib uptake in NSCLC tumours was the 2T3K_vb model. TBR60–90 showed excellent correlation with this model and is the best candidate simplified method. Trial registration https://eudract.ema.europa.eu/ nr 2012-002849-38
Collapse
Affiliation(s)
- E A van de Stadt
- Department of Pulmonology, Amsterdam UMC location VUmc, Amsterdam, the Netherlands. .,Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.
| | - M Yaqub
- Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - A A Lammertsma
- Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - A J Poot
- Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - P R Schober
- Department of Anesthesiology, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - R C Schuit
- Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - E F Smit
- Department of Pulmonology, Amsterdam UMC location VUmc, Amsterdam, the Netherlands.,Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - I Bahce
- Department of Pulmonology, Amsterdam UMC location VUmc, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - N H Hendrikse
- Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUmc, Amsterdam, the Netherlands.,Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| |
Collapse
|
77
|
Jonasson M, Nordeman P, Eriksson J, Wilking H, Wikström J, Takahashi K, Niwa T, Hosoya T, Watanabe Y, Antoni G, Sundström Poromaa I, Lubberink M, Comasco E. Quantification of aromatase binding in the female human brain using [
11
C]cetrozole positron emission tomography. J Neurosci Res 2020; 98:2208-2218. [DOI: 10.1002/jnr.24707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/08/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022]
Affiliation(s)
- My Jonasson
- Department of Surgical Sciences Uppsala University Uppsala Sweden
- Department of Medical Physics Uppsala University Hospital Uppsala Sweden
| | - Patrik Nordeman
- Department of Medicinal Chemistry Uppsala University Uppsala Sweden
- PET centre Uppsala University Hospital Uppsala Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry Uppsala University Uppsala Sweden
- PET centre Uppsala University Hospital Uppsala Sweden
| | | | - Johan Wikström
- Department of Surgical Sciences Uppsala University Uppsala Sweden
| | - Kayo Takahashi
- RIKEN Center for Biosystems Dynamics Research Kobe Japan
| | - Takashi Niwa
- RIKEN Center for Biosystems Dynamics Research Kobe Japan
| | - Takamitsu Hosoya
- RIKEN Center for Biosystems Dynamics Research Kobe Japan
- Institute of Biomaterials and Bioengineering Tokyo Medical and Dental University Tokyo Japan
| | | | - Gunnar Antoni
- Department of Medicinal Chemistry Uppsala University Uppsala Sweden
- PET centre Uppsala University Hospital Uppsala Sweden
| | | | - Mark Lubberink
- Department of Surgical Sciences Uppsala University Uppsala Sweden
- Department of Medical Physics Uppsala University Hospital Uppsala Sweden
| | - Erika Comasco
- Department of Neuroscience Science for Life Laboratory Uppsala University Uppsala Sweden
| |
Collapse
|
78
|
Abstract
Neuroimaging with positron emission tomography (PET) is the most powerful tool for understanding pharmacology, neurochemistry, and pathology in the living human brain. This technology combines high-resolution scanners to measure radioactivity throughout the human body with specific, targeted radioactive molecules, which allow measurements of a myriad of biological processes in vivo. While PET brain imaging has been active for almost 40 years, the pace of development for neuroimaging tools, known as radiotracers, and for quantitative analytical techniques has increased dramatically over the past decade. Accordingly, the fundamental questions that can be addressed with PET have expanded in basic neurobiology, psychiatry, neurology, and related therapeutic development. In this review, we introduce the field of human PET neuroimaging, some of its conceptual underpinnings, and motivating questions. We highlight some of the more recent advances in radiotracer development, quantitative modeling, and applications of PET to the study of the human brain.
Collapse
Affiliation(s)
- Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA;
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| |
Collapse
|
79
|
Tjerkaski J, Cervenka S, Farde L, Matheson GJ. Kinfitr - an open-source tool for reproducible PET modelling: validation and evaluation of test-retest reliability. EJNMMI Res 2020; 10:77. [PMID: 32642865 PMCID: PMC7343683 DOI: 10.1186/s13550-020-00664-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/25/2020] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND In positron emission tomography (PET) imaging, binding is typically estimated by fitting pharmacokinetic models to the series of measurements of radioactivity in the target tissue following intravenous injection of a radioligand. However, there are multiple different models to choose from and numerous analytical decisions that must be made when modelling PET data. Therefore, it is important that analysis tools be adapted to the specific circumstances, and that analyses be documented in a transparent manner. Kinfitr, written in the open-source programming language R, is a tool developed for flexible and reproducible kinetic modelling of PET data, i.e. performing all steps using code which can be publicly shared in analysis notebooks. In this study, we compared outcomes obtained using kinfitr with those obtained using PMOD: a widely used commercial tool. RESULTS Using previously collected test-retest data obtained with four different radioligands, a total of six different kinetic models were fitted to time-activity curves derived from different brain regions. We observed good correspondence between the two kinetic modelling tools both for binding estimates and for microparameters. Likewise, no substantial differences were observed in the test-retest reliability estimates between the two tools. CONCLUSIONS In summary, we showed excellent agreement between the open-source R package kinfitr, and the widely used commercial application PMOD. We, therefore, conclude that kinfitr is a valid and reliable tool for kinetic modelling of PET data.
Collapse
Affiliation(s)
- Jonathan Tjerkaski
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden.
| | - Simon Cervenka
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Lars Farde
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Granville James Matheson
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| |
Collapse
|
80
|
Cui J, Qin Z, Chen S, Chen Y, Liu H. Structure and Tracer Kinetics-Driven Dynamic PET Reconstruction. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2020. [DOI: 10.1109/trpms.2019.2947860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
81
|
Thomsen MB, Schacht AC, Alstrup AKO, Jacobsen J, Lillethorup TP, Bærentzen SL, Noer O, Orlowski D, Elfving B, Müller HK, Brooks DJ, Landau AM. Preclinical PET Studies of [ 11C]UCB-J Binding in Minipig Brain. Mol Imaging Biol 2020; 22:1290-1300. [PMID: 32514885 DOI: 10.1007/s11307-020-01506-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE Loss of neuronal synapse function is associated with a number of brain disorders. The [11C]UCB-J positron emission tomography (PET) tracer allows for in vivo examination of synaptic density, as it binds to synaptic vesicle glycoprotein 2A (SV2A) expressed in presynaptic terminals. Here, we characterise [11C]UCB-J imaging in Göttingen minipigs. PROCEDURES Using PET imaging, we examined tracer specificity and compared kinetic models. We explored the use of a standard blood curve and centrum semiovale white matter as a reference region. We compared in vivo [11C]UCB-J PET imaging to in vitro autoradiography, Western blotting and real-time quantitative polymerase chain reaction. RESULTS The uptake kinetics of [11C]UCB-J could be described using a 1-tissue compartment model and blocking of SV2A availability with levetiracetam showed dose-dependent specific binding. Population-based blood curves resulted in reliable [11C]UCB-J binding estimates, while it was not possible to use centrum semiovale white matter as a non-specific reference region. Brain [11C]UCB-J PET signals correlated well with [3H]UCB-J autoradiography and SV2A protein levels. CONCLUSIONS [11C]UCB-J PET is a valid in vivo marker of synaptic density in the minipig brain, with binding values close to those reported for humans. Minipig models of disease could be valuable for investigating the efficacy of putative neuroprotective agents for preserving synaptic function in future non-invasive, longitudinal studies.
Collapse
Affiliation(s)
- Majken Borup Thomsen
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 165, J, 8200, Aarhus N, Denmark
| | - Anna Christina Schacht
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 165, J, 8200, Aarhus N, Denmark
| | - Aage Kristian Olsen Alstrup
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 165, J, 8200, Aarhus N, Denmark
| | - Jan Jacobsen
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 165, J, 8200, Aarhus N, Denmark
| | - Thea Pinholt Lillethorup
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 165, J, 8200, Aarhus N, Denmark
| | - Simone Larsen Bærentzen
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 165, J, 8200, Aarhus N, Denmark.,Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ove Noer
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 165, J, 8200, Aarhus N, Denmark
| | - Dariusz Orlowski
- Center for Experimental Neuroscience (CENSE), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Heidi Kaastrup Müller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - David J Brooks
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 165, J, 8200, Aarhus N, Denmark.,Institute of Translational and Clinical Research, Faculty of Medical Science, Newcastle upon Tyne University, Newcastle upon Tyne, UK
| | - Anne M Landau
- Department of Nuclear Medicine and PET, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 165, J, 8200, Aarhus N, Denmark. .,Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
82
|
Wilson H, Pagano G, de Natale ER, Mansur A, Caminiti SP, Polychronis S, Middleton LT, Price G, Schmidt KF, Gunn RN, Rabiner EA, Politis M. Mitochondrial Complex 1, Sigma 1, and Synaptic Vesicle 2A in Early Drug-Naive Parkinson's Disease. Mov Disord 2020; 35:1416-1427. [PMID: 32347983 DOI: 10.1002/mds.28064] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/27/2020] [Accepted: 03/30/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Dysfunction of mitochondrial energy generation may contribute to neurodegeneration, leading to synaptic loss in Parkinson's disease (PD). The objective of this study was to find cross-sectional and longitudinal changes in PET markers of synaptic vesicle protein 2A, sigma 1 receptor, and mitochondrial complex 1 in drug-naive PD patients. METHODS Twelve early drug-naive PD patients and 16 healthy controls underwent a 3-Tesla MRI and PET imaging to quantify volume of distribution of [11 C]UCB-J, [11 C]SA-4503, and [18 F]BCPP-EF for synaptic vesicle protein 2A, sigma 1 receptor, and mitochondrial complex 1, respectively. Nine PD patients completed approximately 1-year follow-up assessments. RESULTS Reduced [11 C]UCB-J volume of distribution in the caudate, putamen, thalamus, brain stem, and dorsal raphe and across cortical regions was observed in drug-naive PD patients compared with healthy controls. [11 C]UCB-J volume of distribution was reduced in the locus coeruleus and substantia nigra but did not reach statistical significance. No significant differences were found in [11 C]SA-4503 and [18 F]BCPP-EF volume of distribution in PD compared with healthy controls. Lower brain stem [11 C]UCB-J volume of distribution correlated with Movement Disorder Society Unified Parkinson's Disease Rating Scale part III and total scores. No significant longitudinal changes were identified in PD patients at follow-up compared with baseline. CONCLUSIONS Our findings represent the first in vivo evidence of mitochondrial, endoplasmic reticulum, and synaptic dysfunction in drug-naive PD patients. Synaptic dysfunction likely occurs early in disease pathophysiology and has relevance to symptomatology. Mitochondrial complex 1 and sigma 1 receptor pathology warrants further investigations in PD. Studies in larger cohorts with longer follow-up will determine the validity of these PET markers to track disease progression. © 2020 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Heather Wilson
- Neurodegeneration Imaging Group, University of Exeter Medical School, London, UK.,Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Gennaro Pagano
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Edoardo Rosario de Natale
- Neurodegeneration Imaging Group, University of Exeter Medical School, London, UK.,Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Ayla Mansur
- Invicro, Centre for Imaging Sciences, Hammersmith Hospital, London, UK.,Division of Brain Sciences, Department of Medicine, Imperial College London, UK
| | - Silvia Paola Caminiti
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Sotirios Polychronis
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Lefkos T Middleton
- School of Public Health, Imperial College London, UK.,Public Health Directorate, Imperial College NHS Healthcare Trust, London, UK.,MINDMAPS Consortium, London, UK
| | - Geraint Price
- School of Public Health, Imperial College London, UK.,MINDMAPS Consortium, London, UK
| | | | - Roger N Gunn
- Invicro, Centre for Imaging Sciences, Hammersmith Hospital, London, UK.,Division of Brain Sciences, Department of Medicine, Imperial College London, UK.,MINDMAPS Consortium, London, UK
| | - Eugenii A Rabiner
- Invicro, Centre for Imaging Sciences, Hammersmith Hospital, London, UK.,MINDMAPS Consortium, London, UK.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Marios Politis
- Neurodegeneration Imaging Group, University of Exeter Medical School, London, UK.,Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK.,MINDMAPS Consortium, London, UK
| |
Collapse
|
83
|
Robust nonlinear parameter estimation in tracer kinetic analysis using infinity norm regularization and particle swarm optimization. Phys Med 2020; 72:60-72. [PMID: 32200299 DOI: 10.1016/j.ejmp.2020.03.013] [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] [Received: 11/12/2019] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 11/23/2022] Open
Abstract
In positron emission tomography (PET) studies, the voxel-wise calculation of individual rate constants describing the tracer kinetics is quite challenging because of the nonlinear relationship between the rate constants and PET data and the high noise level in voxel data. Based on preliminary simulations using a standard two-tissue compartment model, we can hypothesize that it is possible to reduce errors in the rate constant estimates when constraining the overestimation of the larger of two exponents in the model equation. We thus propose a novel approach based on infinity-norm regularization for limiting this exponent. Owing to the non-smooth cost function of this regularization scheme, which prevents the use of conventional Jacobian-based optimization methods, we examined a proximal gradient algorithm and the particle swarm optimization (PSO) through a simulation study. Because it exploits multiple initial values, the PSO method shows much better convergence than the proximal gradient algorithm, which is susceptible to the initial values. In the implementation of PSO, the use of a Gamma distribution to govern random movements was shown to improve the convergence rate and stability compared to a uniform distribution. Consequently, Gamma-based PSO with regularization was shown to outperform all other methods tested, including the conventional basis function method and Levenberg-Marquardt algorithm, in terms of its statistical properties.
Collapse
|
84
|
Kuttner S, Wickstrøm KK, Kalda G, Dorraji SE, Martin-Armas M, Oteiza A, Jenssen R, Fenton K, Sundset R, Axelsson J. Machine learning derived input-function in a dynamic 18F-FDG PET study of mice. Biomed Phys Eng Express 2020; 6:015020. [DOI: 10.1088/2057-1976/ab6496] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
85
|
Grimmer T, Shi K, Diehl-Schmid J, Natale B, Drzezga A, Förster S, Förstl H, Schwaiger M, Yakushev I, Wester HJ, Kurz A, Yousefi BH. 18F-FIBT may expand PET for β-amyloid imaging in neurodegenerative diseases. Mol Psychiatry 2020; 25:2608-2619. [PMID: 30120417 PMCID: PMC7515824 DOI: 10.1038/s41380-018-0203-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 06/22/2018] [Accepted: 07/18/2018] [Indexed: 11/09/2022]
Abstract
18F-FIBT, 2-(p-Methylaminophenyl)-7-(2-[18F]fluoroethoxy)imidazo-[2,1-b]benzothiazole, is a new selective PET tracer under clinical investigation to specifically image β-amyloid depositions (Aβ) in humans in-vivo that binds to Aβ with excellent affinity (Kd 0.7 ± 0.2) and high selectivity over tau and α-synuclein aggregates (Ki > 1000 nM). We aimed to characterize 18F-FIBT in a series of patients with different clinical-pathophysiological phenotypes and to compare its binding characteristics to the reference compound PiB. Six patients (mild late-onset and moderate early-onset AD dementia, mild cognitive impairment due to AD, intermediate likelihood, mild behavioral variant of frontotemporal dementia, subjective memory impairment without evidence of neurodegeneration, and mild dementia due to Posterior Cortical Atrophy) underwent PET imaging with 18F-FIBT on PET/MR. With the guidance of MRI, PET images were corrected for partial volume effect, time-activity curves (TACs) of regions of interest (ROIs) were extracted, and non-displaceable binding potentials (BPnd), standardized uptake value ratios (SUVR), and distribution volume ratio (DVR) were compared. Specific binding was detected in the cases with evidence of the AD pathophysiological process visualized in images of BPnd, DVR and SUVR, consistently with patterns of different tracers in previous studies. SUVR showed the highest correlation with clinical severity. The previous preclinical characterization and the results of this case series suggest the clinical usefulness of FIBT as a selective and highly affine next-generation 18F-labeled tracer for amyloid-imaging with excellent pharmacokinetics in the diagnosis of neurodegenerative diseases. The results compare well to the gold standard PiB and hence support further investigation in larger human samples.
Collapse
Affiliation(s)
- Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Kuangyu Shi
- grid.15474.330000 0004 0477 2438Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Str. 22, 81675 Munich, Germany ,grid.5734.50000 0001 0726 5157Department of Nuclear Medicine, University of Bern, Freiburgstr. 10, 3010 Bern, Switzerland
| | - Janine Diehl-Schmid
- grid.15474.330000 0004 0477 2438Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Bianca Natale
- grid.15474.330000 0004 0477 2438Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Alexander Drzezga
- grid.411097.a0000 0000 8852 305XDepartment of Nuclear Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Stefan Förster
- grid.15474.330000 0004 0477 2438Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Hans Förstl
- grid.15474.330000 0004 0477 2438Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Markus Schwaiger
- grid.15474.330000 0004 0477 2438Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Igor Yakushev
- grid.15474.330000 0004 0477 2438Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Hans-Jürgen Wester
- grid.6936.a0000000123222966Pharmaceutical Radiochemistry, Technische Universität München, Walther-Meißner-Str. 3, 85748 Garching, Germany
| | - Alexander Kurz
- grid.15474.330000 0004 0477 2438Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Behrooz Hooshyar Yousefi
- grid.15474.330000 0004 0477 2438Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| |
Collapse
|
86
|
Gallezot JD, Lu Y, Naganawa M, Carson RE. Parametric Imaging With PET and SPECT. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2020. [DOI: 10.1109/trpms.2019.2908633] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
87
|
Vass LD, Lee S, Wilson FJ, Fisk M, Cheriyan J, Wilkinson I. Reproducibility of compartmental modelling of 18F-FDG PET/CT to evaluate lung inflammation. EJNMMI Phys 2019; 6:26. [PMID: 31844995 PMCID: PMC6915187 DOI: 10.1186/s40658-019-0265-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 11/25/2019] [Indexed: 11/18/2022] Open
Abstract
Introduction Compartmental modelling is an established method of quantifying 18F-FDG uptake; however, only recently has it been applied to evaluate pulmonary inflammation. Implementation of compartmental models remains challenging in the lung, partly due to the low signal-to-noise ratio compared to other organs and the lack of standardisation. Good reproducibility is a key requirement of an imaging biomarker which has yet to be demonstrated in pulmonary compartmental models of 18F-FDG; in this paper, we address this unmet need. Methods Retrospective subject data were obtained from the EVOLVE observational study: Ten COPD patients (age =66±9; 8M/2F), 10 α1ATD patients (age =63±8; 7M/3F) and 10 healthy volunteers (age =68±8; 9M/1F) never smokers. PET and CT images were co-registered, and whole lung regions were extracted from CT using an automated algorithm; the descending aorta was defined using a manually drawn region. Subsequent stages of the compartmental analysis were performed by two independent operators using (i) a MIAKATTM based pipeline and (ii) an in-house developed pipeline. We evaluated the metabolic rate constant of 18F-FDG (Kim) and the fractional blood volume (Vb); Bland-Altman plots were used to compare the results. Further, we adjusted the in-house pipeline to identify the salient features in the analysis which may help improve the standardisation of this technique in the lung. Results The initial agreement on a subject level was poor: Bland-Altman coefficients of reproducibility for Kim and Vb were 0.0031 and 0.047 respectively. However, the effect size between the groups (i.e. COPD, α1ATD and healthy subjects) was similar using either pipeline. We identified the key drivers of this difference using an incremental approach: ROI methodology, modelling of the IDIF and time delay estimation. Adjustment of these factors led to improved Bland-Altman coefficients of reproducibility of 0.0015 and 0.027 for Kim and Vb respectively. Conclusions Despite similar methodology, differences in implementation can lead to disparate results in the outcome parameters. When reporting the outcomes of lung compartmental modelling, we recommend the inclusion of the details of ROI methodology, input function fitting and time delay estimation to improve reproducibility.
Collapse
Affiliation(s)
- Laurence D Vass
- Experimental Medicine and Immunotherapeutics, Department of Medicine, Addenbrookes Hospital, Cambridge, UK.
| | | | | | - Marie Fisk
- Experimental Medicine and Immunotherapeutics, Department of Medicine, Addenbrookes Hospital, Cambridge, UK.,Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Joseph Cheriyan
- Experimental Medicine and Immunotherapeutics, Department of Medicine, Addenbrookes Hospital, Cambridge, UK.,GSK R &D, Brentford, UK.,Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Ian Wilkinson
- Experimental Medicine and Immunotherapeutics, Department of Medicine, Addenbrookes Hospital, Cambridge, UK.,Cambridge University Hospitals NHS Trust, Cambridge, UK
| |
Collapse
|
88
|
Direct Parametric Maps Estimation from Dynamic PET Data: An Iterated Conditional Modes Approach. JOURNAL OF HEALTHCARE ENGINEERING 2019; 2018:5942873. [PMID: 30073047 PMCID: PMC6057340 DOI: 10.1155/2018/5942873] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/27/2018] [Accepted: 05/08/2018] [Indexed: 11/24/2022]
Abstract
We propose and test a novel approach for direct parametric image reconstruction of dynamic PET data. We present a theoretical description of the problem of PET direct parametric maps estimation as an inference problem, from a probabilistic point of view, and we derive a simple iterative algorithm, based on the Iterated Conditional Mode (ICM) framework, which exploits the simplicity of a two-step optimization and the efficiency of an analytic method for estimating kinetic parameters from a nonlinear compartmental model. The resulting method is general enough to be flexible to an arbitrary choice of the kinetic model, and unlike many other solutions, it is capable to deal with nonlinear compartmental models without the need for linearization. We tested its performance on a two-tissue compartment model, including an analytical solution to the kinetic parameters evaluation, based on an auxiliary parameter set, with the aim of reducing computation errors and approximations. The new method is tested on simulated and clinical data. Simulation analysis led to the conclusion that the proposed algorithm gives a good estimation of the kinetic parameters in any noise condition. Furthermore, the application of the proposed method to clinical data gave promising results for further studies.
Collapse
|
89
|
Liu H, Wu J, Sun J, Wu T, Fazzone‐Chettiar R, Thorn S, Sinusas AJ, Liu Y. A robust segmentation method with triple‐factor non‐negative matrix factorization for myocardial blood flow quantification from dynamic
82
Rb positron emission tomography. Med Phys 2019; 46:5002-5013. [DOI: 10.1002/mp.13783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 03/25/2019] [Accepted: 08/13/2019] [Indexed: 12/24/2022] Open
Affiliation(s)
- Hui Liu
- Department of Internal Medicine (Cardiology) Yale University New Haven CT 06520USA
| | - Jing Wu
- Department of Radiology and Biomedical Imaging Yale University New Haven CT 06520USA
| | - Jing‐Yi Sun
- Department of Biomedical Imaging and Radiological Sciences National Yang‐Ming University Taipei 11221Taiwan
| | - Tung‐Hsin Wu
- Department of Biomedical Imaging and Radiological Sciences National Yang‐Ming University Taipei 11221Taiwan
| | | | - Stephanie Thorn
- Department of Internal Medicine (Cardiology) Yale University New Haven CT 06520USA
| | - Albert J. Sinusas
- Department of Internal Medicine (Cardiology) Yale University New Haven CT 06520USA
| | - Yi‐Hwa Liu
- Department of Internal Medicine (Cardiology) Yale University New Haven CT 06520USA
- Department of Biomedical Imaging and Radiological Sciences National Yang‐Ming University Taipei 11221Taiwan
- Nuclear Cardiology, Heart and Vascular Center Yale New Haven Hospital New Haven CT 06520USA
- Department of Biomedical Engineering Chung Yuan Christian University Taoyuan 32023Taiwan
| |
Collapse
|
90
|
Heurling K, Smith R, Strandberg OT, Schain M, Ohlsson T, Hansson O, Schöll M. Regional times to equilibria and their impact on semi-quantification of [ 18F]AV-1451 uptake. J Cereb Blood Flow Metab 2019; 39:2223-2232. [PMID: 30073880 PMCID: PMC6827127 DOI: 10.1177/0271678x18791430] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/08/2018] [Accepted: 06/20/2018] [Indexed: 11/17/2022]
Abstract
The semi-quantitative estimate standardised uptake value ratios (SUVR) correlate well with specific binding of the tracer expressed as distribution volume ratios (DVR) for the tau positron emission tomography tracer [18F]AV-1451 uptake and are therefore widely used as proxy for tracer binding. With regard to tracer kinetic modelling, there exists a time point when SUVR deviates minimally from DVR, occurring when the specific binding reaches a transient equilibrium. Here, we have investigated whether the time to equilibrium affects the agreement between SUVR and DVR across different brain regions. We show that the time required to reach equilibrium differs across brain regions, resulting in region-specific biases. However, even though the 80-100 min post-injection time window did not show the smallest bias numerically, the disagreement between SUVR and DVR varied least between regions during this time. In conclusion, our findings suggest a regional component to the bias of SUVR related to the time to transient equilibrium of the specific binding. [18F]AV-1451 uptake should consequently be interpreted with some caution when compared across brain regions using this method of quantification. The commonly used time window 80-100 min post-injection shows the most consistent bias across regions and is recommended for semi-quantification of [18F]AV-1451.
Collapse
Affiliation(s)
- Kerstin Heurling
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Ruben Smith
- Department of Neurology, Lund University, Skåne University Hospital, Lund, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund/Malmö, Sweden
| | - Olof T Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund/Malmö, Sweden
| | - Martin Schain
- Neurobiology Research Unit, Copenhagen University Hospital, Copenhagen, Denmark
| | - Tomas Ohlsson
- Department of Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund/Malmö, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund/Malmö, Sweden
| |
Collapse
|
91
|
Iozzo P, Guzzardi MA. Imaging of brain glucose uptake by PET in obesity and cognitive dysfunction: life-course perspective. Endocr Connect 2019; 8:R169-R183. [PMID: 31590145 PMCID: PMC6865363 DOI: 10.1530/ec-19-0348] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022]
Abstract
The prevalence of obesity has reached epidemic proportions and keeps growing. Obesity seems implicated in the pathogenesis of cognitive dysfunction, Alzheimer's disease and dementia, and vice versa. Growing scientific efforts are being devoted to the identification of central mechanisms underlying the frequent association between obesity and cognitive dysfunction. Glucose brain handling undergoes dynamic changes during the life-course, suggesting that its alterations might precede and contribute to degenerative changes or signaling abnormalities. Imaging of the glucose analog 18F-labeled fluorodeoxyglucose (18FDG) by positron emission tomography (PET) is the gold-standard for the assessment of cerebral glucose metabolism in vivo. This review summarizes the current literature addressing brain glucose uptake measured by PET imaging, and the effect of insulin on brain metabolism, trying to embrace a life-course vision in the identification of patterns that may explain (and contribute to) the frequent association between obesity and cognitive dysfunction. The current evidence supports that brain hypermetabolism and brain insulin resistance occur in selected high-risk conditions as a transient phenomenon, eventually evolving toward normal or low values during life or disease progression. Associative studies suggest that brain hypermetabolism predicts low BDNF levels, hepatic and whole body insulin resistance, food desire and an unfavorable balance between anticipated reward from food and cognitive inhibitory control. Emerging mechanistic links involve the microbiota and the metabolome, which correlate with brain metabolism and cognition, deserving attention as potential future prevention targets.
Collapse
Affiliation(s)
- Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
- Correspondence should be addressed to P Iozzo:
| | | |
Collapse
|
92
|
Golla SS, Verfaillie SC, Boellaard R, Adriaanse SM, Zwan MD, Schuit RC, Timmers T, Groot C, Schober P, Scheltens P, van der Flier WM, Windhorst AD, van Berckel BN, Lammertsma AA. Quantification of [ 18F]florbetapir: A test-retest tracer kinetic modelling study. J Cereb Blood Flow Metab 2019; 39:2172-2180. [PMID: 29897009 PMCID: PMC6826855 DOI: 10.1177/0271678x18783628] [Citation(s) in RCA: 18] [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] [Indexed: 11/29/2022]
Abstract
Accumulation of amyloid beta can be visualized using [18F]florbetapir positron emission tomography. The aim of this study was to identify the optimal model for quantifying [18F]florbetapir uptake and to assess test-retest reliability of corresponding outcome measures. Eight Alzheimer's disease patients (age: 67 ± 6 years, Mini-Mental State Examination (MMSE): 23 ± 3) and eight controls (age: 63 ± 4 years, MMSE: 30 ± 0) were included. Ninety-minute dynamic positron emission tomography scans, together with arterial blood sampling, were acquired immediately following a bolus injection of 294 ± 32 MBq [18F]florbetapir. Several plasma input models and the simplified reference tissue model (SRTM) were evaluated. The Akaike information criterion was used to identify the preferred kinetic model. Compared to controls, Alzheimer's disease patients had lower MMSE scores and evidence for cortical Aβ pathology. A reversible two-tissue compartment model with fitted blood volume fraction (2T4k_VB) was the preferred model for describing [18F]florbetapir kinetics. SRTM-derived non-displaceable binding potential (BPND) correlated well (r2 = 0.83, slope = 0.86) with plasma input-derived distribution volume ratio. Test-retest reliability for plasma input-derived distribution volume ratio, SRTM-derived BPND and SUVr(50-70) were r = 0.88, r = 0.91 and r = 0.86, respectively. In vivo kinetics of [18F]florbetapir could best be described by a reversible two-tissue compartmental model and [18F]florbetapir BPND can be reliably estimated using an SRTM.
Collapse
Affiliation(s)
- Sandeep Sv Golla
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Sander Cj Verfaillie
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands.,Department of Neurology & Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands.,Department of Nuclear Medicine & Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sofie M Adriaanse
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Marissa D Zwan
- Department of Neurology & Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Robert C Schuit
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Tessa Timmers
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands.,Department of Neurology & Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Colin Groot
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Patrick Schober
- Department of Anaesthesiology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Philip Scheltens
- Department of Neurology & Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Wiesje M van der Flier
- Department of Neurology & Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands.,Department of Epidemiology & Biostatistics, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Bart Nm van Berckel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
93
|
Analysis of hypoxia in human glioblastoma tumors with dynamic 18F-FMISO PET imaging. AUSTRALASIAN PHYSICAL & ENGINEERING SCIENCES IN MEDICINE 2019; 42:981-993. [PMID: 31520369 DOI: 10.1007/s13246-019-00797-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/27/2019] [Accepted: 08/31/2019] [Indexed: 02/07/2023]
Abstract
Gliomas are the most common type of primary brain tumors and are classified as grade IV. Necrosis and hypoxia are essential diagnostic features which result in poor prognosis of gliomas. The aim of this study was to report quantitative temporal analyses aiming at determining the hypoxic regions in glioblastoma multiforme and to suggest an optimal time for the clinical single scan of hypoxia. Nine subjects were imaged with PET and 18F-FMISO in dynamic mode for 15 min followed with static scans at 2, 3 and 4 h post-injection. Spectral analysis, tumor-to-blood ratio (TBR) and tumor-to-normal tissue ratio (TNR) were used to delimit perfused and hypoxic tumor regions. TBR and TNR images were further scaled by thresholding at 1.2, 1.4, 2 and 2.5 levels. The images showed a varying tumor volume with time. TBR produced broader images of the tumor than TNR considering the same thresholds on intensity. Spectral analysis reliably determined hypoxia with different degrees of perfusion. By comparing TBR and TNR with spectral analysis images, weak to moderate correlation coefficients were found for most thresholding values and imaging times (range: 0 to 0.69). Hypoxic volume (HV) estimated from the net uptake rate (Ki) were changing among imaging times. The minimum HV changes were found between 3 h and 4 h, confirming that after 3 h, there was a very low exchange of 81F-FMISO between blood and tumor. On the other hand, hypoxia started to dominate the perfused tissue at 90 min, suggesting this time is suitable for a single scan acquisition irrespective of tumor status being highly hypoxic or perfused. At this time, TBR and TNR were respectively found in the nine subjects as 1.72 ± 0.22 and 1.74 ± 0.19.
Collapse
|
94
|
The application of positron emission tomography (PET) imaging in CNS drug development. Brain Imaging Behav 2019; 13:354-365. [PMID: 30259405 DOI: 10.1007/s11682-018-9967-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
As drug discovery and development in Neuroscience push beyond symptom management to disease modification, neuroimaging becomes a key area of translational research that enables measurements of the presence of drugs and downstream physiological consequences of drug action within the living brain. As such, neuroimaging can be used to help optimize decision-making processes throughout the various phases of drug development. Positron Emission Tomography (PET) is a functional imaging technique that allows the quantification and visualization of biochemical processes, by monitoring the time dependent distribution of molecules labelled with short-lived positron-emitting isotopes. This review focuses on the application of PET to support CNS drug development, particularly in the early clinical phases, by allowing us to measure tissue exposure, target engagement, and pharmacological activity. We will also discuss the application of PET imaging as tools to image the pathological hallmarks of disease and evaluate the potential disease-modifying effect of candidate drugs in slowing disease progression.
Collapse
|
95
|
Synthesis and in vivo evaluation of [ 18F]UCB-J for PET imaging of synaptic vesicle glycoprotein 2A (SV2A). Eur J Nucl Med Mol Imaging 2019; 46:1952-1965. [PMID: 31175396 DOI: 10.1007/s00259-019-04357-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/02/2019] [Indexed: 02/05/2023]
Abstract
PURPOSE Synaptic abnormalities have been implicated in a variety of neuropsychiatric disorders, including epilepsy, Alzheimer's disease, and schizophrenia. Hence, PET imaging of the synaptic vesicle glycoprotein 2A (SV2A) may be a valuable in vivo biomarker for neurologic and psychiatric diseases. We previously developed [11C]UCB-J, a PET radiotracer with high affinity and selectivity toward SV2A; however, the short radioactive half-life (20 min for 11C) places some limitations on its broader application. Herein, we report the first synthesis of the longer-lived 18F-labeled counterpart (half-life: 110 min), [18F]UCB-J, and its evaluation in nonhuman primates. METHODS [18F]UCB-J was synthesized from the iodonium precursors. PET imaging experiments with [18F]UCB-J were conducted in rhesus monkeys to assess the pharmacokinetic and in vivo binding properties. Arterial samples were taken for analysis of radioactive metabolites and generation of input functions. Regional time-activity curves were analyzed using the one-tissue compartment model to derive regional distribution volumes and binding potentials for comparison with [11C]UCB-J. RESULTS [18F]UCB-J was prepared in high radiochemical and enantiomeric purity, but low radiochemical yield. Evaluation in nonhuman primates indicated that the radiotracer displayed pharmacokinetic and imaging characteristics similar to those of [11C]UCB-J, with moderate metabolism rate, high brain uptake, fast and reversible binding kinetics, and high specific binding signals. CONCLUSION We have accomplished the first synthesis of the novel SV2A radiotracer [18F]UCB-J. [18F]UCB-J is demonstrated to be an excellent imaging agent and may prove to be useful for imaging and quantification of SV2A expression, and synaptic density, in humans.
Collapse
|
96
|
Baum E, Zhang W, Li S, Cai Z, Holden D, Huang Y. A Novel 18F-Labeled Radioligand for Positron Emission Tomography Imaging of 11β-Hydroxysteroid Dehydrogenase (11β-HSD1): Synthesis and Preliminary Evaluation in Nonhuman Primates. ACS Chem Neurosci 2019; 10:2450-2458. [PMID: 30689943 DOI: 10.1021/acschemneuro.8b00715] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) catalyzes the conversion of cortisone to cortisol and controls a key pathway in the regulation of stress. Studies have implicated 11β-HSD1 in metabolic diseases including type 2 diabetes and obesity, as well as stress-related disorders and neurodegenerative diseases, such as depression and Alzheimer's disease (AD). We have previously developed [11C]AS2471907 as a PET radiotracer to image 11β-HSD1 in the brain of nonhuman primates and humans. However, the radiosynthesis of [11C]AS2471907 was unreliable and low-yielding. Here, we report the development of the 18F-labeled version [18F]AS2471907, including the synthesis of two iodonium ylide precursors and the optimization of 18F-radiosynthesis. Preliminary PET experiments, composed of a baseline scan of [18F]AS2471907 and a blocking scan with the reversible 11β-HSD1 inhibitor ASP3662 (0.3 mg/kg), was also conducted in a rhesus monkey to verify the pharmacokinetics of [18F]AS2471907 and its specific binding in the brain. The iodonium ylide precursors were prepared in a seven-step synthetic route with an optimized overall yield of ∼2%. [18F]AS2471907 was synthesized in good radiochemical purity, with the ortho regioisomer of iodonium ylide providing greater radiochemical yield as compared with the para regioisomer. In monkey brain, [18F]AS2471907 displayed high uptake and heterogeneous distribution, while administration of the 11β-HSD1 inhibitor ASP3662 significantly reduced radiotracer uptake, thus demonstrating the binding specificity of [18F]AS2471907. Given the longer half-life of F-18 and feasibility for central production and distribution, [18F]AS2471907 holds great promise to be a valuable PET radiotracer to image 11β-HSD1 in the brain.
Collapse
Affiliation(s)
- Evan Baum
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, Connecticut 06520-8048, United States
| | - Wenjie Zhang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Songye Li
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, Connecticut 06520-8048, United States
| | - Zhengxin Cai
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, Connecticut 06520-8048, United States
| | - Daniel Holden
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, Connecticut 06520-8048, United States
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, Connecticut 06520-8048, United States
| |
Collapse
|
97
|
Saillant A, Armstrong I, Shah V, Zuehlsdorff S, Hayden C, Declerck J, Saint K, Memmott M, Jenkinson M, Chappell MA. Assessing Reliability of Myocardial Blood Flow After Motion Correction With Dynamic PET Using a Bayesian Framework. IEEE TRANSACTIONS ON MEDICAL IMAGING 2019; 38:1216-1226. [PMID: 30452353 DOI: 10.1109/tmi.2018.2881992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The estimation of myocardial blood flow (MBF) in dynamic PET can be biased by many different processes. A major source of error, particularly in clinical applications, is patient motion. Patient motion, or gross motion, creates displacements between different PET frames as well as between the PET frames and the CT-derived attenuation map, leading to errors in MBF calculation from voxel time series. Motion correction techniques are challenging to evaluate quantitatively and the impact on MBF reliability is not fully understood. Most metrics, such as signal-to-noise ratio (SNR), are characteristic of static images, and are not specific to motion correction in dynamic data. This study presents a new approach of estimating motion correction quality in dynamic cardiac PET imaging. It relies on calculating a MBF surrogate, K1 , along with the uncertainty on the parameter. This technique exploits a Bayesian framework, representing the kinetic parameters as a probability distribution, from which the uncertainty measures can be extracted. If the uncertainty extracted is high, the parameter studied is considered to have high variability - or low confidence - and vice versa. The robustness of the framework is evaluated on simulated time activity curves to ensure that the uncertainties are consistently estimated at the multiple levels of noise. Our framework is applied on 40 patient datasets, divided in 4 motion magnitude categories. Experienced observers manually realigned clinical datasets with 3D translations to correct for motion. K1 uncertainties were compared before and after correction. A reduction of uncertainty after motion correction of up to 60% demonstrates the benefit of motion correction in dynamic PET and as well as provides evidence of the usefulness of the new method presented.
Collapse
|
98
|
Heterogeneous drug tissue binding in brain regions of rats, Alzheimer's patients and controls: impact on translational drug development. Sci Rep 2019; 9:5308. [PMID: 30926941 PMCID: PMC6440985 DOI: 10.1038/s41598-019-41828-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 03/18/2019] [Indexed: 01/08/2023] Open
Abstract
For preclinical and clinical assessment of therapeutically relevant unbound, free, brain concentrations, the pharmacokinetic parameter fraction of unbound drug in brain (fu,brain) is commonly used to compensate total drug concentrations for nonspecific brain tissue binding (BTB). As, homogenous BTB is assumed between species and in health and disease, rat BTB is routinely used. The impact of Alzheimer’s disease (AD) on drug BTB in brain regions of interest (ROI), i.e., fu,brain,ROI, is yet unclear. This study for the first time provides insight into regional drug BTB and the validity of employing rat fu,brain,ROI as a surrogate of human BTB, by investigating five marketed drugs in post-mortem tissue from AD patients (n = 6) and age-matched controls (n = 6). Heterogeneous drug BTB was observed in all within group comparisons independent of disease and species. The findings oppose the assumption of uniform BTB, highlighting the need of case-by-case evaluation of fu,brain,ROI in translational CNS research.
Collapse
|
99
|
Heeman F, Yaqub M, Lopes Alves I, Heurling K, Berkhof J, Gispert JD, Bullich S, Foley C, Lammertsma AA. Optimized dual-time-window protocols for quantitative [ 18F]flutemetamol and [ 18F]florbetaben PET studies. EJNMMI Res 2019; 9:32. [PMID: 30919133 PMCID: PMC6437225 DOI: 10.1186/s13550-019-0499-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/11/2019] [Indexed: 12/12/2022] Open
Abstract
Background A long dynamic scanning protocol may be required to accurately measure longitudinal changes in amyloid load. However, such a protocol results in a lower patient comfort and scanning efficiency compared to static scans. A compromise can be achieved by implementing dual-time-window protocols. This study aimed to optimize these protocols for quantitative [18F]flutemetamol and [18F]florbetaben studies. Methods Rate constants for subjects across the Alzheimer’s disease spectrum (i.e., non-displaceable binding potential (BPND) in the range 0.02–0.77 and 0.02–1.04 for [18F]flutemetamol and [18F]florbetaben, respectively) were established based on clinical [18F]flutemetamol (N = 6) and [18F]florbetaben (N = 20) data, and used to simulate tissue time-activity curves (TACs) of 110 min using a reference tissue and plasma input model. Next, noise was added (N = 50) and data points corresponding to different intervals were removed from the TACs, ranging from 0 (i.e., 90–90 = full-kinetic curve) to 80 (i.e., 10–90) minutes, creating a dual-time-window. Resulting TACs were fitted using the simplified reference tissue method (SRTM) to estimate the BPND, outliers (≥ 1.5 × BPND max) were removed and the bias was assessed using the distribution volume ratio (DVR = BPND + 1). To this end, acceptability curves, which display the fraction of data below a certain bias threshold, were generated and the area under those curves were calculated. Results [18F]Flutemetamol and [18F]florbetaben data demonstrated an increased bias in amyloid estimate for larger intervals and higher noise levels. An acceptable bias (≤ 3.1%) in DVR could be obtained with all except the 10–90 and 20–90-min intervals. Furthermore, a reduced fraction of acceptable data and most outliers were present for these two largest intervals (maximum percentage outliers 48 and 32 for [18F]flutemetamol and [18F]florbetaben, respectively). Conclusions The length of the interval inversely correlates with the accuracy of the BPND estimates. Consequently, a dual-time-window protocol of 0–30 and 90–110 min (=maximum of 60 min interval) allows for accurate estimation of BPND values for both tracers. [18F]flutemetamol: EudraCT 2007-000784-19, registered 8 February 2007, [18F]florbetaben: EudraCT 2006-003882-15, registered 2006. Electronic supplementary material The online version of this article (10.1186/s13550-019-0499-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fiona Heeman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, Netherlands.
| | - Maqsood Yaqub
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, Netherlands
| | - Isadora Lopes Alves
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, Netherlands
| | - Kerstin Heurling
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Johannes Berkhof
- Amsterdam UMC, Vrije Universiteit Amsterdam, Epidemiology and Biostatistics, De Boelelaan, 1117, Amsterdam, Netherlands
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Carrer de Wellington, 30, 08005, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain.,Universitat Pompeu Fabra, Plaça de la Mercè, 10, 08002, Barcelona, Spain
| | - Santiago Bullich
- Life Molecular Imaging GmbH, Tegeler Str. 7, 13353, Berlin, Germany
| | | | - Adriaan A Lammertsma
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, Netherlands
| | | |
Collapse
|
100
|
Li S, Cai Z, Wu X, Holden D, Pracitto R, Kapinos M, Gao H, Labaree D, Nabulsi N, Carson RE, Huang Y. Synthesis and in Vivo Evaluation of a Novel PET Radiotracer for Imaging of Synaptic Vesicle Glycoprotein 2A (SV2A) in Nonhuman Primates. ACS Chem Neurosci 2019; 10:1544-1554. [PMID: 30396272 PMCID: PMC6810685 DOI: 10.1021/acschemneuro.8b00526] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Structural disruption and alterations of synapses are associated with many brain disorders including Alzheimer's disease, epilepsy, depression, and schizophrenia. We have previously developed the PET radiotracer 11C-UCB-J for imaging and quantification of synaptic vesicle glycoprotein 2A (SV2A) and synaptic density in nonhuman primates and humans. Here we report the synthesis of a novel radiotracer 18F-SDM-8 and its in vivo evaluation in rhesus monkeys. The in vitro binding assay of SDM-8 showed high SV2A binding affinity ( Ki = 0.58 nM). 18F-SDM-8 was prepared in high molar activity (241.7 MBq/nmol) and radiochemical purity (>98%). In the brain, 18F-SDM-8 displayed very high uptake with peak standardized uptake value (SVU) greater than 8 and fast and reversible kinetics. A displacement study with levetiracetam and blocking studies with UCB-J and levetiracetam demonstrated its binding reversibility and specificity toward SV2A. Regional binding potential values were calculated and ranged from 0.8 in the brainstem to 4.5 in the cingulate cortex. By comparing to 11C-UCB-J, 18F-SDM-8 displayed the same attractive imaging properties: very high brain uptake, appropriate tissue kinetics, and high levels of specific binding. Given the longer half-life of F-18 and the feasibility for central production and multisite distribution, 18F-SDM-8 holds promise as an excellent radiotracer for SV2A and as a biomarker for synaptic density measurement in neurodegenerative diseases and psychiatric disorders.
Collapse
Affiliation(s)
- Songye Li
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Zhengxin Cai
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Xiaoai Wu
- Department of Nuclear Medicine, West China Hospital, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Daniel Holden
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Richard Pracitto
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Michael Kapinos
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Hong Gao
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - David Labaree
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Nabeel Nabulsi
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Richard E. Carson
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| |
Collapse
|