1
|
Terry-Lorenzo R, Albrecht D, Crouch S, Wong R, Loewen G, Giri N, Skor H, Lin K, Sandiego CM, Pajonas M, Rabiner EA, Gunn RN, Russell DS, Haubenberger D. Quantifying VMAT2 target occupancy at effective valbenazine doses and comparing to a novel VMAT2 inhibitor: a translational PET study. Neuropsychopharmacology 2025; 50:1093-1101. [PMID: 39757283 PMCID: PMC12089418 DOI: 10.1038/s41386-024-02046-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025]
Abstract
Positron emission tomography (PET) is frequently used to obtain target occupancy (%TO) of central nervous system (CNS) drug candidates during clinical development. Obtaining %TO with PET can be particularly powerful when the %TO associated with efficacy is known for a protein target. Using the radiotracer [18F]AV-133, the relationship between plasma concentration (PK) and %TO of NBI-750142, an experimental inhibitor of the vesicular monoamine transporter type 2 (VMAT2) was obtained in both nonhuman primate (NHP) and human. This work established [18F]AV-133 PET as capable of providing a VMAT2 inhibitor PK-%TO relationship that translated from NHP to human. To establish the VMAT2%TO benchmark, PET was performed in NHP with NBI-98782, the main active metabolite of valbenazine, and this PK-%TO relationship was used to estimate VMAT2%TO at NBI-98782 exposures associated with valbenazine therapeutic effects in the treatment of tardive dyskinesia (TD). This work defined 85-90% as the VMAT2%TO achieved by exposures associated with daily dosing with 80 mg valbenazine, a dosing regimen known to exhibit a large effect size in the treatment of TD and in the treatment of chorea associated with Huntington's Disease. NBI-750142 was estimated to achieve 36-78% VMAT2 target occupancy at acceptable doses, indicating potential inferiority in conferring clinical benefit compared to valbenazine. It is recommended that the %TO benchmark of valbenazine derived from [18F]AV-133 PET serve as a gold standard biomarker to evaluate novel VMAT2 inhibitors undergoing clinical development.
Collapse
Affiliation(s)
| | | | | | - Richard Wong
- Neurocrine Biosciences, Inc., San Diego, CA, USA
| | | | - Nagdeep Giri
- Neurocrine Biosciences, Inc., San Diego, CA, USA
| | - Heather Skor
- Neurocrine Biosciences, Inc., San Diego, CA, USA
| | - Kelly Lin
- Neurocrine Biosciences, Inc., San Diego, CA, USA
| | | | | | | | | | | | | |
Collapse
|
2
|
Ai X, Huang B, Chen F, Shi L, Li B, Wang S, Liu Q. RED: Residual estimation diffusion for low-dose PET sinogram reconstruction. Med Image Anal 2025; 102:103558. [PMID: 40121810 DOI: 10.1016/j.media.2025.103558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/08/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Recent advances in diffusion models have demonstrated exceptional performance in generative tasks across various fields. In positron emission tomography (PET), the reduction in tracer dose leads to information loss in sinograms. Using diffusion models to reconstruct missing information can improve imaging quality. Traditional diffusion models effectively use Gaussian noise for image reconstructions. However, in low-dose PET reconstruction, Gaussian noise can worsen the already sparse data by introducing artifacts and inconsistencies. To address this issue, we propose a diffusion model named residual estimation diffusion (RED). From the perspective of diffusion mechanism, RED uses the residual between sinograms to replace Gaussian noise in diffusion process, respectively sets the low-dose and full-dose sinograms as the starting point and endpoint of reconstruction. This mechanism helps preserve the original information in the low-dose sinogram, thereby enhancing reconstruction reliability. From the perspective of data consistency, RED introduces a drift correction strategy to reduce accumulated prediction errors during the reverse process. Calibrating the intermediate results of reverse iterations helps maintain the data consistency and enhances the stability of reconstruction process. In the experiments, RED achieved the best performance across all metrics. Specifically, the PSNR metric showed improvements of 2.75, 5.45, and 8.08 dB in DRF4, 20, and 100 respectively, compared to traditional methods. The code is available at: https://github.com/yqx7150/RED.
Collapse
Affiliation(s)
- Xingyu Ai
- School of Information Engineering, Nanchang University, Nanchang 330031, China
| | - Bin Huang
- School of Mathematics and Computer Sciences, Nanchang University, Nanchang 330031, China
| | - Fang Chen
- School of Information Engineering, Nanchang University, Nanchang 330031, China
| | - Liu Shi
- School of Information Engineering, Nanchang University, Nanchang 330031, China
| | - Binxuan Li
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230000, China
| | - Shaoyu Wang
- School of Information Engineering, Nanchang University, Nanchang 330031, China
| | - Qiegen Liu
- School of Information Engineering, Nanchang University, Nanchang 330031, China
| |
Collapse
|
3
|
Mooney JJ, Jacobs S, Lefebvre ÉA, Cosgrove GP, Clark A, Turner SM, Decaris M, Barnes CN, Jurek M, Williams B, Duan H, Kimura R, Rizzo G, Searle G, Wardak M, Guo HH. Bexotegrast Shows Dose-Dependent Integrin α vβ 6 Receptor Occupancy in Lungs of Participants with Idiopathic Pulmonary Fibrosis: A Phase 2, Open-Label Clinical Trial. Ann Am Thorac Soc 2025; 22:350-358. [PMID: 39499805 PMCID: PMC11892667 DOI: 10.1513/annalsats.202409-969oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/05/2024] [Indexed: 11/07/2024] Open
Abstract
Rationale: Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive disease characterized by dyspnea and loss of lung function. Transforming growth factor-β (TGF-β) activation mediated by αv integrins is central to the pathogenesis of IPF. Bexotegrast (PLN-74809) is an oral, once-daily, dual-selective inhibitor of αvβ6 and αvβ1 integrins under investigation for the treatment of IPF. Positron emission tomography (PET) using an αvβ6-specific PET tracer could confirm target engagement of bexotegrast in the lungs of participants with IPF. Objectives: This Phase 2 study evaluated αvβ6 receptor occupancy in the lung as assessed by changes from baseline in αvβ6 PET tracer uptake, after single-dose administration of bexotegrast to participants with IPF. Methods: In this open-label, single-center study, adults with IPF received up to two single doses of bexotegrast, ranging from 60 to 320 mg with or without background IPF therapy (pirfenidone or nintedanib). At baseline and approximately 4 hours after each orally administered bexotegrast dose, a 60-minute dynamic PET-computed tomography scan was conducted after administration of an αvβ6-specific PET probe ([18F]FP-R01-MG-F2). αvβ6 receptor occupancy by bexotegrast was estimated from the changes in PET tracer uptake after bexotegrast administration. Pharmacokinetics, safety, and tolerability of bexotegrast were also assessed. Results: Eight participants completed the study. Total and unbound plasma bexotegrast concentrations increased in a dose-dependent manner, and regional PET volume of distribution values decreased in a dose- and concentration-dependent manner. The data for volume of distribution fit a simple saturation model, producing an unbound bexotegrast half maximal effective concentration estimate of 3.32 ng/ml. Estimated maximum receptor occupancy was 35%, 53%, 71%, 88%, and 92% after single 60-, 80-, 120-, 240-, and 320-mg doses of bexotegrast, respectively. No treatment-emergent adverse events related to bexotegrast were reported. Conclusions: Dose and concentration-dependent αvβ6 receptor occupancy by bexotegrast was observed by PET imaging, supporting once-daily 160- to 320-mg dosing to evaluate efficacy in clinical trials of IPF. Clinical trial registered with www.clinicaltrials.gov (NCT04072315).
Collapse
Affiliation(s)
- Joshua J. Mooney
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, and
| | - Susan Jacobs
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, and
| | | | | | - Annie Clark
- Pliant Therapeutics, Inc., South San Francisco, California; and
| | - Scott M. Turner
- Pliant Therapeutics, Inc., South San Francisco, California; and
| | - Martin Decaris
- Pliant Therapeutics, Inc., South San Francisco, California; and
| | - Chris N. Barnes
- Pliant Therapeutics, Inc., South San Francisco, California; and
| | - Marzena Jurek
- Pliant Therapeutics, Inc., South San Francisco, California; and
| | - Brittney Williams
- Nuclear Medicine and Molecular Imaging Division, Department of Radiology, Stanford University, Stanford, California
| | - Heying Duan
- Nuclear Medicine and Molecular Imaging Division, Department of Radiology, Stanford University, Stanford, California
| | - Richard Kimura
- Nuclear Medicine and Molecular Imaging Division, Department of Radiology, Stanford University, Stanford, California
| | | | | | - Mirwais Wardak
- Nuclear Medicine and Molecular Imaging Division, Department of Radiology, Stanford University, Stanford, California
| | - H. Henry Guo
- Nuclear Medicine and Molecular Imaging Division, Department of Radiology, Stanford University, Stanford, California
| |
Collapse
|
4
|
Brender B, Burki L, Jeon J, Ng A, Yussefian N, Uribe C, Murrell E, Boileau I, Desmond KL, Narciso L. External delay and dispersion correction of automatically sampled arterial blood with dual flow rates. Biomed Phys Eng Express 2025; 11:025021. [PMID: 39854771 DOI: 10.1088/2057-1976/adae13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/24/2025] [Indexed: 01/26/2025]
Abstract
Objective. Arterial sampling for PET imaging often involves continuously measuring the radiotracer activity concentration in blood using an automatic blood sampling system (ABSS). We proposed and validated an external delay and dispersion correction procedure needed when a change in flow rate occurs during data acquisition. We also measured the external dispersion constant of [11C]CURB, [18F]FDG, [18F]FEPPA, and [18F]SynVesT-1.Approach. External delay and dispersion constants were measured for the flow rates of 350, 300, 180, and 150 ml h-1, using 1-minute-long rectangular inputs (n= 10;18F-fluoride in saline). Resulting constants were used to validate the external delay and dispersion corrections (n= 6;18F-fluoride in saline; flow rate change: 350 to 150 ml h-1and 300 to 180 ml h-1); constants were modelled to transition linearly between flow rates. Corrected curves were assessed using the percent area-under-the-curve (AUC) ratio and a modified model selection criterion (MSC). External delay and dispersion constants were measured for various radiotracers using a blood analog (i.e., similar viscoelastic properties).Main results. ABSS outputs were successfully corrected for external delay and dispersion using our proposed method accounting for a change in flow rate. AUC ratio reduced from ∼10% for the uncorrected 350-150 ml h-1output (∼6% for the 300-180 ml h-1) to < 1% after correction when compared to true input (511 keV energy window); approx. 5-fold increase in MSC. Assuming an internal dispersion constant of 5 s, the dispersion constant (internal + external) for [11C]CURB, [18F]FDG, [18F]FEPPA, and [18F]SynVesT-1 was 13, 9, 16, and 10 s, respectively.Significance. This study presented an external delay and dispersion correction procedure needed when a change in flow rate occurs during ABSS data acquisition. Additionally, this is the first study to measure the external delay and dispersion constants using a blood analog solution, a suitable alternative to blood when estimating external dispersion.
Collapse
Affiliation(s)
- Benjamin Brender
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
| | - Lubna Burki
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Josefina Jeon
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Alvina Ng
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Nikta Yussefian
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Carme Uribe
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Emily Murrell
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Isabelle Boileau
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kimberly L Desmond
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Lucas Narciso
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Kuang X, Li B, Lyu T, Xue Y, Huang H, Xie Q, Zhu W. PET image reconstruction using weighted nuclear norm maximization and deep learning prior. Phys Med Biol 2024; 69:215023. [PMID: 39374634 DOI: 10.1088/1361-6560/ad841d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/07/2024] [Indexed: 10/09/2024]
Abstract
The ill-posed Positron emission tomography (PET) reconstruction problem usually results in limited resolution and significant noise. Recently, deep neural networks have been incorporated into PET iterative reconstruction framework to improve the image quality. In this paper, we propose a new neural network-based iterative reconstruction method by using weighted nuclear norm (WNN) maximization, which aims to recover the image details in the reconstruction process. The novelty of our method is the application of WNN maximization rather than WNN minimization in PET image reconstruction. Meanwhile, a neural network is used to control the noise originated from WNN maximization. Our method is evaluated on simulated and clinical datasets. The simulation results show that the proposed approach outperforms state-of-the-art neural network-based iterative methods by achieving the best contrast/noise tradeoff with a remarkable contrast improvement on the lesion contrast recovery. The study on clinical datasets also demonstrates that our method can recover lesions of different sizes while suppressing noise in various low-dose PET image reconstruction tasks. Our code is available athttps://github.com/Kuangxd/PETReconstruction.
Collapse
Affiliation(s)
- Xiaodong Kuang
- Center for Frontier Fundamental Studies, Zhejiang Lab, Hangzhou, People's Republic of China
| | - Bingxuan Li
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, People's Republic of China
| | - Tianling Lyu
- Center for Frontier Fundamental Studies, Zhejiang Lab, Hangzhou, People's Republic of China
| | - Yitian Xue
- Center for Frontier Fundamental Studies, Zhejiang Lab, Hangzhou, People's Republic of China
| | - Hailiang Huang
- Center for Frontier Fundamental Studies, Zhejiang Lab, Hangzhou, People's Republic of China
| | - Qingguo Xie
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, People's Republic of China
| | - Wentao Zhu
- Center for Frontier Fundamental Studies, Zhejiang Lab, Hangzhou, People's Republic of China
| |
Collapse
|
6
|
Huang B, Li T, Arino-Estrada G, Dulski K, Shopa RY, Moskal P, Stepien E, Qi J. SPLIT: Statistical Positronium Lifetime Image Reconstruction via Time-Thresholding. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:2148-2158. [PMID: 38261489 PMCID: PMC11409919 DOI: 10.1109/tmi.2024.3357659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Positron emission tomography (PET) is a widely utilized medical imaging modality that uses positron-emitting radiotracers to visualize biochemical processes in a living body. The spatiotemporal distribution of a radiotracer is estimated by detecting the coincidence photon pairs generated through positron annihilations. In human tissue, about 40% of the positrons form positroniums prior to the annihilation. The lifetime of these positroniums is influenced by the microenvironment in the tissue and could provide valuable information for better understanding of disease progression and treatment response. Currently, there are few methods available for reconstructing high-resolution lifetime images in practical applications. This paper presents an efficient statistical image reconstruction method for positronium lifetime imaging (PLI). We also analyze the random triple-coincidence events in PLI and propose a correction method for random events, which is essential for real applications. Both simulation and experimental studies demonstrate that the proposed method can produce lifetime images with high numerical accuracy, low variance, and resolution comparable to that of the activity images generated by a PET scanner with currently available time-of-flight resolution.
Collapse
|
7
|
Jagust WJ, Mattay VS, Krainak DM, Wang SJ, Weidner LD, Hofling AA, Koo H, Hsieh P, Kuo PH, Farrar G, Marzella L. Quantitative Brain Amyloid PET. J Nucl Med 2024; 65:670-678. [PMID: 38514082 PMCID: PMC11064834 DOI: 10.2967/jnumed.123.265766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
Since the development of amyloid tracers for PET imaging, there has been interest in quantifying amyloid burden in the brains of patients with Alzheimer disease. Quantitative amyloid PET imaging is poised to become a valuable approach in disease staging, theranostics, monitoring, and as an outcome measure for interventional studies. Yet, there are significant challenges and hurdles to overcome before it can be implemented into widespread clinical practice. On November 17, 2022, the U.S. Food and Drug Administration, Society of Nuclear Medicine and Molecular Imaging, and Medical Imaging and Technology Alliance cosponsored a public workshop comprising experts from academia, industry, and government agencies to discuss the role of quantitative brain amyloid PET imaging in staging, prognosis, and longitudinal assessment of Alzheimer disease. The workshop discussed a range of topics, including available radiopharmaceuticals for amyloid imaging; the methodology, metrics, and analytic validity of quantitative amyloid PET imaging; its use in disease staging, prognosis, and monitoring of progression; and challenges facing the field. This report provides a high-level summary of the presentations and the discussion.
Collapse
Affiliation(s)
| | - Venkata S Mattay
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland;
| | - Daniel M Krainak
- Division of Radiological Imaging and Radiation Therapy Devices, Office of Radiological Health, Office of Product Evaluation and Quality, Centers for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland
| | - Sue-Jane Wang
- Division of Biometrics I, Office of Biostatistics, Office of Translational Sciences, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Lora D Weidner
- Division of Radiological Imaging and Radiation Therapy Devices, Office of Radiological Health, Office of Product Evaluation and Quality, Centers for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland
| | - A Alex Hofling
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Hayoung Koo
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | | | | | | | - Libero Marzella
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
8
|
Pan B, Marsden PK, Reader AJ. Deep learned triple-tracer multiplexed PET myocardial image separation. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1379647. [PMID: 39381030 PMCID: PMC11460302 DOI: 10.3389/fnume.2024.1379647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/21/2024] [Indexed: 10/10/2024]
Abstract
Introduction In multiplexed positron emission tomography (mPET) imaging, physiological and pathological information from different radiotracers can be observed simultaneously in a single dynamic PET scan. The separation of mPET signals within a single PET scan is challenging due to the fact that the PET scanner measures the sum of the PET signals of all the tracers. The conventional multi-tracer compartment modeling (MTCM) method requires staggered injections and assumes that the arterial input functions (AIFs) of each tracer are known. Methods In this work, we propose a deep learning-based method to separate triple-tracer PET images without explicitly knowing the AIFs. A dynamic triple-tracer noisy MLEM reconstruction was used as the network input, and dynamic single-tracer noisy MLEM reconstructions were used as training labels. Results A simulation study was performed to evaluate the performance of the proposed framework on triple-tracer ([ F 18 ]FDG+ Rb 82 +[ Tc 99m ]sestamibi) PET myocardial imaging. The results show that the proposed methodology substantially reduced the noise level compared to the results obtained from single-tracer imaging. Additionally, it achieved lower bias and standard deviation in the separated single-tracer images compared to the MTCM-based method at both the voxel and region of interest (ROI) levels. Discussion As compared to MTCM separation, the proposed method uses spatiotemporal information for separation, which improves the separation performance at both the voxel and ROI levels. The simulation study also demonstrates the feasibility and potential of the proposed DL-based method for the application to pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Bolin Pan
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | | | | |
Collapse
|
9
|
Mohr P, van Sluis J, Lub-de Hooge MN, Lammertsma AA, Brouwers AH, Tsoumpas C. Advances and challenges in immunoPET methodology. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1360710. [PMID: 39355220 PMCID: PMC11440922 DOI: 10.3389/fnume.2024.1360710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/05/2024] [Indexed: 10/03/2024]
Abstract
Immuno-positron emission tomography (immunoPET) enables imaging of specific targets that play a role in targeted therapy and immunotherapy, such as antigens on cell membranes, targets in the disease microenvironment, or immune cells. The most common immunoPET applications use a monoclonal antibody labeled with a relatively long-lived positron emitter such as 89Zr (T 1/2 = 78.4 h), but smaller antibody-based constructs labeled with various other positron emitting radionuclides are also being investigated. This molecular imaging technique can thus guide the development of new drugs and may have a pivotal role in selecting patients for a particular therapy. In early phase immunoPET trials, multiple imaging time points are used to examine the time-dependent biodistribution and to determine the optimal imaging time point, which may be several days after tracer injection due to the slow kinetics of larger molecules. Once this has been established, usually only one static scan is performed and semi-quantitative values are reported. However, total PET uptake of a tracer is the sum of specific and nonspecific uptake. In addition, uptake may be affected by other factors such as perfusion, pre-/co-administration of the unlabeled molecule, and the treatment schedule. This article reviews imaging methodologies used in immunoPET studies and is divided into two parts. The first part summarizes the vast majority of clinical immunoPET studies applying semi-quantitative methodologies. The second part focuses on a handful of studies applying pharmacokinetic models and includes preclinical and simulation studies. Finally, the potential and challenges of immunoPET quantification methodologies are discussed within the context of the recent technological advancements provided by long axial field of view PET/CT scanners.
Collapse
Affiliation(s)
- Philipp Mohr
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Charalampos Tsoumpas
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
10
|
Moradi H, Vashistha R, O'Brien K, Hammond A, Vegh V, Reutens D. A short 18F-FDG imaging window triple injection neuroimaging protocol for parametric mapping in PET. EJNMMI Res 2024; 14:1. [PMID: 38169031 PMCID: PMC10761663 DOI: 10.1186/s13550-023-01061-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND In parametric PET, kinetic parameters are extracted from dynamic PET images. It is not commonly used in clinical practice because of long scan times and the requirement for an arterial input function (AIF). To address these limitations, we designed an 18F-fluorodeoxyglucose (18F-FDG) triple injection dynamic PET protocol for brain imaging with a standard field of view PET scanner using a 24-min imaging window and an input function modeled using measurements from a region of interest placed over the left ventricle. METHODS To test the protocol in 6 healthy participants, we examined the quality of voxel-based maps of kinetic parameters in the brain generated using the two-tissue compartment model and compared estimated parameter values with previously published values. We also utilized data from a 36-min validation imaging window to compare (1) the modeled AIF against the input function measured in the validation window; and (2) the net influx rate ([Formula: see text]) computed using parameter estimates from the short imaging window against the net influx rate obtained using Patlak analysis in the validation window. RESULTS Compared to the AIF measured in the validation window, the input function estimated from the short imaging window achieved a mean area under the curve error of 9%. The voxel-wise Pearson's correlation between [Formula: see text] estimates from the short imaging window and the validation imaging window exceeded 0.95. CONCLUSION The proposed 24-min triple injection protocol enables parametric 18F-FDG neuroimaging with noninvasive estimation of the AIF from cardiac images using a standard field of view PET scanner.
Collapse
Affiliation(s)
- Hamed Moradi
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
- ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, Australia
- Siemens Healthcare Pty Ltd, Melbourne, Australia
| | - Rajat Vashistha
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
- ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, Australia
| | - Kieran O'Brien
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
- ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, Australia
- Siemens Healthcare Pty Ltd, Melbourne, Australia
| | - Amanda Hammond
- ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, Australia
- Siemens Healthcare Pty Ltd, Melbourne, Australia
| | - Viktor Vegh
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia.
- ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, Australia.
| | - David Reutens
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
- ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, Australia
| |
Collapse
|
11
|
Chemli Y, Tétrault MA, Marin T, Normandin MD, Bloch I, El Fakhri G, Ouyang J, Petibon Y. Super-resolution in brain positron emission tomography using a real-time motion capture system. Neuroimage 2023; 272:120056. [PMID: 36977452 PMCID: PMC10122782 DOI: 10.1016/j.neuroimage.2023.120056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/28/2023] [Accepted: 03/25/2023] [Indexed: 03/29/2023] Open
Abstract
Super-resolution (SR) is a methodology that seeks to improve image resolution by exploiting the increased spatial sampling information obtained from multiple acquisitions of the same target with accurately known sub-resolution shifts. This work aims to develop and evaluate an SR estimation framework for brain positron emission tomography (PET), taking advantage of a high-resolution infra-red tracking camera to measure shifts precisely and continuously. Moving phantoms and non-human primate (NHP) experiments were performed on a GE Discovery MI PET/CT scanner (GE Healthcare) using an NDI Polaris Vega (Northern Digital Inc), an external optical motion tracking device. To enable SR, a robust temporal and spatial calibration of the two devices was developed as well as a list-mode Ordered Subset Expectation Maximization PET reconstruction algorithm, incorporating the high-resolution tracking data from the Polaris Vega to correct motion for measured line of responses on an event-by-event basis. For both phantoms and NHP studies, the SR reconstruction method yielded PET images with visibly increased spatial resolution compared to standard static acquisitions, allowing improved visualization of small structures. Quantitative analysis in terms of SSIM, CNR and line profiles were conducted and validated our observations. The results demonstrate that SR can be achieved in brain PET by measuring target motion in real-time using a high-resolution infrared tracking camera.
Collapse
Affiliation(s)
- Yanis Chemli
- Gordon Center for Medical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States; LTCI, Télécom Paris, Institut Polytechnique de Paris, France.
| | - Marc-André Tétrault
- Department of Computer Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Thibault Marin
- Gordon Center for Medical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.
| | - Isabelle Bloch
- Sorbonne Université, CNRS, LIP6, Paris, France; LTCI, Télécom Paris, Institut Polytechnique de Paris, France.
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.
| | - Jinsong Ouyang
- Gordon Center for Medical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.
| | - Yoann Petibon
- Gordon Center for Medical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Edelmann MR. Radiolabelling small and biomolecules for tracking and monitoring. RSC Adv 2022; 12:32383-32400. [PMID: 36425706 PMCID: PMC9650631 DOI: 10.1039/d2ra06236d] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022] Open
Abstract
Radiolabelling small molecules with beta-emitters has been intensively explored in the last decades and novel concepts for the introduction of radionuclides continue to be reported regularly. New catalysts that induce carbon/hydrogen activation are able to incorporate isotopes such as deuterium or tritium into small molecules. However, these established labelling approaches have limited applicability for nucleic acid-based drugs, therapeutic antibodies, or peptides, which are typical of the molecules now being investigated as novel therapeutic modalities. These target molecules are usually larger (significantly >1 kDa), mostly multiply charged, and often poorly soluble in organic solvents. However, in preclinical research they often require radiolabelling in order to track and monitor drug candidates in metabolism, biotransformation, or pharmacokinetic studies. Currently, the most established approach to introduce a tritium atom into an oligonucleotide is based on a multistep synthesis, which leads to a low specific activity with a high level of waste and high costs. The most common way of tritiating peptides is using appropriate precursors. The conjugation of a radiolabelled prosthetic compound to a functional group within a protein sequence is a commonly applied way to introduce a radionuclide or a fluorescent tag into large molecules. This review highlights the state-of-the-art in different radiolabelling approaches for oligonucleotides, peptides, and proteins, as well as a critical assessment of the impact of the label on the properties of the modified molecules. Furthermore, applications of radiolabelled antibodies in biodistribution studies of immune complexes and imaging of brain targets are reported.
Collapse
Affiliation(s)
- Martin R Edelmann
- Department of Pharmacy and Pharmacology, University of Bath Bath BA2 7AY UK
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Therapeutic Modalities, Small Molecule Research, Isotope Synthesis, F. Hoffmann-La Roche Ltd CH-4070 Basel Switzerland
| |
Collapse
|
13
|
Qi J, Huang B. Positronium Lifetime Image Reconstruction for TOF PET. IEEE TRANSACTIONS ON MEDICAL IMAGING 2022; 41:2848-2855. [PMID: 35584079 PMCID: PMC9829407 DOI: 10.1109/tmi.2022.3174561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Positron emission tomography is widely used in clinical and preclinical applications. Positronium lifetime carries information about the tissue microenvironment where positrons are emitted, but such information has not been captured because of two technical challenges. One challenge is the low sensitivity in detecting triple coincidence events. This problem has been mitigated by the recent developments of PET scanners with long (1-2 m) axial field of view. The other challenge is the low spatial resolution of the positronium lifetime images formed by existing methods that is determined by the time-of-flight (TOF) resolution (200-500 ps) of existing PET scanners. This paper solves the second challenge by developing a new image reconstruction method to generate high-resolution positronium lifetime images using existing TOF PET. Simulation studies demonstrate that the proposed method can reconstruct positronium lifetime images at much better spatial resolution than the limit set by the TOF resolution of the PET scanner. The proposed method opens up the possibility of performing positronium lifetime imaging using existing TOF PET scanners. The lifetime information can be used to understand the tissue microenvironment in vivo which could facilitate the study of disease mechanism and selection of proper treatments.
Collapse
|
14
|
Lopes van den Broek S, Shalgunov V, García Vázquez R, Beschorner N, Bidesi NSR, Nedergaard M, Knudsen GM, Sehlin D, Syvänen S, Herth MM. Pretargeted Imaging beyond the Blood-Brain Barrier-Utopia or Feasible? Pharmaceuticals (Basel) 2022; 15:1191. [PMID: 36297303 PMCID: PMC9612205 DOI: 10.3390/ph15101191] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Pretargeting is a promising nuclear imaging technique that allows for the usage of antibodies (Abs) with enhanced imaging contrast and reduced patient radiation burden. It is based on bioorthogonal chemistry with the tetrazine ligation-a reaction between trans-cyclooctenes (TCOs) and tetrazines (Tzs)-currently being the most popular reaction due to its high selectivity and reactivity. As Abs can be designed to bind specifically to currently 'undruggable' targets such as protein isoforms or oligomers, which play a crucial role in neurodegenerative diseases, pretargeted imaging beyond the BBB is highly sought after, but has not been achieved yet. A challenge in this respect is that large molecules such as Abs show poor brain uptake. Uptake can be increased by receptor mediated transcytosis; however, it is largely unknown if the achieved brain concentrations are sufficient for pretargeted imaging. In this study, we investigated whether the required concentrations are feasible to reach. As a model Ab, we used the bispecific anti-amyloid beta (Aβ) anti-transferrin receptor (TfR) Ab 3D6scFv8D3 and conjugated it to a different amount of TCOs per Ab and tested different concentrations in vitro. With this model in hand, we estimated the minimum required TCO concentration to achieve a suitable contrast between the high and low binding regions. The estimation was carried out using pretargeted autoradiography on brain sections of an Alzheimer's disease mouse model. Biodistribution studies in wild-type (WT) mice were used to correlate how different TCO/Ab ratios alter the brain uptake. Pretargeted autoradiography showed that increasing the number of TCOs as well as increasing the TCO-Ab concentration increased the imaging contrast. A minimum brain concentration of TCOs for pretargeting purposes was determined to be 10.7 pmol/g in vitro. Biodistribution studies in WT mice showed a brain uptake of 1.1% ID/g using TCO-3D6scFv8D3 with 6.8 TCO/Ab. According to our estimations using the optimal parameters, pretargeted imaging beyond the BBB is not a utopia. Necessary brain TCO concentrations can be reached and are in the same order of magnitude as required to achieve sufficient contrast. This work gives a first estimate that pretargeted imaging is indeed possible with antibodies. This could allow the imaging of currently 'undruggable' targets and therefore be crucial to monitor (e.g., therapies for intractable neurodegenerative diseases).
Collapse
Affiliation(s)
- Sara Lopes van den Broek
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Rocío García Vázquez
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Natalie Beschorner
- Center for Translational Neuromedicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Natasha S. R. Bidesi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Gitte M. Knudsen
- Neurobiology Research Unit, Rigshospitalet Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Dag Sehlin
- Rudbeck Laboratory, Department of Public Health and Caring Sciences, University of Uppsala, Dag Hammarskjölds väg 20, 75185 Uppsala, Sweden
| | - Stina Syvänen
- Rudbeck Laboratory, Department of Public Health and Caring Sciences, University of Uppsala, Dag Hammarskjölds väg 20, 75185 Uppsala, Sweden
| | - Matthias M. Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
| |
Collapse
|
15
|
Tikhonova MA, Zhanaeva SY, Shvaikovskaya AA, Olkov NM, Aftanas LI, Danilenko KV. Neurospecific Molecules Measured in Periphery in Humans: How Do They Correlate with the Brain Levels? A Systematic Review. Int J Mol Sci 2022; 23:ijms23169193. [PMID: 36012459 PMCID: PMC9409387 DOI: 10.3390/ijms23169193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/09/2022] [Accepted: 08/13/2022] [Indexed: 11/19/2022] Open
Abstract
Human brain state is usually estimated by brain-specific substances in peripheral tissues, but, for most analytes, a concordance between their content in the brain and periphery is unclear. In this systematic review, we summarized the investigated correlations in humans. PubMed was searched up to June 2022. We included studies measuring the same endogenous neurospecific analytes in the central nervous system and periphery in the same subjects. Not eligible were studies of cerebrospinal fluid, with significant blood–brain barrier disruption, of molecules with well-established blood-periphery concordance or measured in brain tumors. Seventeen studies were eligible. Four studies did not report on correlation and four revealed no significant correlation. Four molecules were examined twice. For BDNF, there was no correlation in both studies. For phenylalanine, glutamine, and glutamate, results were contradictory. Strong correlations were found for free tryptophan (r = 0.97) and translocator protein (r = 0.90). Thus, only for three molecules was there some certainty. BDNF in plasma or serum does not reflect brain content, whereas free tryptophan (in plasma) and translocator protein (in blood cells) can serve as peripheral biomarkers. We expect a breakthrough in the field with advanced in vivo metabolomic analyses, neuroimaging techniques, and blood assays for exosomes of brain origin.
Collapse
|
16
|
Takalloobanafshi G, Kukreja A, Hicks JW. Historical efforts to develop 99mTc-based amyloid plaque targeting radiotracers. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2022; 2:963698. [PMID: 39390996 PMCID: PMC11466234 DOI: 10.3389/fnume.2022.963698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/21/2022] [Indexed: 10/12/2024]
Abstract
Imaging biomarkers have changed the way we study Alzheimer's disease and related dementias, develop new therapeutics to treat the disease, and stratify patient populations in clinical trials. With respect to protein aggregates comprised of amyloid-β plaques and tau neurofibrillary tangles, Positron Emission Tomography (PET) has become the gold standard imaging modality for quantitative visualization. Due to high infrastructural costs, the availability of PET remains limited to large urban areas within high income nations. This limits access to leading edge medical imaging, and potentially access to new treatments, by millions of rural and remote residents in those regions as well as billions of people in middle- and low-income countries. Single Photon Emission Computed Tomography (SPECT) is a more widely available imaging alternative with lower infrastructural costs and decades of familiarity amongst nuclear medicine professionals. Recent technological advances have closed the gap in spatial resolution and quantitation between SPECT and PET. If effective SPECT radiotracers were available to visualize amyloid-β plaques, geographic barriers to imaging could be circumvented. In this review, we will discuss past efforts to develop SPECT radiotracers targeting amyloid-β plaques which incorporate the most used radionuclide in nuclear medicine: technetium-99m (99mTc; t 1/2 = 6.01 h; γ = 140 keV). While reviewing the various chemical scaffolds and chelates employed, the focus will be upon the impact to the pharmacological properties of putative 99mTc-based amyloid-targeting radiotracers.
Collapse
Affiliation(s)
- Ghazaleh Takalloobanafshi
- Department of Chemistry, Western University, London, ON, Canada
- Cyclotron and Radiochemistry Facility, Lawson Health Research Institute, London, ON, Canada
| | - Aditi Kukreja
- Department of Medical Biophysics, Western University, London, ON, Canada
| | - Justin W. Hicks
- Cyclotron and Radiochemistry Facility, Lawson Health Research Institute, London, ON, Canada
- Department of Medical Biophysics, Western University, London, ON, Canada
- Saint Joseph's Health Care London, London, ON, Canada
| |
Collapse
|
17
|
Wei S, Joshi N, Salerno M, Ouellette D, Saleh L, Delorenzo C, Woody C, Schlyer D, Purschke ML, Pratte JF, Junnarkar S, Budassi M, Cao T, Fried J, Karp JS, Vaska P. PET Imaging of Leg Arteries for Determining the Input Function in PET/MRI Brain Studies Using a Compact, MRI-Compatible PET System. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2022; 6:583-591. [PMID: 36212108 PMCID: PMC9541963 DOI: 10.1109/trpms.2021.3111841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In this study, we used a compact, high-resolution, and MRI-compatible PET camera (VersaPET) to assess the feasibility of measuring the image-derived input function (IDIF) from arteries in the leg with the ultimate goal of enabling fully quantitative PET brain imaging without blood sampling. We used this approach in five 18F-FDG PET/MRI brain studies in which the input function was also acquired using the gold standard of serial arterial blood sampling. After accounting for partial volume, dispersion, and calibration effects, we compared the metabolic rates of glucose (MRglu) quantified from VersaPET IDIFs in 80 brain regions to those using the gold standard and achieved a bias and variability of <5% which is within the range of reported test-retest values for this type of study. We also achieved a strong linear relationship (R2 >0.97) against the gold standard across regions. The results of this preliminary study are promising and support further studies to optimize methods, validate in a larger cohort, and extend to the modeling of other radiotracers.
Collapse
Affiliation(s)
- Shouyi Wei
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Nandita Joshi
- Department of Electrical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Michael Salerno
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - David Ouellette
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Lemise Saleh
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Christine Delorenzo
- Department of Psychiatry and Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Craig Woody
- Department of Physics, Brookhaven National Laboratory, Upton, NY, USA
| | - David Schlyer
- Department of Physics, Brookhaven National Laboratory, Upton, NY, USA
| | | | - Jean-Francois Pratte
- Interdisciplinary Institute for Technological Innovation, Université de Sherbrooke, Sherbrooke, Canada
| | | | - Michael Budassi
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | | | - Jack Fried
- Department of Physics, Brookhaven National Laboratory, Upton, NY, USA
| | - Joel S. Karp
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Vaska
- Department of Biomedical Engineering and Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
18
|
Vrachliotis A, Kastis GA, Protonotarios NE, Fokas AS, Nekolla SG, Anagnostopoulos CD, Costaridou L, Gaitanis A. Evaluation of the spline reconstruction technique for preclinical PET imaging. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 217:106668. [PMID: 35176596 DOI: 10.1016/j.cmpb.2022.106668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 12/27/2021] [Accepted: 01/26/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND AND OBJECTIVE The Spline Reconstruction Technique (SRT) is a fast algorithm based on a novel numerical implementation of an analytic representation of the inverse Radon transform. The purpose of this study is to provide a comparison between SRT, Filtered Back-Projection (FBP), Ordered Subset Expectation Maximization 2D (2D-OSEM), and the Tera-Tomo 3D algorithm, using phantom data at various acquisition durations as well as small-animal data obtained from the Mediso nanoScan® PET/CT scanner. METHODS For this purpose, the "NEMA NU 4-2008 standards" protocol was employed at five different realizations and acquisition durations. In addition to the image quality metrics described by the NEMA protocol, Cold Region Contrast was also considered as a figure-of-merit. Furthermore, Cold Region Contrast was measured in the myocardial infarction region of six male Wistar rats. The volumetric defect quantification was assessed with dedicated computer software. Lastly, plots of Recovery Coefficient and Spill-Over Ratio as a function of the Percentage Standard Deviation were generated, after smoothing the phantom reconstructions with four different Gaussian filters. Statistical significance was determined by employing the Kruskal-Wallis test or One-way Analysis of Variance depending on the normality of the variable's distribution. RESULTS The present study revealed that, at the expense of slightly increased noise in the reconstructed images, SRT resulted in higher Recovery Coefficient values for small hot regions of interest, when compared with FBP and 2D-OSEM at all acquisition durations. Furthermore, SRT reconstructed images exhibit higher Recovery Coefficient values, for all hot regions of interest, when compared to the other 2D algorithms at short acquisition durations. In both phantom and animal studies, SRT achieved a significant improvement over 2D-OSEM for the Spill-Over Ratio and the Cold Region Contrast. These advantages were maintained even after comparing the algorithms at equal noise levels. The Tera-Tomo 3D algorithm (4 subsets, iterations≥ 13) performed significantly better compared to the other algorithms for all figures-of-merit. No statistically significant differences regarding the myocardial defect size were observed between the algorithms investigated. CONCLUSIONS Overall, SRT appears that could be useful for the quantification of small hot regions of interest, cold regions of interest, as well as in low-count imaging applications.
Collapse
Affiliation(s)
- Alexandros Vrachliotis
- Department of Medical Physics, School of Medicine, University of Patras, Patras 26504, Greece; Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Athens 11527, Greece
| | - George A Kastis
- Mathematics Research Center, Academy of Athens, Athens 11527, Greece; Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research "Demokritos", 15341 Athens, Greece
| | - Nicholas E Protonotarios
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge CB30WA, United Kingdom
| | - Athanasios S Fokas
- Mathematics Research Center, Academy of Athens, Athens 11527, Greece; Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge CB30WA, United Kingdom
| | - Stephan G Nekolla
- Klinikum rechts der Isar, Department of Nuclear Medicine and DZHK (German Centre for Cardiovascular Research), Technical University Munich, Partner Site Munich Heart Alliance, Munich 80336, Germany
| | - Constantinos D Anagnostopoulos
- Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Athens 11527, Greece
| | - Lena Costaridou
- Department of Medical Physics, School of Medicine, University of Patras, Patras 26504, Greece
| | - Anastasios Gaitanis
- Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Athens 11527, Greece.
| |
Collapse
|
19
|
Toyonaga T, Fesharaki-Zadeh A, Strittmatter SM, Carson RE, Cai Z. PET Imaging of Synaptic Density: Challenges and Opportunities of Synaptic Vesicle Glycoprotein 2A PET in Small Animal Imaging. Front Neurosci 2022; 16:787404. [PMID: 35345546 PMCID: PMC8957200 DOI: 10.3389/fnins.2022.787404] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/14/2022] [Indexed: 12/02/2022] Open
Abstract
The development of novel PET imaging agents for synaptic vesicle glycoprotein 2A (SV2A) allowed for the in vivo detection of synaptic density changes, which are correlated with the progression and severity of a variety of neuropsychiatric diseases. While multiple ongoing clinical investigations using SV2A PET are expanding its applications rapidly, preclinical SV2A PET imaging in animal models is an integral component of the translation research and provides supporting and complementary information. Herein, we overview preclinical SV2A PET studies in animal models of neurodegenerative disorders and discuss the opportunities and practical challenges in small animal SV2A PET imaging. At the Yale PET Center, we have conducted SV2A PET imaging studies in animal models of multiple diseases and longitudinal SV2A PET allowed us to evaluate synaptic density dynamics in the brains of disease animal models and to assess pharmacological effects of novel interventions. In this article, we discuss key considerations when designing preclinical SV2A PET imaging studies and strategies for data analysis. Specifically, we compare the brain imaging characteristics of available SV2A tracers, i.e., [11C]UCB-J, [18F]SynVesT-1, [18F]SynVesT-2, and [18F]SDM-16, in rodent brains. We also discuss the limited spatial resolution of PET scanners for small brains and challenges of kinetic modeling. We then compare different injection routes and estimate the maximum throughput (i.e., number of animals) per radiotracer synthesis by taking into account the injectable volume for each injection method, injected mass, and radioactivity half-lives. In summary, this article provides a perspective for designing and analyzing SV2A PET imaging studies in small animals.
Collapse
Affiliation(s)
- Takuya Toyonaga
- Positron Emission Tomography (PET) Center, Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Arman Fesharaki-Zadeh
- Psychiatry, Yale School of Medicine, New Haven, CT, United States
- Neurology, Yale School of Medicine, New Haven, CT, United States
| | - Stephen M. Strittmatter
- Neurology, Yale School of Medicine, New Haven, CT, United States
- Neuroscience, Yale School of Medicine, New Haven, CT, United States
| | - Richard E. Carson
- Positron Emission Tomography (PET) Center, Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Zhengxin Cai
- Positron Emission Tomography (PET) Center, Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
20
|
Norgaard M, Matheson GJ, Hansen HD, Thomas A, Searle G, Rizzo G, Veronese M, Giacomel A, Yaqub M, Tonietto M, Funck T, Gillman A, Boniface H, Routier A, Dalenberg JR, Betthauser T, Feingold F, Markiewicz CJ, Gorgolewski KJ, Blair RW, Appelhoff S, Gau R, Salo T, Niso G, Pernet C, Phillips C, Oostenveld R, Gallezot JD, Carson RE, Knudsen GM, Innis RB, Ganz M. PET-BIDS, an extension to the brain imaging data structure for positron emission tomography. Sci Data 2022; 9:65. [PMID: 35236846 PMCID: PMC8891322 DOI: 10.1038/s41597-022-01164-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/11/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- Martin Norgaard
- Neurobiology Research Unit, Rigshospitalet, and Institute of Clinical Medicine, Univ. Copenhagen, København, Denmark.,Department of Psychology, Stanford University, California, USA
| | - Granville J Matheson
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA.,Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Stockholm, Sweden
| | - Hanne D Hansen
- Neurobiology Research Unit, Rigshospitalet, and Institute of Clinical Medicine, Univ. Copenhagen, København, Denmark.,Athinoula A. Martinos Center for Biomedical Imaging, MGH/HST, Charlestown, MA, USA
| | - Adam Thomas
- Intramural Research Program, NIMH, Bethesda, USA
| | - Graham Searle
- Invicro and Division of Brain Sciences, Imperial College London, London, UK
| | - Gaia Rizzo
- Invicro and Division of Brain Sciences, Imperial College London, London, UK
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, King's College London, London, UK.,Department of Information Engineering, University of Padua, Padua, Italy
| | - Alessio Giacomel
- Centre for Neuroimaging Sciences, King's College London, London, UK
| | - Maqsood Yaqub
- Amsterdam UMC, location VUmc, department of radiology and nuclear medicine, Amsterdam, Netherlands
| | - Matteo Tonietto
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Thomas Funck
- INM-1, Jülich Forschungszentrum, Jülich, Germany
| | - Ashley Gillman
- Aust. e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation, Townsville, Australia
| | - Hugo Boniface
- Centre d'Acquisition et de Traitement des Images, CEA, Paris, France
| | - Alexandre Routier
- Inria, Aramis project-team, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtriére, Paris, France
| | - Jelle R Dalenberg
- Department of Neurology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Tobey Betthauser
- Wisconsin Alzheimer's Disease Research Center, Division of Geriatrics, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | | | | | | | - Ross W Blair
- Department of Psychology, Stanford University, California, USA
| | - Stefan Appelhoff
- Center for Adaptive Rationality, Max Planck Institute for Human Development, Berlin, Germany
| | - Remi Gau
- Institute of psychology, Université catholique de Louvain, Louvain la Neuve, Belgium
| | - Taylor Salo
- Department of Psychology, Florida International University, Miami, FL, USA
| | - Guiomar Niso
- Psychological Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Cyril Pernet
- Neurobiology Research Unit, Rigshospitalet, and Institute of Clinical Medicine, Univ. Copenhagen, København, Denmark
| | - Christophe Phillips
- GIGA Cyclotron Research Centre in vivo imaging, University of Liege, Liege, Belgium
| | - Robert Oostenveld
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands.,NatMEG, Karolinska Institutet, Stockholm, Sweden
| | | | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, USA
| | - Gitte M Knudsen
- Neurobiology Research Unit, Rigshospitalet, and Institute of Clinical Medicine, Univ. Copenhagen, København, Denmark
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Melanie Ganz
- Neurobiology Research Unit, Rigshospitalet, and Institute of Clinical Medicine, Univ. Copenhagen, København, Denmark. .,Department of Computer Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
21
|
Nour MM, Beck K, Liu Y, Arumuham A, Veronese M, Howes OD, Dolan RJ. Relationship Between Replay-Associated Ripples and Hippocampal N-Methyl-D-Aspartate Receptors: Preliminary Evidence From a PET-MEG Study in Schizophrenia. SCHIZOPHRENIA BULLETIN OPEN 2022; 3:sgac044. [PMID: 35911846 PMCID: PMC9334566 DOI: 10.1093/schizbullopen/sgac044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background and Hypotheses Hippocampal replay and associated high-frequency ripple oscillations are among the best-characterized phenomena in resting brain activity. Replay/ripples support memory consolidation and relational inference, and are regulated by N-methyl-D-aspartate receptors (NMDARs). Schizophrenia has been associated with both replay/ripple abnormalities and NMDAR hypofunction in both clinical samples and genetic mouse models, although the relationship between these 2 facets of hippocampal function has not been tested in humans. Study Design Here, we avail of a unique multimodal human neuroimaging data set to investigate the relationship between the availability of (intrachannel) NMDAR binding sites in hippocampus, and replay-associated ripple power, in 16 participants (7 nonclinical participants and 9 people with a diagnosis of schizophrenia, PScz). Each participant had both a [18F]GE-179 positron emission tomography (PET) scan (to measure NMDAR availability, V T ) and a magnetoencephalography (MEG) scan (to measure offline neural replay and associated high-frequency ripple oscillations, using Temporally Delayed Linear Modeling). Study Results We show a positive relationship between hippocampal NMDAR availability and replay-associated ripple power. This linkage was evident across control participants (r(5) = .94, P = .002) and PScz (r(7) = .70, P = .04), with no group difference. Conclusions Our findings provide preliminary evidence for a relationship between hippocampal NMDAR availability and replay-associated ripple power in humans, and haverelevance for NMDAR hypofunction theories of schizophrenia.
Collapse
Affiliation(s)
- Matthew M Nour
- Max Planck University College London Centre for Computational Psychiatry and Ageing Research, London WC1B 5EH, UK
- Wellcome Trust Centre for Human Neuroimaging, University College London, London WC1N 3AR, UK
- Department of Psychosis Studies, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK
| | - Katherine Beck
- Department of Psychosis Studies, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Yunzhe Liu
- Max Planck University College London Centre for Computational Psychiatry and Ageing Research, London WC1B 5EH, UK
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Atheeshaan Arumuham
- Department of Psychosis Studies, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Mattia Veronese
- Department of Psychosis Studies, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Raymond J Dolan
- Max Planck University College London Centre for Computational Psychiatry and Ageing Research, London WC1B 5EH, UK
- Wellcome Trust Centre for Human Neuroimaging, University College London, London WC1N 3AR, UK
| |
Collapse
|
22
|
Vermeulen K, Cools R, Briard E, Auberson Y, Schoepfer J, Koole M, Cawthorne C, Bormans G. Preclinical Evaluation of [ 11C]YC-72-AB85 for In Vivo Visualization of Heat Shock Protein 90 in Brain and Cancer with Positron Emission Tomography. ACS Chem Neurosci 2021; 12:3915-3927. [PMID: 34597516 DOI: 10.1021/acschemneuro.1c00508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Aberrant Hsp90 has been implied in cancer and neurodegenerative disorders. The development of a suitable Hsp90 Positron emission tomography (PET) probe can provide in vivo quantification of the expression levels of Hsp90 as a biomarker for diagnosis and follow-up of cancer and central nervous system (CNS) disease progression. In this respect, [11C]YC-72-AB85 was evaluated as an Hsp90 PET probe in B16.F10 melanoma bearing mice and its brain uptake was determined in rats and nonhuman primate. In vitro binding of [11C]YC-72-AB85 to tissue slices of mouse B16.F10 melanoma, PC3 prostate carcinoma, and rodent brain was evaluated using autoradiography. Biodistribution of [11C]YC-72-AB85 was evaluated in healthy and B16.F10 melanoma mice. In vivo brain uptake was assessed by μPET studies in rats and a rhesus monkey. In vitro binding was deemed Hsp90-specific by blocking studies with heterologous Hsp90 inhibitors onalespib and SNX-0723. Saturable Hsp90 binding was observed in brain, tumor, blood, and blood-rich organs in mice. In combined pretreatment and displacement studies, reversible and Hsp90-specific binding of [11C]YC-72-AB85 was observed in rat brain. Dynamic μPET brain scans in baseline and blocking conditions in a rhesus monkey indicated Hsp90-specific binding. [11C]YC-72-AB85 is a promising PET tracer for in vivo visualization of Hsp90 in tumor and brain. Clear differences of Hsp90 binding to blood and blood-rich organs were observed in tumor vs control mice. Further, we clearly demonstrate, for the first time, binding to a saturable Hsp90 pool in brain of rats and a rhesus monkey.
Collapse
Affiliation(s)
- Koen Vermeulen
- Laboratory for Radiopharmaceutical Research, Department of Pharmacy and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
- Radiobiology Unit & NURA, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| | - Romy Cools
- Laboratory for Radiopharmaceutical Research, Department of Pharmacy and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Emmanuelle Briard
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Yves Auberson
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Joseph Schoepfer
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Michel Koole
- Nuclear Medicine & Molecular Imaging & MoSAIC, Department of Imaging & Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Christopher Cawthorne
- Nuclear Medicine & Molecular Imaging & MoSAIC, Department of Imaging & Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmacy and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
23
|
Wang X, Zhou L, Wang Y, Jiang H, Ye H. Improved low-dose positron emission tomography image reconstruction using deep learned prior. Phys Med Biol 2021; 66. [PMID: 33882466 DOI: 10.1088/1361-6560/abfa36] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/21/2021] [Indexed: 01/18/2023]
Abstract
Positron emission tomography (PET) is a promising medical imaging technology that provides non-invasive and quantitative measurement of biochemical process in the human bodies. PET image reconstruction is challenging due to the ill-poseness of the inverse problem. With lower statistics caused by the limited detected photons, low-dose PET imaging leads to noisy reconstructed images with much quality degradation. Recently, deep neural networks (DNN) have been widely used in computer vision tasks and attracted growing interests in medical imaging. In this paper, we proposed a maximuma posteriori(MAP) reconstruction algorithm incorporating a convolutional neural network (CNN) representation in the formation of the prior. Rather than using the CNN in post-processing, we embedded the neural network in the reconstruction framework for image representation. Using the simulated data, we first quantitatively evaluated our proposed method in terms of the noise-bias tradeoff, and compared with the filtered maximum likelihood (ML), the conventional MAP, and the CNN post-processing methods. In addition to the simulation experiments, the proposed method was further quantitatively validated on the acquired patient brain and body data with the tradeoff between noise and contrast. The results demonstrated that the proposed CNN-MAP method improved noise-bias tradeoff compared with the filtered ML, the conventional MAP, and the CNN post-processing methods in the simulation study. For the patient study, the CNN-MAP method achieved better noise-contrast tradeoff over the other three methods. The quantitative enhancements indicate the potential value of the proposed CNN-MAP method in low-dose PET imaging.
Collapse
Affiliation(s)
- Xinhui Wang
- MinFound Medical Systems Co., Ltd., Hangzhou, People's Republic of China.,Zhejiang MinFound Intelligent Healthcare Technology Co. Ltd., Hangzhou, People's Republic of China
| | - Long Zhou
- MinFound Medical Systems Co., Ltd., Hangzhou, People's Republic of China.,Zhejiang MinFound Intelligent Healthcare Technology Co. Ltd., Hangzhou, People's Republic of China
| | - Yaofa Wang
- MinFound Medical Systems Co., Ltd., Hangzhou, People's Republic of China.,College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Haochuan Jiang
- MinFound Medical Systems Co., Ltd., Hangzhou, People's Republic of China
| | - Hongwei Ye
- MinFound Medical Systems Co., Ltd., Hangzhou, People's Republic of China.,Zhejiang MinFound Intelligent Healthcare Technology Co. Ltd., Hangzhou, People's Republic of China
| |
Collapse
|
24
|
Fan Y, Emvalomenos G, Grazian C, Meikle SR. PET-ABC: fully Bayesian likelihood-free inference for kinetic models. Phys Med Biol 2021; 66. [PMID: 33882476 DOI: 10.1088/1361-6560/abfa37] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 04/21/2021] [Indexed: 11/12/2022]
Abstract
Aims.We describe an intuitive, easy to use method called PET-ABC that enables full Bayesian statistical inference from single subject dynamic PET data. The performance of PET-ABC was compared with weighted non-linear least squares (WNLS) in terms of reliability of kinetic parameter estimation and statistical power for model selection.Methods.Dynamic PET data based on 1-tissue and 2-tissue compartmental models were simulated with 2 noise models and 3 noise levels. PET-ABC was used to evaluate the reliability of parameter estimates under each condition. It was also used to perform model selection for a simulated noisy dataset composed of a mixture of 1- and 2-tissue compartment kinetics. Finally, PET-ABC was used to analyze a non-steady state dynamic [11C] raclopride study performed on a fully conscious rat administered either 2 mg.kg-1amphetamine or saline 20 min after tracer injection.Results.PET-ABC yielded posterior point estimates for model parameters with smaller variance than WNLS, as well as probability density functions indicating confidence intervals for those estimates. It successfully identified the superiority of a 2-tissue compartment model to fit the simulated mixed model data. For the drug challenge study, the post observation probability of striatal displacement of the PET signal was 0.9 for amphetamine and approximately 0 for saline, indicating a high probability of amphetamine-induced endogenous dopamine release in the striatum. PET-ABC also demonstrated superior statistical power to WNLS (0.87 versus 0.09) for selecting the correct model in a simulated ligand displacement study.Conclusions.PET-ABC is a simple and intuitive method that provides complete Bayesian statistical analysis of single subject dynamic PET data, including the extent to which model parameter estimates and model choice are supported by the data. Software for PET-ABC is freely available as part of thePETabcpackagehttps://github.com/cgrazian/PETabc.
Collapse
Affiliation(s)
- Yanan Fan
- School of Mathematics and Statistics, University of New South Wales, Sydney, 2052, Australia.,ARC Centre of Excellence For Mathematical and Statistical Frontiers, ACEMS, Australia
| | - Gaelle Emvalomenos
- Sydney School of Health Sciences, University of Sydney, 2006, Australia.,Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Clara Grazian
- School of Mathematics and Statistics, University of New South Wales, Sydney, 2052, Australia.,ARC Centre of Excellence For Mathematical and Statistical Frontiers, ACEMS, Australia
| | - Steven R Meikle
- Sydney School of Health Sciences, University of Sydney, 2006, Australia.,Brain and Mind Centre, University of Sydney, 2006, Australia
| |
Collapse
|
25
|
Schwarz AJ. The Use, Standardization, and Interpretation of Brain Imaging Data in Clinical Trials of Neurodegenerative Disorders. Neurotherapeutics 2021; 18:686-708. [PMID: 33846962 PMCID: PMC8423963 DOI: 10.1007/s13311-021-01027-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Imaging biomarkers play a wide-ranging role in clinical trials for neurological disorders. This includes selecting the appropriate trial participants, establishing target engagement and mechanism-related pharmacodynamic effect, monitoring safety, and providing evidence of disease modification. In the early stages of clinical drug development, evidence of target engagement and/or downstream pharmacodynamic effect-especially with a clear relationship to dose-can provide confidence that the therapeutic candidate should be advanced to larger and more expensive trials, and can inform the selection of the dose(s) to be further tested, i.e., to "de-risk" the drug development program. In these later-phase trials, evidence that the therapeutic candidate is altering disease-related biomarkers can provide important evidence that the clinical benefit of the compound (if observed) is grounded in meaningful biological changes. The interpretation of disease-related imaging markers, and comparability across different trials and imaging tools, is greatly improved when standardized outcome measures are defined. This standardization should not impinge on scientific advances in the imaging tools per se but provides a common language in which the results generated by these tools are expressed. PET markers of pathological protein aggregates and structural imaging of brain atrophy are common disease-related elements across many neurological disorders. However, PET tracers for pathologies beyond amyloid β and tau are needed, and the interpretability of structural imaging can be enhanced by some simple considerations to guard against the possible confound of pseudo-atrophy. Learnings from much-studied conditions such as Alzheimer's disease and multiple sclerosis will be beneficial as the field embraces rarer diseases.
Collapse
Affiliation(s)
- Adam J Schwarz
- Takeda Pharmaceuticals Ltd., 40 Landsdowne Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
26
|
Meikle SR, Sossi V, Roncali E, Cherry SR, Banati R, Mankoff D, Jones T, James M, Sutcliffe J, Ouyang J, Petibon Y, Ma C, El Fakhri G, Surti S, Karp JS, Badawi RD, Yamaya T, Akamatsu G, Schramm G, Rezaei A, Nuyts J, Fulton R, Kyme A, Lois C, Sari H, Price J, Boellaard R, Jeraj R, Bailey DL, Eslick E, Willowson KP, Dutta J. Quantitative PET in the 2020s: a roadmap. Phys Med Biol 2021; 66:06RM01. [PMID: 33339012 PMCID: PMC9358699 DOI: 10.1088/1361-6560/abd4f7] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Positron emission tomography (PET) plays an increasingly important role in research and clinical applications, catalysed by remarkable technical advances and a growing appreciation of the need for reliable, sensitive biomarkers of human function in health and disease. Over the last 30 years, a large amount of the physics and engineering effort in PET has been motivated by the dominant clinical application during that period, oncology. This has led to important developments such as PET/CT, whole-body PET, 3D PET, accelerated statistical image reconstruction, and time-of-flight PET. Despite impressive improvements in image quality as a result of these advances, the emphasis on static, semi-quantitative 'hot spot' imaging for oncologic applications has meant that the capability of PET to quantify biologically relevant parameters based on tracer kinetics has not been fully exploited. More recent advances, such as PET/MR and total-body PET, have opened up the ability to address a vast range of new research questions, from which a future expansion of applications and radiotracers appears highly likely. Many of these new applications and tracers will, at least initially, require quantitative analyses that more fully exploit the exquisite sensitivity of PET and the tracer principle on which it is based. It is also expected that they will require more sophisticated quantitative analysis methods than those that are currently available. At the same time, artificial intelligence is revolutionizing data analysis and impacting the relationship between the statistical quality of the acquired data and the information we can extract from the data. In this roadmap, leaders of the key sub-disciplines of the field identify the challenges and opportunities to be addressed over the next ten years that will enable PET to realise its full quantitative potential, initially in research laboratories and, ultimately, in clinical practice.
Collapse
Affiliation(s)
- Steven R Meikle
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Brain and Mind Centre, The University of Sydney, Australia
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Canada
| | - Emilie Roncali
- Department of Biomedical Engineering, University of California, Davis, United States of America
| | - Simon R Cherry
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Radiology, University of California, Davis, United States of America
| | - Richard Banati
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Brain and Mind Centre, The University of Sydney, Australia
- Australian Nuclear Science and Technology Organisation, Sydney, Australia
| | - David Mankoff
- Department of Radiology, University of Pennsylvania, United States of America
| | - Terry Jones
- Department of Radiology, University of California, Davis, United States of America
| | - Michelle James
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), CA, United States of America
- Department of Neurology and Neurological Sciences, Stanford University, CA, United States of America
| | - Julie Sutcliffe
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Internal Medicine, University of California, Davis, CA, United States of America
| | - Jinsong Ouyang
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Yoann Petibon
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Chao Ma
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Suleman Surti
- Department of Radiology, University of Pennsylvania, United States of America
| | - Joel S Karp
- Department of Radiology, University of Pennsylvania, United States of America
| | - Ramsey D Badawi
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Radiology, University of California, Davis, United States of America
| | - Taiga Yamaya
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Go Akamatsu
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Georg Schramm
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Ahmadreza Rezaei
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Johan Nuyts
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Roger Fulton
- Brain and Mind Centre, The University of Sydney, Australia
- Department of Medical Physics, Westmead Hospital, Sydney, Australia
| | - André Kyme
- Brain and Mind Centre, The University of Sydney, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Australia
| | - Cristina Lois
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Hasan Sari
- Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
- Athinoula A. Martinos Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
| | - Julie Price
- Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
- Athinoula A. Martinos Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
| | - Ronald Boellaard
- Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, location VUMC, Netherlands
| | - Robert Jeraj
- Departments of Medical Physics, Human Oncology and Radiology, University of Wisconsin, United States of America
- Faculty of Mathematics and Physics, University of Ljubljana, Slovenia
| | - Dale L Bailey
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Science, The University of Sydney, Australia
| | - Enid Eslick
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
| | - Kathy P Willowson
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Science, The University of Sydney, Australia
| | - Joyita Dutta
- Department of Electrical and Computer Engineering, University of Massachusetts Lowell, United States of America
| |
Collapse
|
27
|
Gong Y, Shan H, Teng Y, Tu N, Li M, Liang G, Wang G, Wang S. Parameter-Transferred Wasserstein Generative Adversarial Network (PT-WGAN) for Low-Dose PET Image Denoising. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2021; 5:213-223. [PMID: 35402757 PMCID: PMC8993163 DOI: 10.1109/trpms.2020.3025071] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Due to the widespread use of positron emission tomography (PET) in clinical practice, the potential risk of PET-associated radiation dose to patients needs to be minimized. However, with the reduction in the radiation dose, the resultant images may suffer from noise and artifacts that compromise diagnostic performance. In this paper, we propose a parameter-transferred Wasserstein generative adversarial network (PT-WGAN) for low-dose PET image denoising. The contributions of this paper are twofold: i) a PT-WGAN framework is designed to denoise low-dose PET images without compromising structural details, and ii) a task-specific initialization based on transfer learning is developed to train PT-WGAN using trainable parameters transferred from a pretrained model, which significantly improves the training efficiency of PT-WGAN. The experimental results on clinical data show that the proposed network can suppress image noise more effectively while preserving better image fidelity than recently published state-of-the-art methods. We make our code available at https://github.com/90n9-yu/PT-WGAN.
Collapse
Affiliation(s)
- Yu Gong
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China, and Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hongming Shan
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai 200433, China, and the Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 201210, China
| | - Yueyang Teng
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China, and the Key Laboratory of Intelligent Computing in Medical Images, Ministry of Education, Shenyang 110169, China
| | - Ning Tu
- PET-CT/MRI Center and Molecular Imaging Center, Wuhan University Renmin Hospital, Wuhan, 430060, China
| | - Ming Li
- Neusoft Medical Systems Co., Ltd, Shenyang 110167, China
| | - Guodong Liang
- Neusoft Medical Systems Co., Ltd, Shenyang 110167, China
| | - Ge Wang
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180 USA
| | - Shanshan Wang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
28
|
Xue H, Zhang Q, Zou S, Zhang W, Zhou C, Tie C, Wan Q, Teng Y, Li Y, Liang D, Liu X, Yang Y, Zheng H, Zhu X, Hu Z. LCPR-Net: low-count PET image reconstruction using the domain transform and cycle-consistent generative adversarial networks. Quant Imaging Med Surg 2021; 11:749-762. [PMID: 33532274 PMCID: PMC7779905 DOI: 10.21037/qims-20-66] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 09/25/2020] [Indexed: 11/06/2022]
Abstract
BACKGROUND Reducing the radiation tracer dose and scanning time during positron emission tomography (PET) imaging can reduce the cost of the tracer, reduce motion artifacts, and increase the efficiency of the scanner. However, the reconstructed images to be noisy. It is very important to reconstruct high-quality images with low-count (LC) data. Therefore, we propose a deep learning method called LCPR-Net, which is used for directly reconstructing full-count (FC) PET images from corresponding LC sinogram data. METHODS Based on the framework of a generative adversarial network (GAN), we enforce a cyclic consistency constraint on the least-squares loss to establish a nonlinear end-to-end mapping process from LC sinograms to FC images. In this process, we merge a convolutional neural network (CNN) and a residual network for feature extraction and image reconstruction. In addition, the domain transform (DT) operation sends a priori information to the cycle-consistent GAN (CycleGAN) network, avoiding the need for a large amount of computational resources to learn this transformation. RESULTS The main advantages of this method are as follows. First, the network can use LC sinogram data as input to directly reconstruct an FC PET image. The reconstruction speed is faster than that provided by model-based iterative reconstruction. Second, reconstruction based on the CycleGAN framework improves the quality of the reconstructed image. CONCLUSIONS Compared with other state-of-the-art methods, the quantitative and qualitative evaluation results show that the proposed method is accurate and effective for FC PET image reconstruction.
Collapse
Affiliation(s)
- Hengzhi Xue
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qiyang Zhang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Sijuan Zou
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiguang Zhang
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chao Zhou
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Changjun Tie
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qian Wan
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yueyang Teng
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Yongchang Li
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dong Liang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Liu
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yongfeng Yang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hairong Zheng
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhanli Hu
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
29
|
Brambilla CR, Scheins J, Issa A, Tellmann L, Herzog H, Rota Kops E, Shah NJ, Neuner I, Lerche CW. Bias evaluation and reduction in 3D OP-OSEM reconstruction in dynamic equilibrium PET studies with 11C-labeled for binding potential analysis. PLoS One 2021; 16:e0245580. [PMID: 33481896 PMCID: PMC7822533 DOI: 10.1371/journal.pone.0245580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/05/2021] [Indexed: 11/26/2022] Open
Abstract
Iterative image reconstruction is widely used in positron emission tomography. However, it is known to contribute to quantitation bias and is particularly pronounced during dynamic studies with 11C-labeled radiotracers where count rates become low towards the end of the acquisition. As the strength of the quantitation bias depends on the counts in the reconstructed frame, it can differ from frame to frame of the acquisition. This is especially relevant in the case of neuro-receptor studies with simultaneous PET/MR when a bolus-infusion protocol is applied to allow the comparison of pre- and post-task effects. Here, count dependent changes in quantitation bias may interfere with task changes. We evaluated the impact of different framing schemes on quantitation bias and its propagation into binding potential (BP) using a phantom decay study with 11C and 3D OP-OSEM. Further, we propose a framing scheme that keeps the true counts per frame constant over the acquisition time as constant framing schemes and conventional increasing framing schemes are unlikely to achieve stable bias values during the acquisition time range. For a constant framing scheme with 5 minutes frames, the BP bias was 7.13±2.01% (10.8% to 3.8%) compared to 5.63±2.85% (7.8% to 4.0%) for conventional increasing framing schemes. Using the proposed constant true counts framing scheme, a stabilization of the BP bias was achieved at 2.56±3.92% (3.5% to 1.7%). The change in BP bias was further studied by evaluating the linear slope during the acquisition time interval. The lowest slope values were observed in the constant true counts framing scheme. The constant true counts framing scheme was effective for BP bias stabilization at relevant activity and time ranges. The mean BP bias under these conditions was 2.56±3.92%, which represents the lower limit for the detection of changes in BP during equilibrium and is especially important in the case of cognitive tasks where the expected changes are low.
Collapse
Affiliation(s)
- Cláudia Régio Brambilla
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- * E-mail:
| | - Jürgen Scheins
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Ahlam Issa
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Lutz Tellmann
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Hans Herzog
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Elena Rota Kops
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - N. Jon Shah
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
- Institute of Neuroscience and Medicine, INM-11, Forschungszentrum Jülich GmbH, Jülich, Germany
- JARA–BRAIN–Translational Medicine, RWTH Aachen University, Aachen, Germany
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Irene Neuner
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- JARA–BRAIN–Translational Medicine, RWTH Aachen University, Aachen, Germany
| | - Christoph W. Lerche
- Institute of Neuroscience and Medicine, INM-4, Forschungszentrum Jülich GmbH, Jülich, Germany
| |
Collapse
|
30
|
DPIR-Net: Direct PET Image Reconstruction Based on the Wasserstein Generative Adversarial Network. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2021. [DOI: 10.1109/trpms.2020.2995717] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
31
|
Timmers T, Ossenkoppele R, Visser D, Tuncel H, Wolters EE, Verfaillie SCJ, van der Flier WM, Boellaard R, Golla SSV, van Berckel BNM. Test-retest repeatability of [ 18F]Flortaucipir PET in Alzheimer's disease and cognitively normal individuals. J Cereb Blood Flow Metab 2020; 40:2464-2474. [PMID: 31575335 PMCID: PMC7705644 DOI: 10.1177/0271678x19879226] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/18/2019] [Accepted: 08/08/2019] [Indexed: 11/29/2022]
Abstract
The aim of this study was to investigate the test-retest (TRT) repeatability of various parametric quantification methods for [18F]Flortaucipir positron emission tomography (PET). We included eight subjects with dementia or mild cognitive impairment due to Alzheimer's disease and six cognitively normal subjects. All underwent two 130-min dynamic [18F]Flortaucipir PET scans within 3 ± 1 weeks. Data were analyzed using reference region models receptor parametric mapping (RPM), simplified reference tissue method 2 (SRTM2) and reference logan (RLogan), as well as standardized uptake value ratios (SUVr, time intervals 40-60, 80-100 and 110-130 min post-injection) with cerebellar gray matter as reference region. We obtained distribution volume ratio or SUVr, first for all brain regions and then in three tau-specific regions-of-interest (ROIs). TRT repeatability (%) was defined as |retest-test|/(average (test + retest)) × 100. For all methods and across ROIs, TRT repeatability ranged from (median (IQR)) 0.84% (0.68-2.15) to 6.84% (2.99-11.50). TRT repeatability was good for all reference methods used, although semi-quantitative models (i.e. SUVr) performed marginally worse than quantitative models, for instance TRT repeatability of RPM: 1.98% (0.78-3.58) vs. SUVr80-100: 3.05% (1.28-5.52), p < 0.001. Furthermore, for SUVr80-100 and SUVr110-130, with higher average SUVr, more variation was observed. In conclusion, while TRT repeatability was good for all models used, quantitative methods performed slightly better than semi-quantitative methods.
Collapse
Affiliation(s)
- Tessa Timmers
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Denise Visser
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Hayel Tuncel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Emma E Wolters
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sander CJ Verfaillie
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sandeep SV Golla
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Bart NM van Berckel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
32
|
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: 59] [Impact Index Per Article: 11.8] [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
|
33
|
Knudsen GM, Ganz M, Appelhoff S, Boellaard R, Bormans G, Carson RE, Catana C, Doudet D, Gee AD, Greve DN, Gunn RN, Halldin C, Herscovitch P, Huang H, Keller SH, Lammertsma AA, Lanzenberger R, Liow JS, Lohith TG, Lubberink M, Lyoo CH, Mann JJ, Matheson GJ, Nichols TE, Nørgaard M, Ogden T, Parsey R, Pike VW, Price J, Rizzo G, Rosa-Neto P, Schain M, Scott PJ, Searle G, Slifstein M, Suhara T, Talbot PS, Thomas A, Veronese M, Wong DF, Yaqub M, Zanderigo F, Zoghbi S, Innis RB. Guidelines for the content and format of PET brain data in publications and archives: A consensus paper. J Cereb Blood Flow Metab 2020; 40:1576-1585. [PMID: 32065076 PMCID: PMC7370374 DOI: 10.1177/0271678x20905433] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It is a growing concern that outcomes of neuroimaging studies often cannot be replicated. To counteract this, the magnetic resonance (MR) neuroimaging community has promoted acquisition standards and created data sharing platforms, based on a consensus on how to organize and share MR neuroimaging data. Here, we take a similar approach to positron emission tomography (PET) data. To facilitate comparison of findings across studies, we first recommend publication standards for tracer characteristics, image acquisition, image preprocessing, and outcome estimation for PET neuroimaging data. The co-authors of this paper, representing more than 25 PET centers worldwide, voted to classify information as mandatory, recommended, or optional. Second, we describe a framework to facilitate data archiving and data sharing within and across centers. Because of the high cost of PET neuroimaging studies, sample sizes tend to be small and relatively few sites worldwide have the required multidisciplinary expertise to properly conduct and analyze PET studies. Data sharing will make it easier to combine datasets from different centers to achieve larger sample sizes and stronger statistical power to test hypotheses. The combining of datasets from different centers may be enhanced by adoption of a common set of best practices in data acquisition and analysis.
Collapse
Affiliation(s)
- Gitte M Knudsen
- Neurobiology Research Unit, Rigshospital and University of Copenhagen, Copenhagen, Denmark
| | - Melanie Ganz
- Neurobiology Research Unit, Rigshospital and University of Copenhagen, Copenhagen, Denmark
| | - Stefan Appelhoff
- Center for Adaptive Rationality, Max Planck Institute for Human Development, Berlin, Germany
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, KU, Leuven, Belgium
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, USA
| | - Ciprian Catana
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Department of Radiology, Boston, MA, USA
| | - Doris Doudet
- Department of Medicine/Neurology, Pacific Parkinson Research Center, Vancouver, Canada
| | - Antony D Gee
- Clinical PET Centre, King's College London, London, UK
| | - Douglas N Greve
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Department of Radiology, Boston, MA, USA
| | - Roger N Gunn
- Invicro and Division of Brain Sciences, Imperial College London, London, UK
| | - Christer Halldin
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Peter Herscovitch
- Department of Positron Emission Tomography, National Institutes of Health, Bethesda, USA
| | - Henry Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, USA
| | - Sune H Keller
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Wien, Austria
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | | | - Mark Lubberink
- Uppsala University, Department of Surgical Sciences/Radiology and Nuclear Medicine, Uppsala University Hospital, Department of Medical Physics, Sweden
| | - Chul H Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - J John Mann
- Department of Psychiatry, Molecular Imaging and Neuropathology Division, Columbia University, New York, USA
| | - Granville J Matheson
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Thomas E Nichols
- Oxford Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Population Health, University of Oxford, UK
| | - Martin Nørgaard
- Neurobiology Research Unit, Rigshospital and University of Copenhagen, Copenhagen, Denmark
| | - Todd Ogden
- Columbia Mailman School of Public Health, Columbia University, New York, USA
| | - Ramin Parsey
- Department of Psychiatry, School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Julie Price
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Department of Radiology, Boston, MA, USA
| | - Gaia Rizzo
- Invicro and Division of Brain Sciences, Imperial College London, London, UK
| | - Pedro Rosa-Neto
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Canada.,Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, Douglas Mental Health University Institute, Montreal, Canada
| | - Martin Schain
- Columbia Mailman School of Public Health, Columbia University, New York, USA
| | - Peter Jh Scott
- Department of Radiology, University of Michigan, Ann Arbor, USA
| | - Graham Searle
- Invicro and Division of Brain Sciences, Imperial College London, London, UK
| | - Mark Slifstein
- Department of Psychiatry, School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Tetsuya Suhara
- Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Peter S Talbot
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Adam Thomas
- National Institute of Mental Health, Bethesda, USA
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, King's College London, London, UK
| | - Dean F Wong
- Department of Radiology, Johns Hopkins Hospital, Baltimore, USA
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | | | - Sami Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| |
Collapse
|
34
|
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
|
35
|
Seo JW, Ingham ES, Mahakian L, Tumbale S, Wu B, Aghevlian S, Shams S, Baikoghli M, Jain P, Ding X, Goeden N, Dobreva T, Flytzanis NC, Chavez M, Singhal K, Leib R, James ML, Segal DJ, Cheng RH, Silva EA, Gradinaru V, Ferrara KW. Positron emission tomography imaging of novel AAV capsids maps rapid brain accumulation. Nat Commun 2020; 11:2102. [PMID: 32355221 PMCID: PMC7193641 DOI: 10.1038/s41467-020-15818-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 03/31/2020] [Indexed: 01/07/2023] Open
Abstract
Adeno-associated viruses (AAVs) are typically single-stranded deoxyribonucleic acid (ssDNA) encapsulated within 25-nm protein capsids. Recently, tissue-specific AAV capsids (e.g. PHP.eB) have been shown to enhance brain delivery in rodents via the LY6A receptor on brain endothelial cells. Here, we create a non-invasive positron emission tomography (PET) methodology to track viruses. To provide the sensitivity required to track AAVs injected at picomolar levels, a unique multichelator construct labeled with a positron emitter (Cu-64, t1/2 = 12.7 h) is coupled to the viral capsid. We find that brain accumulation of the PHP.eB capsid 1) exceeds that reported in any previous PET study of brain uptake of targeted therapies and 2) is correlated with optical reporter gene transduction of the brain. The PHP.eB capsid brain endothelial receptor affinity is nearly 20-fold greater than that of AAV9. The results suggest that novel PET imaging techniques can be applied to inform and optimize capsid design. Adeno-associated viruses (AAVs) can be targeted in a tissue-specific manner, but their tissue accumulation cannot be assessed in a non-invasive manner. Here the authors conjugate a multivalent chelator labelled with Cu-64 to the surface of AAVs and image the brain accumulation of the PHB.eB capsid by PET.
Collapse
Affiliation(s)
- Jai Woong Seo
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA.,Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Lisa Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Spencer Tumbale
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Bo Wu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Sadaf Aghevlian
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Shahin Shams
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Mo Baikoghli
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Poorva Jain
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Xiaozhe Ding
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Nick Goeden
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Tatyana Dobreva
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Nicholas C Flytzanis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Michael Chavez
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Ryan Leib
- Stanford University Mass Spectrometry, Stanford, CA, USA
| | - Michelle L James
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - David J Segal
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Eduardo A Silva
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Katherine W Ferrara
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
36
|
Wadhwa P, Thielemans K, Efthimiou N, Wangerin K, Keat N, Emond E, Deller T, Bertolli O, Deidda D, Delso G, Tohme M, Jansen F, Gunn RN, Hallett W, Tsoumpas C. PET image reconstruction using physical and mathematical modelling for time of flight PET-MR scanners in the STIR library. Methods 2020; 185:110-119. [PMID: 32006678 DOI: 10.1016/j.ymeth.2020.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/15/2019] [Accepted: 01/14/2020] [Indexed: 10/25/2022] Open
Abstract
This work demonstrates how computational and physical modelling of the positron emission tomography (PET) image acquisition process for a state-of-the-art integrated PET and magnetic resonance imaging (PET-MR) system can produce images comparable to the manufacturer. The GE SIGNA PET/MR scanner is manufactured by General Electric and has time-of-flight (TOF) capabilities of about 390 ps. All software development took place in the Software for Tomographic Image Reconstruction (STIR: http://stir.sf.net) library, which is a widely used open source software to reconstruct data as exported from emission tomography scanners. The new software developments will be integrated into STIR, providing the opportunity for researchers worldwide to establish and expand their image reconstruction methods. Furthermore, this work is of particular significance as it provides the first validation of TOF PET image reconstruction for real scanner datasets using the STIR library. This paper presents the methodology, analysis, and critical issues encountered in implementing an independent reconstruction software package. Acquired PET data were processed via several appropriate algorithms which are necessary to produce an accurate and precise quantitative image. This included mathematical, physical and anatomical modelling of the patient and simulation of various aspects of the acquisition. These included modelling of random coincidences using 'singles' rates per crystals, detector efficiencies and geometric effects. Attenuation effects were calculated by using the STIR's attenuation correction model. Modelling all these effects within the system matrix allowed the reconstruction of PET images which demonstrates the metabolic uptake of the administered radiopharmaceutical. These implementations were validated using measured phantom and clinical datasets. The developments are tested using the ordered subset expectation maximisation (OSEM) and the more recently proposed kernelised expectation maximisation (KEM) algorithm which incorporates anatomical information from MR images into PET reconstruction.
Collapse
Affiliation(s)
- Palak Wadhwa
- Biomedical Imaging Science Department, School of Medicine, University of Leeds, UK; Invicro, London, UK.
| | - Kris Thielemans
- Institute of Nuclear Medicine, University College London, UK
| | - Nikos Efthimiou
- PET Research Centre, Faculty of Health Sciences, University of Hull, UK
| | | | | | - Elise Emond
- Institute of Nuclear Medicine, University College London, UK
| | | | | | - Daniel Deidda
- Biomedical Imaging Science Department, School of Medicine, University of Leeds, UK; National Physical Laboratory, Teddington, UK
| | | | | | | | | | | | - Charalampos Tsoumpas
- Biomedical Imaging Science Department, School of Medicine, University of Leeds, UK; Invicro, London, UK.
| |
Collapse
|
37
|
Hansson AC, Gründer G, Hirth N, Noori HR, Spanagel R, Sommer WH. Dopamine and opioid systems adaptation in alcoholism revisited: Convergent evidence from positron emission tomography and postmortem studies. Neurosci Biobehav Rev 2019; 106:141-164. [DOI: 10.1016/j.neubiorev.2018.09.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 09/08/2018] [Accepted: 09/14/2018] [Indexed: 12/20/2022]
|
38
|
Yang BY, Telu S, Haskali MB, Morse CL, Pike VW. A Gas Phase Route to [ 18F]fluoroform with Limited Molar Activity Dilution. Sci Rep 2019; 9:14835. [PMID: 31619702 PMCID: PMC6795885 DOI: 10.1038/s41598-019-50747-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/12/2019] [Indexed: 01/03/2023] Open
Abstract
Positron emission tomography (PET) is an important imaging modality for biomedical research and drug development. PET requires biochemically selective radiotracers to realize full potential. Fluorine-18 (t1/2 = 109.8 min) is a major radionuclide for labeling such radiotracers but is only readily available in high activities from cyclotrons as [18F]fluoride ion. [18F]fluoroform has emerged for labeling tracers in trifluoromethyl groups. Prior methods of [18F]fluoroform synthesis used difluoro precursors in solution and led to high dilution with carrier and low molar activity (Am). We explored a new approach for the synthesis of [18F]fluoroform based on the radiosynthesis of [18F]fluoromethane from [18F]fluoride ion and then cobaltIII fluoride mediated gas phase fluorination. We estimate that carrier dilution in this process is limited to about 3-fold and find that moderate to high Am values can be achieved. We show that [18F]fluoroform so produced is highly versatile for rapidly and efficiently labeling various chemotypes that carry trifluoromethyl groups, thereby expanding prospects for developing new PET radiotracers.
Collapse
Affiliation(s)
- Bo Yeun Yang
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, MD, 20892-1003, USA
| | - Sanjay Telu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, MD, 20892-1003, USA
| | - Mohammad B Haskali
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, MD, 20892-1003, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, MD, 20892-1003, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, MD, 20892-1003, USA.
| |
Collapse
|
39
|
Lois C, Gonzalez I, Johnson KA, Price JC. PET imaging of tau protein targets: a methodology perspective. Brain Imaging Behav 2019; 13:333-344. [PMID: 29497982 PMCID: PMC6119534 DOI: 10.1007/s11682-018-9847-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The two neuropathological hallmarks of Alzheimer's disease (AD) are amyloid-[Formula: see text] plaques and neurofibrillary tangles of tau protein. Fifteen years ago, Positron Emission Tomography (PET) with Pittsburgh Compound B (11C-PiB) enabled selective in-vivo visualization of amyloid-[Formula: see text] plaque deposits and has since provided valuable information about the role of amyloid-[Formula: see text] deposition in AD. The progression of tau deposition has been shown to be highly associated with neuronal loss, neurodegeneration, and cognitive decline. Until recently it was not possible to visualize tau deposition in-vivo, but several tau PET tracers are now available in different stages of clinical development. To date, no tau tracer has been approved by the Food and Drug Administration for use in the evaluation of AD or other tauopathies, despite very active research efforts. In this paper we review the recent developments in tau PET imaging with a focus on in-vivo findings in AD and discuss the challenges associated with tau tracer development, the status of development and validation of different tau tracers, and the clinical information these provide.
Collapse
Affiliation(s)
- Cristina Lois
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA.
| | - Ivan Gonzalez
- Athinoula A. Martinos Center for Biomedical Research, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA
| | - Keith A Johnson
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA
| | - Julie C Price
- Athinoula A. Martinos Center for Biomedical Research, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Caravaggio F, Iwata Y, Kim J, Shah P, Gerretsen P, Remington G, Graff-Guerrero A. What proportion of striatal D2 receptors are occupied by endogenous dopamine at baseline? A meta-analysis with implications for understanding antipsychotic occupancy. Neuropharmacology 2019; 163:107591. [PMID: 30940535 DOI: 10.1016/j.neuropharm.2019.03.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/25/2019] [Accepted: 03/27/2019] [Indexed: 11/30/2022]
Abstract
Using molecular imaging techniques - positron emission tomography (PET) and single-photon emission computed tomography (SPECT) - in conjunction with an acute dopamine depletion challenge (alpha-methyl-para-tyrosine) it is possible to estimate endogenous dopamine levels occupying striatal dopamine D2 receptors (D2R) in humans in vivo. However, it is unclear what proportion of striatal D2R are occupied by endogenous dopamine under normal conditions. This is important since it has been suggested that in schizophrenia there may be a substantial proportion of striatal D2R which are occupied by endogenous dopamine and not accessible by therapeutic doses of antipsychotics. In order to clarify these issues, we conducted a meta-analysis of dopamine depletion studies using substituted benzamide radiotracers in healthy persons. This meta-analysis suggests that anywhere from 8 to 21% (weighted average 11%) of striatal D2R may be occupied by endogenous dopamine at baseline. Using these estimates, we propose an updated occupancy model and tentatively suggest that antipsychotics inhibit a smaller proportion of the total pool of striatal D2R in vivo than previously acknowledged. This article is part of the issue entitled 'Special Issue on Antipsychotics'.
Collapse
Affiliation(s)
- Fernando Caravaggio
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada.
| | - Yusuke Iwata
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Julia Kim
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Institute of Medical Science, University of Toronto, 1 Kings College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Parita Shah
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Institute of Medical Science, University of Toronto, 1 Kings College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Philip Gerretsen
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Institute of Medical Science, University of Toronto, 1 Kings College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Gary Remington
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Institute of Medical Science, University of Toronto, 1 Kings College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Ariel Graff-Guerrero
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Institute of Medical Science, University of Toronto, 1 Kings College Circle, Toronto, Ontario, M5S 1A8, Canada
| |
Collapse
|
41
|
Gong K, Guan J, Kim K, Zhang X, Yang J, Seo Y, El Fakhri G, Qi J, Li Q. Iterative PET Image Reconstruction Using Convolutional Neural Network Representation. IEEE TRANSACTIONS ON MEDICAL IMAGING 2019; 38:675-685. [PMID: 30222554 PMCID: PMC6472985 DOI: 10.1109/tmi.2018.2869871] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
PET image reconstruction is challenging due to the ill-poseness of the inverse problem and limited number of detected photons. Recently, the deep neural networks have been widely and successfully used in computer vision tasks and attracted growing interests in medical imaging. In this paper, we trained a deep residual convolutional neural network to improve PET image quality by using the existing inter-patient information. An innovative feature of the proposed method is that we embed the neural network in the iterative reconstruction framework for image representation, rather than using it as a post-processing tool. We formulate the objective function as a constrained optimization problem and solve it using the alternating direction method of multipliers algorithm. Both simulation data and hybrid real data are used to evaluate the proposed method. Quantification results show that our proposed iterative neural network method can outperform the neural network denoising and conventional penalized maximum likelihood methods.
Collapse
Affiliation(s)
- Kuang Gong
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA, and with the Department of Biomedical Engineering, University of California, Davis CA 95616 USA
| | - Jiahui Guan
- Department of Statistics, University of California, Davis, CA 95616 USA
| | - Kyungsang Kim
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA
| | - Xuezhu Zhang
- Department of Biomedical Engineering, University of California, Davis, CA 95616 USA
| | - Jaewon Yang
- Physics Research Laboratory, Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143 USA
| | - Youngho Seo
- Physics Research Laboratory, Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143 USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA
| | - Jinyi Qi
- Department of Biomedical Engineering, University of California, Davis, CA 95616 USA
| | - Quanzheng Li
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
42
|
Boudjelal A, Messali Z, Elmoataz A, Attallah B. Improved Simultaneous Algebraic Reconstruction Technique Algorithm for Positron-Emission Tomography Image Reconstruction via Minimizing the Fast Total Variation. J Med Imaging Radiat Sci 2017; 48:385-393. [PMID: 31047474 DOI: 10.1016/j.jmir.2017.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/05/2017] [Accepted: 09/15/2017] [Indexed: 12/15/2022]
Abstract
CONTEXT There has been considerable progress in the instrumentation for data measurement and computer methods for generating images of measured PET data. These computer methods have been developed to solve the inverse problem, also known as the "image reconstruction from projections" problem. AIM In this paper, we propose a modified Simultaneous Algebraic Reconstruction Technique (SART) algorithm to improve the quality of image reconstruction by incorporating total variation (TV) minimization into the iterative SART algorithm. METHODOLOGY The SART updates the estimated image by forward projecting the initial image onto the sinogram space. Then, the difference between the estimated sinogram and the given sinogram is back-projected onto the image domain. This difference is then subtracted from the initial image to obtain a corrected image. Fast total variation (FTV) minimization is applied to the image obtained in the SART step. The second step is the result obtained from the previous FTV update. The SART and the FTV minimization steps run iteratively in an alternating manner. Fifty iterations were applied to the SART algorithm used in each of the regularization-based methods. In addition to the conventional SART algorithm, spatial smoothing was used to enhance the quality of the image. All images were sized at 128 × 128 pixels. RESULTS The proposed algorithm successfully accomplished edge preservation. A detailed scrutiny revealed that the reconstruction algorithms differed; for example, the SART and the proposed FTV-SART algorithm effectively preserved the hot lesion edges, whereas artifacts and deviations were more likely to occur in the ART algorithm than in the other algorithms. CONCLUSIONS Compared to the standard SART, the proposed algorithm is more robust in removing background noise while preserving edges to suppress the existent image artifacts. The quality measurements and visual inspections show a significant improvement in image quality compared to the conventional SART and Algebraic Reconstruction Technique (ART) algorithms.
Collapse
Affiliation(s)
- Abdelwahhab Boudjelal
- Electronics Department, University of Mohammed Boudiaf-M'sila, M'sila, Algeria; Image Team, GREYC Laboratory, University of Caen Normandy, Caen Cedex, France.
| | - Zoubeida Messali
- Electronics Department, University of Mohamed El Bachir El Ibrahimi-Bordj Bou Arréridj, Bordj Bou Arréridj, Algeria
| | - Abderrahim Elmoataz
- Image Team, GREYC Laboratory, University of Caen Normandy, Caen Cedex, France
| | - Bilal Attallah
- Electronics Department, University of Mohammed Boudiaf-M'sila, M'sila, Algeria; Image Team, GREYC Laboratory, University of Caen Normandy, Caen Cedex, France
| |
Collapse
|
43
|
|
44
|
Lever SZ, Fan KH, Lever JR. Tactics for preclinical validation of receptor-binding radiotracers. Nucl Med Biol 2017; 44:4-30. [PMID: 27755986 PMCID: PMC5161541 DOI: 10.1016/j.nucmedbio.2016.08.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 08/24/2016] [Accepted: 08/24/2016] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Aspects of radiopharmaceutical development are illustrated through preclinical studies of [125I]-(E)-1-(2-(2,3-dihydrobenzofuran-5-yl)ethyl)-4-(iodoallyl)piperazine ([125I]-E-IA-BF-PE-PIPZE), a radioligand for sigma-1 (σ1) receptors, coupled with examples from the recent literature. Findings are compared to those previously observed for [125I]-(E)-1-(2-(2,3-dimethoxy-5-yl)ethyl)-4-(iodoallyl)piperazine ([125I]-E-IA-DM-PE-PIPZE). METHODS Syntheses of E-IA-BF-PE-PIPZE and [125I]-E-IA-BF-PE-PIPZE were accomplished by standard methods. In vitro receptor binding studies and autoradiography were performed, and binding potential was predicted. Measurements of lipophilicity and protein binding were obtained. In vivo studies were conducted in mice to evaluate radioligand stability, as well as specific binding to σ1 sites in brain, brain regions and peripheral organs in the presence and absence of potential blockers. RESULTS E-IA-BF-PE-PIPZE exhibited high affinity and selectivity for σ1 receptors (Ki = 0.43 ± 0.03 nM, σ2/σ1 = 173). [125I]-E-IA-BF-PE-PIPZE was prepared in good yield and purity, with high specific activity. Radioligand binding provided dissociation (koff) and association (kon) rate constants, along with a measured Kd of 0.24 ± 0.01 nM and Bmax of 472 ± 13 fmol/mg protein. The radioligand proved suitable for quantitative autoradiography in vitro using brain sections. Moderate lipophilicity, Log D7.4 2.69 ± 0.28, was determined, and protein binding was 71 ± 0.3%. In vivo, high initial whole brain uptake, >6% injected dose/g, cleared slowly over 24 h. Specific binding represented 75% to 93% of total binding from 15 min to 24 h. Findings were confirmed and extended by regional brain biodistribution. Radiometabolites were not observed in brain (1%). CONCLUSIONS Substitution of dihydrobenzofuranylethyl for dimethoxyphenethyl increased radioligand affinity for σ1 receptors by 16-fold. While high specific binding to σ1 receptors was observed for both radioligands in vivo, [125I]-E-IA-BF-PE-PIPZE displayed much slower clearance kinetics than [125I]-E-IA-DM-PE-PIPZE. Thus, minor structural modifications of σ1 receptor radioligands lead to major differences in binding properties in vitro and in vivo.
Collapse
Affiliation(s)
- Susan Z Lever
- Department of Chemistry, University of Missouri, Columbia, MO, USA; University of Missouri Research Reactor Center, Columbia, MO, USA.
| | - Kuo-Hsien Fan
- Department of Chemistry, University of Missouri, Columbia, MO, USA
| | - John R Lever
- Department of Radiology, University of Missouri, Columbia, MO, USA; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA.
| |
Collapse
|
45
|
De Picker LJ, Morrens M, Chance SA, Boche D. Microglia and Brain Plasticity in Acute Psychosis and Schizophrenia Illness Course: A Meta-Review. Front Psychiatry 2017; 8:238. [PMID: 29201010 PMCID: PMC5696326 DOI: 10.3389/fpsyt.2017.00238] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Schizophrenia poses a tremendous health, social, and economic burden upon patients and society, indicating current treatment options remain inadequate. Recent findings from several lines of evidence have pointed to the importance of immune system involvement in not only premorbid neurodevelopmental but also subsequent symptom generation and aging processes of brain change in schizophrenia. In this meta-review, we use the summarized evidence from recent quantitative systematic reviews (SRs) and meta-analyses of several subspecialties to critically evaluate the hypothesis that immune-related processes shape the symptomatic presentation and illness course of schizophrenia, both directly and indirectly through altered neuroplasticity. METHODS We performed a data search in PubMed for English language SRs and meta-analyses from 2010 to 2017. The methodological quality of the SRs was assessed with the AMSTAR instrument. In addition, we review in this paper 11 original publications on translocator protein (TSPO) positron emission tomography (PET) imaging in schizophrenia. RESULTS We reviewed 26 SRs and meta-analyses. Evidence from clinical observational studies of inflammatory or immunological markers and randomized controlled drug trials of immunomodulatory compounds as add-on in the treatment of schizophrenia suggests psychotic exacerbations are accompanied by immunological changes different from those seen in non-acute states, and that the symptoms of schizophrenia can be modified by compounds such as non-steroidal anti-inflammatory drug and minocycline. Information derived from post-mortem brain tissue analysis and PET neuroimaging studies to evaluate microglial activation have added new perspectives to the available evidence, yet these results are very heterogeneous. Each research domain comes with unique opportunities as well as inherent limitations. A better understanding of the (patho-)physiology of microglial cells and their role in neuroplasticity is key to interpreting the immune-related findings in the context of schizophrenia illness exacerbations and progression. CONCLUSION Evidence from clinical studies analyzing patients' blood and cerebrospinal fluid samples, neuroimaging and post-mortem brain tissue suggests that aberrant immune responses may define schizophrenia illness' course through altered neuroplasticity representing abnormal aging processes. Most findings are however prone to bias and confounding, and often non-specific to schizophrenia, and a multidisciplinary translational approach is needed to consolidate these findings and link them to other schizophrenia hypotheses.
Collapse
Affiliation(s)
- Livia J De Picker
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium.,University Psychiatric Center St. Norbertus, Duffel, Belgium
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium.,University Psychiatric Center St. Norbertus, Duffel, Belgium
| | - Steven A Chance
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
46
|
Sadeghipour N, Davis SC, Tichauer KM. Generalized paired-agent kinetic model for in vivo quantification of cancer cell-surface receptors under receptor saturation conditions. Phys Med Biol 2016; 62:394-414. [PMID: 27997381 DOI: 10.1088/1361-6560/62/2/394] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
New precision medicine drugs oftentimes act through binding to specific cell-surface cancer receptors, and thus their efficacy is highly dependent on the availability of those receptors and the receptor concentration per cell. Paired-agent molecular imaging can provide quantitative information on receptor status in vivo, especially in tumor tissue; however, to date, published approaches to paired-agent quantitative imaging require that only 'trace' levels of imaging agent exist compared to receptor concentration. This strict requirement may limit applicability, particularly in drug binding studies, which seek to report on a biological effect in response to saturating receptors with a drug moiety. To extend the regime over which paired-agent imaging may be used, this work presents a generalized simplified reference tissue model (GSRTM) for paired-agent imaging developed to approximate receptor concentration in both non-receptor-saturated and receptor-saturated conditions. Extensive simulation studies show that tumor receptor concentration estimates recovered using the GSRTM are more accurate in receptor-saturation conditions than the standard simple reference tissue model (SRTM) (% error (mean ± sd): GSRTM 0 ± 1 and SRTM 50 ± 1) and match the SRTM accuracy in non-saturated conditions (% error (mean ± sd): GSRTM 5 ± 5 and SRTM 0 ± 5). To further test the approach, GSRTM-estimated receptor concentration was compared to SRTM-estimated values extracted from tumor xenograft in vivo mouse model data. The GSRTM estimates were observed to deviate from the SRTM in tumors with low receptor saturation (which are likely in a saturated regime). Finally, a general 'rule-of-thumb' algorithm is presented to estimate the expected level of receptor saturation that would be achieved in a given tissue provided dose and pharmacokinetic information about the drug or imaging agent being used, and physiological information about the tissue. These studies suggest that the GSRTM is necessary when receptor saturation exceeds 20% and highlight the potential for GSRTM to accurately measure receptor concentrations under saturation conditions, such as might be required during high dose drug studies, or for imaging applications where high concentrations of imaging agent are required to optimize signal-to-noise conditions. This model can also be applied to PET and SPECT imaging studies that tend to suffer from noisier data, but require one less parameter to fit if images are converted to imaging agent concentration (quantitative PET/SPECT).
Collapse
Affiliation(s)
- N Sadeghipour
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
| | | | | |
Collapse
|
47
|
Abstract
The discovery and development of central nervous system (CNS) drugs is an extremely challenging process requiring large resources, timelines, and associated costs. The high risk of failure leads to high levels of risk. Over the past couple of decades PET imaging has become a central component of the CNS drug-development process, enabling decision-making in phase I studies, where early discharge of risk provides increased confidence to progress a candidate to more costly later phase testing at the right dose level or alternatively to kill a compound through failure to meet key criteria. The so called "3 pillars" of drug survival, namely; tissue exposure, target engagement, and pharmacologic activity, are particularly well suited for evaluation by PET imaging. This review introduces the process of CNS drug development before considering how PET imaging of the "3 pillars" has advanced to provide valuable tools for decision-making on the critical path of CNS drug development. Finally, we review the advances in PET science of biomarker development and analysis that enable sophisticated drug-development studies in man.
Collapse
Affiliation(s)
- Roger N Gunn
- Imanova Ltd, London, United Kingdom; Division of Brain Sciences, Imperial College London, London, United Kingdom; Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom.
| | - Eugenii A Rabiner
- Imanova Ltd, London, United Kingdom; Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, United Kingdom
| |
Collapse
|
48
|
Slifstein M, Abi-Dargham A. Recent Developments in Molecular Brain Imaging of Neuropsychiatric Disorders. Semin Nucl Med 2016; 47:54-63. [PMID: 27987558 DOI: 10.1053/j.semnuclmed.2016.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Molecular imaging with PET or SPECT has been an important research tool in psychiatry for as long as these modalities have been available. Here, we discuss two areas of neuroimaging relevant to current psychiatry research. The first is the use of imaging to study neurotransmission. We discuss the use of pharmacologic probes to induce changes in levels of neurotransmitters that can be inferred through their effects on outcome measures of imaging experiments, from their historical origins focusing on dopamine transmission through recent developments involving serotonin, GABA, and glutamate. Next, we examine imaging of neuroinflammation in the context of psychiatry. Imaging markers of neuroinflammation have been studied extensively in other areas of brain research, but they have more recently attracted interest in psychiatry research, based on accumulating evidence that there may be an inflammatory component to some psychiatric conditions. Furthermore, new probes are under development that would allow unprecedented insights into cellular processes. In summary, molecular imaging would continue to offer great potential as a unique tool to further our understanding of brain function in health and disease.
Collapse
Affiliation(s)
- Mark Slifstein
- Department of Psychiatry, Columbia University Medical Center, New York, NY; New York State Psychiatric Institute, New York, NY; Department of Psychiatry, Stony Brook University, New York, NY.
| | - Anissa Abi-Dargham
- Department of Psychiatry, Columbia University Medical Center, New York, NY; Department of Radiology, Columbia University Medical Center, New York, NY; New York State Psychiatric Institute, New York, NY; Department of Psychiatry, Stony Brook University, New York, NY
| |
Collapse
|
49
|
Caravaggio F, Kegeles LS, Wilson AA, Remington G, Borlido C, Mamo DC, Graff-Guerrero A. Estimating the effect of endogenous dopamine on baseline [(11) C]-(+)-PHNO binding in the human brain. Synapse 2016; 70:453-60. [PMID: 27341789 DOI: 10.1002/syn.21920] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/03/2016] [Accepted: 06/20/2016] [Indexed: 11/10/2022]
Abstract
Endogenous dopamine (DA) levels at dopamine D2/3 receptors (D2/3 R) have been quantified in the living human brain using the agonist radiotracer [(11) C]-(+)-PHNO. As an agonist radiotracer, [(11) C]-(+)-PHNO is more sensitive to endogenous DA levels than antagonist radiotracers. We sought to determine the proportion of the variance in baseline [(11) C]-(+)-PHNO binding to D2/3 Rs which can be accounted for by variation in endogenous DA levels. This was done by computing the Pearson's coefficient for the correlation between baseline binding potential (BPND ) and the change in BPND after acute DA depletion, using previously published data. All correlations were inverse, and the proportion of the variance in baseline [(11) C]-(+)-PHNO BPND that can be accounted for by variation in endogenous DA levels across the striatal subregions ranged from 42-59%. These results indicate that lower baseline values of [(11) C]-(+)-PHNO BPND reflect greater stimulation by endogenous DA. To further validate this interpretation, we sought to examine whether these data could be used to estimate the dissociation constant (Kd) of DA at D2/3 R. In line with previous in vitro work, we estimated the in vivo Kd of DA to be around 20 nM. In summary, the agonist radiotracer [(11) C]-(+)-PHNO can detect the impact of endogenous DA levels at D2/3 R in the living human brain from a single baseline scan, and may be more sensitive to this impact than other commonly employed radiotracers.
Collapse
Affiliation(s)
- Fernando Caravaggio
- Centre for Addiction and Mental Health, Research Imaging Centre, Toronto, Ontario, M5T 1R8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Lawrence S Kegeles
- Department of Psychiatry and Radiology, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, New York
| | - Alan A Wilson
- Centre for Addiction and Mental Health, Research Imaging Centre, Toronto, Ontario, M5T 1R8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.,Department of Psychiatry, University of Toronto, Ontario, M5T 1R8, Canada
| | - Gary Remington
- Centre for Addiction and Mental Health, Research Imaging Centre, Toronto, Ontario, M5T 1R8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.,Department of Psychiatry, University of Toronto, Ontario, M5T 1R8, Canada
| | - Carol Borlido
- Centre for Addiction and Mental Health, Research Imaging Centre, Toronto, Ontario, M5T 1R8, Canada
| | - David C Mamo
- Department of Psychiatry, Faculties of Medicine and Health Science, University of Malta, Msida, Malta
| | - Ariel Graff-Guerrero
- Centre for Addiction and Mental Health, Research Imaging Centre, Toronto, Ontario, M5T 1R8, Canada. .,Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada. .,Department of Psychiatry, University of Toronto, Ontario, M5T 1R8, Canada.
| |
Collapse
|
50
|
Gong K, Majewski S, Kinahan PE, Harrison RL, Elston BF, Manjeshwar R, Dolinsky S, Stolin AV, Brefczynski-Lewis JA, Qi J. Designing a compact high performance brain PET scanner-simulation study. Phys Med Biol 2016; 61:3681-97. [PMID: 27081753 DOI: 10.1088/0031-9155/61/10/3681] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The desire to understand normal and disordered human brain function of upright, moving persons in natural environments motivates the development of the ambulatory micro-dose brain PET imager (AMPET). An ideal system would be light weight but with high sensitivity and spatial resolution, although these requirements are often in conflict with each other. One potential approach to meet the design goals is a compact brain-only imaging device with a head-sized aperture. However, a compact geometry increases parallax error in peripheral lines of response, which increases bias and variance in region of interest (ROI) quantification. Therefore, we performed simulation studies to search for the optimal system configuration and to evaluate the potential improvement in quantification performance over existing scanners. We used the Cramér-Rao variance bound to compare the performance for ROI quantification using different scanner geometries. The results show that while a smaller ring diameter can increase photon detection sensitivity and hence reduce the variance at the center of the field of view, it can also result in higher variance in peripheral regions when the length of detector crystal is 15 mm or more. This variance can be substantially reduced by adding depth-of-interaction (DOI) measurement capability to the detector modules. Our simulation study also shows that the relative performance depends on the size of the ROI, and a large ROI favors a compact geometry even without DOI information. Based on these results, we propose a compact 'helmet' design using detectors with DOI capability. Monte Carlo simulations show the helmet design can achieve four-fold higher sensitivity and resolve smaller features than existing cylindrical brain PET scanners. The simulations also suggest that improving TOF timing resolution from 400 ps to 200 ps also results in noticeable improvement in image quality, indicating better timing resolution is desirable for brain imaging.
Collapse
Affiliation(s)
- Kuang Gong
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|