1
|
Wu H, Liu G, Ruan X, Zhang B, Zhe Z, Shi H. A low-dose protocol in pediatric 18F-FDG scans using 30-cm axis field of view PET/CT. Ann Nucl Med 2025; 39:546-551. [PMID: 40019733 DOI: 10.1007/s12149-025-02030-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVE To explore the feasibility of a low-dose 18F-FDG protocol for the 30-cm standard axial field of view (SAFOV) PET/CT imaging in pediatric patients. METHODS A retrospective analysis was conducted on 112 pediatric patients who underwent a full-dose (3.7 MBq/kg) 18F-FDG PET/CT imaging, and a prospective analysis was performed on 55 patients who received a low-dose (2.5 MBq/kg) imaging. PET images were reconstructed at 1.0-min/bed intervals, and labeled as G1.0, G2.0, G3.0 for the full-dose imaging and G1.0', G2.0', G3.0' for the low-dose imaging. Patients were categorized into three age groups, and the image quality was assessed using the Likert scale and signal-to-noise ratio (SNR); Lesion detectability was evaluated using lesion detection rates and the target-to-liver ratio (TLR). RESULTS In G2.0 and G3.0, all cases (112/112) achieved an image score of ≥ 3 and a lesion detection rate of 100% (98/98). There were no significant differences in SNR between G2.0 and G3.0 (11.09 ± 2.31 vs. 11.88 ± 2.58, p = 0.39), nor between age groups ≤ 5 years and 6-10 years groups (9.52 ± 3.16 vs. 9.53 ± 3.19, p = 0.99). In G3.0', 98.2% of cases (54/55) had an image score ≥ 3 and a lesion detection rate of 100% (43/43). The SNR of every age group for G3.0' was comparable to that of G2.0, and no significant differences between ≤ 5 years and 6-10 years groups (9.32 ± 1.94 vs. 9.99 ± 2.28, p = 0.82). CONCLUSIONS A 2.5 MBq/kg dose with a 3.0 min/bed acquisition protocol is feasible for 18F-FDG 30-cm SAFOV PET/CT imaging in pediatric patients, and SNR and TLR demonstrated age-dependent discrepancies.
Collapse
Affiliation(s)
- Ha Wu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Nuclear Medicine, Children's Hospital, Fudan University, Shanghai, 201102, China
| | - Guobing Liu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiemei Ruan
- Department of Nuclear Medicine, Children's Hospital, Fudan University, Shanghai, 201102, China
| | - Biying Zhang
- Department of Nuclear Medicine, Children's Hospital, Fudan University, Shanghai, 201102, China
| | - Zheng Zhe
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, China.
- Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China.
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Hashimoto F, Ote K, Onishi Y, Tashima H, Akamatsu G, Iwao Y, Takahashi M, Yamaya T. Exploiting network optimization stability for enhanced PET image denoising using deep image prior. Phys Med Biol 2025; 70:105019. [PMID: 40341245 DOI: 10.1088/1361-6560/add63f] [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: 02/16/2025] [Accepted: 05/08/2025] [Indexed: 05/10/2025]
Abstract
Objective. Positron emission tomography (PET) is affected by statistical noise due to constraints on tracer dose and scan duration, impacting both diagnostic performance and quantitative accuracy. While deep learning-based PET denoising methods have been used to improve image quality, they may introduce over-smoothing, which can obscure critical structural details and compromise quantitative accuracy. We propose a method for making a deep learning solution more reliable and apply it to the conditional deep image prior (DIP).Approach. We introduce the idea ofstability informationin the optimization process of conditional DIP, enabling the identification of unstable regions within the network's optimization trajectory. Our method incorporates a stability map, which is derived from multiple intermediate outputs of a moderate neural network at different optimization steps. The final denoised PET image is then obtained by computing a linear combination of the DIP output and the original reconstructed PET image, weighted by the stability map.Main results. We employed eight high-resolution brain PET datasets for comparison. Our method effectively reduces background noise while preserving small structure details in brain [18F]FDG PET images. Comparative analysis demonstrated that our approach outperformed existing methods in terms of peak-to-valley ratio and background noise suppression across various low-dose levels. Additionally, region-of-interest analysis confirmed that the proposed method maintains quantitative accuracy without introducing under- or over-estimation. Furthermore, we applied our method to full-dose PET data to assess its impact on image quality. The results revealed that the proposed method significantly reduced background noise while preserving the peak-to-valley ratio at a level comparable to that of unfiltered full-dose PET images.Significance. The proposed method introduces a robust approach to deep learning-based PET denoising, enhancing its reliability and preserving quantitative accuracy. Furthermore, this strategy can potentially advance performance in high-sensitivity PET scanners and surpass the limit of image quality inherent to PET scanners.
Collapse
Affiliation(s)
- Fumio Hashimoto
- Central Research Laboratory, Hamamatsu Photonics K. K., 5000 Hirakuchi, Hamana-ku, Hamamatsu 434-8601, Japan
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Kibo Ote
- Central Research Laboratory, Hamamatsu Photonics K. K., 5000 Hirakuchi, Hamana-ku, Hamamatsu 434-8601, Japan
| | - Yuya Onishi
- Central Research Laboratory, Hamamatsu Photonics K. K., 5000 Hirakuchi, Hamana-ku, Hamamatsu 434-8601, Japan
| | - Hideaki Tashima
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Go Akamatsu
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Yuma Iwao
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Miwako Takahashi
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Taiga Yamaya
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
3
|
Chung KJ, Chaudhari AJ, Nardo L, Jones T, Chen MS, Badawi RD, Cherry SR, Wang G. Quantitative Total-Body Imaging of Blood Flow with High-Temporal-Resolution Early Dynamic 18F-FDG PET Kinetic Modeling. J Nucl Med 2025:jnumed.124.268706. [PMID: 40306973 DOI: 10.2967/jnumed.124.268706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 04/08/2025] [Indexed: 05/02/2025] Open
Abstract
Past efforts to measure blood flow with the widely available radiotracer 18F-FDG were limited to tissues with high 18F-FDG extraction fraction. In this study, we developed an early dynamic 18F-FDG PET method with high-temporal-resolution (HTR) kinetic modeling to assess total-body blood flow based on deriving the vascular phase of 18F-FDG transit and conducted a pilot comparison study against a 11C-butanol flow-tracer reference. Methods: The first 2 min of dynamic PET scans were reconstructed at HTR (60 × 1 s/frame, 30 × 2 s/frame) to resolve the rapid passage of the radiotracer through blood vessels. In contrast to existing methods that use blood-to-tissue transport rate as a surrogate of blood flow, our method directly estimated blood flow using a distributed kinetic model (adiabatic approximation to tissue homogeneity [AATH] model). To validate our 18F-FDG measurements of blood flow against a reference flow-specific radiotracer, we analyzed total-body dynamic PET images of 6 human participants scanned with both 18F-FDG and 11C-butanol. An additional 34 total-body dynamic 18F-FDG PET images of healthy participants were analyzed for comparison against published blood-flow ranges. Regional blood flow was estimated across the body, and total-body parametric imaging of blood flow was conducted for visual assessment. AATH and standard compartment model fitting was compared using the Akaike information criterion at different temporal resolutions. Results: 18F-FDG blood flow was in quantitative agreement with flow measured from 11C-butanol across same-subject regional measurements (Pearson correlation coefficient, 0.955; P < 0.001; linear regression slope and intercept, 0.973 and -0.012, respectively), which was visually corroborated by total-body blood-flow parametric imaging. Our method resolved a wide range of blood-flow values across the body in broad agreement with published ranges (e.g., healthy cohort values of 0.51 ± 0.12 mL/min/cm3 in the cerebral cortex and 2.03 ± 0.64 mL/min/cm3 in the lungs). HTR (1-2 s/frame) was required for AATH modeling. Conclusion: Total-body blood-flow imaging was feasible using early dynamic 18F-FDG PET with HTR kinetic modeling. This method may be combined with standard 18F-FDG PET methods to enable efficient single-tracer multiparametric flow-metabolism imaging, with numerous research and clinical applications in oncology, cardiovascular disease, pain medicine, and neuroscience.
Collapse
Affiliation(s)
- Kevin J Chung
- Department of Radiology, University of California Davis Health, Sacramento, California;
| | - Abhijit J Chaudhari
- Department of Radiology, University of California Davis Health, Sacramento, California
| | - Lorenzo Nardo
- Department of Radiology, University of California Davis Health, Sacramento, California
| | - Terry Jones
- Department of Radiology, University of California Davis Health, Sacramento, California
| | - Moon S Chen
- Department of Internal Medicine, University of California Davis Health, Sacramento, California; and
| | - Ramsey D Badawi
- Department of Radiology, University of California Davis Health, Sacramento, California
- Department of Biomedical Engineering, University of California at Davis, Davis, California
| | - Simon R Cherry
- Department of Radiology, University of California Davis Health, Sacramento, California
- Department of Biomedical Engineering, University of California at Davis, Davis, California
| | - Guobao Wang
- Department of Radiology, University of California Davis Health, Sacramento, California
| |
Collapse
|
4
|
Dubroff JG, Hsieh CJ, Wiers CE, Lee H, Schmitz A, Li EJ, Schubert EK, Mach RH, Kranzler HR. [ 11C]Carfentanil PET Whole-Body Imaging of μ-Opioid Receptors: A First in-Human Study. J Nucl Med 2025:jnumed.124.269413. [PMID: 40341091 DOI: 10.2967/jnumed.124.269413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/08/2025] [Indexed: 05/10/2025] Open
Abstract
μ-opioid receptors (MORs) are G-coupled receptors widely expressed in the brain and body. MORs have a high affinity for both endogenous opioids such as β-endorphins and exogenous opioids such as fentanyl. They mediate pain and reward and have been implicated in the pathophysiology of opioid, cocaine, and other substance use disorders. Using an instrument with a long axial field of view and the MOR-selective radioligand [11C]carfentanil, we measured the whole-body distribution of MORs in 13 healthy humans. We also examined sex differences in MOR distribution at baseline and after pretreatment with the MOR antagonist naloxone. Methods: Six female and 7 male healthy subjects underwent 2 [11C]carfentanil PET imaging sessions-one at baseline and one immediately after pretreatment with the MOR antagonist naloxone (13 μg/kg). Whole-body PET imaging was performed on an instrument with a 142-cm axial bore. [11C]carfentanil brain distribution volume ratios were determined using the occipital cortex and the visual cortex within it as reference regions. For peripheral organ distribution volume ratios, the descending aorta and proximal-extremity muscle (biceps/triceps) were used as reference regions. Results: Naloxone blockade reduced MOR availability by 40%-50% in the caudate, putamen, thalamus, amygdala, and ventral tegmentum, brain regions known to express high levels of MORs. Women showed greater receptor occupancy in the thalamus, amygdala, hippocampus, and frontal and temporal lobes and a greater naloxone-induced reduction in thalamic MOR availability than men (P < 0.05). For determining brain MOR availability, there was less variance in the visual cortex than in the occipital cortex reference region. For peripheral MOR determination, the descending aorta reference region showed less variance than the extremity muscle, but both showed blocking effects of naloxone. Conclusion: [11C]carfentanil whole-body PET scans are useful for understanding MOR physiology under both baseline and blocking conditions. Extra-central nervous system reference regions may be useful for quantifying radiotracers when a region devoid of binding in the central nervous system is unavailable. The long axial field of view was useful for measuring changes in the short-lived radiotracer [11C]carfentanil, with and without naloxone blocking. Further research is needed to evaluate the behavioral and clinical relevance of sex differences in naloxone-MOR interactions.
Collapse
Affiliation(s)
- Jacob G Dubroff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania;
| | - Chia-Ju Hsieh
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Corinde E Wiers
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Hsiaoju Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexander Schmitz
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth J Li
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Erin K Schubert
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Henry R Kranzler
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, Pennsylvania
| |
Collapse
|
5
|
Razdevšek G, Fakhri GE, Marin T, Dolenec R, Orehar M, Chemli Y, Gola AG, Gascon D, Majewski S, Pestotnik R. Flexible and modular PET: Evaluating the potential of TOF-DOI panel detectors. Med Phys 2025; 52:2845-2860. [PMID: 40089973 PMCID: PMC12059530 DOI: 10.1002/mp.17741] [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: 10/08/2024] [Revised: 02/07/2025] [Accepted: 02/19/2025] [Indexed: 03/18/2025] Open
Abstract
BACKGROUND Panel detectors have the potential to provide a flexible, modular approach to Positron Emission Tomography (PET), enabling customization to meet patient-specific needs and scan objectives. The panel design allows detectors to be positioned close to the patient, aiming to enhance sensitivity and spatial resolution through improved geometric coverage and reduced noncollinearity blurring. Parallax error can be mitigated using depth of interaction (DOI) information. PURPOSE One of the key questions the article addresses is: Do panel detectors offer viable clinical imaging capabilities, or does limited angular sampling restrict their utility by causing image distortions and artifacts? Additionally, this article explores the scalability of panel detectors for constructing scanners with a long axial field of view (LAFOV). METHODS Monte Carlo simulations using GATE software were used to assess the performance of panel detectors with various DOI resolutions and Time-of-Flight (TOF) resolutions as fine as 70 ps. The 30 × $\times$ 30 cm panels comprised pixelated 3 × $\times$ 3 × $\times$ 20 mm LSO crystals. Simulations were run on large high-performance computing clusters (122,000 CPU cores). Open-source CASToR software was used for (TOF MLEM) image reconstruction. The image quality of the scanners was assessed using a range of phantoms (NEMA, Derenzo, XCAT, and a high-resolution brain phantom). The Siemens Biograph Vision PET/CT scanner served as the reference model. The performance of larger 120 × $\times$ 60 cm panels was also evaluated. RESULTS Sensitivity increases over threefold when panel-panel distance is reduced from 80 to 40 cm. The noise equivalent count rate, unmodified by TOF gain, of the panel detectors matches that of the reference clinical scanner at a distance of approximately 50 cm between the panels. Spatial resolution perpendicular to the panels improves from 8.7 to 1.6 mm when the panel-panel distance is reduced, and 70 ps + DOI detectors are used instead of 200 ps, no-DOI detectors. With enhanced TOF and DOI capabilities, panel detectors achieve image quality that matches or surpasses the reference scanner while using about four times less detector material. These detectors can be extended for LAFOV imaging without distortions or artifacts. Additionally, improving TOF and DOI performance enhances contrast-to-noise ratios, thereby improving lesion detection. CONCLUSIONS A compact 2-panel PET scanner can match the performance of conventional scanners, producing high-quality, distortion-free images. Its mobility and flexibility enable novel applications, including bedside imaging and intensive care unitdiagnostics, as well as imaging in positions such as sitting or standing. Furthermore, the modularity of panel detectors offers the potential to construct cost-effective, high-performance total-body imaging systems.
Collapse
Affiliation(s)
- Gašper Razdevšek
- Experimental Particle Physics Department (F9)Jožef Stefan InstituteLjubljanaSlovenia
| | - Georges El Fakhri
- Yale PET CenterYale University School of MedicineNew HavenConnecticutUSA
| | - Thibault Marin
- Yale PET CenterYale University School of MedicineNew HavenConnecticutUSA
| | - Rok Dolenec
- Experimental Particle Physics Department (F9)Jožef Stefan InstituteLjubljanaSlovenia
- Faculty of Mathematics and PhysicsUniversity of LjubljanaLjubljanaSlovenia
| | - Matic Orehar
- Faculty of Mathematics and PhysicsUniversity of LjubljanaLjubljanaSlovenia
| | - Yanis Chemli
- Yale PET CenterYale University School of MedicineNew HavenConnecticutUSA
| | | | - David Gascon
- Institute of Cosmos Sciences (ICCUB)University of BarcelonaBarcelonaSpain
| | - Stan Majewski
- Biomedical EngineeringUniversity of California DavisDavisUSA
| | - Rok Pestotnik
- Experimental Particle Physics Department (F9)Jožef Stefan InstituteLjubljanaSlovenia
| |
Collapse
|
6
|
Baran J, Krzemien W, Parzych S, Raczyński L, Bała M, Coussat A, Chug N, Czerwiński E, Curceanu CO, Dadgar M, Dulski K, Eliyan K, Gajewski J, Gajos A, Hiesmayr BC, Kacprzak K, Kapłon Ł, Klimaszewski K, Korcyl G, Kozik T, Kumar D, Niedźwiecki S, Panek D, Perez Del Rio E, Ruciński A, Sharma S, Shivani, Shopa RY, Skurzok M, Stępień E, Tayefiardebili F, Tayefiardebili K, Wiślicki W, Moskal P. Realistic total-body J-PET geometry optimization: Monte Carlo study. Med Phys 2025; 52:2961-2975. [PMID: 39853786 DOI: 10.1002/mp.17627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/26/2024] [Accepted: 12/28/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Total-body (TB) Positron Emission Tomography (PET) is one of the most promising medical diagnostics modalities, opening new perspectives for personalized medicine, low-dose imaging, multi-organ dynamic imaging or kinetic modeling. The high sensitivity provided by total-body technology can be advantageous for novel tomography methods like positronium imaging, demanding the registration of triple coincidences. Currently, state-of-the-art PET scanners use inorganic scintillators. However, the high acquisition cost reduces the accessibility of TB PET technology. Several efforts are ongoing to mitigate this problem. Among the alternatives, the Jagiellonian PET (J-PET) technology, based on axially arranged plastic scintillator strips, offers a low-cost alternative solution for TB PET. PURPOSE The work aimed to compare five total-body J-PET geometries with plastic scintillators suitable for multi-organ and positronium tomography as a possible next-generation J-PET scanner design. METHODS We present comparative studies of performance characteristics of the cost-effective total-body PET scanners using J-PET technology. We investigated in silico five TB scanner geometries, varying the number of rings, scanner radii, and other parameters. Monte Carlo simulations of the anthropomorphic XCAT phantom, the extended 2-m sensitivity line source and positronium sensitivity phantoms were used to assess the performance of the geometries. Two hot spheres were placed in the lungs and in the liver of the XCAT phantom to mimic the pathological changes. We compared the sensitivity profiles and performed quantitative analysis of the reconstructed images by using quality metrics such as contrast recovery coefficient, background variability and root mean squared error. The studies are complemented by the determination of sensitivity for the positronium lifetime tomography and the relative cost analysis of the studied setups. RESULTS The analysis of the reconstructed XCAT images reveals the superiority of the seven-ring scanners over the three-ring setups. However, the three-ring scanners would be approximately 2-3 times cheaper. The peak sensitivity values for two-gamma vary from 20 to 34 cps/kBq and are dominated by the differences in geometrical acceptance of the scanners. The sensitivity curves for the positronium tomography have a similar shape to the two-gamma sensitivity profiles. The peak values are lower compared to the two-gamma cases, from about 20-28 times, with a maximum value of 1.66 cps/kBq. This can be contrasted with the 50-cm one-layer J-PET modular scanner used to perform the first in-vivo positronium imaging with a sensitivity of 0.06 cps/kBq. CONCLUSIONS The results show the feasibility of multi-organ imaging of all the systems to be considered for the next generation of TB J-PET designs. Among the scanner parameters, the most important ones are related to the axial field-of-view coverage. The two-gamma sensitivity and XCAT image reconstruction analyzes show the advantage of seven-ring scanners. However, the cost of the scintillator materials and SiPMs is more than two times higher for the longer modalities compared to the three-ring solutions. Nevertheless, the relative cost for all the scanners is about 10-4 times lower compared to the cost of the uExplorer. These properties coupled together with J-PET cost-effectiveness and triggerless acquisition mode enabling three-gamma positronium imaging, make the J-PET technology an attractive solution for broad application in clinics.
Collapse
Affiliation(s)
- Jakub Baran
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Wojciech Krzemien
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
- High Energy Physics Division, National Centre for Nuclear Research, Otwock-Świerk, Poland
| | - Szymon Parzych
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Lech Raczyński
- Department of Complex Systems, National Centre for Nuclear Research, Otwock-Świerk, Poland
| | - Mateusz Bała
- Department of Complex Systems, National Centre for Nuclear Research, Otwock-Świerk, Poland
| | - Aurélien Coussat
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Neha Chug
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Eryk Czerwiński
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | | | - Meysam Dadgar
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Kamil Dulski
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Kavya Eliyan
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Jan Gajewski
- Cyclotron Centre Bronowice, Institute of Nuclear Physics Polish Academy of Sciences, Kraków, Poland
| | - Aleksander Gajos
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | | | - Krzysztof Kacprzak
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Łukasz Kapłon
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Konrad Klimaszewski
- Department of Complex Systems, National Centre for Nuclear Research, Otwock-Świerk, Poland
| | - Grzegorz Korcyl
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Tomasz Kozik
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Deepak Kumar
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Szymon Niedźwiecki
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Dominik Panek
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Elena Perez Del Rio
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Antoni Ruciński
- Cyclotron Centre Bronowice, Institute of Nuclear Physics Polish Academy of Sciences, Kraków, Poland
| | - Sushil Sharma
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Shivani
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Roman Y Shopa
- Department of Complex Systems, National Centre for Nuclear Research, Otwock-Świerk, Poland
| | - Magdalena Skurzok
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Ewa Stępień
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Faranak Tayefiardebili
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Keyvan Tayefiardebili
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| | - Wojciech Wiślicki
- Department of Complex Systems, National Centre for Nuclear Research, Otwock-Świerk, Poland
| | - Paweł Moskal
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Kraków, Poland
- Center for Theranostics, Jagiellonian University, Kraków, Poland
| |
Collapse
|
7
|
Zhou L, Li Y, de Leon MJ. PET Imaging of Neurofluids. Neuroimaging Clin N Am 2025; 35:223-238. [PMID: 40210379 DOI: 10.1016/j.nic.2024.12.001] [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] [Indexed: 04/12/2025]
Abstract
Following a brief review of brain neurofluid pathways and the general PET technique, we introduce PET imaging of cerebrospinal fluid and interstitial fluid dynamics. Our summary includes both our published and unpublished observations on the modeling of PET imaging for neurofluid quantification in aging, Alzheimer's disease, and in the presence of amyloid lesions. We identify the limitations of PET imaging and point to validations and potential future directions.
Collapse
Affiliation(s)
- Liangdong Zhou
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st Street Feil-2, New York, NY 10065, USA
| | - Yi Li
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st Street Feil-2, New York, NY 10065, USA
| | - Mony J de Leon
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 E 61st Street Feil-2, New York, NY 10065, USA.
| |
Collapse
|
8
|
Pérez-Medina C, Fisher EA, Fayad ZA, Mulder WJM, Teunissen AJP. Radiolabeling lipoproteins to study and manage disease. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07281-4. [PMID: 40293448 DOI: 10.1007/s00259-025-07281-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE Lipoproteins are endogenous nanoparticles with essential roles in lipid transport and inflammation. Lipoproteins are also valuable in diagnosing and treating disease. For instance, certain lipoproteins are overexpressed in patients with atherosclerotic cardiovascular disease, and reconstituted lipoproteins have been extensively used for drug delivery. Radiolabeling has proven an especially powerful approach for studying and therapeutically exploiting lipoproteins. This review details how radiochemistry and nuclear imaging can facilitate the study of lipoproteins in health and disease. Among other topics, we discuss approaches for radiolabeling lipoproteins and detail how these have helped advance our understanding of lipoprotein biology and the diagnosis and treatment of diseases, including atherosclerosis, cancer, and hypercholesteremia. METHODS We performed an extensive literature search on all peer-reviewed studies involving radiolabeled lipoproteins and selected representative examples to provide a high-level overview of the most important discoveries and technological advancements. RESULTS More than 200 peer-reviewed papers involved radiolabeled lipoproteins, spanning mechanistic, diagnostic, and therapeutic studies across a wide range of diseases. CONCLUSION Radiolabeling has been critical in advancing our understanding of lipoprotein biology and leveraging these nanomaterials for diagnosing and treating disease.
Collapse
Affiliation(s)
| | - Edward A Fisher
- Department of Medicine (Cardiology), New York University Grossman School of Medicine, New York, NY, USA
| | - Zahi A Fayad
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Willem J M Mulder
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Abraham J P Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
9
|
Albano D, Rodella C, Calabrò A, Glaudemans AWJM, Treglia G, Bertagna F. The diagnostic role of 2-[ 18F]FDG PET/CT in critically ill patients with suspected infection of unknown origin: a real-world experience. LA RADIOLOGIA MEDICA 2025:10.1007/s11547-025-02005-y. [PMID: 40257695 DOI: 10.1007/s11547-025-02005-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/14/2025] [Indexed: 04/22/2025]
Abstract
PURPOSE 2-Deoxy-2-[18F]-fluoro-D-glucose (2-[18F]FDG) positron emission tomography/computed tomography (PET/CT) is a hybrid imaging tool with an emerging role in the study of several infectious and inflammatory diseases. Its role in critically ill patients admitted to the intensive care unit (ICU) is only marginally investigated. This study aimed to investigate the diagnostic performance and the clinical impact of 2-[18F]FDG PET/CT in the management of critically ill patients with suspected infections of unknown origin. METHODS We retrospectively included 47 ICU patients (mean age 63.6 years, 33 males) who underwent a 2-[18F]FDG PET/CT to look for infection foci. We calculated the diagnostic performance of PET/CT expressed as sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV) and accuracy. As reference standard, final diagnosis based on microbiological/pathologic data or on clinical follow-up was taken. Moreover, we investigated the impact of PET/CT on clinical management and the association of PET/CT with the main clinical, epidemiological and biochemical parameters. RESULTS The overall 2-[18F]FDG PET/CT sensitivity in detecting infectious foci was 80% (95%CI 64-91%), specificity 75% (95%CI 35-97%), PPV 94% (95%CI 83-93%), NPV 43% (95%CI 26-61%) and accuracy 79% (95%CI 65-90%). The absence of kidney failure, increase leukocyte count and low glucose level at the time of PET were significantly associated with true positive PET/CT. No adverse events during the transport and PET/CT procedure were registered. CONCLUSION PET/CT demonstrated to be an accurate toll for the study of critically ill ICU patients with suspected infections. Kidney failure, leukocyte count and blood glucose were associated with true positive PET/CT.
Collapse
Affiliation(s)
- Domenico Albano
- University of Brescia, Brescia, Italy.
- Nuclear Medicine, ASST Spedali Civili Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy.
| | - Carlo Rodella
- Health Physics Department, ASST Spedali Civili, 25123, Brescia, Italy
| | | | - Andor W J M Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Giorgio Treglia
- Division of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Francesco Bertagna
- University of Brescia, Brescia, Italy
- Nuclear Medicine, ASST Spedali Civili Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy
| |
Collapse
|
10
|
Alberts I, More S, Knapp K, Mei R, Fanti S, Mingels C, Nardo L, Hammond NB, Nagaraj H, Rominger A, Cook GJR, Wilson D. Is Long-Axial-Field-of-View PET/CT Cost-Effective? An International Health-Economic Analysis. J Nucl Med 2025:jnumed.124.269203. [PMID: 40246541 DOI: 10.2967/jnumed.124.269203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Our aim is to assess the cost-effectiveness of long-axial-field-of-view (LAFOV) versus short-axial FOV (SAFOV) PET/CT systems using international data. Methods: Our model compares equipment and operational costs for a PET/CT center and investigates the effect of camera choice (SAFOV vs. LAFOV) and operational models. Variables include scanner, personnel, radiopharmaceuticals, and operational costs. Economic performance was measured as cost per scan per patient, the total maximum number of scans possible, and the incremental cost-effectiveness ratio. The willingness-to-pay threshold (WTPT) was taken as the cost of a PET/CT scan using the baseline scenario. Radiopharmaceutical requirements, radiation dose to staff and patients, and patient time were modeled. Results: An LAFOV system can examine as many patients per day (n = 36) as 2 SAFOV systems but requires fewer technologists (4.5 LAFOV vs. 6.8 SAFOV full-time equivalents) and lower activity (12.5 vs. 35.6 GBq/d), resulting in lower personnel doses (0.9 vs. 2.0 mSv/y). For all countries, LAFOV resulted in lowest per-patient scan costs. The most cost-ineffective method was the use of extended hours. Incremental cost-effectiveness ratio analysis strongly favored LAFOV for all countries, including low-income economies, with WTPT met for all jurisdictions. Net monetary benefit was highest for LAFOV. The minimum number of patients needed to meet WTPT for LAFOV was lowest in lower-income countries, suggesting that high throughput or high per-procedure income is not a prerequisite for cost-effective LAFOV usage. Conclusion: LAFOV was shown to facilitate higher patient throughput at lower per-patient and total lifetime operational costs and with lower radiopharmaceutical requirements. These data suggest that LAFOV systems are not just suited to well-resourced academic centers but also are an economically attractive solution for community and resource-limited settings.
Collapse
Affiliation(s)
- Ian Alberts
- Molecular Imaging and Therapy, BC Cancer, Vancouver, British Columbia, Canada;
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stuart More
- University of Cape Town, Cape Town, South Africa
| | - Karen Knapp
- University of Exeter Medical School, Exeter, United Kingdom
| | - Riccardo Mei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Stefano Fanti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Clemens Mingels
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Switzerland
- Department of Radiology, University of California Davis, Sacramento, California
| | - Lorenzo Nardo
- Department of Radiology, University of California Davis, Sacramento, California
| | - Nii Boye Hammond
- National Centre for Radiotherapy, Oncology and Nuclear Medicine, Korlebu Teaching Hospital, Accra, Ghana
| | - Harish Nagaraj
- Integrated Molecular Imaging Centre, Kenyatta University Teaching, Referral and Research Hospital, Nairobi, Kenya; and
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Switzerland
| | - Gary J R Cook
- PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College, London, United Kingdom
| | - Don Wilson
- Molecular Imaging and Therapy, BC Cancer, Vancouver, British Columbia, Canada
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
11
|
Hicks RJ. Diagnostic Radiopharmaceutical Trial Design: Is It Time to Change Nomenclature? J Nucl Med 2025:jnumed.125.269496. [PMID: 40180565 DOI: 10.2967/jnumed.125.269496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Affiliation(s)
- Rodney J Hicks
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, Victoria, Australia
| |
Collapse
|
12
|
Yusufaly T, Roncali E, Brosch-Lenz J, Uribe C, Jha AK, Currie G, Dutta J, El-Fakhri G, McMeekin H, Pandit-Taskar N, Schwartz J, Shi K, Strigari L, Zaidi H, Saboury B, Rahmim A. Computational Nuclear Oncology Toward Precision Radiopharmaceutical Therapies: Current Tools, Techniques, and Uncharted Territories. J Nucl Med 2025; 66:509-515. [PMID: 39947910 PMCID: PMC11960611 DOI: 10.2967/jnumed.124.267927] [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: 04/22/2024] [Accepted: 01/27/2025] [Indexed: 04/03/2025] Open
Abstract
Radiopharmaceutical therapy (RPT), with its targeted delivery of cytotoxic ionizing radiation, demonstrates significant potential for treating a wide spectrum of malignancies, with particularly unique benefits for metastatic disease. There is an opportunity to optimize RPTs and enhance the precision of theranostics by moving beyond a one-size-fits-all approach and using patient-specific image-based dosimetry for personalized treatment planning. Such an approach, however, requires accurate methods and tools for the mathematic modeling and prediction of dose and clinical outcome. To this end, the SNMMI AI-Dosimetry Working Group is promoting the paradigm of computational nuclear oncology: mathematic models and computational tools describing the hierarchy of etiologic mechanisms involved in RPT dose response. This includes radiopharmacokinetics for image-based internal dosimetry and radiobiology for the mapping of dose response to clinical endpoints. The former area originates in pharmacotherapy, whereas the latter originates in radiotherapy. Accordingly, models and methods developed in these predecessor disciplines serve as a foundation on which to develop a repurposed set of tools more appropriate to RPT. Over the long term, this computational nuclear oncology framework also promises to facilitate widespread cross-fertilization of ideas between nuclear medicine and the greater mathematic and computational oncology communities.
Collapse
Affiliation(s)
- Tahir Yusufaly
- Division of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland;
| | - Emilie Roncali
- Department of Biomedical Engineering, University of California Davis, Davis, California
| | | | - Carlos Uribe
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Abhinav K Jha
- Department of Biomedical Engineering and Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri
| | - Geoffrey Currie
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Joyita Dutta
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts
| | - Georges El-Fakhri
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | | | - Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Jazmin Schwartz
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kuangyu Shi
- Department of Nuclear Medicine, University of Bern, Bern, Switzerland
| | - Lidia Strigari
- Department of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Arman Rahmim
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Physics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
13
|
Shahi A, Poladyan H, Anashkin E, Komarov B, Baldassi B, Rapley M, Babich A, Bubon O, Reznik A. Multi-angle acquisition and 3D composite reconstruction for organ-targeted PET using planar detectors. Med Phys 2025; 52:2507-2519. [PMID: 39729623 PMCID: PMC11972046 DOI: 10.1002/mp.17606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND This study investigates a multi-angle acquisition method aimed at improving image quality in organ-targeted PET detectors with planar detector heads. Organ-targeted PET technologies have emerged to address limitations of conventional whole-body PET/CT systems, such as restricted axial field-of-view (AFOV), limited spatial resolution, and high radiation exposure associated with PET procedures. The AFOV in organ-targeted PET can be adjusted to the organ of interest, minimizing unwanted signals from other parts of the body, thus improving signal collection efficiency and reducing the dose of administered radiotracer. However, while planar detector PET technology allows for quasi-3D image reconstruction due to the separation between detector heads, it suffers from degraded axial spatial resolution and, consequently, reduced recovery coefficients (RCs) along the axial direction perpendicular to the detectors. PURPOSE The purpose of this study was to evaluate the concept of multi-angle image acquisition with two planar PET detectors and composite full 3D image reconstruction. This leverages data collection from multiple polar angles to improve the axial spatial resolution in the direction perpendicular to the detector heads. In such, the concept allows to overcome the intrinsic limitations of planar detectors in axial resolution. METHODS This study evaluates the improvement in the quality of images acquired with the Radialis organ-targeted PET camera through multi-angle image acquisition, in both experimental and simulated imaging scenarios. This includes the use of custom-made phantom with fillable spherical hot inserts, the NEMA NU4-2008 image quality (IQ) phantom, and simulations with a digital brain phantom. The analysis involves the comparison of line profiles drawn through the spherical hot inserts, image uniformity, RCs, and the reduction of smearing observed in the axial planes with and without the multi-angle acquisition strategy. RESULTS Significant improvements were observed in reducing smearing, enhancing image uniformity, and increasing RCs using the evaluated multi-angle acquisition method. In the composite images, the hot spheres appear more symmetrical in all planes. The image uniformity, calculated from the IQ phantom, improves from 7.79% and 10.98%, as measured in the images from the individual acquisitions, to 2.72% in the composite image. There is also an overall improvement in the RCs as measured from the hot rods of the IQ phantom. Furthermore, the simulation study using the digital human brain phantom demonstrates minimal smearing in the four-angle scan, as opposed to a two-angle scan. CONCLUSION The multi-angle acquisition method offers a promising approach to transform planar PET detector technology into a true tomographic organ-targeted PET system and to enable improvement in image quality while preserving a versatility inherent to planar detector technology. Future research will focus on optimizing the multi-angle imaging protocol, including adjustments to detector separations, number of acquisition angles, and reconstruction iterations, alongside incorporating TOF, and reconstruction with point spread function modeling to further improve image quality.
Collapse
Affiliation(s)
- Anirudh Shahi
- Department of PhysicsLakehead UniversityThunder BayOntarioCanada
| | | | | | | | | | - Madeline Rapley
- Department of PhysicsLakehead UniversityThunder BayOntarioCanada
| | | | - Oleksandr Bubon
- Department of PhysicsLakehead UniversityThunder BayOntarioCanada
- Radialis Inc.Thunder BayOntarioCanada
| | - Alla Reznik
- Department of PhysicsLakehead UniversityThunder BayOntarioCanada
- Thunder Bay Regional Health Research InstituteThunder BayOntarioCanada
| |
Collapse
|
14
|
Isayev K, Rasulov O, Sadigova N. Advanced photodetector for hybrid PET-MRI systems. OPEN RESEARCH EUROPE 2025; 4:247. [PMID: 40196757 PMCID: PMC11973626 DOI: 10.12688/openreseurope.18695.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 04/09/2025]
Abstract
Background Currently, a wide variety of silicon photomultipliers (SiPMs) are available, each designed for specific applications in fields such as science, medicine, and industry. Advances in production technology have led to the development of more sensitive and efficient photodiodes, which are critical for applications requiring precision, such as medical imaging. Methods A research group has been working on designing a highly sensitive photodiode to enhance the capabilities of next-generation of hybrid positron emission tomography (PET) and magnetic resonance imaging (MRI) scanners. This involves integrating micropixel avalanche photodiodes (MAPDs) to improve image resolution. The chosen design features deep-immersion MAPDs with a pixel size of 12 microns and a density of 1000 pixels per mm 2, allowing for high-detail photon detection. The 4x4 mm 2 active area is optimized to balance sensitivity and size for high-resolution medical imaging. To produce these photodiodes, the group has outlined a production plan involving 300 mm silicon wafers grown using multiple techniques to enhance material properties. The Malaysian Institute of Microelectronic Systems (MIMOS), renowned for its expertise in optical microelectronics, was selected as the production center. With MIMOS' state-of-the-art facilities, the project aims to meet stringent medical diagnostics standards. Results The experimental results demonstrated that the MAPD-3NM (MAPD design with 12 microns pixel size) photodiode achieved an amplification factor 1.8 times greater than the MAPD-3NK (MAPD design with 10 microns pixel size) under optimal conditions. The both samples size was 4x4 square mm. Its overvoltage range increased by 100%, reaching 4 V, enhancing photon detection and amplification. The MAPD-3NM also showed a significant reduction in dark current, about 3.5 times lower than the MAPD-3NK, improving performance in low-light environments. Additionally, the MAPD-3NM had a capacitance of 200 pF compared to 176 pF for the MAPD-3NK, contributing to its superior performance. These improvements make the MAPD-3NM more efficient and sensitive for scientific and medical applications. Conclusions This project represents a major advancement in photodetector technology for medical diagnostics, aiming to develop more accurate and efficient PET-MRI scanners that enhance patient outcomes with improved imaging capabilities.
Collapse
Affiliation(s)
- K Isayev
- Nuclear Research Department, IDDA, Baku, Azerbaijan
| | - O Rasulov
- Elmek Mozgasban Alapitvany, Budapest, Hungary
| | - N Sadigova
- Azerbaijan University of Architecture and Construction, Baku, Azerbaijan
| |
Collapse
|
15
|
Chung KJ, Abdelhafez YG, Spencer BA, Jones T, Tran Q, Nardo L, Chen MS, Sarkar S, Medici V, Lyo V, Badawi RD, Cherry SR, Wang G. Quantitative PET imaging and modeling of molecular blood-brain barrier permeability. Nat Commun 2025; 16:3076. [PMID: 40159510 PMCID: PMC11955546 DOI: 10.1038/s41467-025-58356-7] [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: 08/02/2024] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
Neuroimaging of blood-brain barrier permeability has been instrumental in identifying its broad involvement in neurological and systemic diseases. However, current methods evaluate the blood-brain barrier mainly as a structural barrier. Here we developed a non-invasive positron emission tomography method in humans to measure the blood-brain barrier permeability of molecular radiotracers that cross the blood-brain barrier through its molecule-specific transport mechanism. Our method uses high-temporal resolution dynamic imaging and kinetic modeling for multiparametric imaging and quantification of the blood-brain barrier permeability-surface area product of molecular radiotracers. We show, in humans, our method can resolve blood-brain barrier permeability across three radiotracers and demonstrate its utility in studying brain aging and brain-body interactions in metabolic dysfunction-associated steatotic liver inflammation. Our method opens new directions to effectively study the molecular permeability of the human blood-brain barrier in vivo using the large catalogue of available molecular positron emission tomography tracers.
Collapse
Affiliation(s)
- Kevin J Chung
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Yasser G Abdelhafez
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Benjamin A Spencer
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Terry Jones
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Quyen Tran
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Lorenzo Nardo
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Moon S Chen
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA, USA
| | - Souvik Sarkar
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA, USA
- Division of Gastroenterology and Hepatology, University of California Davis Health, Sacramento, CA, USA
| | - Valentina Medici
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA, USA
- Division of Gastroenterology and Hepatology, University of California Davis Health, Sacramento, CA, USA
| | - Victoria Lyo
- Department of Surgery, University of California Davis Health, Sacramento, CA, USA
- Center for Alimentary and Metabolic Sciences, University of California Davis Health, Sacramento, CA, USA
| | - Ramsey D Badawi
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, USA
| | - Simon R Cherry
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, USA
| | - Guobao Wang
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA.
| |
Collapse
|
16
|
Zhang X, Xiang Z, Wang F, Pan X, Zhang Q, Wang P, Jiang L, Yuan H. 13N-NH 3 myocardial perfusion imaging with reduced scan duration: a feasibility study in the era of total-body PET/CT. EJNMMI Phys 2025; 12:18. [PMID: 40032742 DOI: 10.1186/s40658-025-00729-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
PURPOSE To explore the feasibility of reducing scan duration of 13N-NH3 myocardial perfusion imaging (MPI) using a total-body PET/CT scanner. METHODS Forty-five patients with known or suspected coronary artery disease (CAD) performing rest 13N-NH3 MPI with total-body PET/CT were retrospectively included. PET data were acquired in list mode for 10 min, and reconstructed into sequence images of different scan duration: 10-min, 7-min, 5-min, 3-min, and 2-min (G10 to G2). Subjective visual evaluation including overall impression, image noise and lesion visibility was evaluated using 5-point Likert scale. Quantitative parameters including perfusion defect extent (Extent), total perfusion defect (TPD), summed rest score (SRS), end-diastolic volume (EDV), end-systolic volume (ESV), ejection fraction (EF), and myocardial blood flow (MBF) were analyzed. The full-time images (G10) were served as the reference. RESULTS There were no significant differences in subjective visual scores between G7-G5 and G10 groups (p > 0.05). A significant decrease in overall impression and image noise of G3-G2 was observed when compared to G10 (p < 0.05). However, no significant difference in lesion visibility was noted between G3 and G10 (p > 0.05). All G3 image quality was clinically acceptable (≥ 3 points). Except for EDV and ESV, other quantitative parameters showed no significant difference between G7-G3 and G10 (p > 0.05) and agreements were good (ICC = 0.974-0.998). For G2, only TPD exhibited no significant difference when compared to G10 (p > 0.05). CONCLUSION Regarding imaging quality and parametric quantification accuracy of 13N-NH3 MPI, a 3-min scan is clinically acceptable, while a 5-min scan is sufficiently reliable.
Collapse
Affiliation(s)
- Xiaochun Zhang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Zeyin Xiang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Fanghu Wang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Xiaoqiang Pan
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Qing Zhang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Peng Wang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Lei Jiang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China.
| | - Hui Yuan
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China.
| |
Collapse
|
17
|
Telli T, Desaulniers M, Pyka T, Caobelli F, Forstmann S, Umutlu L, Fendler WP, Rominger A, Herrmann K, Seifert R. What Role Does PET/MRI Play in Musculoskeletal Disorders? Semin Nucl Med 2025; 55:277-289. [PMID: 38044175 DOI: 10.1053/j.semnuclmed.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 11/13/2023] [Indexed: 12/05/2023]
Abstract
Musculoskeletal disorders of nononcological origin are one of the most frequent reasons for consultation. Patients suffering from musculoskeletal disorders also consult more than once for the same reason. This results in multiple clinical follow-ups after several radiological and serum examinations, the main ones including X-rays targeting the painful anatomical region and inflammatory serum parameters. As part of their work up, patients suffering from musculoskeletal disorders often require multisequence, multi-parameter MRI. PET/MRI is a promising imaging modality for their diagnosis, with the added advantage of being able to be performed in a single visit. PET/MRI is particularly useful for diagnosing osteomyelitis, spondylodiscitis, arthritis, many pediatric pathologies, and a wide range of other musculoskeletal pathologies. PET/MRI is already used to diagnose malignant bone tumors such as osteosarcoma. However, current knowledge of the indications for PET/MRI in nononcological musculoskeletal disorders is based on studies involving only a few patients. This review focuses on the usefulness of PET/MRI for diagnosing nononcological musculoskeletal disorders.
Collapse
Affiliation(s)
- Tugce Telli
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, Essen, Germany.
| | - Mélanie Desaulniers
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, Essen, Germany; Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Thomas Pyka
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Federico Caobelli
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Sophia Forstmann
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, Essen, Germany
| | - Lale Umutlu
- Department of Radiology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Ken Herrmann
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, Essen, Germany; Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| |
Collapse
|
18
|
Gu W, Zhu Z, Liu Z, Wang Y, Li Y, Xu T, Liu W, Luo G, Wang K, Zhou Y. Self-supervised neural network for Patlak-based parametric imaging in dynamic [ 18F]FDG total-body PET. Eur J Nucl Med Mol Imaging 2025; 52:1436-1447. [PMID: 39621094 DOI: 10.1007/s00259-024-07008-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/25/2024] [Indexed: 02/20/2025]
Abstract
PURPOSE The objective of this study is to generate reliable Ki parametric images from a shortened [18F]FDG total-body PET for clinical applications using a self-supervised neural network algorithm. METHODS We proposed a self-supervised neural network algorithm with Patlak graphical analysis (SN-Patlak) to generate Ki images from shortened dynamic [18F]FDG PET without 60-min full-dynamic PET-based training. The algorithm deeply integrates neural network architecture with a Patlak method, employing the fitting error of the Patlak plot as the neural network's loss function. As the 0-60 min blood time activity curve (TAC) required by the standard Patlak plot is unobtainable from shortened dynamic PET scans, a population-based "normalized time" (integral-to-instantaneous blood concentration ratio) was used for the linear fitting of Patlak plot of t* to 60 min, and the modified Patlak plot equation was then incorporated into the neural network. Ki images were generated by minimizing the difference between the input layer (measured tissue-to-blood concentration ratios) and the output layer (predicted tissue-to-blood concentration ratios). The effects of t* (20 to 50 min post injection) on the Ki images generated from the SN-Patlak and standard Patlak was evaluated using the normalized mean square error (NMSE), and Pearson's correlation coefficient (Pearson's r). RESULTS The Ki images generated by the SN-Patlak are robust to the dynamic PET scan duration, and the Ki images generated by the SN-Patlak from just a 10-minute (50-60 min post-injection) dynamic [18F]FDG total-body PET scan are comparable to those generated by the standard Patlak method from 40-min (20-60 min post injection) with NMSE = 0.15 ± 0.03 and Pearson's r = 0.93 ± 0.01. CONCLUSIONS The SN-Patlak parametric imaging algorithm is robust and reliable for quantification of 10-min dynamic [18F]FDG total-body PET.
Collapse
Affiliation(s)
- Wenjian Gu
- Faculty of Computing, Harbin Institute of Technology, Harbin, China
- United Imaging Healthcare Technology Group Co., Ltd, Shanghai, China
| | - Zhanshi Zhu
- Faculty of Computing, Harbin Institute of Technology, Harbin, China
- United Imaging Healthcare Technology Group Co., Ltd, Shanghai, China
| | - Ze Liu
- United Imaging Healthcare Technology Group Co., Ltd, Shanghai, China
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China
| | - Yihan Wang
- United Imaging Healthcare Technology Group Co., Ltd, Shanghai, China
| | - Yanxiao Li
- United Imaging Healthcare Technology Group Co., Ltd, Shanghai, China
| | - Tianyi Xu
- United Imaging Healthcare Technology Group Co., Ltd, Shanghai, China
| | - Weiping Liu
- United Imaging Healthcare Technology Group Co., Ltd, Shanghai, China
- Institute of Biomedical Manufacturing and Life Quality Engineering, School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Gongning Luo
- Faculty of Computing, Harbin Institute of Technology, Harbin, China.
| | - Kuanquan Wang
- Faculty of Computing, Harbin Institute of Technology, Harbin, China.
| | - Yun Zhou
- United Imaging Healthcare Technology Group Co., Ltd, Shanghai, China.
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
19
|
Yi M, Lee D, Gola A, Merzi S, Penna M, Lee JS, Cherry SR, Kwon SI. Segmented SiPM Readout for Cherenkov Time-of-Flight Positron Emission Tomography Detectors Based on Bismuth Germanate. ACS PHOTONICS 2025; 12:1125-1136. [PMID: 39989930 PMCID: PMC11843713 DOI: 10.1021/acsphotonics.4c02265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/25/2025]
Abstract
Positron emission tomography (PET) is the most sensitive biomedical imaging modality for noninvasively detecting and visualizing positron-emitting radiopharmaceuticals within a subject. In PET, measuring the time-of-flight (TOF) information for each pair of 511 keV annihilation photons improves effective sensitivity but requires high timing resolution. Hybrid materials that emit both scintillation and Cherenkov photons, such as bismuth germanate, recently offer the potential for more precise timing information from Cherenkov photons while maintaining adequate energy resolution from scintillation photons. However, a significant challenge in using such hybrid materials for TOF PET applications lies in the event-dependent timing spread caused by the mixed detection of Cherenkov and scintillation photons due to relatively lower production of Cherenkov photons. This study introduces an innovative approach by segmenting silicon photomultiplier (SiPM) pixels coupled to a single crystal, rather than using traditional SiPMs that are as large as or larger than the crystals they read. We demonstrated that multiple timestamps and photon counts obtained from the segmented SiPM can classify events by providing temporal photon density, effectively addressing this challenge. The approach and findings would lead to new opportunities in applications that require precise timing and photon counting.
Collapse
Affiliation(s)
- Minseok Yi
- Department
of Biomedical Engineering, University of
California, Davis, One Shields Avenue, Davis, California 95616, United States
- Interdisciplinary
Program in Bioengineering, Seoul National University College of Engineering, Seoul 03080, Republic of Korea
- Integrated
Major in Innovative Medical Science, Seoul
National University, Seoul 03080, Republic
of Korea
| | - Daehee Lee
- Department
of Biomedical Engineering, University of
California, Davis, One Shields Avenue, Davis, California 95616, United States
| | - Alberto Gola
- Fondazione
Bruno Kessler, via Sommarive
18, Trento I-38123, Italy
| | - Stefano Merzi
- Fondazione
Bruno Kessler, via Sommarive
18, Trento I-38123, Italy
| | - Michele Penna
- Fondazione
Bruno Kessler, via Sommarive
18, Trento I-38123, Italy
| | - Jae Sung Lee
- Interdisciplinary
Program in Bioengineering, Seoul National University College of Engineering, Seoul 03080, Republic of Korea
- Integrated
Major in Innovative Medical Science, Seoul
National University, Seoul 03080, Republic
of Korea
- Brightonix
Imaging Inc., Seoul 04782, Republic of Korea
| | - Simon R. Cherry
- Department
of Biomedical Engineering, University of
California, Davis, One Shields Avenue, Davis, California 95616, United States
| | - Sun Il Kwon
- Department
of Biomedical Engineering, University of
California, Davis, One Shields Avenue, Davis, California 95616, United States
| |
Collapse
|
20
|
Nadig V, Gundacker S, Herweg K, Naunheim S, Schug D, Weissler B, Schulz V. ASICs in PET: what we have and what we need. EJNMMI Phys 2025; 12:16. [PMID: 39939493 PMCID: PMC11822191 DOI: 10.1186/s40658-025-00717-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Designing positron emission tomography (PET) scanners involves several significant challenges. These include the precise measurement of the time of arrival of signals, accurate integration of the pulse shape, maintaining low power consumption, and supporting the readout of thousands of channels. To address these challenges, researchers and engineers frequently develop application-specific integrated circuits (ASICs), which are custom-designed readout electronics optimized for specific tasks. As a result, a wide range of ASIC solutions has emerged in PET applications. However, there is currently no comprehensive or standardized comparison of these ASIC designs across the field. METHODS In this paper, we evaluate the requirements posed to readout electronics in the field of PET, give an overview of the most important ASICs available for PET applications and discuss how to characterize their essential features and performance parameters. We thoroughly review the hardware characteristics of the different circuits, such as the number of readout channels provided, their power consumption, input and output design. Furthermore, we summarize their performance as characterized in literature. RESULTS While the ASICs described show common trends towards lower power consumption or a higher number of readout channels over the past two decades, their characteristics and also their performance assessment by the developers, producers and vendors differ in many aspects. To cope with the challenge of selecting a suitable ASIC for a given purpose and PET application from the varying information available, this article suggests a protocol to assess an ASIC's performance parameters and characteristics. CONCLUSION ASICs developed for PET applications are versatile. With novel benchmarks set for the impact of scintillator and photosensor on the time-of-flight performance, the pressure on ASICs to deliver higher timing resolution and cope with an even higher data rate is enormous. Latest developments promise new circuits and improvements in time-of-flight performance. This article provides an overview on existing and emerging readout solutions in PET over the past 20 years, which is currently lacking in literature.
Collapse
Affiliation(s)
- Vanessa Nadig
- University Hospital Aaachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Stefan Gundacker
- University Hospital Aaachen, Pauwelsstrasse 30, 52074, Aachen, Germany
- Institute of High Energy Physics, Austrian Academy of Sciences, Nikolsdorfer Gasse 18, 1050, Vienna, AT, Austria
| | - Katrin Herweg
- University Hospital Aaachen, Pauwelsstrasse 30, 52074, Aachen, Germany
- Institute for Imaging and Computer Vision, RWTH Aachen University, Kopernikusstraße 16, 52074, Aachen, GER, Germany
| | - Stephan Naunheim
- University Hospital Aaachen, Pauwelsstrasse 30, 52074, Aachen, Germany
- Institute for Imaging and Computer Vision, RWTH Aachen University, Kopernikusstraße 16, 52074, Aachen, GER, Germany
| | - David Schug
- University Hospital Aaachen, Pauwelsstrasse 30, 52074, Aachen, Germany
- Hyperion Hybrid Imaging Systems GmbH, Pauwelsstrasse 19, 52074, Aachen, GER, Germany
- Institute for Imaging and Computer Vision, RWTH Aachen University, Kopernikusstraße 16, 52074, Aachen, GER, Germany
| | - Bjoern Weissler
- University Hospital Aaachen, Pauwelsstrasse 30, 52074, Aachen, Germany
- Hyperion Hybrid Imaging Systems GmbH, Pauwelsstrasse 19, 52074, Aachen, GER, Germany
- Institute for Imaging and Computer Vision, RWTH Aachen University, Kopernikusstraße 16, 52074, Aachen, GER, Germany
| | - Volkmar Schulz
- Hyperion Hybrid Imaging Systems GmbH, Pauwelsstrasse 19, 52074, Aachen, GER, Germany.
- III. Physikalisches Institut B, RWTH Aachen University, Otto-Blumenthal-Straße, 52074, Aachen, GER, Germany.
- Institute for Imaging and Computer Vision, RWTH Aachen University, Kopernikusstraße 16, 52074, Aachen, GER, Germany.
| |
Collapse
|
21
|
Dar D, Rodak M, Da Pieve C, Gorczewska I, Sharma G, Chmielik E, Niedbala M, Bzowski P, d’Amico A, Bobek-Billewicz B, Nowicka E, Tarnawski R, Kaspera W, Kramer-Marek G. Imaging PD-L1 in the brain-Journey from the lab to the clinic. Neuro Oncol 2025; 27:567-582. [PMID: 39470381 PMCID: PMC11812043 DOI: 10.1093/neuonc/noae190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICPIs) have proven to restore adaptive anti-tumor immunity in many cancers; however, no noteworthy therapeutic schedule has been established for patients with glioblastoma (GBM). High programmed death-ligand 1 (PD-L1) expression is associated with immunosuppressive and aggressive phenotypes in GBM. Presently, there is no standardized protocol for assessing PD-L1 expression levels to select patients and monitor their response to ICPI therapy. The aim of this study was to investigate the use of 89Zr-DFO-Atezolizumab to image the spatio-temporal distribution of PD-L1 in preclinical mouse models and in patients with newly diagnosed GBM treated with/without neoadjuvant Pembrolizumab. METHODS The immunoreactivity, binding affinity, and specificity of 89Zr-DFO-Atezolizumab were confirmed in vitro. Mice-bearing orthotopic GBM tumors or patients with newly diagnosed GBM treated with/without Pembrolizumab were intravenously injected with 89Zr-DFO-Atezolizumab, and PET/CT images were acquired 24, 48, and 72 hours in mice and at 48 and 72 post-injection in patients. Radioconjugate uptake was quantified in the tumor and healthy tissues. Ex vivo immunohistochemistry (IHC) and immunophenotyping were performed on mouse tumor samples or resected human tumors. RESULTS 89Zr-DFO-Atezolizumab was prepared with high radiochemical purity (RCP > 99%). In vitro cell-associated radioactivity of 89Zr-DFO-Atezolizumab corroborated cell line PD-L1 expression. PD-L1 in mouse GBM tumors was detected with high specificity using 89Zr-DFO-Atezolizumab and radioconjugate uptake correlated with IHC. Patients experienced no 89Zr-DFO-Atezolizumab-related side effects. High 89Zr-DFO-Atezolizumab uptake was observed in patient tumors at 48 hours post-injection, however, the uptake varied between patients treated with/without Pembrolizumab. CONCLUSIONS 89Zr-DFO-Atezolizumab can visualize distinct PD-L1 expression levels with high specificity in preclinical mouse models and in patients with GBM, whilst complementing ex vivo analysis.
Collapse
Affiliation(s)
- Dawoud Dar
- Department of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Magdalena Rodak
- Department of Radiopharmacy and Preclinical PET Imaging, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Chiara Da Pieve
- Department of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Izabela Gorczewska
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Gitanjali Sharma
- Department of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Ewa Chmielik
- Department of Tumor Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Marcin Niedbala
- Department of Neurosurgery, Medical University of Silesia, Katowice, Poland
| | - Pawel Bzowski
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Andrea d’Amico
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Barbara Bobek-Billewicz
- Department of Radiology and Imaging Diagnostic, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Elzbieta Nowicka
- Department of IIIrd Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Rafal Tarnawski
- Department of IIIrd Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Wojciech Kaspera
- Department of Neurosurgery, Medical University of Silesia, Katowice, Poland
| | - Gabriela Kramer-Marek
- Department of Radiopharmacy and Preclinical PET Imaging, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
- Department of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| |
Collapse
|
22
|
Gao M, Daube-Witherspoon ME, Karp JS, Surti S. Total-Body PET System Designs with Axial and Transverse Gaps: A Study of Lesion Quantification and Detectability. J Nucl Med 2025; 66:323-329. [PMID: 39819688 PMCID: PMC11800738 DOI: 10.2967/jnumed.124.267769] [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: 09/03/2024] [Accepted: 12/16/2024] [Indexed: 01/19/2025] Open
Abstract
High-sensitivity total-body PET enables faster scans, lower doses, and dynamic multiorgan imaging. However, the higher system cost of a scanner with a long axial field of view (AFOV) hinders its wider application. This paper investigates the impact on the lesion quantification and detectability of cost-effective total-body PET sparse designs. Methods: Using the PennPET Explorer (PPEx) as a model, 3 sparse configurations with the same 142-cm AFOV were considered, including designs with only axial gaps (AGs), only transverse gaps (TGs), and a mixture of AGs and TGs (MG), with retained detector fractions (DFs) ranging from 71% to 40%. Human data from the PPEx were used to emulate sparse designs by discarding lines of response as a proxy for missing detectors. We embedded lesion events in the resultant list data with varying uptakes in the lung and liver before reconstruction. A generalized scan statistics methodology was used to measure lesion detectability and quantification as a function of lesion uptake and scan duration. Results: Relative to a fully populated system, an AG design with 71% performs well but is susceptible to image artifacts as the DF decreases to 58%. A TG design performs well with a DF of 58% but requires twice the scan time to achieve similar lesion detectability and is susceptible to transverse field-of-view truncation below 60 cm as the DF is further decreased. An MG design with a DF of 58% requires 3 times the scan time to achieve similar lesion detectability, and with no evidence of artifacts even as the DF is decreased to 40%. Conclusion: Sparse designs with artifact-free images can provide comparable lesion quantification and detectability to the fully populated PPEx after compensating for the reduced sensitivity with increased scan time. Because an AG design is more susceptible to image artifacts with a lower DF, a system with only AGs is not an optimal choice for dramatic cost reduction. A TG design provides a higher relative sensitivity than AG or MG designs for a given DF, leading to a shorter scan time to achieve comparable lesion detectability. However, the increased truncation of the transverse field of view with decreasing DF limits this design choice. An MG design allows for the greatest cost reduction (lowest DF) if the scan duration is increased to compensate for the higher loss in sensitivity. Sparse designs of PET with a long AFOV provide a technologic solution for introducing such systems at reduced cost into routine clinical use.
Collapse
Affiliation(s)
- Min Gao
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Joel S Karp
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Suleman Surti
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
23
|
Das KJ, Meena JK, Kumar D. Applicability and performance of 18F-FDG PET-based modalities for whole-body cancer screening: a systematic review and meta-analysis. Jpn J Radiol 2025; 43:266-281. [PMID: 39302525 DOI: 10.1007/s11604-024-01659-4] [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: 04/23/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE Screening tests are the cornerstone for early detection and optimal management of cancers. Most of the present cancer-screening tests are intrusive, time-consuming, and specifically target a particular anatomical site or cancer type. Only a few studies have reported the objective measures of 18F-FDG PET-based cancer screening in asymptomatic individuals. This review and meta-analysis is an attempt to assess the applicability and performance of 18F-FDG PET-based modalities for whole-body cancer screening. MATERIALS AND METHODS The systematic review and meta-analysis were performed following PRISMA guidelines. Literature searches in PubMed, Scopus, and Embase were conducted using relevant MeSH terms and keywords, for articles published in the last 2 decades (2000-2022). Pooled estimates of diagnostic test accuracy-including sensitivity, specificity, positive-likelihood ratio, negative-likelihood ratio, and hierarchical summary ROC (HSROC) curve were generated using bivariate random-effects meta-analysis. RESULTS Seventeen studies were included in the systematic review and 13 studies were deemed eligible for meta-analysis. The mean estimates of pooled sensitivity, specificity, positive-likelihood ratio, negative-likelihood ratio, and Odds ratio using 18F-FDG PET with a 95% confidence interval were 0.47 (0.25-0.69), 0.97 (0.95-0.98), 18.8 (6.8-51.5), 0.45 (0.27-0.76), 41.0 (7.9-211.8) and for 18F-FDG PET/CT were 0.83 (0.75-0.88), 0.98 (0.97-0.99), 49.7 (29.2-84.5), 0.15 (0.8-0.28), 329.9 (125.0-870.8), respectively. Among screening modalities, 18F-FDG PET/CT had a higher accuracy i.e., the area under the HSROC curve (AUC): 0.91 (0.87-0.95) compared to 18F-FDG PET: 0.72 (0.61-0.82). CONCLUSION This study demonstrates that currently 18F-FDG PET-based screening has limited applicability for population-based cancer-screening programs. However, it has a promising role as a combined screening strategy for at-risk individuals and allows for comprehensive diagnostic and prognostic evaluation in high-resource settings.
Collapse
Affiliation(s)
- K J Das
- Dept. of Nuclear Medicine, National Cancer Institute (NCI-AIIMS), All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - J K Meena
- Dept. of Preventive Oncology, National Cancer Institute (NCI-AIIMS), All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| | - D Kumar
- Dept. of Nuclear Medicine, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| |
Collapse
|
24
|
Alberts IL, Xue S, Sari H, Cavinato L, Prenosil G, Afshar-Oromieh A, Mingels C, Shi K, Caobelli F, Rahmim A, Pyka T, Rominger A. Long-axial field-of-view PET/CT improves radiomics feature reliability. Eur J Nucl Med Mol Imaging 2025; 52:1004-1016. [PMID: 39477863 DOI: 10.1007/s00259-024-06921-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/11/2024] [Indexed: 01/23/2025]
Abstract
PURPOSE To assess the influence of long-axial field-of-view (LAFOV) PET/CT systems on radiomics feature reliability, to assess the suitability for short-duration or low-activity acquisitions for textural feature analysis and to investigate the influence of acceptance angle. METHODS 34 patients were analysed: twelve patients underwent oncological 2-[18F]-FDG PET/CT, fourteen [18F]PSMA-1007 and eight [68Ga]Ga-DOTATOC. Data were obtained using a 106 cm LAFOV system for 10 min. Sinograms were generated from list-mode data corresponding to scan durations of 2, 5, 10, 20, 30, 60, 120, 240, 360 and 600s using both standard (minimum ring difference MRD 85 crystals) and maximum acceptance angles (MRD 322). Target lesions were segmented and radiomics features were calculated. To assess feature correlation, Pearson's product-moment correlation coefficient (PPMCC) was calculated with respect to the full duration acquisition for MRD 85 and 322 respectively. The number of features with excellent (r > 0.9), moderate (r > 0.7 and < 0.9) and poor (r ≤ 0.7) correlation was compared as a measure of feature stability. Intra-class heterogeneity was assessed by means of the quartile coefficient of dispersion. RESULTS As expected, PPMCC improved with acquisition time for all features. By 240s almost all features showed at least moderate agreement with the full count (C100%) data, and by 360s almost all showed excellent agreement. Compared to standard-axial field of view (SAFOV) equivalent scans, fewer features showed moderate or poor agreement, and this was most pronounced for [68Ga]Ga-DOTATOC. Data obtained at C100% at MRD 322 were better able to capture between-patient heterogeneities. CONCLUSION The improved feature reliability at longer acquisition times and higher MRD demonstrate the advantages of high sensitivity LAFOV systems for reproducible and low-noise data. High fidelity between MRD 85 and MRD 322 was seen at all scan durations > 2s. When contrasted with data comparable to a simulated SAFOV acquisition, full-count and full-MRD data were better able to capture underlying feature heterogeneities.
Collapse
Affiliation(s)
- Ian L Alberts
- Molecular Imaging and Therapy, BC Cancer - Vancouver, 600 West 10th Ave, Vancouver, BC, V5Z 1H5, Canada.
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland.
| | - Song Xue
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Hasan Sari
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
- Advanced Clinical Imaging Technology, Siemens Healthcare AG, Lausanne, Switzerland
| | - Lara Cavinato
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
- Laboratory for Modelling and Scientific Computing, Department of Mathematics, Politecnico di Milano, Milan, 20133, Italy
| | - George Prenosil
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Clemens Mingels
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Federico Caobelli
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Arman Rahmim
- Molecular Imaging and Therapy, BC Cancer - Vancouver, 600 West 10th Ave, Vancouver, BC, V5Z 1H5, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Thomas Pyka
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| |
Collapse
|
25
|
Liu Z, Mungai S, Kuang Z, Ren N, Xie S, Peng Q, Williams C, Yang Y. High-resolution TOF-DOI PET detectors through the implementation of dual-ended readout with SiPM arrays of different pixel sizes on the two ends. Med Phys 2025; 52:867-879. [PMID: 39607086 DOI: 10.1002/mp.17544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND An organ-specific Positron emission tomography (PET) scanner can achieve the same sensitivity with much fewer detectors as compared to a whole-body PET scanner, thereby substantially reducing the system cost. It can also achieve much better spatial resolution as compared to a whole-body PET scanner since the photon noncollinearity effect is reduced by using smaller detector ring diameter. Consequently, the development of organ-specific PET scanners with high spatial resolution, high sensitivity, and low cost has been a major focus of international research in PET instrument development for many years. PURPOSE The focus of this work is to develop high-resolution depth encoding PET detectors with high timing resolution and a reduced number of signal processing electronic channels. Consequently, PET scanners tailored for specific organs can be developed with high spatial and timing resolutions, enhanced sensitivity, and affordable cost. METHODS An 8 × 8 silicon photomultiplier (SiPM) array with a pixel size of 3 × 3 mm2 and a multiplexed signal readout circuit is coupled to one end of the lutetium yttrium orthosilicate (LYSO) array with a glass light guide between them to achieve a good crystal identification of small crystals by using only four position-encoding energy signals. A 4 × 4 SiPM array with a pixel size of 6 × 6 mm2 and an individual readout circuit is coupled to the other end of the crystal array without a light guide to achieve a good coincidence timing resolution (CTR). The depth of interaction (DOI) of the detector is measured by ratio of the energies measured by the two SiPM arrays and can be used to correct the depth dependency of the timing. The flood histograms, energy resolutions (ERs), DOI resolutions, and CTRs of two detectors utilizing LYSO arrays with different crystal sizes were measured with each of the two SiPM arrays alternately placed at the front of the detectors. RESULTS A better flood histogram was obtained by placing the 8 × 8 SiPM array in front of the detector. The depth dependency of timing was larger when the 4 × 4 SiPM array was placed at the front of the detector. A better CTR was obtained by placing the 4 × 4 SiPM array at the back of the detector as compared to placing it at the front of the detector if the depth-dependent timing correction was not performed. If the depth-dependent timing correction was performed, a better CTR can be obtained by placing the 4 × 4 SiPM array at the front of the detector. The first detector using a 12 × 12 LYSO crystal array with a crystal size of 1.95 × 1.95 × 20 mm3 provided a flood histogram with all crystals clearly resolved, an ER of 11.7%, a DOI resolution of 2.9 mm, and a CTR of 275 ps with the depth-dependent timing correction. The second detector using a 23 × 23 LYSO crystal array with a crystal size of 0.95 × 0.95 × 20 mm3 provided a flood histogram with all but the edge crystals clearly resolved, an ER of 17.6%, a DOI resolution of 2.3 mm, and a CTR of 293 ps with the depth-dependent timing correction. CONCLUSIONS PET detectors with small crystal cross-sectional sizes, good DOI and timing resolutions and a reduced number of electronics channels were developed. The detectors can be used to develop high performance organ-specific PET scanners.
Collapse
Affiliation(s)
- Zheng Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Samuel Mungai
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhonghua Kuang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ning Ren
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Siwei Xie
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Qiyu Peng
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Crispin Williams
- European Centre for Nuclear Research (CERN), Geneva, Switzerland
| | - Yongfeng Yang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
26
|
Cook GJR, Alberts IL, Wagner T, Fischer BM, Nazir MS, Lilburn D. The impact of long axial field of view (LAFOV) PET on oncologic imaging. Eur J Radiol 2025; 183:111873. [PMID: 39647272 PMCID: PMC11904125 DOI: 10.1016/j.ejrad.2024.111873] [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: 08/30/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
The development of long axial field of view (LAFOV) positron emission tomography coupled with computed tomography (PET/CT) scanners might be considered the biggest step forward in PET imaging since it became a mainstream clinical modality. Despite increased capital and maintenance costs and data storage requirements, the improvement in image quality, significantly faster acquisition times and lower radiopharmaceutical administered activities, allow a high quality and more efficient clinical service. This step change in technology overcomes some of the limitations of standard short axial field of view scanners. It allows simultaneous imaging of all body systems, and with the ability to obtain high temporal resolution data, it increases potential research applications, particularly in multisystem disease or for dosimetry measurements of novel radiopharmaceuticals. The improvements in sensitivity and signal-to-noise facilitates the use of tracers with long half-lives and low administered activity (e.g. [89Zr]-labelled monoclonal antibodies) or very short half-lives (e.g. [82Rb]), opening up applications that hitherto have been challenging. It is early in the evolution of LAFOV PET/CT and the advantages these systems offer have still to be fully realised in providing additional impact in clinical practice. In this article we describe the potential advantages of LAFOV PET technology and some of the clinical and research applications where it has been applied as well as some of the future developments that may enhance the modality further.
Collapse
Affiliation(s)
- Gary J R Cook
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College, London SE1 7EH, UK.
| | - Ian L Alberts
- Molecular Imaging and Therapy, BC Cancer Agency, Vancouver, BC, Canada; Department of Radiology, University of British Columbia, Vancouver, BC, Canada.
| | - Thomas Wagner
- Department of Nuclear Medicine, Royal Free London NHS Trust, London NW3 1TX, UK.
| | - B Malene Fischer
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, 2100 Copenhagen, Denmark.
| | - Muhummad Sohaib Nazir
- Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK; Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| | - David Lilburn
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College, London SE1 7EH, UK.
| |
Collapse
|
27
|
Benitez‐Aurioles J, Clegg PS, Alcaide‐Corral CJ, Wimberley C, Tavares AAS. Multi-organ kinetic modeling for Na[ 18F]F pre-clinical total-body PET studies. Med Phys 2025; 52:924-937. [PMID: 39499788 PMCID: PMC11788250 DOI: 10.1002/mp.17499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 09/20/2024] [Accepted: 10/11/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND Total-body positron emission tomography (PET), already well-established in the pre-clinical setting, makes it possible to study multi-parameters in biological systems as a whole, rather than focusing on single tissues analysis. Simultaneous kinetic analysis of multiple organs poses some daunting new challenges. PURPOSE To explore quantifying the pharmacokinetics of Na[18F]F in multiple dissimilar murine organs simultaneously in vivo with total-body PET imaging using different compartmental models for each organ and a shared cardiovascular system. METHODS Six mice underwent a 60-min total-body PET scan following intravenous bolus injection of Na[18F]F. Compartmental models were constructed for each organ (heart, lungs, liver, kidneys, and bone) using an image derived input function. Non-linear least squares fitting of a model that connects the five organs to a shared cardiovascular system was used to analyze both the first 3 min of data and the full hour. Analysis was repeated 5000 times using different initial parameter values for each duration, permitting analysis of correlations between parameters. RESULTS The models give a good qualitative account of the activity curves irrespective of the duration of the data; however, the quality of the fits to 3 min of data (averageχ ν 2 $\chi _\nu ^2$ is 2.72) was generally better. Comparison of perfusion values to literature values was possible for the liver and lungs with the former (liver, 0.540 ± 0.177 mL/ml/min) being well-above expectations and the latter (lungs, 0.184 ± 0.413 mL/ml/min) in rough agreement. Correlations between microparameter values (especially affecting k2) caused very noticeable problems for data modeling from both the kidneys and the femur. CONCLUSION The present study demonstrates an approach to performing kinetic modeling for multiple organs simultaneously with Na[18F]F. The observed correlations between microparameter values remain a challenge. Nonetheless, many microparameters can be estimated reliably with a quantitative analysis of perfusion being possible for some organs.
Collapse
Affiliation(s)
- Jose Benitez‐Aurioles
- School of Physics & AstronomyUniversity of EdinburghEdinburghUK
- University of Manchester, Division of InformaticsImaging and Data ScienceManchesterUK
| | - Paul S. Clegg
- School of Physics & AstronomyUniversity of EdinburghEdinburghUK
| | - Carlos J. Alcaide‐Corral
- Edinburgh ImagingUniversity of EdinburghEdinburghUK
- University/BHF Centre for Cardiovascular ScienceUniversity of EdinburghEdinburghUK
| | - Catriona Wimberley
- School of Physics & AstronomyUniversity of EdinburghEdinburghUK
- Edinburgh ImagingUniversity of EdinburghEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Adriana A. S. Tavares
- Edinburgh ImagingUniversity of EdinburghEdinburghUK
- University/BHF Centre for Cardiovascular ScienceUniversity of EdinburghEdinburghUK
| |
Collapse
|
28
|
Cheng Z, Deng X, Song S, Wu Y, Tang H, Zou S, Zhu Y, Liu A, Zhu X. Leveraging small voxel with optimal acquisition time for [ 18F]mFBG total-body PET/CT imaging in pediatric patients with neuroblastoma: a preliminary study. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07098-1. [PMID: 39888420 DOI: 10.1007/s00259-025-07098-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 01/14/2025] [Indexed: 02/01/2025]
Abstract
PURPOSE The advent of total-body PET/CT presents an opportunity for significant advancements in imaging of neuroblastoma with [18F]meta-fluorobenzylguanidine ([18F]mFBG). Small voxel imaging has proven to have better lesion detectability but need enough radioactivity counts. This study aims to balance shortened acquisition times and small voxel reconstruction to keep sufficient image quality and diagnostic confidence on [18F]mFBG total-body PET for neuroblastoma. METHODS We retrospectively enrolled 33 pediatric patients with neuroblastoma who underwent 37 [18F]mFBG total-body uEXPLORER PET/CT scans of 10-min duration. PET images were reconstructed with varying acquisition times (0.5-10 min) and three matrix sizes (192 × 192, 512 × 512 and 1024 × 1024). The subjective (scored on a 5-point scale) and objective image quality (signal-to-noise ratio, SNR) of all the sets of reconstructed images were analyzed by nuclear medicine physicians. For indeterminate lesions identified in the group of 192 × 192 matrix with the 10-min scan (G192-10), diagnostic confidence was further evaluated in images reconstructed with the 512 × 512 and 1024 × 1024 matrices (G512 and G1024). RESULTS Of the 33 patients with 37 [18F]mFBG PET/CT scans, 17 patients with 20 scans had positive [18F]mFBG PET/CT findings. Sufficient subjective image quality was achieved with at least 2-min acquisition of 192 × 192 matrix and 4-min acquisition of 512 × 512 matrix (with all scores ≥ 3). SNR increased with longer acquisition times for the same voxel size, while decreased as voxel size shrunk. Although the Curie and SIOPEN scores remained consistent across G192, G512, and G1024-10 groups, the G512 groups with at least 2-min acquisition and G1024-10 showed significantly higher confidence scores for characterizing indeterminate lesions on the G192-10 images, with almost all indeterminate lesions being rated as very confident. CONCLUSIONS A matrix of 512 × 512 with a minimum of 4-min acquisition on [18F]mFBG total-body PET/CT is recommended for sufficient image quality and improved diagnostic confidence, particularly in detecting indeterminate lesions.
Collapse
Affiliation(s)
- Zhaoting Cheng
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- National Center for Major Public Health Events, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Xiaoyun Deng
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- National Center for Major Public Health Events, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Shuang Song
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- National Center for Major Public Health Events, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Yang Wu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- National Center for Major Public Health Events, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Hongmei Tang
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- National Center for Major Public Health Events, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Sijuan Zou
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- National Center for Major Public Health Events, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Yuankai Zhu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- National Center for Major Public Health Events, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Aiguo Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
- National Center for Major Public Health Events, 1095 Jiefang Ave, Wuhan, 430030, China.
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
- National Center for Major Public Health Events, 1095 Jiefang Ave, Wuhan, 430030, China.
| |
Collapse
|
29
|
McAteer MA, McGowan DR, Cook GJR, Leung HY, Ng T, O'Connor JPB, Aloj L, Barnes A, Blower PJ, Brindle KM, Braun J, Buckley C, Darian D, Evans P, Goh V, Grainger D, Green C, Hall MG, Harding TA, Hines CDG, Hollingsworth SJ, Cristinacce PLH, Illing RO, Lee M, Leurent B, Mallett S, Neji R, Norori N, Pashayan N, Patel N, Prior K, Reiner T, Retter A, Taylor A, van der Aart J, Woollcott J, Wong WL, van der Meulen J, Punwani S, Higgins GS. Translation of PET radiotracers for cancer imaging: recommendations from the National Cancer Imaging Translational Accelerator (NCITA) consensus meeting. BMC Med 2025; 23:37. [PMID: 39849494 PMCID: PMC11756105 DOI: 10.1186/s12916-024-03831-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND The clinical translation of positron emission tomography (PET) radiotracers for cancer management presents complex challenges. We have developed consensus-based recommendations for preclinical and clinical assessment of novel and established radiotracers, applied to image different cancer types, to improve the standardisation of translational methodologies and accelerate clinical implementation. METHODS A consensus process was developed using the RAND/UCLA Appropriateness Method (RAM) to gather insights from a multidisciplinary panel of 38 key stakeholders on the appropriateness of preclinical and clinical methodologies and stakeholder engagement for PET radiotracer translation. Panellists independently completed a consensus survey of 57 questions, rating each on a 9-point Likert scale. Subsequently, panellists attended a consensus meeting to discuss survey outcomes and readjust scores independently if desired. Survey items with median scores ≥ 7 were considered 'required/appropriate', ≤ 3 'not required/inappropriate', and 4-6 indicated 'uncertainty remained'. Consensus was determined as ~ 70% participant agreement on whether the item was 'required/appropriate' or 'not required/not appropriate'. RESULTS Consensus was achieved for 38 of 57 (67%) survey questions related to preclinical and clinical methodologies, and stakeholder engagement. For evaluating established radiotracers in new cancer types, in vitro and preclinical studies were considered unnecessary, clinical pharmacokinetic studies were considered appropriate, and clinical dosimetry and biodistribution studies were considered unnecessary, if sufficient previous data existed. There was 'agreement without consensus' that clinical repeatability and reproducibility studies are required while 'uncertainty remained' regarding the need for comparison studies. For novel radiotracers, in vitro and preclinical studies, such as dosimetry and/or biodistribution studies and tumour histological assessment were considered appropriate, as well as comprehensive clinical validation. Conversely, preclinical reproducibility studies were considered unnecessary and 'uncertainties remained' regarding preclinical pharmacokinetic and repeatability evaluation. Other consensus areas included standardisation of clinical study protocols, streamlined regulatory frameworks and patient and public involvement. While a centralised UK clinical imaging research infrastructure and open access federated data repository were considered necessary, there was 'agreement without consensus' regarding the requirement for a centralised UK preclinical imaging infrastructure. CONCLUSIONS We provide consensus-based recommendations, emphasising streamlined methodologies and regulatory frameworks, together with active stakeholder engagement, for improving PET radiotracer standardisation, reproducibility and clinical implementation in oncology.
Collapse
Affiliation(s)
| | - Daniel R McGowan
- Department of Oncology, University of Oxford, Oxford, UK
- Department of Medical Physics and Clinical Engineering, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Gary J R Cook
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- King's College London and Guy's and St Thomas' PET Centre, St Thomas' Hospital, London, UK
| | - Hing Y Leung
- CRUK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Tony Ng
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
- Oncology Translational Research, GSK, Stevenage, UK
| | - James P B O'Connor
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Luigi Aloj
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Anna Barnes
- Southeast Region, Office of the Chief Scientific Officer, NHS-England, England, UK
- King's Technology Evaluation Centre (KiTEC), School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - Phil J Blower
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - John Braun
- RMH Radiotherapy Focus Group & RMH Biomedical Research Centre Consumer Group, Sutton, UK
| | | | | | - Paul Evans
- GE HealthCare, Pharmaceutical Diagnostics, Chalfont St. Giles, UK
| | - Vicky Goh
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Department of Radiology, NHS Foundation Trust, Guy's and St Thomas, London, UK
| | - David Grainger
- Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Carol Green
- Patient and Public Representative, Oxford, UK
| | - Matt G Hall
- National Physical Laboratory, Teddington, UK
| | - Thomas A Harding
- Prostate Cancer UK, London, UK
- Population Health Sciences, University of Bristol, Bristol, UK
| | | | | | | | - Rowland O Illing
- Department of Surgery & Interventional Science, University College London, London, UK
| | - Martin Lee
- Clinical Trial and Statistics Unit, Institute of Cancer Research, Sutton, UK
- The Royal Marsden Clinical Research Facility, London, UK
| | - Baptiste Leurent
- Department of Statistical Science, University College London, London, UK
| | - Sue Mallett
- Centre for Medical Imaging, University College London, London, UK
| | - Radhouene Neji
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Siemens Healthcare Limited, Camberley, UK
| | | | - Nora Pashayan
- Department of Applied Health Research, University College London, London, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Neel Patel
- Department of Radiology, Churchill Hospital, Oxford University NHS Foundation Trust, Oxford, UK
- Telix Pharmaceuticals Limited, North Melbourne, Australia
| | | | - Thomas Reiner
- Evergreen Theragnostics, Springfield, NJ, 07081, USA
| | - Adam Retter
- Centre for Medical Imaging, University College London, London, UK
| | - Alasdair Taylor
- University Hospitals of Morecambe Bay NHS Foundation Trust, Royal Lancaster Infirmary, Lancaster, UK
| | | | | | - Wai-Lup Wong
- PET CT Department, Strickland Scanner Centre Mount Vernon Hospital, Northwood, UK
| | - Jan van der Meulen
- Department of Health Services Research & Policy, London School of Hygiene & Tropical Medicine, London, UK
| | - Shonit Punwani
- Centre for Medical Imaging, University College London, London, UK
| | | |
Collapse
|
30
|
Katal S, Patel P, Lee J, Taubman K, Gholamrezanezhad A. Total Body PET/CT: A Role in Musculoskeletal Diseases. Semin Nucl Med 2025; 55:86-97. [PMID: 38944556 DOI: 10.1053/j.semnuclmed.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 07/01/2024]
Abstract
Recent advancements in PET technology have culminated in the development of total-body PET (TB-PET) systems, which overcome many limitations of traditional scanners. These TB-PET scanners, while still becoming widely available, represent the forefront of clinical imaging across numerous medical institutions worldwide. Early clinical applications have demonstrated their enhanced image quality, precise lesion quantification, and overall superior performance relative to conventional scanners. The capabilities of TB-PET technology, including extended scan range, ultrahigh sensitivity, exceptional temporal resolution, and dynamic imaging, offer significant potential to tackle unresolved clinical challenges in medical imaging. In this discussion, we aim to explore the emerging applications, opportunities, and future perspectives of TB-PET/CT in musculoskeletal disorders (MSDs). Clinical applications for both oncologic and non-oncologic musculoskeletal diseases are discussed, including inflammatory arthritis, infections, osteoarthritis, osteoporosis, and skeletal muscle disorders. From the ability to visualize small musculoskeletal structures and the entire axial and appendicular skeleton, TB-PET shows significant potential in the diagnosis and management of MSD conditions as it becomes more widely available.
Collapse
Affiliation(s)
- Sanaz Katal
- Department of Medical Imaging, St. Vincent's Hospital, Melbourne, Victoria, Australia; Melborune Theranostic Innovation Centre (MTIC), Melbourne, Victoria, Australia
| | - Parth Patel
- Department of Radiology, University of Central Florida, Orlando, FL
| | - Jonathan Lee
- Department of Radiology, Keck School of Medicine, Los Angeles, CA
| | - Kim Taubman
- Department of Medical Imaging, St. Vincent's Hospital, Melbourne, Victoria, Australia; University of Melbourne, Melbourne, Victoria, Australia
| | | |
Collapse
|
31
|
Dubroff JG, Hsieh CJ, Wiers CE, Lee H, Li EJ, Schubert EK, Mach RH, Kranzler HR. [ 11 C]Carfentanil PET Whole-Body Imaging of Mu-Opioid Receptors: A First In-Human Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.12.31.24319819. [PMID: 39802787 PMCID: PMC11722493 DOI: 10.1101/2024.12.31.24319819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Introduction Mu-opioid receptors (MORs) are G-coupled protein receptors with a high affinity for both endogenous and exogenous opioids. MORs are widely expressed in the central nervous system (CNS), peripheral organs, and the immune system. They mediate pain and reward and have been implicated in the pathophysiology of opioid, cocaine, and other substance use disorders. Using the long axial field-of-view (LAFOV) PennPET Explorer instrument and the MOR selective radioligand [ 11 C]carfentanil ([ 11 C]CFN), we measured the whole-body distribution of MORs in 13 healthy humans. We also examined sex differences in MOR distribution at baseline and after pretreatment with the MOR antagonist naloxone. Methods Six female and seven male healthy subjects underwent two [ 11 C]CFN PET imaging sessions-one at baseline and one immediately following pre-treatment with the MOR antagonist naloxone (13 mcg/kg). Whole-body PET imaging was performed on the PennPET Explorer, a 142-cm axial bore instrument. [ 11 C]CFN brain distribution volume ratios (DVRs) were determined using the occipital cortex and the visual cortex within it as reference regions. For peripheral organ DVRs, the descending aorta and proximal extremity muscle (biceps/triceps) were used as reference regions. Results Naloxone blockade reduced MOR availability by 40-50% in the caudate, putamen, thalamus, amygdala, and ventral tegmentum, brain regions known to express high levels of MORs. Women showed greater receptor occupancy in the thalamus, amygdala, hippocampus and frontal and temporal lobes and a greater naloxone-induced reduction in thalamic MOR availability than men (p's <0.05). For determining brain MOR availability, there was less variance in the visual cortex than the occipital cortex reference region. For peripheral MOR determination, the descending aorta reference region showed less variance than the extremity muscle, but both showed blocking effects of naloxone. Conclusions [ 11 C]CFN whole- body PET scans are useful for understanding MOR physiology under both baseline and blocking conditions. Extra-CNS reference regions may be useful for quantifying radiotracers when a region devoid of binding in the CNS is unavailable. The LAFOV PET instrument was useful for measuring changes in the short-lived radiotracer [ 11 C]CFN, with and without naloxone blocking. Further research is needed to evaluate the behavioral and clinical relevance of sex differences in naloxone-MOR interactions.
Collapse
|
32
|
Wei Q, Wang Y, Huang X, Li D, Zheng Y, Wang P, Sun X, Chai P, Han X, Liu S, Feng B, Zhou W, Zeng X, Zhu M, Zhang Z, Wei L. Performance evaluation of a small-animal PET scanner with 213 mm axis using NEMA NU 4-2008. Med Phys 2025; 52:530-541. [PMID: 39432708 DOI: 10.1002/mp.17469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Long-axis positron emission tomography (PET) has emerged as one of the recent research directions in PET due to its ability to significantly enhance sensitivity and counting performance for low-dose imaging, rapid imaging, and whole-body dynamic imaging. PURPOSE The PET system presented in this study is a long-axis animal PET based on lutetium-yttrium orthosilicate and silicon photomultiplier, designed for whole-body imaging in rats. It features a diameter of 143 mm and an axial length of 213.3 mm. This study evaluated the performance of this PET system in accordance with the National Electrical Manufacturers Association (NEMA) NU 4-2008 standards. METHODS The performance evaluation was conducted according to the NEMA NU 4-2008 standards in terms of spatial resolution, sensitivity, counting rate performance, scatter fraction (SF) and image quality. In addition, a rat imaging study was conducted to assess the imaging capability of this PET system. RESULTS The average energy resolution of the PET system was 12.87%, the average coincidence timing resolution was 751 ps. The FWHM of spatial resolution reconstructed by filtered back projection and 3D-OSEM-PSF algorithm at 5 mm radial offset from the axial center were 1.65 and 0.88 mm. The peak absolute sensitivity measured by a point source at the center of the field of view was evaluated as 6.71% (361-661 keV) and 10.31% (250-750 keV). For the mouse-like phantom, the SF was 11.0% and the peak noise equivalent counting rate (NECR) was 1193 kcps at 94.2 MBq (2.54 mCi). For the rat-like phantom, the SF was 26.8% and the NECR was 682.5 kcps at 78.6 MBq (2.12 mCi). CONCLUSIONS The performance measurement results demonstrate that this PET system exhibits high sensitivity and count rate performance, making it potential for high-quality whole-body dynamic imaging of rats.
Collapse
Affiliation(s)
- Qing Wei
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Yingjie Wang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Xianchao Huang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Daowu Li
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Yushuang Zheng
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Peilin Wang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Xiaoli Sun
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Pei Chai
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Xiaorou Han
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Shuangquan Liu
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Baotong Feng
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Wei Zhou
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Xiangtao Zeng
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Meiling Zhu
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Zhiming Zhang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| | - Long Wei
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
- Jinan Laboratory of Applied Nuclear Science, Jinan, China
- CAEA center of Excellence on Nuclear Technology Applications for Nuclear Detection and Imaging, Beijing, China
| |
Collapse
|
33
|
Zoghi S, Mingels C, Badawi RD, Spencer BA, Yarbrough TL, Nardo L, Chaudhari AJ. Role of Total Body PET/CT in Inflammatory Disorders. Semin Nucl Med 2025; 55:41-51. [PMID: 39578110 PMCID: PMC11645246 DOI: 10.1053/j.semnuclmed.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/24/2024]
Abstract
Inflammatory disorders historically have been difficult to monitor with conventional PET imaging due to limitations including radiation exposure, lack of validated imaging biomarkers, low spatial resolution, and long acquisition durations. However, the recent development of long-axial field-of-view (LAFOV) PET/CT scanners may allow utilization of novel noninvasive biomarkers to diagnose, predict outcomes, and monitor therapeutic response of inflammatory conditions. LAFOV PET scanners can image most of the human body (if not the entire body) simultaneously in one bed position, with improved signal collection efficiency compared to conventional PET scanners. This allows for imaging with shorter acquisition durations, decreased injected radiotracer dose, prolonged uptake times, or a combination of any of these. In addition, LAFOV PET scanners enable whole-body dynamic imaging. Altogether, these intrinsically superior capabilities in assessing both local and systemic diseases, have allowed these scanners to make increasingly significant contributions to the assessment of inflammatory conditions. This review aims to further explore the role and benefits of LAFOV scanners for imaging various inflammatory conditions while addressing future developments and challenges faced by this technology.
Collapse
Affiliation(s)
- Shervin Zoghi
- Department of Radiology, University of California Davis, Sacramento, CA, USA.
| | - Clemens Mingels
- Department of Radiology, University of California Davis, Sacramento, CA, USA; Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ramsey D Badawi
- Department of Radiology, University of California Davis, Sacramento, CA, USA
| | - Benjamin A Spencer
- Department of Radiology, University of California Davis, Sacramento, CA, USA
| | - Tracy L Yarbrough
- Department of Radiology, University of California Davis, Sacramento, CA, USA
| | - Lorenzo Nardo
- Department of Radiology, University of California Davis, Sacramento, CA, USA
| | - Abhijit J Chaudhari
- Department of Radiology, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
34
|
Godinez F, Mingels C, Bayerlein R, Mehadji B, Nardo L. Total Body PET/CT: Future Aspects. Semin Nucl Med 2025; 55:107-115. [PMID: 39542814 PMCID: PMC11977673 DOI: 10.1053/j.semnuclmed.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 11/17/2024]
Abstract
Total-body (TB) positron emission tomography (PET) scanners are classified by their axial field of view (FOV). Long axial field of view (LAFOV) PET scanners can capture images from eyes to thighs in a one-bed position, covering all major organs with an axial FOV of about 100 cm. However, they often miss essential areas like distal lower extremities, limiting their use beyond oncology.TB-PET is reserved for scanners with a FOV of 180 cm or longer, allowing coverage of most of the body. LAFOV PET technology emerged about 40 years ago but gained traction recently due to advancements in data acquisition and cost. Early research highlighted its benefits, leading to the first FDA-cleared TB-PET/CT device in 2019 at UC Davis. Since then, various LAFOV scanners with enhanced capabilities have been developed, improving image quality, reducing acquisition times, and allowing for dynamic imaging. The uEXPLORER, the first LAFOV scanner, has a 194 cm active PET AFOV, far exceeding traditional scanners. The Panorama GS and others have followed suit in optimizing FOVs. Despite slow adoption due to the COVID pandemic and costs, over 50 LAFOV scanners are now in use globally. This review explores the future of LAFOV technology based on recent literature and experiences, covering its clinical applications, implications for radiation oncology, challenges in managing PET data, and expectations for technological advancements.
Collapse
Affiliation(s)
- Felipe Godinez
- Department of Radiology, University of California Davis, Sacramento, CA.
| | - Clemens Mingels
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Reimund Bayerlein
- Department of Biomedical Engineering, University of California Davis, Davis, CA
| | - Brahim Mehadji
- Department of Radiology, University of California Davis, Sacramento, CA
| | - Lorenzo Nardo
- Department of Radiology, University of California Davis, Sacramento, CA
| |
Collapse
|
35
|
Wu H, Liu G, Yu H, Zheng Z, He Y, Shi H. Feasibility of ultra-low-activity 18F-FDG PET/CT imaging in children. Br J Radiol 2025; 98:136-142. [PMID: 39423099 DOI: 10.1093/bjr/tqae208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 12/26/2023] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
OBJECTIVES To investigate the feasibility of paediatric 18F-FDG total-body PET/CT imaging with an ultra-low activity and explore an optimized acquisition time range. METHODS A total of 38 paediatric patients were prospectively enrolled and underwent dynamic total-body PET/CT imaging using ultra-low 18F-FDG activity (0.37 MBq/kg). The 60-minute list-mode raw data were acquired and then reconstructed as static PET images by using 50-51, 50-52, 50-53, 50-54, 50-55, 50-58, 50-60, and 45-60 minutes data, which were noted as G1, G2, G3, G4, G5, G8, G10, and G15, respectively. Image qualities were subjectively evaluated using the Likert scale and were objectively evaluated by the quantitative metrics including standard uptake value (SUV), signal-to-noise ratio (SNR), target-to-background ratio (TBR), and contrast-to-noise ratio (CNR). RESULTS The injected activity of FDG was 13.38 ± 5.68 MBq (4.40-28.16 MBq) and produced 0.58 ± 0.19 mSv (0.29-1.04 mSv) of effective dose. The inter-reader agreement of subjective image quality was excellent (kappa = 0.878; 95% CI, 0.845-0.910). The average scores of image quality for G1-G15 were 1.10 ± 0.20, 2.03 ± 0.26, 2.66 ± 0.35, 3.00 ± 0.27, 3.32 ± 0.34, 4.25 ± 0.30, 4.49 ± 0.36, and 4.70 ± 0.37, respectively. All image scores are above 3, and all lesions are detectable starting from G8. SNRs of backgrounds, TBRs, and CNRs were significant differences from the control group before G8 (all P < 0.05). CONCLUSION The image quality of the 8 min acquisition for paediatric 18F-FDG total-body PET/CT with an ultra-low activity could meet the diagnostic requirements. ADVANCES IN KNOWLEDGE This study confirms the feasibility of ultra-low dose PET imaging in children, and its methods and findings may guide clinical practice. Paediatric patients will benefit from reduced radiation doses.
Collapse
Affiliation(s)
- Ha Wu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Nuclear Medicine, Children's Hospital, Fudan University, Shanghai, 201102, China
| | - Guobing Liu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Haojun Yu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhe Zheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yibo He
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
36
|
González Trotter D, McGinniss J, Mohammadi K, Musser BJ, Herman GA, Mellis S, Economides AN. Comparison of PET/CT versus CT only in the assessment of new heterotopic ossification bone lesions in patients with fibrodysplasia ossificans progressiva. Bone 2025; 190:117280. [PMID: 39395655 DOI: 10.1016/j.bone.2024.117280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an ultra-rare disorder characterized by the deposition of bone in soft tissues, known as heterotopic ossification (HO). This post hoc analysis compared the performance of two imaging modalities for the detection and volumetric measurement of new HO lesions. LUMINA-1, a phase 2, randomized, double-blind study (NCT03188666), evaluated the safety and efficacy of garetosmab, an anti-activin A antibody, versus placebo in adult patients with FOP. From baseline through to week 28, 18F-labeled sodium fluoride positron emission tomography (PET)/X-ray computed tomography (CT) and CT-only scans prospectively acquired during the initial placebo-controlled period of the study were independently reviewed by two sets of fixed blinded readers plus an adjudicator for the presence and volume of new HO lesions. The number of patients with new lesions was 14/44 (31.8 %) and 12/44 (27.3 %) as detected by PET/CT and CT only, respectively. The aggregate number/volume of new lesions were very similar both for the placebo and the garetosmab group between PET/CT (27/245.0 cm3 and 3/21.3 cm3, respectively) and CT only (37/261.8 cm3 and 1/0.1 cm3, respectively). The mean (standard deviation) number of new lesions per patient by PET/CT through week 28 was 0.68 (1.57) versus 0.86 (1.95) as detected by CT only. Through week 28, the mean (standard deviation) volume of new lesions per patient detected by PET/CT was 6.05 (14.88) cm3 versus 5.94 (21.13) cm3 by CT only. Moderate agreement between PET/CT and CT-only detection was observed when identifying patients with new lesions, with a kappa coefficient of 0.46 (standard error, 0.146; 95 % confidence interval, 0.17-0.74). CT-only imaging showed similar performance to PET/CT in the detection and characterization of new HO lesions. CT-only imaging therefore is a viable option for the assessment of therapies on new HO in patients with FOP.
Collapse
Affiliation(s)
| | | | | | | | | | - Scott Mellis
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | |
Collapse
|
37
|
Rathod N, Jutidamrongphan W, Bosbach WA, Chen Y, Penner JL, Sari H, Zeimpekis K, Montes AL, Moskal P, Stepien E, Shi K, Rominger A, Seifert R. Total Body PET/CT: Clinical Value and Future Aspects of Quantification in Static and Dynamic Imaging. Semin Nucl Med 2025; 55:98-106. [PMID: 39616013 DOI: 10.1053/j.semnuclmed.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 12/15/2024]
Abstract
Total body (TB) Positron Emission Tomography (PET) / Computed Tomography (CT) scanners have revolutionized nuclear medicine by enabling whole-body imaging in a single bed position.1 This review assesses the physical and clinical value of TB-PET/CT, with a focus on the advancements in both static and dynamic imaging, as well as the evolving quantification techniques. The significantly enhanced sensitivity of TB scanners can reduce radiation exposure and scan time, offering improved patient comfort and making it particularly useful for pediatric imaging and various other scenarios. Shorter scan times also decrease motion artifacts, leading to higher-quality images and better diagnostic accuracy. Dynamic PET imaging with TB scanners extends these advantages by capturing temporal changes in tracer uptake over time, providing real-time insights into both structural and functional assessment, and promoting the ability to monitor disease progression and treatment response. We also present CT-free attenuation correction methods that utilize the increased sensitivity of TB-PET as a potential improvement for dynamic TB-PET protocols. In static imaging, emerging quantification techniques such as dual-tracer PET using TB scanners allow imaging of two biological pathways, simultaneously, for a more comprehensive assessment of disease. In addition, positronium imaging, a novel technique utilizing positronium lifetime measurements, is introduced as a promising aspect for providing structural information alongside functional quantification. Finally, the potential of expanding clinical applications with the increased sensitivity of TB-PET/CT scanners is discussed.
Collapse
Affiliation(s)
- Narendra Rathod
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland.
| | - Warissara Jutidamrongphan
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Wolfram Andreas Bosbach
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Yizhou Chen
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Jan Luca Penner
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Hasan Sari
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Konstantinos Zeimpekis
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Alejandro López Montes
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Pawel Moskal
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland and Centre for Theranostics, Jagiellonian University, Krakow, Poland
| | - Ewa Stepien
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland and Centre for Theranostics, Jagiellonian University, Krakow, Poland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Robert Seifert
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| |
Collapse
|
38
|
Hicks RJ, Ware RE, Callahan J. Total-Body PET/CT: Pros and Cons. Semin Nucl Med 2025; 55:11-20. [PMID: 39289090 DOI: 10.1053/j.semnuclmed.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 09/19/2024]
Abstract
PET/CT devices with an axial field-of-view (FOV) of 1 m allow simultaneous imaging from the head to the upper thighs, the typical axial extent of many "whole-body" oncological studies acquired by moving a patient sequentially through a conventional FOV device, or rapid total-body imaging using the same approach. Increasing the FOV to around 2 m provides true simultaneous total-body imaging. Either approach dramatically increases the sensitivity for detection of annihilation events arising within the body. For the purposes of this review, both configurations are considered to represent "total-body" PET/CT devices because they share both advantages and disadvantages. These pros and cons are discussed in the context of both clinical and research applications from a patient and institutional perspective.
Collapse
Affiliation(s)
- Rodney J Hicks
- The Melbourne Theranostic Innovation Centre, North Melbourne, Victoria 3051, Australia; St Vincent's Hospital, Department of Medicine, The University of Melbourne, Fitzroy, Victoria 3065, Australia.
| | - Robert E Ware
- The Melbourne Theranostic Innovation Centre, North Melbourne, Victoria 3051, Australia
| | - Jason Callahan
- The Melbourne Theranostic Innovation Centre, North Melbourne, Victoria 3051, Australia
| |
Collapse
|
39
|
Sachpekidis C, Dimitrakopoulou-Strauss A. Long Axial Field-of-View (LAFOV) PET/CT in Prostate Cancer. Semin Nucl Med 2025; 55:67-75. [PMID: 38825439 DOI: 10.1053/j.semnuclmed.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 05/16/2024] [Indexed: 06/04/2024]
Abstract
PSMA-targeted PET/CT is currently considered the most effective non-invasive diagnostic technique for imaging PSMA-positive lesions in prostate cancer (PC), and its introduction has significantly enhanced the role of nuclear medicine in both the diagnosis and therapy (theranostics) of this oncological entity. In line with developments in radiopharmaceuticals, significant progress has been made in the development of PET/CT systems. In particular, the advent of long axial field-of-view (LAFOV) PET/CT scanners has represented a major leap forward in molecular imaging, with early results from clinical applications of these systems showing significant improvements over previous standard axial field-of-view systems in terms of sensitivity, image quality and lesion quantification, while enabling whole-body dynamic PET imaging. In this context, the introduction of the new LAFOV scanners may further enhance the use and potential of PSMA-ligand PET/CT in the diagnosis and management of PC. The initial but steadily growing literature on the application of the new technology in the field of PSMA-ligand PET/CT has already yielded encouraging results regarding the detection of PC lesions with high sensitivity while providing the possibility of ultra-fast or ultra-low dose examinations. Moreover, whole-body dynamic PET has rendered for the first time feasible to capture the pharmacokinetics PSMA-ligands in all major organs and most tumor lesions with high temporal resolution. The main results of these studies are presented in this review.
Collapse
Affiliation(s)
- Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | | |
Collapse
|
40
|
Hricak H, Mayerhoefer ME, Herrmann K, Lewis JS, Pomper MG, Hess CP, Riklund K, Scott AM, Weissleder R. Advances and challenges in precision imaging. Lancet Oncol 2025; 26:e34-e45. [PMID: 39756454 DOI: 10.1016/s1470-2045(24)00395-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 01/07/2025]
Abstract
Technological innovations in genomics and related fields have facilitated large sequencing efforts, supported new biological discoveries in cancer, and spawned an era of liquid biopsy biomarkers. Despite these advances, precision oncology has practical constraints, partly related to cancer's biological diversity and spatial and temporal complexity. Advanced imaging technologies are being developed to address some of the current limitations in early detection, treatment selection and planning, drug delivery, and therapeutic response, as well as difficulties posed by drug resistance, drug toxicity, disease monitoring, and metastatic evolution. We discuss key areas of advanced imaging for improving cancer outcomes and survival. Finally, we discuss practical challenges to the broader adoption of precision imaging in the clinic and the need for a robust translational infrastructure.
Collapse
Affiliation(s)
- Hedvig Hricak
- Department of Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marius E Mayerhoefer
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Jason S Lewis
- Department of Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology and Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
| | - Martin G Pomper
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher P Hess
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, CA, USA
| | - Katrine Riklund
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
| | - Andrew M Scott
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia; School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia; Faculty of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Ralph Weissleder
- Department of Radiology and Center for Systems Biology, Massachusetts General Brigham, Boston, MA, USA; Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Greenwood HE, Barber AR, Edwards RS, Tyrrell WE, George ME, Dos Santos SN, Baark F, Tanc M, Khalil E, Falzone A, Ward NP, DeBlasi JM, Torrente L, Soni PN, Pearce DR, Firth G, Smith LM, Vilhelmsson Timmermand O, Huebner A, Swanton C, Hynds RE, DeNicola GM, Witney TH. Imaging NRF2 activation in non-small cell lung cancer with positron emission tomography. Nat Commun 2024; 15:10484. [PMID: 39690148 PMCID: PMC11652680 DOI: 10.1038/s41467-024-54852-4] [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: 03/04/2024] [Accepted: 11/21/2024] [Indexed: 12/19/2024] Open
Abstract
Mutations in the NRF2-KEAP1 pathway are common in non-small cell lung cancer (NSCLC) and confer broad-spectrum therapeutic resistance, leading to poor outcomes. Currently, there is no means to non-invasively identify NRF2 activation in living subjects. Here, we show that positron emission tomography imaging with the system xc- radiotracer, [18F]FSPG, provides a sensitive and specific marker of NRF2 activation in orthotopic, patient-derived, and genetically engineered mouse models of NSCLC. We found a NRF2-related gene expression signature in a large cohort of NSCLC patients, suggesting an opportunity to preselect patients prior to [18F]FSPG imaging. Furthermore, we reveal that system xc- is a metabolic vulnerability that can be therapeutically targeted with an antibody-drug conjugate for sustained tumour growth suppression. Overall, our results establish [18F]FSPG as a predictive marker of therapy resistance in NSCLC and provide the basis for the clinical evaluation of both imaging and therapeutic agents that target this important antioxidant pathway.
Collapse
Affiliation(s)
- Hannah E Greenwood
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Abigail R Barber
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Richard S Edwards
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Will E Tyrrell
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Madeleine E George
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Sofia N Dos Santos
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Friedrich Baark
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Muhammet Tanc
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Eman Khalil
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Aimee Falzone
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Nathan P Ward
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Janine M DeBlasi
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Laura Torrente
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Pritin N Soni
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - David R Pearce
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - George Firth
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Lydia M Smith
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK
| | | | - Ariana Huebner
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Charles Swanton
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Robert E Hynds
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Gina M DeNicola
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Timothy H Witney
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, UK.
| |
Collapse
|
42
|
Zhu J, Zhou W, Yao Y, Zhou X, Ma X, Zhang B, Yang Z, Tang B, Zhu H, Li N. Targeted Positron Emission Tomography-Tracked Biomimetic Codelivery Synergistically Amplifies Ferroptosis and Pyroptosis for Inducing Lung Cancer Regression and Anti-PD-L1 Immunotherapy Efficacy. ACS NANO 2024; 18:31401-31420. [PMID: 39475541 DOI: 10.1021/acsnano.4c11278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2024]
Abstract
The chemoresistance and systemic toxicity of cisplatin (CDDP) severely limit its application in the treatment of non-small cell lung cancer (NSCLC). Here, I-124 labeled cancer cell membrane biomimetic nanovesicles loading Polyphyllin VI (PPVI) and CDDP (termed 124I-P/C@CMLvs) were constructed to enhance the sensitivity and efficacy of CDDP. The radiochemical purity (RCP) of 124I-P/C@CMLvs reached more than 99% and maintained reliable stability in vitro. Micro-positron emission tomography (micro-PET) imaging of I-124 quantitatively revealed the distribution and specific homologous tumor targeting ability of 124I-P/C@CMLvs in vivo with superior diagnosis performance, beneficial for dynamically monitoring the efficacy against NSCLC. Loaded PPVI significantly strengthened the sensitivity of NSCLC to CDDP therapy and exerted synergistic anti-tumor effect in vitro and in vivo, which was achieved by PPVI promoting p53 deubiquitination and stimulating reactive oxygen species (ROS) production to trigger the crosstalk between the amplification of GPX4 signaling-mediated ferroptosis and NLRP3/GSDMD/Caspase-1 axis-mediated pyroptosis. 124I-P/C@CMLvs also significantly stimulated the infiltration of immune cells including dendritic cells, CD8+ T cells, and CD4+ T cells in tumor tissues (P < 0.05). The combination of 124I-P/C@CMLvs and anti-PD-L1 therapy further synergistically promoted NSCLC regression. Altogether, 124I-P/C@CMLvs provide a transformational solution to the challenge of improving CDDP sensitivity and realizing the integration of diagnosis, treatment, and monitoring of NSCLC.
Collapse
Affiliation(s)
- Jinyu Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Wenyuan Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Yuan Yao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Xin Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Xiaokun Ma
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Baohui Zhang
- Department of Physiology, School of Life Science, China Medical University, Shenyang,Liaoning 110122, China
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Bufu Tang
- Department of Radiation Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032,China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| | - Nan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, China
| |
Collapse
|
43
|
Kim SM, Lee JS. A comprehensive review on Compton camera image reconstruction: from principles to AI innovations. Biomed Eng Lett 2024; 14:1175-1193. [PMID: 39465108 PMCID: PMC11502649 DOI: 10.1007/s13534-024-00418-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/09/2024] [Accepted: 08/20/2024] [Indexed: 10/29/2024] Open
Abstract
Compton cameras have emerged as promising tools in biomedical imaging, offering sensitive gamma-ray imaging capabilities for diverse applications. This review paper comprehensively overviews the latest advancements in Compton camera image reconstruction technologies. Beginning with a discussion of the fundamental principles of Compton scattering and its relevance to gamma-ray imaging, the paper explores the key components and design considerations of Compton camera systems. We then review various image reconstruction algorithms employed in Compton camera systems, including analytical, iterative, and statistical approaches. Recent developments in machine learning-based reconstruction methods are also discussed, highlighting their potential to enhance image quality and reduce reconstruction time in biomedical applications. In particular, we focus on the challenges posed by conical back-projection in Compton camera image reconstruction, and how innovative signal processing techniques have addressed these challenges to improve image accuracy and spatial resolution. Furthermore, experimental validations of Compton camera imaging in preclinical and clinical settings, including multi-tracer and whole-gamma imaging studies are introduced. In summary, this review provides potentially useful information about the current state-of-the-art Compton camera image reconstruction technologies, offering a helpful guide for investigators new to this field.
Collapse
Affiliation(s)
- Soo Mee Kim
- Maritime ICT & Mobility Research Department, Korea Institute of Ocean Science and Technology, Busan, Korea
| | - Jae Sung Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- Brightonix Imaging Inc., Seoul, Korea
| |
Collapse
|
44
|
Zhou X, Xue S, Li L, Seifert R, Dong S, Chen R, Huang G, Rominger A, Liu J, Shi K. Sedation-free pediatric [ 18F]FDG imaging on totalbody PET/CT with the assistance of artificial intelligence. Eur J Nucl Med Mol Imaging 2024; 51:4062-4072. [PMID: 38958680 DOI: 10.1007/s00259-024-06818-3] [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: 03/27/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE While sedation is routinely used in pediatric PET examinations to preserve diagnostic quality, it may result in side effects and may affect the radiotracer's biodistribution. This study aims to investigate the feasibility of sedation-free pediatric PET imaging using ultra-fast total-body (TB) PET scanners and deep learning (DL)-based attenuation and scatter correction (ASC). METHODS This retrospective study included TB PET (uExplorer) imaging of 35 sedated pediatric patients under four years old to determine the minimum effective scanning time. A DL-based ASC method was applied to enhance PET quantification. Both quantitative and qualitative assessments were conducted to evaluate the image quality of ultra-fast DL-ASC PET. Five non-sedated pediatric patients were subsequently used to validate the proposed approach. RESULTS Comparisons between standard 300-second and ultra-fast 15-second imaging, CT-ASC and DL-ASC ultra-fast 15-second images, as well as DL-ASC ultra-fast 15-second images in non-sedated and sedated patients, showed no significant differences in qualitative scoring, lesion detectability, and quantitative Standard Uptake Value (SUV) (P = ns). CONCLUSIONS This study demonstrates that pediatric PET imaging can be effectively performed without sedation by combining ultra-fast imaging techniques with a DL-based ASC. This advancement in sedation-free ultra-fast PET imaging holds potential for broader clinical adoption.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Song Xue
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Street Freiburgstr. 18, Bern, 3010, Switzerland
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Lianghua Li
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Robert Seifert
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Street Freiburgstr. 18, Bern, 3010, Switzerland
| | - Shunjie Dong
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruohua Chen
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Street Freiburgstr. 18, Bern, 3010, Switzerland
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Street Freiburgstr. 18, Bern, 3010, Switzerland
| |
Collapse
|
45
|
Liu G, Gu T, Chen S, Gu Y, Yu H, Shi H. Total-body dynamic PET/CT imaging reveals kinetic distribution of [ 13N]NH 3 in normal organs. Eur J Nucl Med Mol Imaging 2024; 51:3888-3899. [PMID: 38976037 DOI: 10.1007/s00259-024-06826-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/30/2024] [Indexed: 07/09/2024]
Abstract
PURPOSE To systematically investigate kinetic metrics and metabolic trapping of [13N]NH3 in organs. METHODS Eleven participants performed total-body [13N]NH3 dynamic positron emission tomography (PET). Regions of interest were drawn in organs to obtain time-to-activity curves (TACs), which were fitted with an irreversible two-tissue compartment model (2TC) to investigate constant rates K1, k2 and k3, and to calculate Ki. Additionally, one-tissue compartment model using full data (1TCfull) and the first four minutes of data (1TC4min) were fitted to TAC data. K1 and k2 were compared among different models to assess [13N]NH3 trapping in organs. RESULTS Kinetic rates of [13N]NH3 varied significantly among organs. The mean K1 ranged from 0.049 mL/cm3/min in the muscle to 2.936 mL/cm3/min in the kidney. The k2 and k3 were lowest in the liver (0.001 min- 1) and in the pituitary (0.009 min- 1), while highest in the kidney (0.587 min- 1) and in the liver (0.800 min- 1), respectively. The Ki was largest in the myocardium (0.601 ± 0.259 mL/cm3/min) while smallest in the bone marrow (0.028 ± 0.022 mL/cm3/min). Three groups of organs with similar kinetic characteristics were revealed: (1) the thyroid, the lung, the spleen, the pancreas, and the kidney; (2) the liver and the muscle; and (3) the cortex, the white matter, the cerebellum, the pituitary, the parotid, the submandibular gland, the myocardium, the bone, and the bone marrow. Obvious k3 was identified in multiple organs, and significant changes of K1 in multiple organs and k2 in most organs were found between 2TC and 1TCfull, but both K1 and k2 were comparable between 2TC and 1TC4min. CONCLUSION The kinetic rates of [13N]NH3 differed among organs with some have obvious 13N-anmmonia trapping. The normal distribution of kinetic metrics of 13N-anmmonia in organs can serve as a reference for its potential use in tumor imaging.
Collapse
Affiliation(s)
- Guobing Liu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Institute of Nuclear Medicine, Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Taoying Gu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Institute of Nuclear Medicine, Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuguang Chen
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Institute of Nuclear Medicine, Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yushen Gu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Institute of Nuclear Medicine, Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haojun Yu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Institute of Nuclear Medicine, Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China.
- Institute of Nuclear Medicine, Fudan University, Shanghai, China.
- Shanghai Institute of Medical Imaging, Shanghai, China.
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
46
|
Cui J, Luo Y, Chen D, Shi K, Su X, Liu H. IE-CycleGAN: improved cycle consistent adversarial network for unpaired PET image enhancement. Eur J Nucl Med Mol Imaging 2024; 51:3874-3887. [PMID: 39042332 DOI: 10.1007/s00259-024-06823-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/30/2024] [Indexed: 07/24/2024]
Abstract
PURPOSE Technological advances in instruments have greatly promoted the development of positron emission tomography (PET) scanners. State-of-the-art PET scanners such as uEXPLORER can collect PET images of significantly higher quality. However, these scanners are not currently available in most local hospitals due to the high cost of manufacturing and maintenance. Our study aims to convert low-quality PET images acquired by common PET scanners into images of comparable quality to those obtained by state-of-the-art scanners without the need for paired low- and high-quality PET images. METHODS In this paper, we proposed an improved CycleGAN (IE-CycleGAN) model for unpaired PET image enhancement. The proposed method is based on CycleGAN, and the correlation coefficient loss and patient-specific prior loss were added to constrain the structure of the generated images. Furthermore, we defined a normalX-to-advanced training strategy to enhance the generalization ability of the network. The proposed method was validated on unpaired uEXPLORER datasets and Biograph Vision local hospital datasets. RESULTS For the uEXPLORER dataset, the proposed method achieved better results than non-local mean filtering (NLM), block-matching and 3D filtering (BM3D), and deep image prior (DIP), which are comparable to Unet (supervised) and CycleGAN (supervised). For the Biograph Vision local hospital datasets, the proposed method achieved higher contrast-to-noise ratios (CNR) and tumor-to-background SUVmax ratios (TBR) than NLM, BM3D, and DIP. In addition, the proposed method showed higher contrast, SUVmax, and TBR than Unet (supervised) and CycleGAN (supervised) when applied to images from different scanners. CONCLUSION The proposed unpaired PET image enhancement method outperforms NLM, BM3D, and DIP. Moreover, it performs better than the Unet (supervised) and CycleGAN (supervised) when implemented on local hospital datasets, which demonstrates its excellent generalization ability.
Collapse
Affiliation(s)
- Jianan Cui
- The Institute of Information Processing and Automation, College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Yi Luo
- The State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, China
| | - Donghe Chen
- The PET Center, Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Kuangyu Shi
- The Department of Nuclear Medicine, Bern University Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Xinhui Su
- The PET Center, Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China.
| | - Huafeng Liu
- The State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
47
|
Wang D, Mo Y, Liu F, Zheng S, Liu H, Li H, Guo J, Fan W, Qiu B, Zhang X, Liu H. Repeated dynamic [ 18F]FDG PET/CT imaging using a high-sensitivity PET/CT scanner for assessing non-small cell lung cancer patients undergoing induction immuno-chemotherapy followed by hypo-fractionated chemoradiotherapy and consolidative immunotherapy: report from a prospective observational study (GASTO-1067). Eur J Nucl Med Mol Imaging 2024; 51:4083-4098. [PMID: 38953934 DOI: 10.1007/s00259-024-06819-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024]
Abstract
OBJECTIVE The study aims to investigate the role of dynamic [18F]FDG PET/CT imaging by high-sensitivity PET/CT scanner for assessing patients with locally advanced non-small cell lung cancer (LA-NSCLC) who undergo induction immuno-chemotherapy, followed by concurrent hypo-fractionated chemoradiotherapy (hypo-CCRT) and consolidative immunotherapy. METHODS Patients with unresectable LA-NSCLC are prospectively recruited. Dynamic [18F]FDG PET/CT scans are conducted at four timepoints: before treatment (Baseline), after induction immuno-chemotherapy (Post-IC), during hypo-CCRT (Mid-hypo-CCRT) and after hypo-CCRT (Post-hypo-CCRT). The primary lung tumors (PTs) are manually delineated, and the metabolic features, including the Patlak-Ki (Ki), maximum SUV (SUVmax), metabolic tumor volume (MTV) and total lesion glycolysis (TLG) have been evaluated. The expressions of CD3, CD8, CD68, CD163, CD34 and Ki67 in primary lung tumors at baseline are assayed by immunohistochemistry. The levels of blood lymphocytes at four timepoints are analyzed with flow cytometry. RESULTS Fifteen LA-NSCLC patients are enrolled between December 2020 and December 2022. Baseline Ki of primary tumor yields the highest AUC values of 0.722 and 0.796 for predicting disease progression and patient death, respectively. Patients are classified into the High FDG Ki group (n = 8, Ki > 2.779 ml/min/100 g) and the Low FDG Ki group (n = 7, Ki ≤ 2.779 ml/min/100 g). The High FDG Ki group presents better progression-free survival (P = 0.01) and overall survival (P = 0.025). The High FDG Ki group exhibits more significant reductions in Ki after hypo-CCRT compared to the Low FDG Ki group. Patients with a reduction in Ki > 73.1% exhibit better progression-free survival than those with a reduction ≤ 73.1% in Ki (median: not reached vs. 7.33 months, P = 0.12). The levels of CD3+ T cells (P = 0.003), CD8+ T cells (P = 0.002), CD68+ macrophages (P = 0.071) and CD163+ macrophages (P = 0.012) in primary tumor tissues are higher in the High FDG Ki group. The High FDG Ki group has higher CD3+CD8+ lymphocytes in blood at baseline (P = 0.108), post-IC (P = 0.023) and post-hypo-CCRT (P = 0.041) than the Low FDG Ki group. CONCLUSIONS The metabolic features in the High FDG Ki group significantly decrease during the treatment, particularly after induction immuno-chemotherapy. The Ki value of primary tumor shows significant relationship with the treatment response and survival in LA-NSCLC patients by the combined immuno-chemoradiotherapy regimen. TRIAL REGISTRATION ClinicalTrials.gov. NCT04654234. Registered 4 December 2020.
Collapse
Affiliation(s)
- DaQuan Wang
- Department of Radiation Oncology, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China
| | - YiWen Mo
- Department of Nuclear Medicine, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China
| | - FangJie Liu
- Department of Radiation Oncology, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China
| | - ShiYang Zheng
- Department of Radiation Oncology, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China
| | - Hui Liu
- United Imaging Healthcare, Shanghai, China
| | - HongDi Li
- United Imaging Healthcare, Shanghai, China
| | - JinYu Guo
- Department of Radiation Oncology, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China
| | - Wei Fan
- Department of Nuclear Medicine, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China
| | - Bo Qiu
- Department of Radiation Oncology, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China.
| | - Xu Zhang
- Department of Nuclear Medicine, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Hui Liu
- Department of Radiation Oncology, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China.
| |
Collapse
|
48
|
Yi M, Lee D, Gola A, Merzi S, Penna M, Lee JS, Cherry SR, Kwon SI. Segmented readout for Cherenkov time-of-flight positron emission tomography detectors based on bismuth germanate. ARXIV 2024:arXiv:2410.12161v1. [PMID: 39483343 PMCID: PMC11527102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Positron emission tomography (PET) is the most sensitive biomedical imaging modality for non-invasively detecting and visualizing positron-emitting radiopharmaceuticals within a subject. In PET, measuring the time-of-flight (TOF) information for each pair of 511-keV annihilation photons improves effective sensitivity but requires high timing resolution. Hybrid materials that emit both scintillation and Cherenkov photons, such as bismuth germanate (BGO), recently offer the potential for more precise timing information from Cherenkov photons while maintaining adequate energy resolution from scintillation photons. However, a significant challenge in using such hybrid materials for TOF PET applications lies in the event-dependent timing spread caused by the mixed detection of Cherenkov and scintillation photons due to relatively lower production of Cherenkov photons. This study introduces an innovative approach by segmenting silicon photomultiplier (SiPM) pixels coupled to a single crystal, rather than using traditional SiPMs that are as large as or larger than the crystals they read. We demonstrated that multiple time stamps and photon counts obtained from the segmented SiPM can classify events by providing temporal photon density, effectively addressing this challenge. The approach and findings would lead to new opportunities in applications that require precise timing and photon counting, spanning the fields of medical imaging, high-energy physics, and optical physics.
Collapse
Affiliation(s)
- Minseok Yi
- Department of Biomedical Engineering, University of California, Davis, USA
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National Graduate School, Republic of Korea
- Integrated Major in Innovative Medical Science, Seoul National Graduate School, Republic of Korea
| | - Daehee Lee
- Department of Biomedical Engineering, University of California, Davis, USA
| | | | | | | | - Jae Sung Lee
- Brightonix Imaging Inc., Republic of Korea
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National Graduate School, Republic of Korea
- Integrated Major in Innovative Medical Science, Seoul National Graduate School, Republic of Korea
| | - Simon R Cherry
- Department of Biomedical Engineering, University of California, Davis, USA
| | - Sun Il Kwon
- Department of Biomedical Engineering, University of California, Davis, USA
| |
Collapse
|
49
|
Zhang X, Xiang Z, Wang F, Han C, Zhang Q, Liu E, Yuan H, Jiang L. Feasibility of shortening scan duration of 18F-FDG myocardial metabolism imaging using a total-body PET/CT scanner. EJNMMI Phys 2024; 11:83. [PMID: 39390229 PMCID: PMC11467154 DOI: 10.1186/s40658-024-00689-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024] Open
Abstract
PURPOSE To evaluate 18F-FDG myocardial metabolism imaging (MMI) using a total-body PET/CT scanner and explore the feasible scan duration to guide the clinical practice. METHODS A retrospective analysis was conducted on 41 patients who underwent myocardial perfusion-metabolism imaging to assess myocardial viability. The patients underwent 18F-FDG MMI with a total-body PET/CT scanner using a list-mode for 600 s. PET data were trimmed and reconstructed to simulate images of 600-s, 300-s, 120-s, 60-s, and 30-s acquisition time (G600-G30). Images among different groups were subjectively evaluated using a 5-point Likert scale. Semi-quantitative evaluation was performed using standardized uptake value (SUV), myocardial to background activity ratio (M/B), signal to noise ratio (SNR), contrast to noise ratio (CNR), contrast ratio (CR), and coefficient of variation (CV). Myocardial viability analysis included indexes of Mismatch and Scar. G600 served as the reference. RESULTS Subjective visual evaluation indicated a decline in the scores of image quality with shortening scan duration. All the G600, G300, and G120 images were clinically acceptable (score ≥ 3), and their image quality scores were 4.9 ± 0.3, 4.8 ± 0.4, and 4.5 ± 0.8, respectively (P > 0.05). Moreover, as the scan duration reduced, the semi-quantitative parameters M/B, SNR, CNR, and CR decreased, while SUV and CV increased, and significant difference was observed in G300-G30 groups when comparing to G600 group (P < 0.05). For myocardial viability analysis of left ventricular and coronary segments, the Mismatch and Scar values of G300-G30 groups were almost identical to G600 group (ICC: 0.968-1.0, P < 0.001). CONCLUSION Sufficient image quality for clinical diagnosis could be achieved at G120 for MMI using a total-body PET/CT scanner, while the image quality of G30 was acceptable for myocardial viability analysis.
Collapse
Affiliation(s)
- Xiaochun Zhang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Zeyin Xiang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Fanghu Wang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Chunlei Han
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Qing Zhang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Entao Liu
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Hui Yuan
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China.
| | - Lei Jiang
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China.
| |
Collapse
|
50
|
Michail C, Liaparinos P, Kalyvas N, Kandarakis I, Fountos G, Valais I. Radiation Detectors and Sensors in Medical Imaging. SENSORS (BASEL, SWITZERLAND) 2024; 24:6251. [PMID: 39409289 PMCID: PMC11478476 DOI: 10.3390/s24196251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024]
Abstract
Medical imaging instrumentation design and construction is based on radiation sources and radiation detectors/sensors. This review focuses on the detectors and sensors of medical imaging systems. These systems are subdivided into various categories depending on their structure, the type of radiation they capture, how the radiation is measured, how the images are formed, and the medical goals they serve. Related to medical goals, detectors fall into two major areas: (i) anatomical imaging, which mainly concerns the techniques of diagnostic radiology, and (ii) functional-molecular imaging, which mainly concerns nuclear medicine. An important parameter in the evaluation of the detectors is the combination of the quality of the diagnostic result they offer and the burden of the patient with radiation dose. The latter has to be minimized; thus, the input signal (radiation photon flux) must be kept at low levels. For this reason, the detective quantum efficiency (DQE), expressing signal-to-noise ratio transfer through an imaging system, is of primary importance. In diagnostic radiology, image quality is better than in nuclear medicine; however, in most cases, the dose is higher. On the other hand, nuclear medicine focuses on the detection of functional findings and not on the accurate spatial determination of anatomical data. Detectors are integrated into projection or tomographic imaging systems and are based on the use of scintillators with optical sensors, photoconductors, or semiconductors. Analysis and modeling of such systems can be performed employing theoretical models developed in the framework of cascaded linear systems analysis (LCSA), as well as within the signal detection theory (SDT) and information theory.
Collapse
Affiliation(s)
| | | | | | - Ioannis Kandarakis
- Radiation Physics, Materials Technology and Biomedical Imaging Laboratory, Department of Biomedical Engineering, University of West Attica, Ag. Spyridonos, 12210 Athens, Greece; (C.M.); (P.L.); (N.K.); (G.F.); (I.V.)
| | | | | |
Collapse
|