1
|
Goldschmidt-Clermont PJ, Sevilla BA. Redox and actin, a fascinating story. Redox Biol 2025; 83:103630. [PMID: 40328105 DOI: 10.1016/j.redox.2025.103630] [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: 03/29/2025] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
Actin is an extraordinarily complex protein whose functions are essential to cell motility, division, contraction, signaling, transport, tissular structures, DNA repair, and many more cellular activities critical to life for both animals and plants. It is one of the most abundant and conserved proteins and it exists in either a soluble, globular (monomeric, G-actin) or an insoluble, self-assembled (polymerized or filamentous actin, F-actin) conformation as a key component of the cytoskeleton. In the early 1990's little, if anything, was known about the impact of reactive oxygen species (ROS) on the biology of actin except that ROS could disrupt the actin cytoskeleton. Instructively, G-actin is susceptible to alteration by ROS, and thus, purification of G-actin is typically performed in the presence of strong antioxidants (like dithiothreitol) to limit its oxidative degradation. In contrast, F-actin is a more stable conformation and thus actin can be kept relatively intact in purified preparations as filaments at low temperature for extended periods of time. Both G- and F-actin interact with a myriad of intracellular proteins and at least with a couple of extracellular proteins, and these interactions are essential to the many actin functions. This review will show how, over the past 30 years, our understanding of the role of ROS for actin biology has evolved from noxious denaturizing agents to remarkable regulators of the actin cytoskeleton in cells and consequent cellular functions.
Collapse
|
2
|
Huang M, Wang W, Wang R, Tian R. The prognostic value of pretreatment [ 18F]FDG PET/CT parameters in esophageal cancer: a meta-analysis. Eur Radiol 2025; 35:3396-3408. [PMID: 39570366 DOI: 10.1007/s00330-024-11207-3] [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: 06/20/2024] [Revised: 08/19/2024] [Accepted: 10/14/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVES This study aims to evaluate the prognostic implications of pretreatment [18F]FDG-PET metrics in esophageal cancer patients through a meta-analysis of the existing literature. METHODS We carefully searched electronic databases, including PubMed and Embase, from inception to April 1, 2024, to identify studies describing the prognostic value of pretreatment PET metrics for advanced esophageal cancer. Clinical endpoints examined were overall survival (OS), recurrence-free survival (RFS)/disease-free survival (DFS), and progression-free survival (PFS). Hazard ratios (HRs) for PFS and OS were taken directly from the original reports. RESULTS Forty-seven publications, including 5504 patients, were included in our analysis. OS and PFS were analyzed in 31 and nine studies, respectively, and DFS/RFS was analyzed in 16 studies. The comprehensive pooled analysis revealed significant associations between metabolic parameters derived from positron emission tomography (PET) imaging and clinical outcomes. Expressly, the pooled HR indicated that patients with higher SUVmax were significantly associated with poor PFS (HR: 1.06; 95% CI: 1.01-1.12, p = 0.011) and poor RFS/DFS (HR: 1.09; 95% CI: 1.02-1.18, p = 0.019). Patients with higher SUVmean were significantly associated with poorer OS (HR: 1.07; 95% CI: 1.01-1.14, p = 0.025). High MTV was significantly associated with inferior OS (HR: 1.02; 95% CI: 1.00-1.05, p = 0.049). High TLG was significantly associated with poorer RFS/DFS (HR: 2.02; 95% CI: 1.11-3.68, p = 0.022). CONCLUSION This study unveiled pretreatment FDG-derived parameters as valuable prognostic indicators in assessing esophageal cancer outcomes. Specifically, SUVmax is associated with PFS and RFS/DFS. SUVmean and MTV were correlated with OS, and TLG was only associated with RFS/DFS. KEY POINTS Question Inconsistent findings on the prognostic value of pretreatment [18F]FDG PET parameters in esophageal cancer require comprehensive analysis to clarify their role in outcome prediction. Findings Higher pretreatment [18F]FDG-PET metrics (SUVmax, SUVmean, MTV, TLG) are associated with poor survival outcomes, emphasizing their potential value in enhancing prognostic assessments for esophageal cancer. Clinical relevance This study highlights the prognostic significance of pretreatment [18F]FDG-PET metrics in esophageal cancer, providing valuable insights for patient outcome prediction and potentially guiding personalized treatment strategies.
Collapse
Affiliation(s)
- Mingxing Huang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Weichen Wang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rang Wang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rong Tian
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
3
|
Li H, Li X, Yang L, Chen L, Fu C, Liang J, Zhen Z, XuanYuan Y, Gao Y, Xu J. Sequential synthesis of [ 18F]FDG and [ 68Ga]Ga-DOTA-TATE on the AllinOne radiosynthesizer: A fully automated dual-radionuclides protocol with single cassette and reagents loading. Appl Radiat Isot 2025; 223:111899. [PMID: 40349649 DOI: 10.1016/j.apradiso.2025.111899] [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: 11/13/2024] [Revised: 05/04/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND The development of a unified and efficient protocol for the automated synthesis of PET imaging agents is crucial for enhancing operational efficiency and safety. This study aimed to streamline the sequential production process of 18F-fluorodeoxyglucose ([18F]FDG) and [68Ga]Ga-DOTA-TATE with only single cassette and reagents loading using the AllinOne 36-valve synthesis module, with the goal of improving hot cell utilization and minimizing radiation exposure for operators. The protocol was designed to allow for sequential synthesis without the need for repeated hot cell access or waiting for radiation levels to decrease, thereby reducing the time and resources required for PET imaging agent production. RESULTS Our study demonstrated the stability and reliability of the newly designed synthesis protocol. The activity yields for two batches of [18F]FDG were 73 ± 6.2 % and 64 ± 4.7 % (n = 3), respectively, all with synthesis times about 23 min, and with a radiochemical purity consistently over 96 %. For [68Ga]Ga-DOTA-TATE, the yield was 71 ± 5.8 % with synthesis times about 18 min (n = 3), with a purity exceeding 97 %. The synthesized products met all quality control criteria, including appearance, pH value, radioactivity concentration, sterility, endotoxin levels, and solvent residue. CONCLUSION The single-cassette protocol significantly improved efficiency and reduced radiation exposure. High yields and purities confirm its clinical feasibility, ensuring PET imaging agent availability. This scalable solution enhances patient care by simplifying the production process and meeting the demands of various patients for different PET imaging agents within a single day, demonstrating its potential in clinical settings.
Collapse
Affiliation(s)
- Huiqiang Li
- Department of Nuclear Medicine, Henan Key Laboratory of Novel Molecular Probes and Clinical Translation in Nuclear Medicine, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Xiaochen Li
- Department of Nuclear Medicine, Henan Key Laboratory of Novel Molecular Probes and Clinical Translation in Nuclear Medicine, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Lingyue Yang
- Department of Nuclear Medicine, Henan Key Laboratory of Novel Molecular Probes and Clinical Translation in Nuclear Medicine, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Lijuan Chen
- Department of Nuclear Medicine, Henan Key Laboratory of Novel Molecular Probes and Clinical Translation in Nuclear Medicine, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Chang Fu
- Department of Nuclear Medicine, Henan Key Laboratory of Novel Molecular Probes and Clinical Translation in Nuclear Medicine, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Jianfei Liang
- Beijing PET Technology CO., LTD., Beijing, 100093, China
| | - Zhifei Zhen
- Department of Nuclear Medicine, Henan Key Laboratory of Novel Molecular Probes and Clinical Translation in Nuclear Medicine, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Yirui XuanYuan
- West China School of Medicine, Sichuan University, Chengdu, 610041, China
| | - Yongju Gao
- Department of Nuclear Medicine, Henan Key Laboratory of Novel Molecular Probes and Clinical Translation in Nuclear Medicine, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| | - Junling Xu
- Department of Nuclear Medicine, Henan Key Laboratory of Novel Molecular Probes and Clinical Translation in Nuclear Medicine, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| |
Collapse
|
4
|
Chen Z, Yang H, Qi M, Chen W, Liu F, Song S, Zhang J. Enhancing 18F-FDG PET image quality and lesion diagnostic performance across different body mass index using the deep progressive learning reconstruction algorithm. Cancer Imaging 2025; 25:58. [PMID: 40312739 PMCID: PMC12044768 DOI: 10.1186/s40644-025-00877-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: 12/09/2024] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND As body mass index (BMI) increases, the quality of 2-deoxy-2-[fluorine-18]fluoro-D-glucose (18F-FDG) positron emission tomography (PET) images reconstructed with ordered subset expectation maximization (OSEM) declines, negatively impacting lesion diagnostics. It is crucial to identify methods that ensure consistent diagnostic accuracy and maintain image quality. Deep progressive learning (DPL) algorithm, an Artificial Intelligence(AI)-based PET reconstruction technique, offers a promising solution. METHODS 150 patients underwent 18F-FDG PET/CT scans and were categorized by BMI into underweight, normal, and overweight groups. PET images were reconstructed using both OSEM and DPL and their image quality was assessed both visually and quantitatively. Visual assessment employed a 5-point Likert scale to evaluate overall score, image sharpness, image noise, and diagnostic confidence. Quantitative assessment parameters included the background liver image-uniformity-index ([Formula: see text]) and signal-to-noise ratio ([Formula: see text]). Additionally, 466 identifiable lesions were categorized by size: sub-centimeter and larger. We compared maximum standard uptake value ([Formula: see text]), signal-to-background ratio ([Formula: see text]), [Formula: see text], contrast-to-background ratio ([Formula: see text]), and contrast-to-noise ratio ([Formula: see text]) of these lesions to evaluate the diagnostic performance of the DPL and OSEM algorithms across different lesion sizes and BMI categories. RESULTS DPL produced superior PET image quality compared to OSEM across all BMI groups. The visual quality of DPL showed a slight decline with increasing BMI, while OSEM exhibited a more significant decline. DPL maintained a stable [Formula: see text] across BMI increases, whereas OSEM exhibited increased noise. In the DPL group, quantitative image quality for overweight patients matched that of normal patients with minimal variance from underweight patients. In contrast, OSEM demonstrated significant declines in quantitative image quality with rising BMI. DPL yielded significantly higher contrast ([Formula: see text], [Formula: see text],[Formula: see text]) and [Formula: see text] than OSEM for all lesions across all BMI categories. CONCLUSION DPL consistently provided superior image quality and lesion diagnostic performance compared to OSEM across all BMI categories in 18F-FDG PET/CT scans. Therefore, we recommend using the DPL algorithm for 18F-FDG PET/CT image reconstruction in all BMI patients.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai, 200032, China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, Shanghai, 200433, China
| | - Hongxing Yang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai, 200032, China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, Shanghai, 200433, China
| | - Ming Qi
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai, 200032, China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, Shanghai, 200433, China
| | - Wen Chen
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai, 200032, China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, Shanghai, 200433, China
| | - Fei Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai, 200032, China
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, Shanghai, 200433, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai, 200032, China.
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, Shanghai, 200433, China.
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai, 200032, China.
- Key Laboratory of Nuclear Physics and Ion-beam Application (MOE), Fudan University, Shanghai, 200433, China.
- Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
5
|
Juweid ME, Al‐Qasem SF, Khuri FR, Gallamini A, Lohmann P, Ziellenbach H, Mottaghy FM. Beyond fluorodeoxyglucose: Molecular imaging of cancer in precision medicine. CA Cancer J Clin 2025; 75:226-242. [PMID: 40183513 PMCID: PMC12061632 DOI: 10.3322/caac.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Cancer molecular imaging is the noninvasive visualization of a process unique to or altered in neoplasia, such as proliferation, glucose metabolism, and receptor expression, which is relevant to patient management. Several molecular imaging modalities are now available, including magnetic resonance, optical, and nuclear imaging. Nuclear imaging, particularly using fluorine-18-fluorodeoxyglucose positron emission tomography, is widely used in the staging and response assessment of multiple cancer types. However, at this writing, new nuclear medicine probes, especially positron emission tomography tracers, are increasingly used or are being investigated for cancer evaluation. This review focuses on these probes, their biologic targets, and the applications or potential applications for their use in the assessment of various neoplasms, including both probes available for commercial use-such as somatostatin receptor ligands in neuroendocrine tumors, prostate-specific membrane antigen ligands in prostate cancer, norepinephrine analogs in neural crest tumors like neuroblastoma, and estrogen analogs in breast cancer-and others in clinical development, such as fibroblast-activating protein inhibitors, C-X-C chemokine receptor type 4 ligands, and monoclonal antibodies targeting receptor tyrosine kinases, CD4-positive or CD8-positive tumor-infiltrating lymphocytes, tumor-associated macrophages, and cancer stem cell biomarkers. These developments represent a major step toward the integration of molecular imaging as a powerful tool in precision medicine, with an expectedly significant impact on patient management and outcome.
Collapse
Affiliation(s)
- Malik E. Juweid
- Department of Radiology and Nuclear MedicineSchool of MedicineUniversity of JordanAmmanJordan
- The National Center for Diabetes, Endocrinology, and GeneticsUniversity of JordanAmmanJordan
| | - Soud F. Al‐Qasem
- Department of Radiology and Nuclear MedicineSchool of MedicineUniversity of JordanAmmanJordan
| | - Fadlo R. Khuri
- Division of OncologyDepartment of Internal MedicineAmerican University of BeirutBeirutLebanon
| | - Andrea Gallamini
- Research and Innovation DepartmentAntoine Lacassagne Cancer CenterNiceFrance
| | - Philipp Lohmann
- Department of Nuclear MedicineUniversity Hospital AachenRWTH Aachen UniversityAachenGermany
- Medical Imaging Physics (INM‐4)Institute of Neuroscience and Medicine, Research Center JuelichJuelichGermany
| | | | - Felix M. Mottaghy
- Department of Nuclear MedicineUniversity Hospital AachenRWTH Aachen UniversityAachenGermany
- Department of Radiology and Nuclear MedicineMaastricht University Medical CenterMaastrichtthe Netherlands
- Center of Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)CologneGermany
| |
Collapse
|
6
|
Schmidt M, Binder H, Schneider MR. The metabolic underpinnings of sebaceous lipogenesis. Commun Biol 2025; 8:670. [PMID: 40289206 PMCID: PMC12034822 DOI: 10.1038/s42003-025-08105-9] [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: 02/24/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Sebaceous glands synthesize and secrete sebum, a mélange of lipids and other cellular products that safeguards the mammalian integument. Differentiating sebocytes delaminate from the basal membrane and dislodge towards the gland's middle, where they eventually undergo a poorly understood death mode in which the whole cell becomes a secretion product (holocrine secretion). Supported by recent transcriptomics data, this review examines the idea that peripheral sebocytes have a remarkable ability to draw nutrients from the blood and become committed to unrestrainedly invest all available resources into synthetic processes for accomplishing sebum synthesis, thereby exploiting core metabolic fluxes as glycogen turnover, glutamine-directed anaplerosis, the pentose phosphate pathway and de novo lipogenesis. Finally, we propose that metabolic-driven processes are an important mechanistic component of holocrine secretion. A deeper understanding of these metabolic adaptations could indicate novel strategies for modulating sebum synthesis, a key pathogenic factor in acne and other skin diseases.
Collapse
Affiliation(s)
- Maria Schmidt
- Interdisciplinary Institute for Bioinformatics (IZBI), University of Leipzig, Leipzig, Germany
| | - Hans Binder
- Interdisciplinary Institute for Bioinformatics (IZBI), University of Leipzig, Leipzig, Germany
- Armenian Bioinformatics Institute (ABI), Yerevan, Armenia
| | - Marlon R Schneider
- Institute of Veterinary Physiology, Veterinary Faculty, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
7
|
Urakawa N, Kanaji S, Sawada R, Koterazawa Y, Ikeda T, Harada H, Goto H, Hasegawa H, Yamashita K, Matsuda T, Kakeji Y. Efficacy of 18F-Fluoro-2-Deoxyglucose Positron Emission Tomography as a Predictor of Treatment Response to Neoadjuvant S-1 + Oxaliplatin Chemotherapy for Gastric Cancer. Cancer Rep (Hoboken) 2025; 8:e70190. [PMID: 40275469 PMCID: PMC12021666 DOI: 10.1002/cnr2.70190] [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: 06/17/2024] [Revised: 02/27/2025] [Accepted: 03/12/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy is widely recognized as the established treatment for advanced gastric cancer. However, predicting its efficacy before surgery remains challenging. AIM The present study aimed to evaluate the effectiveness of 18F-fluoro-2-deoxyglucose positron emission tomography (FDG-PET) as a predictor of treatment response to the S-1+Oxaliplatin regimen (SOX). METHODS AND RESULTS Thirty patients who underwent gastrectomy following neoadjuvant SOX between January 2021 and July 2023 were included. Patients underwent FDG-PET pre- and postsurgery. The maximum standardized uptake value (SUVmax) from FDG-PET was examined in relation to histological tumor response and prognosis. SUVmax decreased significantly after chemotherapy in all patients (p < 0.001), especially in those with Grade 1a, 2, and 3 tumors (p < 0.05). SUV reduction increased stepwise with the histological response grade. Optimal cut-off values for the percentage decrease in SUVmax (ΔSUVmax) predictive of histologic efficacy were identified as 53% (area under curve 0.855, p = 0.0018) for Grade 1b or higher and 75% (area under curve 0.806, p = 0.0044) for Grade 2 or higher. Patients with ΔSUVmax > 50% had improved recurrence-free survival (p = 0.027). CONCLUSION FDG-PET may be useful as a predictor of treatment response in neoadjuvant SOX therapy for gastric cancer. The determination of the optimal ΔSUVmax value may enhance the precision of histological tumor response prediction.
Collapse
Affiliation(s)
- Naoki Urakawa
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Shingo Kanaji
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Ryuichiro Sawada
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Yasufumi Koterazawa
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Taro Ikeda
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Hitoshi Harada
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Hironobu Goto
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Hiroshi Hasegawa
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Kimihiro Yamashita
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Takeru Matsuda
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Yoshihiro Kakeji
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| |
Collapse
|
8
|
Jaber N, Saadani H, Schats W, Aalbersberg EA, Stokkel MPM. Novel Clinical PET Tracers in the Pipeline for Melanoma. Curr Oncol Rep 2025; 27:458-471. [PMID: 40072700 DOI: 10.1007/s11912-025-01659-1] [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] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
PURPOSE The aim of this review is to provide an overview of novel clinical PET tracers in the pipeline for melanoma. Secondarily, to provide a head-to-head comparison with the current clinical standard used in clinical practice, [18F]FDG, if available. RECENT FINDINGS [18F]FDG PET/CT has become important in the clinical setting for melanoma as it serves many purposes, but lacks other important qualities due its nonspecific nature. There is an increased clinical need for specific tracers. Many new PET tracers, such as melanin-targeted and antibody-based probes, have been studied in melanoma with the intention of achieving high sensitivity detection of metastases and small lesions. There are four main groups of PET tracers in de pipeline for melanoma: melanin-, FAP-, PD-1/PD-L1- and CD8+ T cell-tracers. Melanin-targeted tracers and FAP inhibitors revealed potential for diagnostic application, whilst PD-1/PD-L1 and CD8+ T cell tracers demonstrated potential for response assessment and prediction. In conclusion, research has revealed promising results from current (ongoing) studies; however, more melanoma patients need to be included to further assess the value of these tracers.
Collapse
Affiliation(s)
- Nora Jaber
- Department of Nuclear Medicine, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Hanna Saadani
- Department of Nuclear Medicine, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands.
| | - Winnie Schats
- Department of Scientific Information Service, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Else A Aalbersberg
- Department of Nuclear Medicine, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Marcel P M Stokkel
- Department of Nuclear Medicine, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| |
Collapse
|
9
|
Ailawadhi S, Pafundi D, Peterson J. Advances and future directions in radiopharmaceutical delivery for cancer treatment. Expert Rev Anticancer Ther 2025; 25:351-361. [PMID: 40019019 DOI: 10.1080/14737140.2025.2472859] [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: 09/09/2024] [Revised: 01/29/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
INTRODUCTION Targeted radiopharmaceutical therapies (RPTs) have emerged as a promising approach for the precise treatment of various cancers. Delivering ionizing radiation directly to cancer cells while sparing surrounding healthy tissue, radiopharmaceuticals offer enhanced efficacy and reduced toxicity compared to conventional external beam radiation therapy (i.e. photons and electrons). AREAS COVERED In the current era of personalized cancer care, the appropriate choice of RPTs for a clinical condition and the specific patient's care needs to be better understood. Several available RPT agents with their respective clinical applicability along with rapidly ongoing research in this field have now given RPTs the ability to lend themselves to a personalized medicine focus. This review provides an overview of recent advancements in RPT, including nuclide selection and development, molecular targeting strategies, radiopharmaceutical development, and clinical applications. EXPERT OPINION We discuss the underlying principles, challenges, and opportunities for future development. Furthermore, we explore emerging technologies and future directions in the field, highlighting the potential impact on personalized cancer care.
Collapse
Affiliation(s)
| | - Deanna Pafundi
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Jennifer Peterson
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
10
|
Huang JW, Zeng H, Zhang Q, Liu XY, Feng C. Advances in the clinical diagnosis of lung cancer using contrast-enhanced ultrasound. Front Med (Lausanne) 2025; 12:1543033. [PMID: 40177283 PMCID: PMC11961447 DOI: 10.3389/fmed.2025.1543033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Lung cancer (LC) remains one of the leading causes of cancer-related mortality worldwide, emphasizing the urgent need for innovative diagnostic tools to improve early detection and patient outcomes. Contrast-enhanced ultrasound (CEUS) has emerged as a promising complement to conventional imaging modalities, offering distinct advantages such as real-time dynamic imaging, cost-effectiveness, and the absence of ionizing radiation. By enhancing the visualization of tumor vascularization, CEUS enables differentiation between benign and malignant pulmonary nodules while providing valuable insights into tumor angiogenesis, a hallmark of malignancy, and therapeutic response. Additionally, CEUS demonstrates utility in assessing regional lymph nodes, detecting distant metastases, and analyzing blood flow dynamics through quantitative methods such as time-intensity curve analysis. Despite these benefits, certain limitations persist, including reduced efficacy in imaging deep-seated lesions, variability due to patient-specific physiological factors, and dependency on operator expertise. However, advancements in targeted contrast agents, integration with multimodal imaging techniques, and the application of artificial intelligence hold significant potential to address these challenges. This review systematically evaluates the clinical applications, advantages, and limitations of CEUS in LC diagnosis, providing a comprehensive understanding of its role in modern precision oncology. Furthermore, it highlights future research directions aimed at enhancing diagnostic accuracy, improving clinical workflows, and expanding the adoption of CEUS in routine practice.
Collapse
Affiliation(s)
- Jian-wei Huang
- Department of Chest Surgery, The Affiliated Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Hai Zeng
- Department of General Surgery, The Affiliated Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Quan Zhang
- Department of Chest Surgery, The Affiliated Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Xiao-yu Liu
- Department of Chest Surgery, The Affiliated Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Chong Feng
- Department of Ultrasound, The Affiliated Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| |
Collapse
|
11
|
Mitsea A, Christoloukas N, Koutsipetsidou S, Papavasileiou P, Oikonomou G, Angelopoulos C. Positron Emission Tomography-Magnetic Resonance Imaging, a New Hybrid Imaging Modality for Dentomaxillofacial Malignancies-A Systematic Review. Diagnostics (Basel) 2025; 15:654. [PMID: 40149996 PMCID: PMC11941154 DOI: 10.3390/diagnostics15060654] [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: 01/05/2025] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Emerging hybrid imaging modalities, like Positron Emission Tomography/Computed Tomography (PET/CT) and Positron Emission Tomography/Magnetic Resonance Imaging (PET/MRI), are useful for assessing head and neck cancer (HNC) and its prognosis during follow-up. PET/MRI systems enable simultaneous PET and MRI scans within a single session. These combined PET/MRI scanners merge MRI's better soft tissue contrast and the molecular metabolic information offered by PET. Aim: To review scientific articles on the use of hybrid PET/MRI techniques in diagnosing dentomaxillofacial malignancies. Method: The available literature on the use of PET/MRI for the diagnosis of dentomaxillofacial malignancies in four online databases (Scopus, PubMed, Web of Science, and the Cochrane Library) was searched. Eligible for this review were original full-text articles on PET/MRI imaging, published between January 2010 and November 2024, based on experimental or clinical research involving humans. Results: Out of the 783 articles retrieved, only twelve articles were included in this systematic review. Nearly half of the articles (5 out of 12) concluded that PET/MRI is superior to PET, MRI, and PET/CT imaging in relation to defining malignancies' size. Six articles found no statistically significant results and the diagnostic accuracy presented was similar in PET/MRI versus MRI and PET/CT images. Regarding the overall risk of bias, most articles had a moderate risk. Conclusions: The use of PET/MRI in HNC cases provides a more accurate diagnosis regarding dimensions of the tumor and thus a more accurate surgical approach if needed. Further prospective studies on a larger cohort of patients are required to obtain more accurate results on the application of hybrid PET/MRI.
Collapse
Affiliation(s)
- Anastasia Mitsea
- Department of Oral Diagnosis & Radiology, School of Dentistry, National and Kapodistrian University of Athens, 2 Thivon Str., 11527 Athens, Greece
| | - Nikolaos Christoloukas
- Department of Oral Diagnosis & Radiology, School of Dentistry, National and Kapodistrian University of Athens, 2 Thivon Str., 11527 Athens, Greece
| | - Spyridoula Koutsipetsidou
- Biomedical Sciences, Division of Radiology and Radiotherapy, University of West Attica, 28 Agiou Spiridonos Str., 12243 Athens, Greece
| | - Periklis Papavasileiou
- Biomedical Sciences, Division of Radiology and Radiotherapy, University of West Attica, 28 Agiou Spiridonos Str., 12243 Athens, Greece
| | - Georgia Oikonomou
- Biomedical Sciences, Division of Radiology and Radiotherapy, University of West Attica, 28 Agiou Spiridonos Str., 12243 Athens, Greece
| | - Christos Angelopoulos
- Department of Oral Diagnosis & Radiology, School of Dentistry, National and Kapodistrian University of Athens, 2 Thivon Str., 11527 Athens, Greece
| |
Collapse
|
12
|
Guo R, Wang J, Miao Y, Zhang X, Xue S, Zhang Y, Shi K, Li B, Zheng G. 3D full-dose brain-PET volume recovery from low-dose data through deep learning: quantitative assessment and clinical evaluation. Eur Radiol 2025; 35:1133-1145. [PMID: 39609283 DOI: 10.1007/s00330-024-11225-1] [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: 06/13/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVES Low-dose (LD) PET imaging would lead to reduced image quality and diagnostic efficacy. We propose a deep learning (DL) method to reduce radiotracer dosage for PET studies while maintaining diagnostic quality. METHODS This retrospective study was performed on 456 participants respectively scanned by three different PET scanners with two different tracers. A DL method called spatially aware noise reduction network (SANR) was proposed to recover 3D full-dose (FD) PET volumes from LD data. The performance of SANR was compared with a 2D DL method taking regular FD PET volumes as the reference. Wilcoxon signed-rank test was conducted to compare the image quality metrics across different DL denoising methods. For clinical evaluation, two nuclear medicine physicians examined the recovered FD PET volumes using a 5-point grading scheme (5 = excellent) and gave a binary decision (negative or positive) for diagnostic quality assessment. RESULTS Statistically significant differences (p < 0.05) were found in terms of image quality metrics when SANR was compared with the 2D DL method. For clinical evaluation, SANR achieved a lesion detection accuracy of 95.3% (95% CI: 90.1%, 100%), while the reference full-dose PET volumes obtained a lesion detection accuracy of 98.4% (95% CI: 95.4%, 100%). In Alzheimer's disease diagnosis, both the reference FD PET volumes and the FD PET volumes recovered by SANR exhibited the same accuracy. CONCLUSION Compared with reference FD PET, LD PET denoised by the proposed approach significantly reduced radiotracer dosage and showed noninferior diagnostic performance in brain lesion detection and Alzheimer's disease diagnosis. KEY POINTS Question The current trend in PET imaging is to reduce injected dosage, which leads to low-quality PET images and reduces diagnostic efficacy. Findings The proposed deep learning method could recover diagnostic quality PET images from data acquired with a markedly reduced radiotracer dosage. Clinical relevance The proposed method would enhance the utility of PET scanning at lower radiotracer dosage and inform future workflows for brain lesion detection and Alzheimer's disease diagnosis, especially for those patients who need multiple examinations.
Collapse
Affiliation(s)
- Rui Guo
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiale Wang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Miao
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinyu Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Song Xue
- Department of Nuclear Medicine, University of Bern, Bern, Switzerland
| | - Yu Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Kuangyu Shi
- Department of Nuclear Medicine, University of Bern, Bern, Switzerland
- Department of Informatics, Technical University of Munich, Munich, Germany
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Guoyan Zheng
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
13
|
Fatima N, Laiq M, Rafay M, Azam SM, Zaman MU. Role of 18 FDG PET/CT in Detecting Primary Tumors in Patients with Carcinoma of Unknown Primary: Single-Center Cross-Sectional Study from 2017 to 2023 (Extension Study). World J Nucl Med 2025; 24:57-63. [PMID: 39959146 PMCID: PMC11828639 DOI: 10.1055/s-0044-1795101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025] Open
Abstract
Background Carcinoma of unknown primary (CUP) is a diverse group of cancers in which the primary tumor site remains occult despite detailed investigations. This is an extension of a published parent study with a smaller cohort, to further validate the published facts of detection efficiency of 18F-fluorodeoxyglucose positron emission tomography/computed tomography ( 18 FDG PET/CT) in patients with CUP over a larger sample from 2017 to 2023. Methods Patients with CUP referred for 18 FDG PET/CT scan for detection of primary sites during the study period were recruited. 18 FDG PET/CT scan was acquired using a standardized protocol, and patients with suspected primary sites underwent biopsies. Scan findings and biopsy results were analyzed to find the detection rate, sensitivity, area under the curve (AUC), and positive predictive value (PPV). As no biopsy was performed in cases with negative scan, these cases were considered false negatives (FNs). Results Total 230 patients with CUP were included with similar demographic trend (mean age: 58 ± 14 years; 63% male and 37% female; mean body mass index: 26.82 ± 5.4 kg/m 2 ); 138/230 (60 vs. 74% in parent study) patients were found to have a hypermetabolic focus suggestive of primary tumor sites and subjected to biopsy which turned out positive in 127/138 (true positive [TP]: 92 vs. 76% in parent study) and negative in 11/138 (true negative [TN]: 8 vs. 24% in parent study). Sensitivity and PPV of 18 FDG PET/CT were 58 and 92%, respectively (68 and 76%, respectively, in parent study). The remaining 92/230 (40%) patients with negative 18 FDG PET/CT for primary focus did not have biopsy. No significant demographic difference was seen in patients with TP and FN studies ( p > 0.05). Receiver operating characteristics (ROC) curve revealed fair diagnostic strength of 18 FDG PET/CT for detecting unknown primary (AUC 0.710; p ≤ 0.05; standard error = 0.0167; confidence interval: 0.647-0.768; vs. nonsignificant in parent study). Conclusion We conclude that this extension study with a larger cohort compared with the parent study has found a similar detection efficiency of 18 FDG PET/CT for identifying primary tumor in patients with CUP (58 vs. 57%) but with better PPV and sensitivity. Upfront use of 18 FDG PET/CT in CUP could preclude the use of many futile diagnostic procedures. Furthermore, the use of tumor-specific PET tracers, higher resolution scanners, and acquiring delayed images in patients with negative 18 FDG study could reduce FN results in patients with CUP.
Collapse
Affiliation(s)
- Nosheen Fatima
- Section of PET/CT Imaging Services, Department of Radiology, Aga Khan University Hospital, Karachi, Pakistan
| | - Mina Laiq
- Section of PET/CT Imaging Services, Department of Radiology, Aga Khan University Hospital, Karachi, Pakistan
| | - Muhammad Rafay
- Section of PET/CT Imaging Services, Department of Radiology, Aga Khan University Hospital, Karachi, Pakistan
| | - Sara Muhammad Azam
- Section of PET/CT Imaging Services, Department of Radiology, Aga Khan University Hospital, Karachi, Pakistan
| | - Maseeh uz Zaman
- Section of PET/CT Imaging Services, Department of Radiology, Aga Khan University Hospital, Karachi, Pakistan
| |
Collapse
|
14
|
Viohl N, Steinert M, Freesmeyer M, Kühnel C, Drescher R. 18F-FDG PET/CT in the Preoperative Diagnostic and Staging of Lung Cancer and as a Predictor of Lymph Node Involvement. J Clin Med 2025; 14:1324. [PMID: 40004854 PMCID: PMC11856622 DOI: 10.3390/jcm14041324] [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: 01/14/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The aim of this study was to evaluate the efficacy and accuracy of PET imaging and performance in defining the preoperative TNM classification, especially the intrathoracic lymph node staging, of patients with lung cancer. Methods: A retrospective, single-institution study of consecutive patients with surgical therapy of lung cancer that were undergoing preoperative PET/CT scanning at the same center was conducted. A total of 104 patients were included. All patients underwent surgical evaluation with mediastinal and hilar lymph node sampling. Five patients with preoperative suspicion of N3 nodal status who were only tested for N2 were excluded from the observations and analyses of nodal status. Results: PET/CT staged the nodal status correctly in 85 out of 99 patients (85.9%); overstaging occurred in 7 patients (7.1%) and understaging in 7 patients (7.1%). The overall prevalence of lymph node metastases was 42.3%. When preoperative T classification was compared with postoperative histopathological T classification, 75% patients were correctly staged, 13.5% were overstaged, and 11.5% were understaged by PET/CT. In univariate analysis, lymph node involvement was significantly associated (p < 0.05) with the following primary tumor characteristics: increasing diameter (>35 mm), a maximum standardized uptake value > 9.5, and higher grading. The tumor diameter and the degree of differentiation were found to be factors influencing the SUVmax of the primary tumor as well. Conclusions: Our data show that integrated PET/CT provides high accuracy in the intrathoracic nodal staging and tumor expansion of lung cancer patients and emphasizes the continued need for surgical staging.
Collapse
Affiliation(s)
- Nathalie Viohl
- Clinic of Nuclear Medicine, Jena University Hospital, 07743 Jena, Germany; (N.V.); (C.K.); (R.D.)
| | - Matthias Steinert
- Clinic of Thoracic Surgery, Leipzig University Hospital, 04103 Leipzig, Germany;
| | - Martin Freesmeyer
- Clinic of Nuclear Medicine, Jena University Hospital, 07743 Jena, Germany; (N.V.); (C.K.); (R.D.)
| | - Christian Kühnel
- Clinic of Nuclear Medicine, Jena University Hospital, 07743 Jena, Germany; (N.V.); (C.K.); (R.D.)
| | - Robert Drescher
- Clinic of Nuclear Medicine, Jena University Hospital, 07743 Jena, Germany; (N.V.); (C.K.); (R.D.)
| |
Collapse
|
15
|
Hindso TG, Martinussen T, Bjerrum CW, Keller SH, Loft A, Sjøl MB, Nissen K, Faber C, Donia M, Svane IM, Ellebaek E, Heegaard S, Kiilgaard JF, Madsen K. 18F-FDG PET/CT assessment of metabolic tumor burden predicts survival in patients with metastatic posterior uveal melanoma. Sci Rep 2025; 15:4110. [PMID: 39901052 PMCID: PMC11790917 DOI: 10.1038/s41598-025-88625-w] [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: 11/27/2024] [Accepted: 01/29/2025] [Indexed: 02/05/2025] Open
Abstract
The prognostic value of metabolic tumor burden parameters obtained from 18F-FDG PET/CT imaging was evaluated in this retrospective national multicenter study of patients with metastatic posterior uveal melanoma (PUM) and compared to the largest diameter of the largest metastatic lesion (LDLM) and the American Joint Committee on Cancer (AJCC) staging system. The Maximal Standard Uptake Value (SUVmax), Metabolic Tumor Volume (MTV), and Total Lesion Glycolysis (TLG) were obtained in 106 patients. Higher values of SUVmax (p = 0.007, log-rank), MTV (p < 0.001, log-rank), and TLG (p < 0.001, long-rank) were associated with shorter survival. The three parameters were also independent predictors in the multivariate Cox model, while the AJCC staging turned insignificant. Time-dependent positive predictive value (PPV) analysis and Receiver Operating Characteristics (ROC) curves showed that MTV (Area Under the Curve (AUC) = 0.78), TLG (AUC = 0.78), and LDLM (AUC = 0.76) were good predictors of 1-year survival. For the subset of 97 patients with liver metastases, the corresponding regional measurements in the liver tended to be even better predictors. In conclusion, MTV and TLG were found to be better predictors of survival in metastatic PUM than the AJCC staging system, but when LDLM was used as a continuous variable it showed an equally good prediction of 1-year survival.
Collapse
Affiliation(s)
- Tine Gadegaard Hindso
- Department of Ophthalmology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, Copenhagen Ø, 2100, Denmark.
| | - Torben Martinussen
- Department of Biostatistics, University of Copenhagen, Øster Farimagsgade 5, Copenhagen K, 1014, Denmark
| | - Camilla Wium Bjerrum
- Department of Radiology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, Copenhagen Ø, 2100, Denmark
| | - Sune Høgild Keller
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, Copenhagen Ø, 2100, Denmark
| | - Annika Loft
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, Copenhagen Ø, 2100, Denmark
| | - Mette Bagger Sjøl
- Department of Ophthalmology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, Copenhagen Ø, 2100, Denmark
| | - Kristoffer Nissen
- Department of Ophthalmology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, Copenhagen Ø, 2100, Denmark
| | - Carsten Faber
- Department of Ophthalmology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, Copenhagen Ø, 2100, Denmark
| | - Marco Donia
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital - Herlev and Gentofte, Borgmester Ib Juuls Vej 13, Herlev, 2730, Denmark
| | - Inge Marie Svane
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital - Herlev and Gentofte, Borgmester Ib Juuls Vej 13, Herlev, 2730, Denmark
| | - Eva Ellebaek
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital - Herlev and Gentofte, Borgmester Ib Juuls Vej 13, Herlev, 2730, Denmark
| | - Steffen Heegaard
- Department of Ophthalmology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, Copenhagen Ø, 2100, Denmark
- Department of Pathology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, Copenhagen Ø, 2100, Denmark
| | - Jens Folke Kiilgaard
- Department of Ophthalmology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, Copenhagen Ø, 2100, Denmark
| | - Karine Madsen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, Copenhagen Ø, 2100, Denmark
| |
Collapse
|
16
|
Ellingson BM, Okobi Q, Chong R, Plawat R, Zhao E, Gafita A, Sonni I, Chun S, Filka E, Yao J, Telesca D, Li S, Li G, Lai A, Nghiemphu P, Czernin J, Nathanson DA, Cloughesy TF. A comparative study of preclinical and clinical molecular imaging response to EGFR inhibition using osimertinib in glioblastoma. Neurooncol Adv 2025; 7:vdaf022. [PMID: 40051661 PMCID: PMC11883343 DOI: 10.1093/noajnl/vdaf022] [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] [Indexed: 03/09/2025] Open
Abstract
Background To demonstrate the potential value of 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) as a rapid, non-invasive metabolic imaging surrogate for pharmacological modulation of EGFR signaling in EGFR-driven GBM, we synchronously conducted a preclinical imaging study using patient-derived orthotopic xenograft (PDOX) models and validated it in a phase II molecular imaging study in recurrent GBM (rGBM) patients using osimertinib. Methods A GBM PDOX mouse model study was performed concurrently with an open-label, single-arm, single-center, phase II study of osimertinib (NCT03732352) that enrolled 12 patients with rGBM with EGFR alterations. Patients received osimertinib daily and 3 18F-FDG PET scans: two 24 h apart prior to dosing, and one 48 h after dosing. Results GBM PDOX models suggest osimertinib has limited impact on both 18F-FDG uptake (+ 9.8%-+25.9%) and survival (+ 15.5%; P = .01), which may be explained by insufficient exposure in the brain (Kpuu: 0.30) required to robustly inhibit the EGFR alterations found in GBM. Treatment with osimertinib had subtle, but measurable decreases in the linear rate of change of 18F-FDG nSUV growth rate averaging -4.5% per day (P = .01) and change in 18F-FDG uptake was correlated with change in tumor growth rate (R2 = 0.4719, P = .0195). No metabolic (PERCIST) or radiographic (RANO) responses were seen, and no improvements in PFS or OS were observed. Conclusions This study demonstrated the feasibility of using FDG PET as a clinically reliable imaging biomarker for assessing EGFR inhibition in GBM, while revealing osimertinib's limited impact on both metabolic activity and tumor growth in GBM, findings that were concordant between preclinical and clinical observations.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- UCLA Brain Tumor Imaging Laboratory (BTIL), Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Qunicy Okobi
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Robert Chong
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Rhea Plawat
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Eva Zhao
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Andrei Gafita
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Ida Sonni
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Saewon Chun
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Emese Filka
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory (BTIL), Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Donatello Telesca
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, California, USA
| | - Shanpeng Li
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, California, USA
| | - Gang Li
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, California, USA
| | - Albert Lai
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Phioanh Nghiemphu
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Johannes Czernin
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - David A Nathanson
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Timothy F Cloughesy
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
17
|
Jiang Y, Huang M, Zhao Y, Dai J, Yang Q, Tang X, Li X, Cui Y, Zhang J, Sun J, Fu L, Mao H, Peng XG. A [ 18F]FDG PET based nomogram to predict cancer-associated cachexia and survival outcome: A multi-center study. Nutrition 2025; 129:112593. [PMID: 39426212 DOI: 10.1016/j.nut.2024.112593] [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: 04/16/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVES Cancer patients with cachexia face poor prognosis and shortened survival. Early diagnosis and accurate prognosis prediction remain challenging. This multi-center study aims to develop and externally validate a nomogram integrating [18F]fluoro-2-deoxy-D-glucose ([18F]FDG) PET findings and routine clinical biochemistry tests for predicting cancer-associated cachexia, while also assessing its potential prognostic value. RESEARCH METHODS & PROCEDURES A retrospective analysis of 658 cancer patients (390 in the development cohort, 268 in the validation cohort) utilized [18F]FDG PET/CT data from two centers. Logistic regression identified organ-specific standardized uptake values (SUVs) and clinical variables associated with cancer-associated cachexia. Diagnostic accuracy, discriminative ability, and clinical effectiveness were assessed using area under the curve (AUC), calibration curve, and decision curve. Nomogram predictability for overall survival was evaluated through Cox regression and Kaplan-Meier curves. RESULTS The combined nomogram incorporating age (odds ratio [OR] = 1.893; P = 0.012), hemoglobin (OR = 2.591; P < 0.001), maximum SUV of the liver (OR = 3.646; P < 0.001), and minimum SUV of the subcutaneous fat (OR = 5.060; P < 0.001) achieved good performance in predicting cancer-associated cachexia (AUC = 0.807/0.726, development/validation). Calibration and decision curve analyses confirmed its clinical effectiveness. Kaplan-Meier curves analysis showed that overall survival can be categorized using the combined nomogram (P < 0.001). CONCLUSION Combining radiological information from clinical standard [18F]FDG PET data from cancer patients with biochemical results in their routine clinical blood tests through a well-constructed nomogram enables predicting cachexia and its effect on the prognosis of cancer patients.
Collapse
Affiliation(s)
- Yang Jiang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Mouqing Huang
- Department of Nuclear Medicine, Ganzhou People's Hospital, Ganzhou, China
| | - Yufei Zhao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jingyue Dai
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Qingwen Yang
- Department of Internal Medicine, Ulm University & Ulm University Hospital, Ulm, Germany
| | - Xingzhe Tang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xinxiang Li
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ying Cui
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jingqi Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jialu Sun
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lin Fu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia, USA
| | - Xin-Gui Peng
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Department of Radiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China.
| |
Collapse
|
18
|
Chung YH, Park S, Lee M, Lee J, Ji Y, Song YJ, Woo TG, Shin E, Baek S, Hwang YJ, Kim Y, Kim M, Han J, Kim HR, Choi J, Kim BH, Park BJ. Therapeutic effect of novel drug candidate, PRG-N-01, on NF2 syndrome-related tumor. Neuro Oncol 2024:noae282. [PMID: 39731295 DOI: 10.1093/neuonc/noae282] [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: 11/06/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND NF2-related schwannomatosis (NF2-SWN) is associated with multiple benign tumors in the nervous system. NF2-SWN, caused by mutations in the NF2 gene, has developed into intracranial and spinal schwannomas. Because of the high surgical risk and frequent recurrence of multiple tumors, targeted therapy is necessary. However, there are no approved drugs. METHODS We examined the action mechanism of PRG-N-01, a candidate molecule for NF2-SWN, through the direct binding assay and mass spectrometry. For in vitro anti-proliferative experiments, primary cells derived from NF2 mouse model and patient tumors, were treated with PRG-N-01. The in vivo therapeutic and preventive efficacy was validated via intraperitoneal and oral administration in NF2 mouse model (Postn-Cre; Nf2f/f). Gene expression profile in the DRG of mouse model was explored by RNA sequencing. The pharmacological properties of PRG-N-01 were analyzed through the preclinical study. RESULTS PRG-N-01 binds to the N-terminal extremity of TGFβR1 (TβR1) kinase domain, where TβR1 and RKIP interact, inhibiting the binding and preventing degradation of RKIP. In vivo administration in the mouse model suppressed schwannoma progression in the DRG. Early oral administration of the PRG-N-01 also demonstrated preventive effects on NF2-SWN. PRG-N-01 treatment suppressed tumor growth genes while upregulating genes related to for normal cell metabolism and schwann cell differentiation in DRG. PRG-N-01 showed druggable properties through the preclinical study including ADME, pharmacodynamics, pharmacokinetics and toxicology. CONCLUSIONS Together, our study provides the rationale and critical data for a prospective clinical trial of PRG-N-01 in NF2-SWN patients indicating PRG-N-01 as a promising candidate for the treatment.
Collapse
Affiliation(s)
- Yeon-Ho Chung
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Soyoung Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Moonyoung Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jongwon Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yeongseon Ji
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Yi Jin Song
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Tae-Gyun Woo
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Eunbyeol Shin
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Songyoung Baek
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Young Jun Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Yuju Kim
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Minju Kim
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Jin Han
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Hong-Rae Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Bae-Hoon Kim
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Bum-Joon Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| |
Collapse
|
19
|
Kenny TC, Birsoy K. Mitochondria and Cancer. Cold Spring Harb Perspect Med 2024; 14:a041534. [PMID: 38692736 PMCID: PMC11610758 DOI: 10.1101/cshperspect.a041534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Mitochondria are semiautonomous organelles with diverse metabolic and cellular functions including anabolism and energy production through oxidative phosphorylation. Following the pioneering observations of Otto Warburg nearly a century ago, an immense body of work has examined the role of mitochondria in cancer pathogenesis and progression. Here, we summarize the current state of the field, which has coalesced around the position that functional mitochondria are required for cancer cell proliferation. In this review, we discuss how mitochondria influence tumorigenesis by impacting anabolism, intracellular signaling, and the tumor microenvironment. Consistent with their critical functions in tumor formation, mitochondria have become an attractive target for cancer therapy. We provide a comprehensive update on the numerous therapeutic modalities targeting the mitochondria of cancer cells making their way through clinical trials.
Collapse
Affiliation(s)
- Timothy C Kenny
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York 10065, USA
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
20
|
Padilha DMH, Mendes MCS, Takahashi MES, Lascala F, Silveira MN, Pozzuto L, Carrilho LAO, Guerra LD, Moreira RCL, Branbilla SR, Ramos CD, Carvalheira JBC. Subcutaneous adipose tissue radiodensity: An emerging risk factor for severe COVID-19. Nutrition 2024; 128:112561. [PMID: 39277984 DOI: 10.1016/j.nut.2024.112561] [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: 04/17/2024] [Revised: 07/13/2024] [Accepted: 07/25/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND Adipose tissue radiodensity and metabolic activity may influence COVID-19 outcomes. This study evaluated the association between adipose tissue characteristics and clinical outcomes in COVID-19 patients. METHODS Two retrospective cohorts of hospitalized COVID-19 patients were analyzed. Subcutaneous adipose tissue radiodensity (SATR) and visceral adipose tissue radiodensity were assessed by computed tomography. Fluorine-18-labelled fluorodeoxyglucose PET/computed tomography measured adipose tissue metabolic activity. Associations with mortality, length of stay, ventilation requirement, and complications were examined using regression analyses. RESULTS High SATR was independently associated with increased mortality risk (OR: 2.70; P = 0.033), longer hospitalization (P < 0.001), higher rates of mechanical ventilation (P = 0.007), and complications: acute kidney injury (P = 0.001), secondary infection (P = 0.007), shock (P = 0.010), and pulmonary embolism (P = 0.011). SATR positively correlated with SAT glucose uptake (ρ = 0.52) and negatively with leptin levels (ρ = -0.48). CONCLUSIONS Elevated SATR at COVID-19 diagnosis predicts disease severity and worse outcomes. SATR is a potential prognostic biomarker for acute and chronic inflammatory conditions.
Collapse
Affiliation(s)
- Daniela M H Padilha
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil; Nestlé Health Science, Lausanne, Switzerland
| | - Maria C S Mendes
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil; Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Maria E S Takahashi
- Institute of Physics "Gleb Wataghin", University of Campinas, Campinas SP, Brazil
| | - Fabiana Lascala
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Marina N Silveira
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Lara Pozzuto
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Larissa A O Carrilho
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Lívia D Guerra
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Rafaella C L Moreira
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Sandra R Branbilla
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Celso Darío Ramos
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - José B C Carvalheira
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil.
| |
Collapse
|
21
|
Yamada S, Kotani T, Tamaki N, Nakai Y, Toyama Y, Nishimura M, Nakamura Y, Nii T, Yamada K. Dynamic FDG PET/CT for differentiating focal pelvic uptake in patients with gynecological cancer. Sci Rep 2024; 14:29499. [PMID: 39604525 PMCID: PMC11603040 DOI: 10.1038/s41598-024-81236-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: 09/21/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Abstract
This study aimed to evaluate the ability of serial whole-body dynamic PET/CT to differentiate physiological from abnormal 18F-FDG uptake in the abdomen and pelvis of gynecological cancer patients. We conducted a retrospective study of 61 18F-FDG PET/CT examinations for suspected gynecological malignancies or metastases between March 2018 and January 2020. Our protocol included four-phase dynamic whole-body scans. High-uptake foci with SUVmax > 2.5 in the abdominopelvic region caudal to the renal portal were picked up and visually evaluated as "changed" (disappeared during any phase or morphological changes in more than half of the foci) or "unchanged" in motion on the serial dynamic images. Focal 18F-FDG uptake was observed in 84 foci. Of the 58 foci determined pathologically or clinically to have pathological uptake, no change was observed on serial dynamic imaging in 54 foci (sensitivity, 93%). Of the 26 foci of physiological uptake, temporal changes in uptake were observed in 20 foci using dynamic imaging (specificity, 77%). The positive and negative predictive values were 90% and 83%, respectively, with an accuracy of 88%. Dynamic whole-body 18F-FDG PET/CT imaging allows for differentiation between pathological and physiological uptake in the abdominopelvic region of patients with gynecological cancer.
Collapse
Affiliation(s)
- Sachimi Yamada
- Department of Radiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.
- Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan.
| | - Tomoya Kotani
- Department of Radiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nagara Tamaki
- Department of Radiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Kyoto College of Medical Science, Nantan, Kyoto, Japan
| | - Yoshitomo Nakai
- Department of Radiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuchiyo Toyama
- Department of Radiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Motoki Nishimura
- Department of Radiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Kyoto Chubu Medical Center, Nantan, Kyoto, Japan
| | - Yasunori Nakamura
- Kyoto College of Medical Science, Nantan, Kyoto, Japan
- Department of Radiological Technology, University Hospital, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Nii
- Department of Radiological Technology, University Hospital, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kei Yamada
- Department of Radiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
22
|
Zeng T, Chen Y, Zhu D, Huang Y, Huang Y, Chen Y, Shi J, Ding B, Huang J. AI diagnostics in bone oncology for predicting bone metastasis in lung cancer patients using DenseNet-264 deep learning model and radiomics. J Bone Oncol 2024; 48:100640. [PMID: 39399584 PMCID: PMC11470571 DOI: 10.1016/j.jbo.2024.100640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/14/2024] [Accepted: 09/21/2024] [Indexed: 10/15/2024] Open
Abstract
This study aims to predict bone metastasis in lung cancer patients using radiomics and deep learning. Early prediction of bone metastasis is crucial for timely intervention and personalized treatment plans. This can improve patient outcomes and quality of life. By integrating advanced imaging techniques with artificial intelligence, this study seeks to enhance predictive accuracy and clinical decision-making. Methods We included 189 lung cancer patients, comprising 89 with non-bone metastasis and 100 with confirmed bone metastasis. Radiomic features were extracted from CT images, and feature selection was performed using Minimum Redundancy Maximum Relevance (mRMR) and Least Absolute Shrinkage and Selection Operator (LASSO). We developed and validated a radiomics model and a deep learning model using DenseNet-264. Model performance was evaluated using the area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, and specificity. Statistical comparisons were made using the DeLong test. Results The radiomics model achieved an AUC of 0.815 on the training set and 0.778 on the validation set. The DenseNet-264 model demonstrated superior performance with an AUC of 0.990 on the training set and 0.971 on the validation set. The DeLong test confirmed that the AUC of the DenseNet-264 model was significantly higher than that of the radiomics model (p < 0.05). Conclusions The DenseNet-264 model significantly outperforms the radiomics model in predicting bone metastasis in lung cancer patients. The early and accurate prediction provided by the deep learning model can facilitate timely interventions and personalized treatment planning, potentially improving patient outcomes. Future studies should focus on validating these findings in larger, multi-center cohorts and integrating clinical data to further enhance predictive accuracy.
Collapse
Affiliation(s)
- Taisheng Zeng
- Faculty of Mathematics and Computer Science, Quanzhou Normal University, Quanzhou 362000, China
- Fujian Provincial Key Laboratory of Data Intensive Computing, Quanzhou 362000, China
- Key Laboratory of Intelligent Computing and Information Processing, Fujian Province University, Quanzhou 362000, China
| | - Yusi Chen
- Faculty of Mathematics and Computer Science, Quanzhou Normal University, Quanzhou 362000, China
- Fujian Provincial Key Laboratory of Data Intensive Computing, Quanzhou 362000, China
- Key Laboratory of Intelligent Computing and Information Processing, Fujian Province University, Quanzhou 362000, China
| | - Daxin Zhu
- Faculty of Mathematics and Computer Science, Quanzhou Normal University, Quanzhou 362000, China
- Fujian Provincial Key Laboratory of Data Intensive Computing, Quanzhou 362000, China
- Key Laboratory of Intelligent Computing and Information Processing, Fujian Province University, Quanzhou 362000, China
| | - Yifeng Huang
- Department of Diagnostic Radiology, Huaqiao University Affliated Strait Hospital, Quanzhou, Fujian 362000, China
| | - Ying Huang
- Department of Diagnostic Radiology, Huaqiao University Affliated Strait Hospital, Quanzhou, Fujian 362000, China
| | - Yijie Chen
- Department of General Surgery, Huaqiao University Affliated Strait Hospital, Quanzhou, Fujian 362000, China
| | - Jianshe Shi
- Department of General Surgery, Huaqiao University Affliated Strait Hospital, Quanzhou, Fujian 362000, China
| | - Bijiao Ding
- Department of Diagnostic Radiology, Huaqiao University Affliated Strait Hospital, Quanzhou, Fujian 362000, China
| | - Jianlong Huang
- Faculty of Mathematics and Computer Science, Quanzhou Normal University, Quanzhou 362000, China
- Fujian Provincial Key Laboratory of Data Intensive Computing, Quanzhou 362000, China
- Key Laboratory of Intelligent Computing and Information Processing, Fujian Province University, Quanzhou 362000, China
| |
Collapse
|
23
|
Kim D, Kang SK, Shin SA, Choi H, Lee JS. Improving 18F-FDG PET Quantification Through a Spatial Normalization Method. J Nucl Med 2024; 65:1645-1651. [PMID: 39209545 PMCID: PMC11448607 DOI: 10.2967/jnumed.123.267360] [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: 12/29/2023] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Quantification of 18F-FDG PET images is useful for accurate diagnosis and evaluation of various brain diseases, including brain tumors, epilepsy, dementia, and Parkinson disease. However, accurate quantification of 18F-FDG PET images requires matched 3-dimensional T1 MRI scans of the same individuals to provide detailed information on brain anatomy. In this paper, we propose a transfer learning approach to adapt a pretrained deep neural network model from amyloid PET to spatially normalize 18F-FDG PET images without the need for 3-dimensional MRI. Methods: The proposed method is based on a deep learning model for automatic spatial normalization of 18F-FDG brain PET images, which was developed by fine-tuning a pretrained model for amyloid PET using only 103 18F-FDG PET and MR images. After training, the algorithm was tested on 65 internal and 78 external test sets. All T1 MR images with a 1-mm isotropic voxel size were processed with FreeSurfer software to provide cortical segmentation maps used to extract a ground-truth regional SUV ratio using cerebellar gray matter as a reference region. These values were compared with those from spatial normalization-based quantification methods using the proposed method and statistical parametric mapping software. Results: The proposed method showed superior spatial normalization compared with statistical parametric mapping, as evidenced by increased normalized mutual information and better size and shape matching in PET images. Quantitative evaluation revealed a consistently higher SUV ratio correlation and intraclass correlation coefficients for the proposed method across various brain regions in both internal and external datasets. The remarkably good correlation and intraclass correlation coefficient values of the proposed method for the external dataset are noteworthy, considering the dataset's different ethnic distribution and the use of different PET scanners and image reconstruction algorithms. Conclusion: This study successfully applied transfer learning to a deep neural network for 18F-FDG PET spatial normalization, demonstrating its resource efficiency and improved performance. This highlights the efficacy of transfer learning, which requires a smaller number of datasets than does the original network training, thus increasing the potential for broader use of deep learning-based brain PET spatial normalization techniques for various clinical and research radiotracers.
Collapse
Affiliation(s)
- Daewoon Kim
- Interdisciplinary Program of Bioengineering, Seoul National University, Seoul, South Korea
- Artificial Intelligence Institute, Seoul National University, Seoul, South Korea
| | - Seung Kwan Kang
- Brightonix Imaging Inc., Seoul, South Korea;
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea; and
| | | | - Hongyoon Choi
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea; and
- Department of Nuclear Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, South Korea
| | - Jae Sung Lee
- Interdisciplinary Program of Bioengineering, Seoul National University, Seoul, South Korea;
- Artificial Intelligence Institute, Seoul National University, Seoul, South Korea
- Brightonix Imaging Inc., Seoul, South Korea
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea; and
- Department of Nuclear Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
24
|
Dwivedi P, Kumar Jha A, Mithun S, Sawant V, Vajarkar V, Chauhan M, Choudhury S, Rangarajan V. Dose estimation in patients from different protocols of 18F-FDG PET/CT studies and analysis of optimization strategies. RADIATION PROTECTION DOSIMETRY 2024; 200:1384-1390. [PMID: 39213637 DOI: 10.1093/rpd/ncae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/10/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024]
Abstract
This study aimed to evaluate the dose in different protocols of 18F-2-fluoro-2-deoxy-D-glucose positron emission tomography/computed tomography (PET/CT) procedure. The retrospective study involves 207 patients with confirmed malignancies who underwent PET/CT. Effective dose (E) from PET was estimated based on injected activity and dose coefficient as per International Commission on Radiation Protection (ICRP) 128. Estimation of E from CT was done utilizing the dose length product (DLP) method and conversion factors as per ICRP 102. There was a significant statistical difference observed in E between different PET/CT protocols (P < .001). E of PET in the whole body (WB) was found to be 4.9 ± 0.9 mSv, whereas mean volume computed tomography dose indexvol, DLP, and E of CT in WB were 7.0 ± 0.2 mGy, 674.3 ± 80.7 mGy.cm, and 10.1 ± 1.2 mSv, respectively. No linear correlation was seen between the size-specific dose estimate and E of CT (r = -0.003; P = .978). The total mean E in WB PET/CT was 17.0 ± 1.7 mSv. CT dose was contributing more than PET dose in all protocols except brain PET/CT. Optimization strategies can be evaluated only if monitored periodically.
Collapse
Affiliation(s)
- Pooja Dwivedi
- Department of Nuclear Medicine and Molecular Imaging, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Mumbai 400094, India
| | - Ashish Kumar Jha
- Homi Bhabha National Institute, Mumbai 400094, India
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Tata Memorial Centre, Dr Ernest Borges Rd, Parel, Mumbai, Maharashtra 400012, India
| | - Sneha Mithun
- Homi Bhabha National Institute, Mumbai 400094, India
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Tata Memorial Centre, Dr Ernest Borges Rd, Parel, Mumbai, Maharashtra 400012, India
| | - Viraj Sawant
- Department of Nuclear Medicine and Molecular Imaging, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Mumbai 400094, India
| | - Vishal Vajarkar
- Department of Nuclear Medicine and Molecular Imaging, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Mumbai 400094, India
| | - Manoj Chauhan
- Department of Nuclear Medicine and Molecular Imaging, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Mumbai 400094, India
| | - Sayak Choudhury
- Department of Nuclear Medicine and Molecular Imaging, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Mumbai 400094, India
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine and Molecular Imaging, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Mumbai 400094, India
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Tata Memorial Centre, Dr Ernest Borges Rd, Parel, Mumbai, Maharashtra 400012, India
| |
Collapse
|
25
|
Wang Q, Abdelhafez YG, Nalbant H, Spencer BA, Bayerlein R, Qi J, Cherry SR, Nardo L, Badawi RD. Refining penalty parameter selection in whole-body PET image reconstruction for lung cancer patients using the cross-validation log-likelihood method. Phys Med Biol 2024; 69:10.1088/1361-6560/ad7222. [PMID: 39168154 PMCID: PMC11500750 DOI: 10.1088/1361-6560/ad7222] [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: 05/31/2024] [Accepted: 08/21/2024] [Indexed: 08/23/2024]
Abstract
Objective.Penalty parameters in penalized likelihood positron emission tomography (PET) reconstruction are typically determined empirically. The cross-validation log-likelihood (CVLL) method has been introduced to optimize these parameters by maximizing a CVLL function, which assesses the likelihood of reconstructed images using one subset of a list-mode dataset based on another subset. This study aims to validate the efficacy of the CVLL method in whole-body imaging for cancer patients using a conventional clinical PET scanner.Approach.Fifteen lung cancer patients were injected with 243.7 ± 23.8 MBq of [18F]FDG and underwent a 22 min PET scan on a Biograph mCT PET/CT scanner, starting at 60 ± 5 min post-injection. The PET list-mode data were partitioned by subsampling without replacement, with 20 minutes of data for image reconstruction using an in-house ordered subset expectation maximization algorithm and the remaining 2 minutes of data for cross-validation. Two penalty parameters, penalty strengthβand Fair penalty function parameterδ, were subjected to optimization. Whole-body images were reconstructed, and CVLL values were computed across various penalty parameter combinations. The optimal image corresponding to the maximum CVLL value was selected by a grid search for each patient.Main results.Theδvalue required to maximize the CVLL value was notably small (⩽10-6in this study). The influences of voxel size and scan duration on image optimization were investigated. A correlation analysis revealed a significant inverse relationship between optimalβand scan count level, with a correlation coefficient of -0.68 (p-value = 3.5 × 10-5). The optimal images selected by the CVLL method were compared with those chosen by two radiologists based on their diagnostic preferences. Differences were observed in the selection of optimal images.Significance.This study demonstrates the feasibility of incorporating the CVLL method into routine imaging protocols, potentially allowing for a wide range of combinations of injected radioactivity amounts and scan durations in modern PET imaging.
Collapse
Affiliation(s)
- Qian Wang
- Department of Biomedical Engineering, University of California, Davis, California, USA
- Department of Radiology, University of California, Davis, California, USA
| | - Yasser G Abdelhafez
- Department of Radiology, University of California, Davis, California, USA
- Department of Radiotherapy and Nuclear Medicine, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Hande Nalbant
- Department of Radiology, University of California, Davis, California, USA
| | - Benjamin A Spencer
- Department of Biomedical Engineering, University of California, Davis, California, USA
- Department of Radiology, University of California, Davis, California, USA
| | - Reimund Bayerlein
- Department of Biomedical Engineering, University of California, Davis, California, USA
- Department of Radiology, University of California, Davis, California, USA
| | - Jinyi Qi
- Department of Biomedical Engineering, University of California, Davis, California, USA
| | - Simon R. Cherry
- Department of Biomedical Engineering, University of California, Davis, California, USA
- Department of Radiology, University of California, Davis, California, USA
| | - Lorenzo Nardo
- Department of Radiology, University of California, Davis, California, USA
| | - Ramsey D Badawi
- Department of Biomedical Engineering, University of California, Davis, California, USA
- Department of Radiology, University of California, Davis, California, USA
| |
Collapse
|
26
|
Feng Y, Cheng B, Zhan S, Liu H, Li J, Chen P, Wang Z, Huang X, Fu X, Ye W, Wang R, Wang Q, Xiang Y, Wang H, Zhu F, Zheng X, Fu W, Hu G, Chen Z, He J, Liang W. The impact of PET/CT and brain MRI for metastasis detection among patients with clinical T1-category lung cancer: Findings from a large-scale cohort study. Eur J Nucl Med Mol Imaging 2024; 51:3400-3416. [PMID: 38722381 PMCID: PMC11369054 DOI: 10.1007/s00259-024-06740-8] [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: 01/03/2024] [Accepted: 04/26/2024] [Indexed: 09/03/2024]
Abstract
PURPOSE [18F]-FDG PET/CT and brain MRI are common approaches to detect metastasis in patients of lung cancer. Current guidelines for the use of PET/CT and MRI in clinical T1-category lung cancer lack risk-based stratification and require optimization. This study stratified patients based on metastatic risk in terms of the lesions' size and morphological characteristics. METHODS The detection rate of metastasis was measured in different sizes and morphological characteristics (solid and sub-solid) of tumors. To confirm the cut-off value for discriminating metastasis and overall survival (OS) prediction, the receiver operating characteristic (ROC) analysis was performed based on PET/CT metabolic parameters (SUVmax/SUVmean/SULpeak/MTV/TLG), followed by Kaplan-Meier analysis for survival in post-operation patients with and without PET/CT plus MRI. RESULTS 2,298 patients were included. No metastasis was observed in patients with solid nodules < 8.0 mm and sub-solid nodules < 10.0 mm. The cut-off of PET/CT metabolic parameters on discriminating metastasis were 1.09 (SUVmax), 0.26 (SUVmean), 0.31 (SULpeak), 0.55 (MTV), and 0.81 (TLG), respectively. Patients undergoing PET/CT plus MRI exhibited longer OS compared to those who did not receive it in solid nodules ≥ 8.0 mm & sub-solid nodules ≥ 10.0 mm (HR, 0.44; p < 0.001); in solid nodules ≥ 8.0 mm (HR, 0.12; p<0.001) and in sub-solid nodules ≥ 10.0 mm (HR; 0.61; p=0.075), respectively. Compared to patients with metabolic parameters lower than cut-off values, patients with higher metabolic parameters displayed shorter OS: SUVmax (HR, 12.94; p < 0.001), SUVmean (HR, 11.33; p <0.001), SULpeak (HR, 9.65; p < 0.001), MTV (HR, 9.16; p = 0.031), and TLG (HR, 12.06; p < 0.001). CONCLUSION The necessity of PET/CT and MRI should be cautiously evaluated in patients with solid nodules < 8.0 mm and sub-solid nodules < 10.0 mm, however, these examinations remained essential and beneficial for patients with solid nodules ≥ 8.0 mm and sub-solid nodules ≥ 10.0 mm.
Collapse
Affiliation(s)
- Yi Feng
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Bo Cheng
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Shuting Zhan
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Haiping Liu
- PET/CT Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianfu Li
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Peiling Chen
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Zixun Wang
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Xiaoyan Huang
- The Radiology Department of the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Xiuxia Fu
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Wenjun Ye
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Runchen Wang
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Qixia Wang
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Yang Xiang
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Huiting Wang
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Feng Zhu
- Detroit Medical Center Sinai-Grace Hospital, Internal Medicine Department, 6071 Outer Dr W, Detroit, MI, 48235, USA
| | - Xin Zheng
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Wenhai Fu
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
| | - Guodong Hu
- Department of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523108, China
| | - Zhuxing Chen
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China
- Pulmonary Nodule Surgical Department, The First People's Hospital of Foshan, Foshan, 528000, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China.
- Department of Thoracic Surgery, NANFANG Hospital of Southern Medical University, Guangzhou, China.
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, China.
- Department of Oncology Medical Center, The First People's Hospital of Zhaoqing, Zhaoqing City, Guangdong Province, China.
| |
Collapse
|
27
|
Li W, Huang Z, Chen Z, Jiang Y, Zhou C, Zhang X, Fan W, Zhao Y, Zhang L, Wan L, Yang Y, Zheng H, Liang D, Hu Z. Learning CT-free attenuation-corrected total-body PET images through deep learning. Eur Radiol 2024; 34:5578-5587. [PMID: 38355987 DOI: 10.1007/s00330-024-10647-1] [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: 10/08/2023] [Revised: 11/30/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024]
Abstract
OBJECTIVES Total-body PET/CT scanners with long axial fields of view have enabled unprecedented image quality and quantitative accuracy. However, the ionizing radiation from CT is a major issue in PET imaging, which is more evident with reduced radiopharmaceutical doses in total-body PET/CT. Therefore, we attempted to generate CT-free attenuation-corrected (CTF-AC) total-body PET images through deep learning. METHODS Based on total-body PET data from 122 subjects (29 females and 93 males), a well-established cycle-consistent generative adversarial network (Cycle-GAN) was employed to generate CTF-AC total-body PET images directly while introducing site structures as prior information. Statistical analyses, including Pearson correlation coefficient (PCC) and t-tests, were utilized for the correlation measurements. RESULTS The generated CTF-AC total-body PET images closely resembled real AC PET images, showing reduced noise and good contrast in different tissue structures. The obtained peak signal-to-noise ratio and structural similarity index measure values were 36.92 ± 5.49 dB (p < 0.01) and 0.980 ± 0.041 (p < 0.01), respectively. Furthermore, the standardized uptake value (SUV) distribution was consistent with that of real AC PET images. CONCLUSION Our approach could directly generate CTF-AC total-body PET images, greatly reducing the radiation risk to patients from redundant anatomical examinations. Moreover, the model was validated based on a multidose-level NAC-AC PET dataset, demonstrating the potential of our method for low-dose PET attenuation correction. In future work, we will attempt to validate the proposed method with total-body PET/CT systems in more clinical practices. CLINICAL RELEVANCE STATEMENT The ionizing radiation from CT is a major issue in PET imaging, which is more evident with reduced radiopharmaceutical doses in total-body PET/CT. Our CT-free PET attenuation correction method would be beneficial for a wide range of patient populations, especially for pediatric examinations and patients who need multiple scans or who require long-term follow-up. KEY POINTS • CT is the main source of radiation in PET/CT imaging, especially for total-body PET/CT devices, and reduced radiopharmaceutical doses make the radiation burden from CT more obvious. • The CT-free PET attenuation correction method would be beneficial for patients who need multiple scans or long-term follow-up by reducing additional radiation from redundant anatomical examinations. • The proposed method could directly generate CT-free attenuation-corrected (CTF-AC) total-body PET images, which is beneficial for PET/MRI or PET-only devices lacking CT image poses.
Collapse
Affiliation(s)
- Wenbo Li
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Zhenxing Huang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zixiang Chen
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Yongluo Jiang
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Chao Zhou
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xu Zhang
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wei Fan
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yumo Zhao
- Central Research Institute, United Imaging Healthcare Group, Shanghai, 201807, China
| | - Lulu Zhang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liwen Wan
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yongfeng Yang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hairong Zheng
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Dong Liang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zhanli Hu
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
28
|
Mirvald C, Garaz R, Sinescu I, Preda A, Labanaris A, Yossepowitch O, Tsaur I, Surcel C. Current Role of PET CT in Staging and Management of Penile Cancers. J Clin Med 2024; 13:4879. [PMID: 39201021 PMCID: PMC11355205 DOI: 10.3390/jcm13164879] [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: 07/03/2024] [Revised: 07/30/2024] [Accepted: 08/16/2024] [Indexed: 09/02/2024] Open
Abstract
Penile cancer (PeCa) is a rare urological malignancy characterized by significant geographical variations in both incidence and mortality rates. Due to its rarity and the consequent lack of randomized trials, current management is based on retrospective studies and small prospective trials. In addition, both the diagnostic pathways and treatment strategies exhibit substantial heterogeneity, differing significantly between less-developed and well-developed countries. The prognosis of PeCas is determined by the presence and extent of regional lymph node (LN) involvement. Therefore, the early detection and treatment of LN metastasis is paramount to ensure better outcomes. In recent decades, overall survival of PeCas has increased, mainly due to advancements in imaging techniques and risk stratification. We aim to provide an overview of the current role of PET CT imaging in the management of patients with PeCa.
Collapse
Affiliation(s)
- Cristian Mirvald
- Department of Urology, Fundeni Clinical Institute, 022328 Bucharest, Romania (A.P.)
- “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Radion Garaz
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Ioanel Sinescu
- Department of Urology, Fundeni Clinical Institute, 022328 Bucharest, Romania (A.P.)
- “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Adrian Preda
- Department of Urology, Fundeni Clinical Institute, 022328 Bucharest, Romania (A.P.)
| | - Apostolos Labanaris
- Department of Urology, Interbalkan Medical Center, 57001 Thessaloniki, Greece
| | - Ofer Yossepowitch
- Department of Urology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
| | - Igor Tsaur
- Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Cristian Surcel
- Department of Urology, Fundeni Clinical Institute, 022328 Bucharest, Romania (A.P.)
- “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
29
|
Cheon M, Yi H, Ha JY, Kim MA. Atypical 18F-FDG PET-CT Findings in a Rare Case of Primary Hepatic Leiomyosarcoma. Diagnostics (Basel) 2024; 14:1502. [PMID: 39061638 PMCID: PMC11275497 DOI: 10.3390/diagnostics14141502] [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: 06/11/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
The primary hepatic leiomyosarcoma is a rare malignant tumor arising from the smooth muscle cells in the hepatic vessels, bile ducts, and ligamentum teres. It is considered a subtype of hepatic sarcomas. We report awkward 18F-FDG PET-CT findings of a primary hepatic leiomyosarcoma masquerading as a benign hepatic tumor, which were confirmed by histopathological and immunohistochemical examinations in a 78-year-old woman.
Collapse
Affiliation(s)
- Miju Cheon
- Department of Nuclear Medicine, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea
| | - Hyunkyung Yi
- Department of Nuclear Medicine, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea
| | - Joo Young Ha
- Division of Hematology and Oncology, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea
| | - Min A Kim
- Department of Pathology, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea
| |
Collapse
|
30
|
Reed MB, Handschuh PA, Schmidt C, Murgaš M, Gomola D, Milz C, Klug S, Eggerstorfer B, Aichinger L, Godbersen GM, Nics L, Traub-Weidinger T, Hacker M, Lanzenberger R, Hahn A. Validation of cardiac image-derived input functions for functional PET quantification. Eur J Nucl Med Mol Imaging 2024; 51:2625-2637. [PMID: 38676734 PMCID: PMC11224076 DOI: 10.1007/s00259-024-06716-8] [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: 01/15/2024] [Accepted: 04/14/2024] [Indexed: 04/29/2024]
Abstract
PURPOSE Functional PET (fPET) is a novel technique for studying dynamic changes in brain metabolism and neurotransmitter signaling. Accurate quantification of fPET relies on measuring the arterial input function (AIF), traditionally achieved through invasive arterial blood sampling. While non-invasive image-derived input functions (IDIF) offer an alternative, they suffer from limited spatial resolution and field of view. To overcome these issues, we developed and validated a scan protocol for brain fPET utilizing cardiac IDIF, aiming to mitigate known IDIF limitations. METHODS Twenty healthy individuals underwent fPET/MR scans using [18F]FDG or 6-[18F]FDOPA, utilizing bed motion shuttling to capture cardiac IDIF and brain task-induced changes. Arterial and venous blood sampling was used to validate IDIFs. Participants performed a monetary incentive delay task. IDIFs from various blood pools and composites estimated from a linear fit over all IDIF blood pools (3VOI) and further supplemented with venous blood samples (3VOIVB) were compared to the AIF. Quantitative task-specific images from both tracers were compared to assess the performance of each input function to the gold standard. RESULTS For both radiotracer cohorts, moderate to high agreement (r: 0.60-0.89) between IDIFs and AIF for both radiotracer cohorts was observed, with further improvement (r: 0.87-0.93) for composite IDIFs (3VOI and 3VOIVB). Both methods showed equivalent quantitative values and high agreement (r: 0.975-0.998) with AIF-derived measurements. CONCLUSION Our proposed protocol enables accurate non-invasive estimation of the input function with full quantification of task-specific changes, addressing the limitations of IDIF for brain imaging by sampling larger blood pools over the thorax. These advancements increase applicability to any PET scanner and clinical research setting by reducing experimental complexity and increasing patient comfort.
Collapse
Affiliation(s)
- Murray Bruce Reed
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Patricia Anna Handschuh
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Clemens Schmidt
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Matej Murgaš
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - David Gomola
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Christian Milz
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Sebastian Klug
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Benjamin Eggerstorfer
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Lisa Aichinger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Godber Mathis Godbersen
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Tatjana Traub-Weidinger
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria.
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria.
| | - Andreas Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| |
Collapse
|
31
|
Goodson BM, Chekmenev EY. Toward next-generation molecular imaging. Proc Natl Acad Sci U S A 2024; 121:e2405380121. [PMID: 38657055 PMCID: PMC11067020 DOI: 10.1073/pnas.2405380121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Affiliation(s)
- Boyd M. Goodson
- School of Chemical & Biomolecular Sciences and Materials Technology Center, Southern Illinois University, Carbondale, IL62901
| | - Eduard Y. Chekmenev
- Department of Chemistry, Integrative Biosciences, Karmanos Cancer Institute, Wayne State University, Detroit, MI48202
| |
Collapse
|
32
|
Zhong Z, Yang K, Li Y, Zhou S, Yao H, Zhao Y, Huang Y, Zou J, Li Y, Jiajia Li, Lian G, Huang K, Chen S. Tumor-associated macrophages drive glycolysis through the IL-8/STAT3/GLUT3 signaling pathway in pancreatic cancer progression. Cancer Lett 2024; 588:216784. [PMID: 38458594 DOI: 10.1016/j.canlet.2024.216784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/07/2024] [Accepted: 03/03/2024] [Indexed: 03/10/2024]
Abstract
Glycolytic metabolism is a hallmark of pancreatic ductal adenocarcinoma (PDAC), and tumor-associated stromal cells play important roles in tumor metabolism. We previously reported that tumor-associated macrophages (TAMs) facilitate PDAC progression. However, little is known about whether TAMs are involved in regulating glycolysis in PDAC. Here, we found a positive correlation between CD68+ TAM infiltration and FDG maximal standardized uptake (FDG SUVmax) on PET-CT images of PDAC. We discovered that the glycolytic gene set was prominently enriched in the high TAM infiltration group through Gene Set Enrichment Analysis using The Cancer Genome Atlas database. Mechanistically, TAMs secreted IL-8 to promote GLUT3 expression in PDAC cells, enhancing tumor glycolysis both in vitro and in vivo, whereas this effect could be blocked by the IL-8 receptor inhibitor reparixin. Furthermore, IL-8 promoted the translocation of phosphorylated STAT3 into the nucleus to activate the GLUT3 promoter. Overall, we demonstrated that TAMs boosted PDAC cell glycolysis through the IL-8/STAT3/GLUT3 signaling pathway. Our cumulative findings suggest that the abrogation of TAM-induced tumor glycolysis by reparixin might exhibit an antitumor impact and offer a potential therapeutic target for PDAC.
Collapse
Affiliation(s)
- Ziyi Zhong
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Kege Yang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Yunlong Li
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Shurui Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, PR China
| | - Hanming Yao
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Yue Zhao
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Yuzhou Huang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Jinmao Zou
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Yaqing Li
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Jiajia Li
- Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China
| | - Guoda Lian
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Kaihong Huang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| | - Shaojie Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| |
Collapse
|
33
|
Kaji T, Osanai K, Takahashi A, Kinoshita A, Satoh D, Nakata T, Tamaki N. Improvement of motion artifacts using dynamic whole-body 18F-FDG PET/CT imaging. Jpn J Radiol 2024; 42:374-381. [PMID: 38093138 PMCID: PMC10980605 DOI: 10.1007/s11604-023-01513-z] [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: 08/14/2023] [Accepted: 11/05/2023] [Indexed: 04/01/2024]
Abstract
PURPOSE Serial dynamic whole-body PET imaging is valuable for assessing serial changes in tracer uptake. The purpose of this study was to evaluate the improvement of motion artifacts in patients using serial dynamic whole-body 18F-fluorodeoxyglyucose (FDG) PET/CT imaging. MATERIALS AND METHODS In 797 consecutive patients, serial 3-min dynamic whole-body FDG PET imaging was performed seven times, at 60 or 90 min after FDG administration. In cases with large body motion during imaging, we tried to improve the images by summing the images before body motion. An image quality study was performed on another 50 patients without obvious body motion using the same acquisition mode. RESULTS Obvious body movement was observed in 106 of 797 cases (13.3%), and severe motion artifacts which interfered image interpretation were observed in 18 (2.3%). In these 18 cases, summation of the images before the body movement enabled us to obtain images that excluded the effect of the body motion. In the visual evaluation of the image quality in another 50 patients studied, acceptable image quality was obtained when 2 or more times the serial 3-min image data were added. CONCLUSION Serial dynamic whole-body FDG PET imaging can minimize body motion artifacts by summation of the images before the body motion. Such serial dynamic study may be a choice for PET imaging to eliminate motion artifacts.
Collapse
Affiliation(s)
- Tomohito Kaji
- Department of Radiology, Division of Nuclear Medicine and PET Center, Hakodate Goryokaku Hospital, 38-3 Goryokaku-Cho, Hakodate, Hokkaido, 040-8611, Japan.
| | - Kouji Osanai
- Department of Radiology, Division of Nuclear Medicine and PET Center, Hakodate Goryokaku Hospital, 38-3 Goryokaku-Cho, Hakodate, Hokkaido, 040-8611, Japan
| | - Atsushi Takahashi
- Department of Radiology, Division of Nuclear Medicine and PET Center, Hakodate Goryokaku Hospital, 38-3 Goryokaku-Cho, Hakodate, Hokkaido, 040-8611, Japan
| | - Atsushi Kinoshita
- Department of Radiology, Division of Nuclear Medicine and PET Center, Hakodate Goryokaku Hospital, 38-3 Goryokaku-Cho, Hakodate, Hokkaido, 040-8611, Japan
| | - Daiki Satoh
- Department of Radiology, Division of Nuclear Medicine and PET Center, Hakodate Goryokaku Hospital, 38-3 Goryokaku-Cho, Hakodate, Hokkaido, 040-8611, Japan
| | - Tomoaki Nakata
- Department of Radiology, Division of Nuclear Medicine and PET Center, Hakodate Goryokaku Hospital, 38-3 Goryokaku-Cho, Hakodate, Hokkaido, 040-8611, Japan
| | - Nagara Tamaki
- Department of Radiology, Kyoto Prefectural University of Medicine, 465 Kajii-Cho, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
- Kyoto College of Medical Science, Oyama-Higashi, Sonobe, Nantan, Kyoto, 622-0041, Japan
| |
Collapse
|
34
|
Lee ST, Kovaleva N, Senko C, Kee D, Scott AM. Positron Emission Tomography/Computed Tomography Transformation of Oncology: Melanoma and Skin Malignancies. PET Clin 2024; 19:231-248. [PMID: 38233284 DOI: 10.1016/j.cpet.2023.12.009] [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: 01/19/2024]
Abstract
Skin cancers are the most common cancers, with melanoma resulting in the highest cause of death in this category. Accurate clinical, histologic, and imaging staging with fludeoxyglucose positron emission tomography (FDG PET) is most important to guide patient management. Whilst surgical excision with clear margins is the gold-standard treatment for primary cutaneous melanoma, targeted therapies have generated remarkable and rapid clinical responses in melanoma, for which FDG PET also plays an important role in assessment of treatment response and post-therapy surveillance. Non-FDG PET tracers, advanced PET technology, and PET radiomics may potentially change the landscape of the utilization of PET in the imaging of patients with cutaneous malignancies.
Collapse
Affiliation(s)
- Sze-Ting Lee
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia; Olivia Newton-John Cancer Research Institute, and La Trobe University, Heidelberg, Australia; Department of Surgery, University of Melbourne, Melbourne, Australia; School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Natalia Kovaleva
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Australia
| | - Clare Senko
- Olivia Newton-John Cancer Research Institute, and La Trobe University, Heidelberg, Australia; Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, Australia
| | - Damien Kee
- Olivia Newton-John Cancer Research Institute, and La Trobe University, Heidelberg, Australia; Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, Australia; Department of Medical Oncology, Peter MacCallum Cancer Center, Melbourne, Australia
| | - Andrew M Scott
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia; Olivia Newton-John Cancer Research Institute, and La Trobe University, Heidelberg, Australia.
| |
Collapse
|
35
|
Fu M, Zhang N, Huang Z, Zhou C, Zhang X, Yuan J, He Q, Yang Y, Zheng H, Liang D, Wu FX, Fan W, Hu Z. OIF-Net: An Optical Flow Registration-Based PET/MR Cross-Modal Interactive Fusion Network for Low-Count Brain PET Image Denoising. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:1554-1567. [PMID: 38096101 DOI: 10.1109/tmi.2023.3342809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The short frames of low-count positron emission tomography (PET) images generally cause high levels of statistical noise. Thus, improving the quality of low-count images by using image postprocessing algorithms to achieve better clinical diagnoses has attracted widespread attention in the medical imaging community. Most existing deep learning-based low-count PET image enhancement methods have achieved satisfying results, however, few of them focus on denoising low-count PET images with the magnetic resonance (MR) image modality as guidance. The prior context features contained in MR images can provide abundant and complementary information for single low-count PET image denoising, especially in ultralow-count (2.5%) cases. To this end, we propose a novel two-stream dual PET/MR cross-modal interactive fusion network with an optical flow pre-alignment module, namely, OIF-Net. Specifically, the learnable optical flow registration module enables the spatial manipulation of MR imaging inputs within the network without any extra training supervision. Registered MR images fundamentally solve the problem of feature misalignment in the multimodal fusion stage, which greatly benefits the subsequent denoising process. In addition, we design a spatial-channel feature enhancement module (SC-FEM) that considers the interactive impacts of multiple modalities and provides additional information flexibility in both the spatial and channel dimensions. Furthermore, instead of simply concatenating two extracted features from these two modalities as an intermediate fusion method, the proposed cross-modal feature fusion module (CM-FFM) adopts cross-attention at multiple feature levels and greatly improves the two modalities' feature fusion procedure. Extensive experimental assessments conducted on real clinical datasets, as well as an independent clinical testing dataset, demonstrate that the proposed OIF-Net outperforms the state-of-the-art methods.
Collapse
|
36
|
Shi X, Liu T, Pei P, Shen W, Hu L, Zhu R, Wang F, Chen C, Yang K. Radionuclide-Labeled Antisilencing Function 1a Inhibitory Peptides for Tumor Identification and Individualized Therapy. ACS NANO 2024; 18:9114-9127. [PMID: 38477305 DOI: 10.1021/acsnano.4c00081] [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: 03/14/2024]
Abstract
Immune checkpoint blockade (ICB) therapy is promising to revolutionize cancer regimens, but the low response rate and the lack of a suitable patient stratification method have impeded universal profit to cancer patients. Noninvasive positron emission tomography (PET) imaging in the whole body, upon coupling with specific biomarkers closely related to the immune response, could provide spatiotemporal information to prescribe cancer therapy. Herein, we demonstrate that antisilencing function 1a (ASF1a) could serve as a biomarker target to delineate tumor immune microenvironments by immune PET (iPET). The iPET radiotracer (68Ga-AP1) is designed to target ASF1a in tumors and predict immune response, and the signal intensity predicts anti-PD-1 (αPD-1) therapy response in a negative correlation manner. The ICB-resistant tumors with a high level of ASF1a as revealed by iPET (ASF1aHigh-iPET) are prescribed to be treated by either the combined 177Lu-labeled AP1 and αPD-1 or the standalone α particle-emitting 225Ac-labeled AP1, both achieving enhanced therapeutic efficacy and prolonged survival time. Our study not only replenishes the iPET arsenal for immune-related response evaluation by designing a reliable biomarker and a facile radiotracer but also provides optional therapeutic strategies for ICB-resistant tumors with versatile radionuclide-labeled AP1 peptides, which is promising for real-time clinical diagnosis and individualized therapy planning simultaneously.
Collapse
Affiliation(s)
- Xiumin Shi
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Teng Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pei Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wenhao Shen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Ran Zhu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
37
|
Ciavattone NG, Guan N, Farfel A, Stauff J, Desmond T, Viglianti BL, Scott PJ, Brooks AF, Luker GD. Evaluating immunotherapeutic outcomes in triple-negative breast cancer with a cholesterol radiotracer in mice. JCI Insight 2024; 9:e175320. [PMID: 38502228 PMCID: PMC11141879 DOI: 10.1172/jci.insight.175320] [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: 08/31/2023] [Accepted: 03/13/2024] [Indexed: 03/21/2024] Open
Abstract
Evaluating the response to immune checkpoint inhibitors (ICIs) remains an unmet challenge in triple-negative breast cancer (TNBC). The requirement for cholesterol in the activation and function of T cells led us to hypothesize that quantifying cellular accumulation of this molecule could distinguish successful from ineffective checkpoint immunotherapy. To analyze accumulation of cholesterol by T cells in the immune microenvironment of breast cancer, we leveraged the PET radiotracer, eFNP-59. eFNP-59 is an analog of cholesterol that our group validated as an imaging biomarker for cholesterol uptake in preclinical models and initial human studies. In immunocompetent mouse models of TNBC, we found that elevated uptake of exogenous labeled cholesterol analogs functions as a marker for T cell activation. When comparing ICI-responsive and -nonresponsive tumors directly, uptake of fluorescent cholesterol and eFNP-59 increased in T cells from ICI-responsive tumors. We discovered that accumulation of cholesterol by T cells increased in ICI-responding tumors that received anti-PD-1 checkpoint immunotherapy. In patients with TNBC, tumors containing cycling T cells had features of cholesterol uptake and trafficking within those populations. These results suggest that uptake of exogenous cholesterol analogs by tumor-infiltrating T cells allows detection of T cell activation and has potential to assess the success of ICI therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Gary D Luker
- Department of Radiology, and
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
38
|
Salehjahromi M, Karpinets TV, Sujit SJ, Qayati M, Chen P, Aminu M, Saad MB, Bandyopadhyay R, Hong L, Sheshadri A, Lin J, Antonoff MB, Sepesi B, Ostrin EJ, Toumazis I, Huang P, Cheng C, Cascone T, Vokes NI, Behrens C, Siewerdsen JH, Hazle JD, Chang JY, Zhang J, Lu Y, Godoy MCB, Chung C, Jaffray D, Wistuba I, Lee JJ, Vaporciyan AA, Gibbons DL, Gladish G, Heymach JV, Wu CC, Zhang J, Wu J. Synthetic PET from CT improves diagnosis and prognosis for lung cancer: Proof of concept. Cell Rep Med 2024; 5:101463. [PMID: 38471502 PMCID: PMC10983039 DOI: 10.1016/j.xcrm.2024.101463] [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: 02/01/2023] [Revised: 09/07/2023] [Accepted: 02/15/2024] [Indexed: 03/14/2024]
Abstract
[18F]Fluorodeoxyglucose positron emission tomography (FDG-PET) and computed tomography (CT) are indispensable components in modern medicine. Although PET can provide additional diagnostic value, it is costly and not universally accessible, particularly in low-income countries. To bridge this gap, we have developed a conditional generative adversarial network pipeline that can produce FDG-PET from diagnostic CT scans based on multi-center multi-modal lung cancer datasets (n = 1,478). Synthetic PET images are validated across imaging, biological, and clinical aspects. Radiologists confirm comparable imaging quality and tumor contrast between synthetic and actual PET scans. Radiogenomics analysis further proves that the dysregulated cancer hallmark pathways of synthetic PET are consistent with actual PET. We also demonstrate the clinical values of synthetic PET in improving lung cancer diagnosis, staging, risk prediction, and prognosis. Taken together, this proof-of-concept study testifies to the feasibility of applying deep learning to obtain high-fidelity PET translated from CT.
Collapse
Affiliation(s)
| | | | - Sheeba J Sujit
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | - Mohamed Qayati
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | - Pingjun Chen
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | - Muhammad Aminu
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | - Maliazurina B Saad
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Lingzhi Hong
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, MD Anderson Cancer Center, Houston, TX USA
| | - Julie Lin
- Department of Pulmonary Medicine, MD Anderson Cancer Center, Houston, TX USA
| | - Mara B Antonoff
- Department of Thoracic and Cardiovascular Surgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin J Ostrin
- Department of General Internal Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Iakovos Toumazis
- Department of Health Services Research, MD Anderson Cancer Center, Houston, TX, USA
| | - Peng Huang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Chao Cheng
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Tina Cascone
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Natalie I Vokes
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey H Siewerdsen
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA; Institute for Data Science in Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - John D Hazle
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
| | - Joe Y Chang
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Lu
- Department of Nuclear Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Myrna C B Godoy
- Department of Thoracic Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA; Institute for Data Science in Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - David Jaffray
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA; Institute for Data Science in Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - J Jack Lee
- Department of Biostatistics, MD Anderson Cancer Center, Houston, TX, USA
| | - Ara A Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory Gladish
- Department of Thoracic Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Carol C Wu
- Department of Thoracic Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Zhang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA; Lung Cancer Genomics Program, MD Anderson Cancer Center, Houston, TX, USA; Lung Cancer Interception Program, MD Anderson Cancer Center, Houston, TX, USA
| | - Jia Wu
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA; Institute for Data Science in Oncology, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
39
|
Khessib T, Jha P, Davidzon GA, Iagaru A, Shah J. Nuclear Medicine and Molecular Imaging Applications in Gynecologic Malignancies: A Comprehensive Review. Semin Nucl Med 2024; 54:270-292. [PMID: 38342655 DOI: 10.1053/j.semnuclmed.2024.01.003] [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: 10/09/2023] [Revised: 01/16/2024] [Accepted: 01/16/2024] [Indexed: 02/13/2024]
Abstract
Gynecologic malignancies, consisting of endometrial, cervical, ovarian, vulvar, and vaginal cancers, pose significant diagnostic and management challenges due to their complex anatomic location and potential for rapid progression. These tumors cause substantial morbidity and mortality, often because of their delayed diagnosis and treatment. An estimated 19% of newly diagnosed cancers among women are gynecologic in origin. In recent years, there has been growing evidence supporting the integration of nuclear medicine imaging modalities in the diagnostic work-up and management of gynecologic cancers. The sensitivity of fluorine-18 fluorodeoxyglucose positron emission tomography (18F-FDG PET) combined with the anatomical specificity of computed tomography (CT) and magnetic resonance imaging (MRI) allows for the hybrid evaluation of metabolic activity and structural abnormalities that has become an indispensable tool in oncologic imaging. Lymphoscintigraphy, using technetium 99m (99mTc) based radiotracers along with single photon emission computed tomography/ computed tomography (SPECT/CT), holds a vital role in the identification of sentinel lymph nodes to minimize the surgical morbidity from extensive lymph node dissections. While not yet standard for gynecologic malignancies, promising therapeutic nuclear medicine agents serve as specialized treatment options for patients with advanced or recurrent disease. This article aims to provide a comprehensive review on the nuclear medicine applications in gynecologic malignancies through the following objectives: 1) To describe the role of nuclear medicine in the initial staging, lymph node mapping, response assessment, and recurrence/surveillance imaging of common gynecologic cancers, 2) To review the limitations of 18F-FDG PET/CT and promising applications of 18F-FDG PET/MRI in gynecologic malignancy, 3) To underscore the promising theragnostic applications of nuclear medicine, 4) To highlight the current role of nuclear medicine imaging in gynecologic cancers as per the National Comprehensive Cancer Network (NCCN), European Society of Surgical Oncology (ESGO), and European Society of Medical Oncology (ESMO) guidelines.
Collapse
Affiliation(s)
- Tasnim Khessib
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford Health Care; 300 Pasteur Drive, Palo Alto, CA 94305
| | - Priyanka Jha
- Division of Body Imaging, Department of Radiology, Stanford Health Care; 300 Pasteur Drive, Palo Alto, CA 94035
| | - Guido A Davidzon
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford Health Care; 300 Pasteur Drive, Palo Alto, CA 94305
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford Health Care; 300 Pasteur Drive, Palo Alto, CA 94305
| | - Jagruti Shah
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford Health Care; 300 Pasteur Drive, Palo Alto, CA 94305.
| |
Collapse
|
40
|
Nantogma S, de Maissin H, Adelabu I, Abdurraheem A, Nelson C, Chukanov NV, Salnikov OG, Koptyug IV, Lehmkuhl S, Schmidt AB, Appelt S, Theis T, Chekmenev EY. Carbon-13 Radiofrequency Amplification by Stimulated Emission of Radiation of the Hyperpolarized Ketone and Hemiketal Forms of Allyl [1- 13C]Pyruvate. ACS Sens 2024; 9:770-780. [PMID: 38198709 PMCID: PMC10922715 DOI: 10.1021/acssensors.3c02075] [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: 01/12/2024]
Abstract
13C hyperpolarized pyruvate is an emerging MRI contrast agent for sensing molecular events in cancer and other diseases with aberrant metabolic pathways. This metabolic contrast agent can be produced via several hyperpolarization techniques. Despite remarkable success in research settings, widespread clinical adoption faces substantial roadblocks because the current sensing technology utilized to sense this contrast agent requires the excitation of 13C nuclear spins that also need to be synchronized with MRI field gradient pulses. Here, we demonstrate sensing of hyperpolarized allyl [1-13C]pyruvate via the stimulated emission of radiation that mitigates the requirements currently blocking broader adoption. Specifically, 13C Radiofrequency Amplification by Stimulated Emission of Radiation (13C RASER) was obtained after pairwise addition of parahydrogen to a pyruvate precursor, detected in a commercial inductive detector with a quality factor (Q) of 32 for sample concentrations as low as 0.125 M with 13C polarization of 4%. Moreover, parahydrogen-induced polarization allowed for the preparation of a mixture of ketone and hemiketal forms of hyperpolarized allyl [1-13C]pyruvate, which are separated by 10 ppm in 13C NMR spectra. This is a good model system to study the simultaneous 13C RASER signals of multiple 13C species. This system models the metabolic production of hyperpolarized [1-13C]lactate from hyperpolarized [1-13C]pyruvate, which has a similar chemical shift difference. Our results show that 13C RASER signals can be obtained from both species simultaneously when the emission threshold is exceeded for both species. On the other hand, when the emission threshold is exceeded only for one of the hyperpolarized species, 13C stimulated emission is confined to this species only, therefore enabling the background-free detection of individual hyperpolarized 13C signals. The reported results pave the way to novel sensing approaches of 13C hyperpolarized pyruvate, potentially unlocking hyperpolarized 13C MRI on virtually any MRI system─an attractive vision for the future molecular imaging and diagnostics.
Collapse
Affiliation(s)
- Shiraz Nantogma
- Department of Chemistry, Integrative Bio-Sciences (IBIO), Karmanos Cancer Institute (KCI), Wayne State University, Detroit, Michigan 48202, United States
| | - Henri de Maissin
- Division of Medical Physics, Department of Radiology, Medical Center, University of Freiburg, Freiburg 79106, Germany
- Faculty of Medicine, University of Freiburg, Killianstr. 5a, Freiburg 79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Isaiah Adelabu
- Department of Chemistry, Integrative Bio-Sciences (IBIO), Karmanos Cancer Institute (KCI), Wayne State University, Detroit, Michigan 48202, United States
| | - Abubakar Abdurraheem
- Department of Chemistry, Integrative Bio-Sciences (IBIO), Karmanos Cancer Institute (KCI), Wayne State University, Detroit, Michigan 48202, United States
| | - Christopher Nelson
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | | | - Oleg G Salnikov
- International Tomography Center SB RAS, 630090 Novosibirsk, Russia
| | - Igor V Koptyug
- International Tomography Center SB RAS, 630090 Novosibirsk, Russia
- Boreskov Institute of Catalysis SB RAS, 630090 Novosibirsk, Russia
| | - Sören Lehmkuhl
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Karlsruhe 76344, Germany
| | - Andreas B Schmidt
- Department of Chemistry, Integrative Bio-Sciences (IBIO), Karmanos Cancer Institute (KCI), Wayne State University, Detroit, Michigan 48202, United States
- Division of Medical Physics, Department of Radiology, Medical Center, University of Freiburg, Freiburg 79106, Germany
- Faculty of Medicine, University of Freiburg, Killianstr. 5a, Freiburg 79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Stephan Appelt
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen 52056, Germany
- Central Institute for Engineering, Electronics and Analytics - Electronic Systems (ZEA-2), Forschungszentrum Jülich GmbH, Jülich D-52425, Germany
| | - Thomas Theis
- Department of Physics, North Carolina State University, Raleigh, North Carolina 27606, United States
- Joint UNC & NC State Department of Biomedical Engineering, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Eduard Y Chekmenev
- Department of Chemistry, Integrative Bio-Sciences (IBIO), Karmanos Cancer Institute (KCI), Wayne State University, Detroit, Michigan 48202, United States
- Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
41
|
Wang W, Zhen S, Ping Y, Wang L, Zhang Y. Metabolomic biomarkers in liquid biopsy: accurate cancer diagnosis and prognosis monitoring. Front Oncol 2024; 14:1331215. [PMID: 38384814 PMCID: PMC10879439 DOI: 10.3389/fonc.2024.1331215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/26/2024] [Indexed: 02/23/2024] Open
Abstract
Liquid biopsy, a novel detection method, has recently become an active research area in clinical cancer owing to its unique advantages. Studies on circulating free DNA, circulating tumor cells, and exosomes obtained by liquid biopsy have shown great advances and they have entered clinical practice as new cancer biomarkers. The metabolism of the body is dynamic as cancer originates and progresses. Metabolic abnormalities caused by cancer can be detected in the blood, sputum, urine, and other biological fluids via systemic or local circulation. A considerable number of recent studies have focused on the roles of metabolic molecules in cancer. The purpose of this review is to provide an overview of metabolic markers from various biological fluids in the latest clinical studies, which may contribute to cancer screening and diagnosis, differentiation of cancer typing, grading and staging, and prediction of therapeutic response and prognosis.
Collapse
Affiliation(s)
- Wenqian Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province, Zhengzhou, Henan, China
| | - Shanshan Zhen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province, Zhengzhou, Henan, China
| | - Yu Ping
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province, Zhengzhou, Henan, China
| | - Liping Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
42
|
Xu M, Gu B, Zhang J, Xu X, Qiao Y, Hu S, Song S. Differentiation of cancer of unknown primary and lymphoma in head and neck metastatic poorly differentiated cancer using 18 F-FDG PET/CT tumor metabolic heterogeneity index. Nucl Med Commun 2024; 45:148-154. [PMID: 38095143 DOI: 10.1097/mnm.0000000000001797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
OBJECTIVE To explore the value of 18 F-FDG PET/CT tumor metabolic heterogeneity index (HI) and establish and validate a nomogram model for distinguishing head and neck cancer of unknown primary (HNCUP) from lymphoma with head and neck metastatic poorly differentiated cancer. METHODS This retrospective analysis was conducted on 1242 patients with cervical metastatic poorly differentiated cancer. 108 patients, who were clinically and pathologically confirmed as HNCUP or lymphoma, were finally enrolled. Two independent sample t-tests and χ 2 test were used to compare the clinical and imaging features. Binary logistic regression was used to screen for independent predictive factors. RESULTS Among the 108 patients), 65 patients were diagnosed with HNCUP and 43 were lymphoma. Gender ( P = 0.001), SUV max ( P < 0.001), SUV mean ( P < 0.001), TLG ( P = 0.012), and HI ( P < 0.001) had statistical significance in distinguishing HNCUP and lymphoma. Female ( OR = 4.546, P = 0.003) and patients with HI ≥ 2.37 ( OR = 3.461, P = 0.047) were more likely to be diagnosed as lymphoma. CONCLUSION For patients with cervical metastatic poorly differentiated cancer, gender and HI were independent predictors of pathological type. For such patients, clinical attention should be paid to avoid misdiagnosing lymphoma as HNCUP, which may delay treatment.
Collapse
Affiliation(s)
- Mingzhen Xu
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000)
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, China
| | - Bingxin Gu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, China
| | - Xiaoping Xu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, China
| | - Ying Qiao
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, China
| | - Silong Hu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, China
| | - Shaoli Song
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000)
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, China
| |
Collapse
|
43
|
Yang H, Chen S, Qi M, Chen W, Kong Q, Zhang J, Song S. Investigation of PET image quality with acquisition time/bed and enhancement of lesion quantification accuracy through deep progressive learning. EJNMMI Phys 2024; 11:7. [PMID: 38195785 PMCID: PMC10776545 DOI: 10.1186/s40658-023-00607-x] [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: 10/16/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024] Open
Abstract
OBJECTIVE To improve the PET image quality by a deep progressive learning (DPL) reconstruction algorithm and evaluate the DPL performance in lesion quantification. METHODS We reconstructed PET images from 48 oncological patients using ordered subset expectation maximization (OSEM) and deep progressive learning (DPL) methods. The patients were enrolled into three overlapped studies: 11 patients for image quality assessment (study 1), 34 patients for sub-centimeter lesion quantification (study 2), and 28 patients for imaging of overweight or obese individuals (study 3). In study 1, we evaluated the image quality visually based on four criteria: overall score, image sharpness, image noise, and diagnostic confidence. We also measured the image quality quantitatively using the signal-to-background ratio (SBR), signal-to-noise ratio (SNR), contrast-to-background ratio (CBR), and contrast-to-noise ratio (CNR). To evaluate the performance of the DPL algorithm in quantifying lesions, we compared the maximum standardized uptake values (SUVmax), SBR, CBR, SNR and CNR of 63 sub-centimeter lesions in study 2 and 44 lesions in study 3. RESULTS DPL produced better PET image quality than OSEM did based on the visual evaluation methods when the acquisition time was 0.5, 1.0 and 1.5 min/bed. However, no discernible differences were found between the two methods when the acquisition time was 2.0, 2.5 and 3.0 min/bed. Quantitative results showed that DPL had significantly higher values of SBR, CBR, SNR, and CNR than OSEM did for each acquisition time. For sub-centimeter lesion quantification, the SUVmax, SBR, CBR, SNR, and CNR of DPL were significantly enhanced, compared with OSEM. Similarly, for lesion quantification in overweight and obese patients, DPL significantly increased these parameters compared with OSEM. CONCLUSION The DPL algorithm dramatically enhanced the quality of PET images and enabled more accurate quantification of sub-centimeters lesions in patients and lesions in overweight or obese patients. This is particularly beneficial for overweight or obese patients who usually have lower image quality due to the increased attenuation.
Collapse
Affiliation(s)
- Hongxing Yang
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, No. 220, Handan Road, Yangpu District, Shanghai, 200433, People's Republic of China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 130, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
- Center for Biomedical Imaging, Fudan University, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Engineering Research Center for Molecular Imaging Probes, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
- Institute of Modern Physics, Fudan University, No. 220, Handan Road, Yangpu District, Shanghai, 200433, People's Republic of China
| | - Shihao Chen
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, No. 220, Handan Road, Yangpu District, Shanghai, 200433, People's Republic of China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 130, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
- Center for Biomedical Imaging, Fudan University, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Engineering Research Center for Molecular Imaging Probes, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Ming Qi
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, No. 220, Handan Road, Yangpu District, Shanghai, 200433, People's Republic of China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 130, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
- Center for Biomedical Imaging, Fudan University, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Engineering Research Center for Molecular Imaging Probes, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Wen Chen
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, No. 220, Handan Road, Yangpu District, Shanghai, 200433, People's Republic of China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 130, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
- Center for Biomedical Imaging, Fudan University, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Engineering Research Center for Molecular Imaging Probes, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Qing Kong
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, No. 220, Handan Road, Yangpu District, Shanghai, 200433, People's Republic of China
- Shanghai Engineering Research Center for Molecular Imaging Probes, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 130, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China.
- Center for Biomedical Imaging, Fudan University, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China.
- Shanghai Engineering Research Center for Molecular Imaging Probes, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China.
- Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, No. 220, Handan Road, Yangpu District, Shanghai, 200032, People's Republic of China.
| | - Shaoli Song
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, No. 220, Handan Road, Yangpu District, Shanghai, 200433, People's Republic of China.
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 130, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China.
- Center for Biomedical Imaging, Fudan University, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China.
- Shanghai Engineering Research Center for Molecular Imaging Probes, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
44
|
Schmitz-Peiffer F, Lukas M, Mohan AM, Albrecht J, Aschenbach JR, Brenner W, Beindorff N. Effects of isoflurane anaesthesia depth and duration on renal function measured with [ 99mTc]Tc-mercaptoacetyltriglycine SPECT in mice. EJNMMI Res 2024; 14:4. [PMID: 38180547 PMCID: PMC10769950 DOI: 10.1186/s13550-023-01065-3] [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: 10/23/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND The influence of anaesthetic depth and the potential influence of different anaesthetic beds and thus different handling procedures were investigated in 86 severe combined immunodeficient (SCID) mice using semi-stationary dynamic single photon emission computed tomography (SPECT) for kidney scintigraphy. Therefore, isoflurane concentrations were adjusted using respiratory rate for low (80-90 breath/min) and deep anaesthesia (40-45 breath/min). At low anaesthesia, we additionally tested the influence of single bed versus 3-mouse bed hotel; the hotel mice were anaesthetized consecutively at ~ 30, 20, and 10 min before tracer injections for positions 1, 2, and 3, respectively. Intravenous [99mTc]Tc-MAG3 injection of ~ 28 MBq was performed after SPECT start. Time-activity curves were used to calculate time-to-peak (Tmax), T50 (50% clearance) and T25 (75% clearance). RESULTS Low and deep anaesthesia corresponded to median isoflurane concentrations of 1.3% and 1.5%, respectively, with no significant differences in heart rate (p = 0.74). Low anaesthesia resulted in shorter aortic blood clearance half-life (p = 0.091) and increased relative renal tracer influx rate (p = 0.018). A tendency toward earlier Tmax occurred under low anaesthesia (p = 0.063) with no differences in T50 (p = 0.40) and T25 (p = 0.24). Variance increased with deep anaesthesia. Compared to single mouse scans, hotel mice in position 1 showed a delayed Tmax, T50, and T25 (p < 0.05 each). Furthermore, hotel mice in position 1 showed delayed Tmax versus position 3, and delayed T50 and T25 versus position 2 and 3 (p < 0.05 each). No difference occurred between single bed and positions 2 (p = 1.0) and 3 (p = 1.0). CONCLUSIONS Deep anaesthesia and prolonged low anaesthesia should be avoided during renal scintigraphy because they result in prolonged blood clearance half-life, delayed renal influx and/or later Tmax. Vice versa, low anaesthesia with high respiratory rates of 80-90 rpm and short duration (≤ 20 min) should be preferred to obtain representative data with low variance.
Collapse
Affiliation(s)
- Fabian Schmitz-Peiffer
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Mathias Lukas
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ajay-Mohan Mohan
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Jakob Albrecht
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Radiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg R Aschenbach
- Institute of Veterinary Physiology, School of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Winfried Brenner
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
45
|
Wang D, Jiang C, He J, Teng Y, Qin H, Liu J, Yang X. M 3S-Net: multi-modality multi-branch multi-self-attention network with structure-promoting loss for low-dose PET/CT enhancement. Phys Med Biol 2024; 69:025001. [PMID: 38086073 DOI: 10.1088/1361-6560/ad14c5] [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: 09/17/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024]
Abstract
Objective.PET (Positron Emission Tomography) inherently involves radiotracer injections and long scanning time, which raises concerns about the risk of radiation exposure and patient comfort. Reductions in radiotracer dosage and acquisition time can lower the potential risk and improve patient comfort, respectively, but both will also reduce photon counts and hence degrade the image quality. Therefore, it is of interest to improve the quality of low-dose PET images.Approach.A supervised multi-modality deep learning model, named M3S-Net, was proposed to generate standard-dose PET images (60 s per bed position) from low-dose ones (10 s per bed position) and the corresponding CT images. Specifically, we designed a multi-branch convolutional neural network with multi-self-attention mechanisms, which first extracted features from PET and CT images in two separate branches and then fused the features to generate the final generated PET images. Moreover, a novel multi-modality structure-promoting term was proposed in the loss function to learn the anatomical information contained in CT images.Main results.We conducted extensive numerical experiments on real clinical data collected from local hospitals. Compared with state-of-the-art methods, the proposed M3S-Net not only achieved higher objective metrics and better generated tumors, but also performed better in preserving edges and suppressing noise and artifacts.Significance.The experimental results of quantitative metrics and qualitative displays demonstrate that the proposed M3S-Net can generate high-quality PET images from low-dose ones, which are competable to standard-dose PET images. This is valuable in reducing PET acquisition time and has potential applications in dynamic PET imaging.
Collapse
Affiliation(s)
- Dong Wang
- School of Mathematics/S.T.Yau Center of Southeast University, Southeast University, 210096, People's Republic of China
- Nanjing Center of Applied Mathematics, Nanjing, 211135, People's Republic of China
| | - Chong Jiang
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Yue Teng
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Hourong Qin
- Department of Mathematics, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Jijun Liu
- School of Mathematics/S.T.Yau Center of Southeast University, Southeast University, 210096, People's Republic of China
- Nanjing Center of Applied Mathematics, Nanjing, 211135, People's Republic of China
| | - Xiaoping Yang
- Department of Mathematics, Nanjing University, Nanjing, 210093, People's Republic of China
| |
Collapse
|
46
|
Sagara H, Inoue K, Yaku H, Ohsawa A, Mano C, Morita T, Hiyama T, Muramatsu Y, Inaki A, Fujii H. A new simpler image quality index based on body size for FDG-PET/CT. Nucl Med Commun 2024; 45:93-101. [PMID: 37901919 DOI: 10.1097/mnm.0000000000001787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Noise equivalent count density (NEC density ) is often used to evaluate the image quality of whole-body fluorodeoxyglucose tomography tests. However, this index is calculated using the patient volume, which is difficult to obtain at every facility. In this study, we proposed new image quality indices that can be evaluated at all facilities. In total, 94 patients were enrolled in the study. The correlations of patients' body weight and BMI with volume were examined. New image quality indices normalized by body weight and BMI were defined as NEC bw and NEC bmi , respectively. Correlations between NEC bw , NEC bmi , and NEC density were examined. Further, the correlations between these two new indices and visual scores were evaluated. Good correlations were observed between volume and body weight (r = 0.861, P < 0.001) and between volume and BMI (r = 0.728, P < 0.001). NEC bw and NEC bmi correlated well with NEC density (r = 0.954 for NEC bw and r = 0.897 for NEC bmi , P < 0.001). These correlations improved when the examined bed positions were set to the same number. Additionally, the correlations of visual scores with NEC bw and NEC bmi were similar to those between the visual score and NEC density . Our investigation indicated that the newly proposed image quality metrics, NEC bw and NEC bmi , were easily calculated and as useful as NEC density for evaluating image quality when subjects had similar physiques.
Collapse
Affiliation(s)
- Hiroaki Sagara
- Division of Functional Imaging, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center,
- Department of Radiologic Technology, National Cancer Center Hospital East, Kashiwa,
- Department of Radiological Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo,
| | - Kazumasa Inoue
- Department of Radiological Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo,
| | - Hideki Yaku
- RYUKYU ISG Co., Ltd, Kyoto,
- Optical Information Engineering, Systems Innovation Engineering, Graduate School of Advanced Technology and Science, Tokushima University 2-1 Minamijyousanjima-cho, Tokushima and
| | - Amon Ohsawa
- Department of Radiologic Technology, National Cancer Center Hospital East, Kashiwa,
| | - Chikara Mano
- Department of Radiologic Technology, National Cancer Center Hospital East, Kashiwa,
| | - Takahiro Morita
- Department of Diagnostic Radiology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takashi Hiyama
- Department of Diagnostic Radiology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshihisa Muramatsu
- Department of Radiologic Technology, National Cancer Center Hospital East, Kashiwa,
| | - Anri Inaki
- Division of Functional Imaging, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center,
| | - Hirofumi Fujii
- Division of Functional Imaging, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center,
| |
Collapse
|
47
|
Mirshahvalad SA, Kohan A, Metser U, Hinzpeter R, Ortega C, Farag A, Veit-Haibach P. Diagnostic performance of whole-body [ 18F]FDG PET/MR in cancer M staging: A systematic review and meta-analysis. Eur Radiol 2024; 34:673-685. [PMID: 37535156 DOI: 10.1007/s00330-023-10009-3] [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: 01/17/2023] [Revised: 05/14/2023] [Accepted: 06/07/2023] [Indexed: 08/04/2023]
Abstract
OBJECTIVES To calculate the pooled diagnostic performances of whole-body [18F]FDG PET/MR in M staging of [18F]FDG-avid cancer entities. METHODS A diagnostic meta-analysis was conducted on the [18F]FDG PET/MR in M staging, including studies: (1) evaluated [18F]FDG PET/MR in detecting distant metastasis; (2) compared[ 18F]FDG PET/MR with histopathology, follow-up, or asynchronous multimodality imaging as the reference standard; (3) provided data for the whole-body evaluation; (4) provided adequate data to calculate the meta-analytic performances. Pooled performances were calculated with their confidence interval. In addition, forest plots, SROC curves, and likelihood ratio scatterplots were drawn. All analyses were performed using STATA 16. RESULTS From 52 eligible studies, 2289 patients and 2072 metastases were entered in the meta-analysis. The whole-body pooled sensitivities were 0.95 (95%CI: 0.91-0.97) and 0.97 (95%CI: 0.91-0.99) at the patient and lesion levels, respectively. The pooled specificities were 0.99 (95%CI: 0.97-1.00) and 0.97 (95%CI: 0.90-0.99), respectively. Additionally, subgroup analyses were performed. The calculated pooled sensitivities for lung, gastrointestinal, breast, and gynecological cancers were 0.90, 0.93, 1.00, and 0.97, respectively. The pooled specificities were 1.00, 0.98, 0.97, and 1.00, respectively. Furthermore, the pooled sensitivities for non-small cell lung, colorectal, and cervical cancers were 0.92, 0.96, and 0.86, respectively. The pooled specificities were 1.00, 0.95, and 1.00, respectively. CONCLUSION [18F]FDG PET/MR was a highly accurate modality in M staging in the reported [18F]FDG-avid malignancies. The results showed high sensitivity and specificity in each reviewed malignancy type. Thus, our findings may help clinicians and patients to be confident about the performance of [18F]FDG PET/MR in the clinic. CLINICAL RELEVANCE STATEMENT Although [18F]FDG PET/MR is not a routine imaging technique in current guidelines, mostly due to its availability and logistic issues, our findings might add to the limited evidence regarding its performance, showing a sensitivity of 0.95 and specificity of 0.97. KEY POINTS • The whole-body [18F]FDG PET/MR showed high accuracy in detecting distant metastases at both patient and lesion levels. • The pooled sensitivities were 95% and 97% and pooled specificities were 99% and 97% at patient and lesion levels, respectively. • The results suggested that 18F-FDG PET/MR was a strong modality in the exclusion and confirmation of distant metastases.
Collapse
Affiliation(s)
- Seyed Ali Mirshahvalad
- Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, Toronto General Hospital, 585 University Avenue, Toronto, Ontario, M5G 2N2, Canada.
| | - Andres Kohan
- Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, Toronto General Hospital, 585 University Avenue, Toronto, Ontario, M5G 2N2, Canada
| | - Ur Metser
- Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, Toronto General Hospital, 585 University Avenue, Toronto, Ontario, M5G 2N2, Canada
| | - Ricarda Hinzpeter
- Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, Toronto General Hospital, 585 University Avenue, Toronto, Ontario, M5G 2N2, Canada
| | - Claudia Ortega
- Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, Toronto General Hospital, 585 University Avenue, Toronto, Ontario, M5G 2N2, Canada
| | - Adam Farag
- Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, Toronto General Hospital, 585 University Avenue, Toronto, Ontario, M5G 2N2, Canada
| | - Patrick Veit-Haibach
- Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, Toronto General Hospital, 585 University Avenue, Toronto, Ontario, M5G 2N2, Canada
| |
Collapse
|
48
|
Cetani F, Pardi E, Torregrossa L, Borsari S, Pierotti L, Dinoi E, Marcocci C. Approach to the Patient With Parathyroid Carcinoma. J Clin Endocrinol Metab 2023; 109:256-268. [PMID: 37531615 DOI: 10.1210/clinem/dgad455] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 06/27/2023] [Accepted: 07/31/2023] [Indexed: 08/04/2023]
Abstract
Parathyroid carcinoma (PC) is usually associated with severe symptomatic primary hyperparathyroidism (PHPT) and accounts for less than 1% of all cases of PHPT and approximately 0.005% of all cancers. PC most commonly occurs as a sporadic disease and somatic CDC73 mutations can be detected in up to 80% of cases. Approximately 30% of patients harbor a germline mutation of the CDC73 gene. Preoperative diagnosis of PC is difficult because no disease-specific markers are available, and PC should be suspected in patients with severe hypercalcemia and end-organ complications. The diagnosis is based on the evidence of invasive tumor growth at histology and/or metastases. En bloc resection of the tumor, together with the ipsilateral thyroid lobe and adjacent structures, should be performed by an experienced surgeon when PC is suspected. This surgical approach reduces the risk of recurrence and metastasis and offers the highest chance of cure. Nonetheless, PC has a recurrence rate of 40% to 60% and, if feasible, multiple surgical procedures should be performed. When surgery is no longer an option, medical treatment is aimed to reduce hypercalcemia and target organ complications. Targeted agents have been effectively used in a few cases. We describe herein a patient with severe PHPT due to PC and provide a systematic diagnostic and treatment approach. A thorough review of the medical history, a typical clinical and biochemical phenotype and, in some cases, the revision of the histological examination provide the clues for the diagnosis of PC.
Collapse
Affiliation(s)
- Filomena Cetani
- Unit of Endocrinology, University Hospital of Pisa, 56124 Pisa, Italy
| | - Elena Pardi
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy
| | - Liborio Torregrossa
- Department of Surgical, Medical, Molecular Pathology and Clinical Area, University of Pisa, 56126 Pisa, Italy
| | - Simona Borsari
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy
| | - Laura Pierotti
- Unit of Endocrinology, University Hospital of Pisa, 56124 Pisa, Italy
| | - Elisa Dinoi
- Unit of Endocrinology, University Hospital of Pisa, 56124 Pisa, Italy
| | - Claudio Marcocci
- Unit of Endocrinology, University Hospital of Pisa, 56124 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy
| |
Collapse
|
49
|
Huang Z, Li W, Wu Y, Guo N, Yang L, Zhang N, Pang Z, Yang Y, Zhou Y, Shang Y, Zheng H, Liang D, Wang M, Hu Z. Short-axis PET image quality improvement based on a uEXPLORER total-body PET system through deep learning. Eur J Nucl Med Mol Imaging 2023; 51:27-39. [PMID: 37672046 DOI: 10.1007/s00259-023-06422-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 08/30/2023] [Indexed: 09/07/2023]
Abstract
PURPOSE The axial field of view (AFOV) of a positron emission tomography (PET) scanner greatly affects the quality of PET images. Although a total-body PET scanner (uEXPLORER) with a large AFOV is more sensitive, it is more expensive and difficult to widely use. Therefore, we attempt to utilize high-quality images generated by uEXPLORER to optimize the quality of images from short-axis PET scanners through deep learning technology while controlling costs. METHODS The experiments were conducted using PET images of three anatomical locations (brain, lung, and abdomen) from 335 patients. To simulate PET images from different axes, two protocols were used to obtain PET image pairs (each patient was scanned once). For low-quality PET (LQ-PET) images with a 320-mm AFOV, we applied a 300-mm FOV for brain reconstruction and a 500-mm FOV for lung and abdomen reconstruction. For high-quality PET (HQ-PET) images, we applied a 1940-mm AFOV during the reconstruction process. A 3D Unet was utilized to learn the mapping relationship between LQ-PET and HQ-PET images. In addition, the peak signal-to-noise ratio (PSNR) and structural similarity index measure (SSIM) were employed to evaluate the model performance. Furthermore, two nuclear medicine doctors evaluated the image quality based on clinical readings. RESULTS The generated PET images of the brain, lung, and abdomen were quantitatively and qualitatively compatible with the HQ-PET images. In particular, our method achieved PSNR values of 35.41 ± 5.45 dB (p < 0.05), 33.77 ± 6.18 dB (p < 0.05), and 38.58 ± 7.28 dB (p < 0.05) for the three beds. The overall mean SSIM was greater than 0.94 for all patients who underwent testing. Moreover, the total subjective quality levels of the generated PET images for three beds were 3.74 ± 0.74, 3.69 ± 0.81, and 3.42 ± 0.99 (the highest possible score was 5, and the minimum score was 1) from two experienced nuclear medicine experts. Additionally, we evaluated the distribution of quantitative standard uptake values (SUV) in the region of interest (ROI). Both the SUV distribution and the peaks of the profile show that our results are consistent with the HQ-PET images, proving the superiority of our approach. CONCLUSION The findings demonstrate the potential of the proposed technique for improving the image quality of a PET scanner with a 320 mm or even shorter AFOV. Furthermore, this study explored the potential of utilizing uEXPLORER to achieve improved short-axis PET image quality at a limited economic cost, and computer-aided diagnosis systems that are related can help patients and radiologists.
Collapse
Affiliation(s)
- Zhenxing Huang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wenbo Li
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yaping Wu
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Nannan Guo
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- School of Mathematics and Statistics, Henan University, Kaifeng, 475004, China
| | - Lin Yang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- School of Mathematics and Statistics, Henan University, Kaifeng, 475004, China
| | - Na Zhang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zhifeng Pang
- School of Mathematics and Statistics, Henan University, Kaifeng, 475004, China
| | - Yongfeng Yang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yun Zhou
- Central Research Institute, United Imaging Healthcare Group, Shanghai, 201807, China
| | - Yue Shang
- Performance Strategy & Analytics, UCLA Health, Los Angeles, CA, 90001, USA
| | - Hairong Zheng
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Dong Liang
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Meiyun Wang
- School of Mathematics and Statistics, Henan University, Kaifeng, 475004, China.
| | - Zhanli Hu
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
50
|
Tonda K, Iwabuchi Y, Shiga T, Owaki Y, Fujita A, Nakahara T, Sakurai R, Shimizu A, Yamada Y, Okada M, Jinzaki M. Impact of patient characteristic factors on the dynamics of liver glucose metabolism: Evaluation of multiparametric imaging with dynamic whole-body 18 F-fluorodeoxyglucose-positron emission tomography. Diabetes Obes Metab 2023; 25:3521-3528. [PMID: 37589247 DOI: 10.1111/dom.15247] [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: 06/11/2023] [Revised: 07/20/2023] [Accepted: 07/30/2023] [Indexed: 08/18/2023]
Abstract
AIMS To assess the impact of various patient characteristics on the dynamics of liver glucose metabolism using automated multiparametric imaging with whole-body dynamic 18 F-fluorodeoxyglucose (FDG)-positron emission tomography (PET). MATERIALS AND METHODS We retrospectively enrolled 540 patients who underwent whole-body dynamic FDG-PET. Three quantitative indices representing hepatic glucose metabolism [mean standardized uptake value normalized by lean body mass (SULmean), metabolic glucose rate (kinetic index) and distribution volume (DV)] were measured from multiparametric PET images produced automatically based on the Patlak plot model. Patient characteristics including age, sex, body mass index, fasting time, blood glucose level, and the presence of diabetes mellitus (DM) or hepatic steatosis (HS) were collected. We examined the correlations between the characteristic factors and three quantitative indices using multiple regression analysis. RESULTS The success rate of kinetic analysis using multiparametric PET imaging was 93.3% (504/540). Hepatic SULmean was significantly correlated with age (p < .001), sex (p < .001) and blood glucose level (p = .002). DV was significantly correlated with age (p = .033), sex (p < .001), body mass index (p = .002), fasting time (p = .043) and the presence of HS (p = .002). The kinetic index was significantly correlated with age (p < .001) and sex (p = .004). In the comparison of the healthy, DM and HS groups, patients with DM had a significantly increased SULmean, whereas patients with HS had a significantly decreased DV. CONCLUSIONS Our results showed that liver glucose metabolism was influenced by various patient characteristic factors. Multiparametric FDG-PET imaging can be used to analyse the kinetics of liver glucose metabolism in routine clinical practice.
Collapse
Affiliation(s)
- Kai Tonda
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Yu Iwabuchi
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Tohru Shiga
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, Fukushima, Japan
| | - Yoshiki Owaki
- Office of Radiation Technology, Keio University Hospital, Tokyo, Japan
| | - Arashi Fujita
- Office of Radiation Technology, Keio University Hospital, Tokyo, Japan
| | - Takehiro Nakahara
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Ryosuke Sakurai
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Atsushi Shimizu
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshitake Yamada
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Okada
- Department of Radiology, Nihon University School of Medicine, Tokyo, Japan
| | - Masahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|