1
|
Samim A, Suurd DPD, van Rooij R, van Noesel MM, Lam MGEH, Braat AJAT, Tolboom N, Borgwardt L, Tytgat GAM, de Keizer B. SUV normalisation and reference tissue selection for [ 1⁸F]mFBG PET-CT in paediatric and adult patients. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07242-x. [PMID: 40172693 DOI: 10.1007/s00259-025-07242-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025]
Abstract
PURPOSE Meta-[1⁸F]fluorobenzylguanidine ([1⁸F]mFBG) PET-CT is a novel imaging modality for norepinephrine transporter-expressing tumours, such as neuroblastoma and phaeochromocytoma, enabling quantitative assessment and improved diagnostic accuracy compared to meta-[123I]iodobenzylguanidine ([123I]mIBG) scintigraphy. This study aims to: 1) Identify the optimal standardised uptake value (SUV) normalisation method: body weight (BW) or lean body mass (LBM); 2) Determine the most stable reference tissue with SUV uptake below pathological levels. METHODS We analysed 63 [1⁸F]mFBG PET-CTs from 35 patients (20 paediatric neuroblastoma, 15 adult phaeochromocytoma). SUVmean was measured in the liver, blood pool, bone marrow, and muscle, normalised using BW (SUVBW), LBM via James (SUVLBMJames), and LBM via Janmahasatian (SUVLBMJanma). Variability of SUVs and their correlation with weight were assessed. RESULTS LBM-based normalisation reduced SUV variability compared to BW-based normalisation. Bone marrow demonstrated the lowest variability and least weight dependency (r2 0.45 for SUVBW versus 0.31 for SUVLBMJanma). The liver had the highest SUVs, increasing the risk of false negatives if used as reference tissue, while the blood pool had the lowest SUVs, raising the risk of false positives. Muscle showed relatively stable SUVs with increasing weight but higher variability than bone marrow. CONCLUSION LBM-based SUV normalisation reduces weight dependency for [1⁸F]mFBG PET-CT. Bone marrow is the most reliable reference tissue due to its low variability and balanced SUVs, while muscle may serve as an alternative if diffuse bone marrow uptake is present. These findings support standardising LBM-adjusted SUV methods and using bone marrow as the primary reference tissue to enhance diagnostic accuracy. CLINICAL TRIAL REGISTRATION EudraCT Number: 2019-003713-33; EU Clinical Trials Number: 2024-513622-35-00.
Collapse
Affiliation(s)
- Atia Samim
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Nuclear Medicine and Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Diederik P D Suurd
- Department of Nuclear Medicine and Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rob van Rooij
- Department of Nuclear Medicine and Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Nuclear Medicine and Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marnix G E H Lam
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Nuclear Medicine and Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Arthur J A T Braat
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Nuclear Medicine and Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nelleke Tolboom
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Nuclear Medicine and Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lise Borgwardt
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Godelieve A M Tytgat
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bart de Keizer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Department of Nuclear Medicine and Radiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
2
|
de Vries ISA, Lodema S, Braat AJAT, Merks JHM, van Rooij R, de Keizer B. Improving accuracy of SUV estimates in paediatric oncology: Recommending against the use of body weight corrected SUV in [ 18F]FDG PET. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07104-6. [PMID: 39920266 DOI: 10.1007/s00259-025-07104-6] [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] [Accepted: 01/20/2025] [Indexed: 02/09/2025]
Abstract
PURPOSE Few studies have assessed body weight dependency of Standardised Uptake Value (SUV) formulations in paediatric patients. This study aims to compare different SUV formulations measured in reference tissues in paediatric patients and determine which correction method shows the least dependency on body weight. METHODS A single-centre, retrospective analysis of [18F]FDG PET/CT scans was performed. SUV measurements were taken from liver and blood pool using EARL1 reconstructions. SUV measurements were corrected for body weight (SUVBW), lean body mass (LBM) according to James (SUVLBMJames) and Janmahasatian (SUVLBMJanma), and body surface area (BSA) according to DuBois (SUVBSADuBois) and Haycock (SUVBSAHaycock). The coefficient of determination (r2) was used to assess the correlation between SUV and body weight. RESULTS In total, 461 scans were analysed, including 185 (40%) from female patients. The median age of patients was 12 years (IQR 8-15.5 years). SUVBW exhibited the strongest correlation with body weight, with r2 = 0.65 for the liver and r2 = 0.50 for the blood pool. In contrast, SUVBSADuBois and SUVBSAHaycock had the weakest correlation, with r2 = 0.09 for the liver and r2 = 0.06 for the blood pool. SUVLBMJames and SUVLBMJanma had moderate correlations, with r2 = 0.51 and r2 = 0.44 for the liver and blood pool, respectively, and r2 = 0.47 and r2 = 0.42, respectively. CONCLUSION In paediatric [18F]FDG PET/CT scans, SUVBW should be avoided due to elevating values in heavier patients. SUVBSA presents the least dependency on body weight and provides the most consistent assessments of metabolic activity.
Collapse
Affiliation(s)
| | - Silke Lodema
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Arthur J A T Braat
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Johannes H M Merks
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rob van Rooij
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Bart de Keizer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
3
|
Acar Tayyar MN, Tamam MÖ, Babacan GB, Şahin MC, Özçevik H, Gürdal N, Atakır K. [ 18F]FDG PET/CT beyond staging: Prognostic significance of sarcopenia and adipose tissue metabolism in esophageal carcinomas. Rev Esp Med Nucl Imagen Mol 2024:500090. [PMID: 39732336 DOI: 10.1016/j.remnie.2024.500090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/02/2024] [Indexed: 12/30/2024]
Abstract
AIM This study aimed to investigate the relationship between PET and CT parameters and sarcopenia, adipose tissue, and tumor metabolism in esophageal carcinoma (EC) and its impact on survival in EC. METHOD Our study included 122 EC patients who underwent PET/CT for staging. Muscle and adipose tissue characteristics were evaluated, including lumbar (L3) and cervical (C3) muscle areas, psoas major (PM) and sternocleidomastoid muscle (SCM) parameters, and PET parameters for visceral and subcutaneous adipose tissue (SAT). Sarcopenia was determined using CT images, with a threshold for muscle tissue at the L3 vertebral level, and its impact on overall survival (OS) was investigated. RESULTS Sarcopenia was detected in 48 patients. SULmax in the primary tumor (PT) was significantly higher in sarcopenic patients (SP). The frequency of distant metastasis was higher in SP and OS was significantly lower. In the locally advanced stage, sarcopenia status decreased survival. L3, PM, C3, and SCM muscle areas were highly correlated. Subcutaneous adipose tissue SUVmax was significantly increased in SP and those with distant metastasis. Univariate analysis identified PT SULmax, PT SUVmean, PT TLG, lymph node and distant metastasis, SAT SUVmax, and sarcopenia as poor prognostic factors, while multivariate analysis confirmed BMI, distant metastasis, PT SUVmean, PT TLG as independent predictors of OS. CONCLUSION This study demonstrated that sarcopenia, linked to reduced survival, correlates with primary tumor SULmax, distant metastasis, and subcutaneous tissue PET parameters, exerting a notable impact on survival, particularly in locally advanced stages. Attenuation-corrected CT can be used instead of diagnostic CT, and sarcopenia can be diagnosed using not only L3 but also C3 slices.
Collapse
Affiliation(s)
- Merve Nur Acar Tayyar
- Department of Nuclear Medicine, University of Health Sciences, Prof. Dr. Cemil Tascioglu City Hospital, Istanbul, Turkey.
| | - Müge Öner Tamam
- Department of Nuclear Medicine, University of Health Sciences, Prof. Dr. Cemil Tascioglu City Hospital, Istanbul, Turkey
| | - Gündüzalp Buğrahan Babacan
- Department of Nuclear Medicine, University of Health Sciences, Prof. Dr. Cemil Tascioglu City Hospital, Istanbul, Turkey
| | - Mehmet Can Şahin
- Department of Nuclear Medicine, University of Health Sciences, Prof. Dr. Cemil Tascioglu City Hospital, Istanbul, Turkey
| | - Halim Özçevik
- Department of Nuclear Medicine, University of Health Sciences, Başaksehir Çam and Sakura City Hospital, Istanbul, Turkey
| | - Necla Gürdal
- Department of Radiation Oncology, University of Health Sciences, Prof. Dr. Cemil Tascioglu City Hospital, Istanbul, Turkey
| | - Kadir Atakır
- Department of Radiology, University of Health Sciences, Prof. Dr. Cemil Tascioglu City Hospital, Istanbul, Turkey
| |
Collapse
|
4
|
Hou X, He Y, Liu G, Chen S, Shi H. SPECT/CT imaging: quantifying 99mTc-MDP concentration in the spine and pelvis. Ann Nucl Med 2024; 38:933-942. [PMID: 39154304 DOI: 10.1007/s12149-024-01967-9] [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: 05/21/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
OBJECTIVE This study aimed to identify a relatively robust SUV for guiding clinical practice through quantitative measurement and comparison of various normalization methods based on the SUV of 99mTc-MDP in the normal spine and pelvis using an integrated SPECT/CT scanner. METHODS Between June 2017 and September 2019, a total of 500 oncology patients (mean age, 60.9; men, 66.0%) who underwent bone SPECT/CT scans with 99mTc-MDP were enrolled in this retrospective study. The mean SUV (SUVmean) of 4962 spinal and pelvic bones was calculated based on the patients' body weight (BW), lean body mass (LBM), bone mineral content (BMC), body surface area (BSA), and body mass index (BMI), defined as SUVbw, SUVlbm, SUVbmc, SUVbsa, and SUVbmi, respectively. The coefficients of variation (CoVs) of the aforementioned parameters were compared, and the correlation and multiple linear regression analyses were used to compare the extent to which these parameters were affected by sex, age, height, weight, BMI, and CT values. RESULTS The average SUVs in the normal spine and pelvis displayed a relatively wide variability: 4.573 ± 1.972 for SUVbw, 3.555 ± 1.517 for SUVlbm, 0.163 ± 0.071 for SUVbmc, 0.124 ± 0.052 for SUVbsa, and 1.668 ± 0.732 for SUVbmi. In general, SUVbsa had relatively lowest CoV (42.1%) in all vertebrae and pelvis compared with other SUVs. For correlation analyses, all SUVs displayed weak but significant correlations with age and CT values. For regression analyses, SUVbsa was influenced only by age, BMI, and CT values independently. The effects of these variables on SUVbsa were all smaller than those on conventional SUVbw. CONCLUSIONS The SUVs of 99mTc-MDP in normal bone derived from quantitative bone SPECT/CT could serve as a reference for evaluating tumor bone metastasis, but it should be assessed on a site-specific basis. SUVbsa exhibited superior robustness among all the SUV normalization variations, indicating potential clinical applications.
Collapse
Affiliation(s)
- Xiaoguang Hou
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
| | - Yibo He
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
| | - Guobing Liu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
| | - Shuguang Chen
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Nuclear Medicine Institute of Fudan University, Shanghai, China.
- Shanghai Institute of Medical Imaging, Shanghai, China.
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Nuclear Medicine Institute of Fudan University, Shanghai, China.
- Shanghai Institute of Medical Imaging, Shanghai, China.
| |
Collapse
|
5
|
Fraum TJ, Sari H, Dias AH, Munk OL, Pyka T, Smith AM, Mawlawi OR, Laforest R, Wang G. Whole-Body Multiparametric PET in Clinical Oncology: Current Status, Challenges, and Opportunities. AJR Am J Roentgenol 2024; 223:e2431712. [PMID: 39230403 DOI: 10.2214/ajr.24.31712] [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: 09/05/2024]
Abstract
The interpretation of clinical oncologic PET studies has historically used static reconstructions based on SUVs. SUVs and SUV-based images have important limitations, including dependence on uptake times and reduced conspicuity of tracer-avid lesions in organs with high background uptake. The acquisition of dynamic PET images enables additional PET reconstructions via Patlak modeling, which assumes that a tracer is irreversibly trapped by tissues of interest. The resulting multiparametric PET images capture a tracer's net trapping rate and apparent volume of distribution, separating the contributions of bound and free tracer fractions to the PET signal captured in the SUV. Potential benefits of multiparametric PET include higher quantitative stability, superior lesion conspicuity, and greater accuracy for differentiating malignant and benign lesions. However, the imaging protocols necessary for multiparametric PET are inherently more complex and time intensive, despite the recent introduction of automated or semiautomated scanner-based reconstruction packages. In this Review, we examine the current state of multiparametric PET in whole-body oncologic imaging. We summarize the Patlak method and relevant tracer kinetics, discuss clinical workflows and protocol considerations, and highlight clinical challenges and opportunities. We aim to help oncologic imagers make informed decisions about whether to implement multiparametric PET in their clinical practices.
Collapse
Affiliation(s)
- Tyler J Fraum
- Department of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, St. Louis, MO 63110
| | - Hasan Sari
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Siemens Healthineers International AG, Zurich, Switzerland
| | - André H Dias
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Ole L Munk
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thomas Pyka
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- TUM School of Medicine and Health, Munich, Germany
| | | | - Osama R Mawlawi
- Department of Imaging Physics, UT MD Anderson Cancer Center, Houston, TX
| | - Richard Laforest
- Department of Radiology, Washington University School of Medicine, 510 S Kingshighway Blvd, St. Louis, MO 63110
| | - Guobao Wang
- Department of Radiology, University of California Davis Health, Sacramento, CA
| |
Collapse
|
6
|
Ustsinau U, Kulterer OC, Rausch I, Krššák M, Kiefer FW, Hacker M, Philippe C. A PET/MRI study on the effect of obesity and NAFLD on hepatic [ 18F]FDG uptake. Eur J Radiol 2024; 177:111552. [PMID: 38861905 DOI: 10.1016/j.ejrad.2024.111552] [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: 03/08/2024] [Revised: 04/25/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024]
Abstract
PURPOSE The potential limitations of hepatic [18F]FDG-PET imaging for individuals with obesity and excessive liver fat (NAFLD) are being investigated. In this study, we aim to determine the reliability of standardized uptake values (SUVs) focusing on adjustment for liver fat content (LFC) derived from DIXON images and the effects of whole-body normalizations. METHODS Lean and with obesity volunteers who underwent [18F]FDG-PET/MRI were reviewed retrospectively. DIXON fat images were used to determine LFC and for adjustment of SUVmean. The hepatic SUVs (mean, fat adjusted mean and max) were normalized to body weight, lean body mass and body surface area. Blood samples were analysed for glucose, serological liver enzymes and lipoproteins for further correlation of [18F]FDG uptake. RESULTS Out of 11 volunteers with obesity (M:8, F:3, BMI:30-39 kg/m2), 9 confirmed the presence of NAFLD (>5.6 % fat). 22 age-matched lean volunteers (M:10, F:11, BMI:19-26 kg/m2) were used as control group. Both SUVmean, before and after adjustment to LFC, did not provide any difference between lean and with obesity groups under BW, LBM and BSA. SUVmax BW showed a difference between groups (p = 0.05). SUVs were independent of levels of GPT, GOT, gGT, insulin, HOMA-IR, triglycerides, cholesterol and LDL. Volunteers with low HDL were clustered with an increased hepatic [18F]FDG uptake. CONCLUSION Our method for adjustment of hepatic [18F]FDG-PET with DIXON fat images allows to achieve accurate results for individuals with NAFLD and obesity. For homogenic results, raw SUVmean should be combined with adjustment for liver fat, appropriate normalization and consideration of HDL levels.
Collapse
Affiliation(s)
- Usevalad Ustsinau
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Oana C Kulterer
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Ivo Rausch
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Martin Krššák
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Florian W Kiefer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Cecile Philippe
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Shimizu K, Nakata M, Saisho S, Inubushi M, Okumura N, Murakawa T, Yamashita M, Nakamura H, Sano Y, Kataoka K, Toyooka S. Predictive Value of PERCIST for Locally Advanced Non-Small Cell Lung Cancer Treated with Preoperative Induction Therapy - A Multicenter Study in Japan. Cancer Manag Res 2024; 16:965-976. [PMID: 39099761 PMCID: PMC11297483 DOI: 10.2147/cmar.s464265] [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: 03/28/2024] [Accepted: 07/23/2024] [Indexed: 08/06/2024] Open
Abstract
Background Induction therapy followed by surgery is recommended as an alternative treatment strategy for locally advanced non-small cell lung cancer (NSCLC). Patients who achieve pathologic response after induction therapy have better outcomes than non-responders; therefore, therapeutic response must be evaluated. Recently, new approaches for monitoring therapeutic responses, which are based on 18F-Fluorodeoxyglucose positron emission tomography (FDG-PET), have been developed. In this study, we evaluated the predictive value of Positron Emission Tomography Response Criteria in Solid Tumors (PERCIST), which uses standardized uptake values corrected for lean body mass (SUL) and total lesion glycolysis (TLG). Methods A total of 130 patients in the Setouchi Lung Cancer Group who underwent FDG-PET imaging before and after induction therapy prior to a planned surgical resection for NSCLC between 2007 and 2016 were studied retrospectively. The pathologic responses of the primary lung tumors and metastatic lymph nodes were compared with their responses based on evaluation using PERCIST. Results Postoperative pathologic studies revealed pathologic complete response (pCR) in 42 (32.3%) patients. PERCIST was significantly correlated with pathologic response (p < 0.001). The sensitivity, specificity, and accuracy of PERCIST for predicting pCR were 16.7% (7/42), 88.6% (78/88), and 65.4% (85/130), respectively. Patients with pCR had significantly higher reduction rates in SULpeak for both primary lung tumors and metastatic lymph nodes and TLG for primary tumors than non-responders. In a multivariate Cox regression analysis, tumor site in upper lobes, reduction rate of TLG in primary tumor, and pathologic N0 were independent predictors of favorable recurrence-free survival (RFS). Conclusion Our study suggested that PERCIST, especially the rate of TLG reduction rate, are useful to predict the pathological response and prognosis after induction therapy. Although improvement is necessary, PERCIST can be a promising method of the post-induction status in lung cancer. Further research is needed to confirm our findings.
Collapse
Affiliation(s)
- Katsuhiko Shimizu
- Department of General Thoracic Surgery, Kawasaki Medical School, Kurashiki, Japan
| | - Masao Nakata
- Department of General Thoracic Surgery, Kawasaki Medical School, Kurashiki, Japan
| | - Shinsuke Saisho
- Department of General Thoracic Surgery, Kawasaki Medical School, Kurashiki, Japan
| | - Masayuki Inubushi
- Division of Nuclear Medicine, Department of Radiology, Kawasaki Medical School, Kurashiki, Japan
| | - Norihito Okumura
- Department of Thoracic Surgery, Kurashiki Central Hospital, Kurashiki, Japan
| | - Tomohiro Murakawa
- Department of Thoracic Surgery, Kansai Medical University, Osaka, Japan
| | | | - Hiroshige Nakamura
- Division of General Thoracic Surgery, Tottori University Hospital, Yonago, Japan
| | - Yoshifumi Sano
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon, Japan
| | - Kazuhiko Kataoka
- Department of Thoracic Surgery, National Hospital Organization Iwakuni Clinical Center, Iwakuni, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
8
|
Zhu YM, Peng P, Liu X, Qi SN, Wang SL, Fang H, Song YW, Liu YP, Jin J, Li N, Lu NN, Jing H, Tang Y, Chen B, Zhang WW, Zhai YR, Yang Y, Liang B, Zheng R, Li YX. Optimizing the prognostic capacity of baseline 18F-FDG PET/CT metabolic parameters in extranodal natural killer/T-cell lymphoma by using relative and absolute thresholds. Heliyon 2024; 10:e25184. [PMID: 38322946 PMCID: PMC10844272 DOI: 10.1016/j.heliyon.2024.e25184] [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: 08/29/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Objectives To investigate the prognostic capacity of baseline 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) metabolic parameters in extranodal natural killer/T-cell lymphoma (ENKTCL), and the influence of relative thresholds (RT) and absolute thresholds (AT) selection on prognostic capacity. Materials and methods Metabolic tumor volume (MTV)-based parameters were defined using RTs (41 % or 25 % of maximum standardized uptake value [SUVmax]), ATs (SUV 2.5, 3.0, 4.0, or mean liver uptake) in 133 patients. Metabolic parameters were classified into avidity-related parameters (SUVmax, mean SUV [SUVmean], standard deviation of SUV [SUVsd]), volume-related parameters (RT-MTV), and avidity- and volume-related parameters (total lesion glycolysis [TLG] and AT-MTV). The prognostic capacity of the metabolic parameters and the effects of different threshold types (RT vs. AT) were evaluated. Results All metabolic parameters were moderately associated with prognosis. However, the area under the receiver operating characteristic curve of MTV and TLG was slightly higher than that of avidity-related parameters for predicting 5-year progression-free survival (PFS) (0.614-0.705 vs. 0.563-0.609) and overall survival (OS) (0.670-0.748 vs. 0.562-0.593). Correlations of MTV and avidity-related parameters differed between RTs (r < 0.06, P = 0.324-0.985) and ATs (r 0.56-0.84, P ≤ 0.001). AT-MTV was the optimal predictor for PFS and OS, while RT-TLG was the optimal predictor for PFS, and the combination of RT-MTV with SUVmax was the optimal predictor for OS. Conclusion The incorporation of volume and avidity significantly improved the prognostic capacity of PET in ENKTCL. Composite parameters that encompassed both avidity and volume were recommended.
Collapse
Affiliation(s)
- Ying-Ming Zhu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Pan Peng
- Department of Nuclear Medicine, National Cancer Center/Cancer Hospital, CAMS and PUMC, Beijing, China
| | - Xin Liu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Shu-Nan Qi
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Shu-Lian Wang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Hui Fang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Yong-Wen Song
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Yue-Ping Liu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital & Shenzhen Hospital, CAMS and PUMC, Shenzhen, China
| | - Ning Li
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Ning-Ning Lu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Hao Jing
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Yuan Tang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Bo Chen
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Wen-Wen Zhang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Yi-Rui Zhai
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Yong Yang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bin Liang
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Rong Zheng
- Department of Nuclear Medicine, National Cancer Center/Cancer Hospital, CAMS and PUMC, Beijing, China
| | - Ye-Xiong Li
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| |
Collapse
|
9
|
Yang Y, Wei H, Fu F, Wei W, Wu Y, Bai Y, Li Q, Wang M. Preoperative prediction of lymphovascular invasion of colorectal cancer by radiomics based on 18F-FDG PET-CT and clinical factors. FRONTIERS IN RADIOLOGY 2023; 3:1212382. [PMID: 37614530 PMCID: PMC10442652 DOI: 10.3389/fradi.2023.1212382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023]
Abstract
Purpose The purpose of this study was to investigate the value of a clinical radiomics model based on Positron emission tomography-computed tomography (PET-CT) radiomics features combined with clinical predictors of Lymphovascular invasion (LVI) in predicting preoperative LVI in patients with colorectal cancer (CRC). Methods A total of 95 CRC patients who underwent preoperative 18F-fluorodeoxyglucose (FDG) PET-CT examination were retrospectively enrolled. Univariate and multivariate logistic regression analyses were used to analyse clinical factors and PET metabolic data in the LVI-positive and LVI-negative groups to identify independent predictors of LVI. We constructed four prediction models based on radiomics features and clinical data to predict LVI status. The predictive efficacy of different models was evaluated according to the receiver operating characteristic curve. Then, the nomogram of the best model was constructed, and its performance was evaluated using calibration and clinical decision curves. Results Mean standardized uptake value (SUVmean), maximum tumour diameter and lymph node metastasis were independent predictors of LVI in CRC patients (P < 0.05). The clinical radiomics model obtained the best prediction performance, with an Area Under Curve (AUC) of 0.922 (95%CI 0.820-0.977) and 0.918 (95%CI 0.782-0.982) in the training and validation cohorts, respectively. A nomogram based on the clinical radiomics model was constructed, and the calibration curve fitted well (P > 0.05). Conclusion The clinical radiomics prediction model constructed in this study has high value in the preoperative individualized prediction of LVI in CRC patients.
Collapse
Affiliation(s)
- Yan Yang
- Department of Medical Imaging, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Huanhuan Wei
- Department of Medical Imaging, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Fangfang Fu
- Henan Key Laboratory of Neurological Imaging, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Wei Wei
- Henan Key Laboratory of Neurological Imaging, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yaping Wu
- Henan Key Laboratory of Neurological Imaging, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yan Bai
- Henan Key Laboratory of Neurological Imaging, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Qing Li
- Department of Medical Imaging, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Meiyun Wang
- Henan Key Laboratory of Neurological Imaging, Henan Provincial People’s Hospital, Zhengzhou, China
| |
Collapse
|
10
|
Galvez T, Berkane I, Thézenas S, Eberlé MC, Flori N, Guillemard S, Ilonca AD, Santoro L, Kotzki PO, Senesse P, Deshayes E. Identification of 18F-FDG PET/CT Parameters Associated with Weight Loss in Patients with Esophageal Cancer. Nutrients 2023; 15:3042. [PMID: 37447367 DOI: 10.3390/nu15133042] [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: 05/08/2023] [Revised: 06/16/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023] Open
Abstract
18F-FDG PET-CT is routinely performed as part of the initial staging of numerous cancers. Other than having descriptive, predictive and prognostic values for tumors, 18F-FDG PET-CT provides full-body data, which could inform on concurrent pathophysiological processes such as malnutrition. To test this hypothesis, we measured the 18F-FDG uptake in several organs and evaluated their association with weight loss in patients at diagnosis of esophageal cancer. Forty-eight patients were included in this retrospective monocentric study. 18F-FDG uptake quantification was performed in the brain, the liver, the spleen, bone marrow, muscle and the esophageal tumor itself and was compared between patients with different amounts of weight loss. We found that Total Lesion Glycolysis (TLG) and peak Standardized Uptake Values (SUVpeak) measured in the brain correlated with the amount of weight loss: TLG was, on average, higher in patients who had lost more than 5% of their usual weight, whereas brain SUVpeak were, on average, lower in patients who had lost more than 10% of their weight. Higher TLG and lower brain SUVpeak were associated with worse OS in the univariate analysis. This study reports a new and significant association between 18F-FDG uptake in the brain and initial weight loss in patients with esophageal cancer.
Collapse
Affiliation(s)
- Thierry Galvez
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
- Department of Endocrinology, Diabetes and Nutrition, CHU de Montpellier, Université de Montpellier, 34295 Montpellier, France
| | - Ikrame Berkane
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Simon Thézenas
- Biometry Unit, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Marie-Claude Eberlé
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Nicolas Flori
- Department of Clinical Nutrition and Gastroenterology, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, 34298 Montpellier, France
| | - Sophie Guillemard
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Alina Diana Ilonca
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Lore Santoro
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Pierre-Olivier Kotzki
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, 34298 Montpellier, France
| | - Pierre Senesse
- Department of Clinical Nutrition and Gastroenterology, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, 34298 Montpellier, France
| | - Emmanuel Deshayes
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, 34298 Montpellier, France
| |
Collapse
|
11
|
Weyts K, Quak E, Licaj I, Ciappuccini R, Lasnon C, Corroyer-Dulmont A, Foucras G, Bardet S, Jaudet C. Deep Learning Denoising Improves and Homogenizes Patient [ 18F]FDG PET Image Quality in Digital PET/CT. Diagnostics (Basel) 2023; 13:1626. [PMID: 37175017 PMCID: PMC10177812 DOI: 10.3390/diagnostics13091626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/18/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
Given the constant pressure to increase patient throughput while respecting radiation protection, global body PET image quality (IQ) is not satisfactory in all patients. We first studied the association between IQ and other variables, in particular body habitus, on a digital PET/CT. Second, to improve and homogenize IQ, we evaluated a deep learning PET denoising solution (Subtle PETTM) using convolutional neural networks. We analysed retrospectively in 113 patients visual IQ (by a 5-point Likert score in two readers) and semi-quantitative IQ (by the coefficient of variation in the liver, CVliv) as well as lesion detection and quantification in native and denoised PET. In native PET, visual and semi-quantitative IQ were lower in patients with larger body habitus (p < 0.0001 for both) and in men vs. women (p ≤ 0.03 for CVliv). After PET denoising, visual IQ scores increased and became more homogeneous between patients (4.8 ± 0.3 in denoised vs. 3.6 ± 0.6 in native PET; p < 0.0001). CVliv were lower in denoised PET than in native PET, 6.9 ± 0.9% vs. 12.2 ± 1.6%; p < 0.0001. The slope calculated by linear regression of CVliv according to weight was significantly lower in denoised than in native PET (p = 0.0002), demonstrating more uniform CVliv. Lesion concordance rate between both PET series was 369/371 (99.5%), with two lesions exclusively detected in native PET. SUVmax and SUVpeak of up to the five most intense native PET lesions per patient were lower in denoised PET (p < 0.001), with an average relative bias of -7.7% and -2.8%, respectively. DL-based PET denoising by Subtle PETTM allowed [18F]FDG PET global image quality to be improved and homogenized, while maintaining satisfactory lesion detection and quantification. DL-based denoising may render body habitus adaptive PET protocols unnecessary, and pave the way for the improvement and homogenization of PET modalities.
Collapse
Affiliation(s)
- Kathleen Weyts
- Department of Nuclear Medicine, Baclesse Cancer Centre, 14076 Caen, France
| | - Elske Quak
- Department of Nuclear Medicine, Baclesse Cancer Centre, 14076 Caen, France
| | - Idlir Licaj
- Department of Biostatistics, Baclesse Cancer Centre, 14076 Caen, France
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway
| | - Renaud Ciappuccini
- Department of Nuclear Medicine, Baclesse Cancer Centre, 14076 Caen, France
| | - Charline Lasnon
- Department of Nuclear Medicine, Baclesse Cancer Centre, 14076 Caen, France
| | - Aurélien Corroyer-Dulmont
- Department of Medical Physics, Baclesse Cancer Centre, 14076 Caen, France
- ISTCT Unit, CNRS, UNICAEN, Normandy University, GIP CYCERON, 14074 Caen, France
| | - Gauthier Foucras
- Department of Nuclear Medicine, Baclesse Cancer Centre, 14076 Caen, France
| | - Stéphane Bardet
- Department of Nuclear Medicine, Baclesse Cancer Centre, 14076 Caen, France
| | - Cyril Jaudet
- Department of Nuclear Medicine, Baclesse Cancer Centre, 14076 Caen, France
- Department of Medical Physics, Baclesse Cancer Centre, 14076 Caen, France
| |
Collapse
|
12
|
Sponagel J, Jones JK, Frankfater C, Zhang S, Tung O, Cho K, Tinkum KL, Gass H, Nunez E, Spitz DR, Chinnaiyan P, Schaefer J, Patti GJ, Graham MS, Mauguen A, Grkovski M, Dunphy MP, Krebs S, Luo J, Rubin JB, Ippolito JE. Sex differences in brain tumor glutamine metabolism reveal sex-specific vulnerabilities to treatment. MED 2022; 3:792-811.e12. [PMID: 36108629 PMCID: PMC9669217 DOI: 10.1016/j.medj.2022.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 07/08/2022] [Accepted: 08/22/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Brain cancer incidence and mortality rates are greater in males. Understanding the molecular mechanisms that underlie those sex differences could improve treatment strategies. Although sex differences in normal metabolism are well described, it is currently unknown whether they persist in cancerous tissue. METHODS Using positron emission tomography (PET) imaging and mass spectrometry, we assessed sex differences in glioma metabolism in samples from affected individuals. We assessed the role of glutamine metabolism in male and female murine transformed astrocytes using isotope labeling, metabolic rescue experiments, and pharmacological and genetic perturbations to modulate pathway activity. FINDINGS We found that male glioblastoma surgical specimens are enriched for amino acid metabolites, including glutamine. Fluoroglutamine PET imaging analyses showed that gliomas in affected male individuals exhibit significantly higher glutamine uptake. These sex differences were well modeled in murine transformed astrocytes, in which male cells imported and metabolized more glutamine and were more sensitive to glutaminase 1 (GLS1) inhibition. The sensitivity to GLS1 inhibition in males was driven by their dependence on glutamine-derived glutamate for α-ketoglutarate synthesis and tricarboxylic acid (TCA) cycle replenishment. Females were resistant to GLS1 inhibition through greater pyruvate carboxylase (PC)-mediated TCA cycle replenishment, and knockdown of PC sensitized females to GLS1 inhibition. CONCLUSION Our results show that clinically important sex differences exist in targetable elements of metabolism. Recognition of sex-biased metabolism may improve treatments through further laboratory and clinical research. FUNDING This work was supported by NIH grants, Joshua's Great Things, the Siteman Investment Program, and the Barnard Research Fund.
Collapse
Affiliation(s)
- Jasmin Sponagel
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jill K Jones
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cheryl Frankfater
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Biomedical Mass Spectrometry Resource, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shanshan Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Olivia Tung
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kevin Cho
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kelsey L Tinkum
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hannah Gass
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Elena Nunez
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52246, USA; Holden Comprehensive Cancer Center, Department of Pathology, University of Iowa, Iowa City, IA 52246, USA
| | - Prakash Chinnaiyan
- Department of Radiation Oncology, Beaumont Health, Royal Oak, MI 48073, USA; Oakland University William Beaumont School of Medicine, Rochester, MI 48073, USA
| | - Jacob Schaefer
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Gary J Patti
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maya S Graham
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Milan Grkovski
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mark P Dunphy
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Simone Krebs
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jingqin Luo
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Joseph E Ippolito
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
13
|
Badawe EM, Abdel Gawad H, El-Nagdy MS, Khalil MM. Variations induced by body weight and background lesion normalization in standardized uptake value estimated by F18-FDG PET/CT. Eur J Hybrid Imaging 2022; 6:22. [PMID: 36224438 PMCID: PMC9556683 DOI: 10.1186/s41824-022-00142-5] [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: 05/25/2022] [Accepted: 07/12/2022] [Indexed: 11/10/2022] Open
Abstract
AIM This work aims to study the impact of different SUV variants in terms of mean and maximum measures as well as various normalization methods with respect to body weight, body mass index, body surface area, and lean body mass in patients with lymphoma. METHODS Sixty-nine patients (34 male-35 female) were retrospectively selected. All patients had undergone F18-FDG PET/CT using the standard imaging protocol. In the first part of this study, SUVmean and SUVmax of patients' lesions and three background sites including liver, aorta, and muscle were determined. Then, the normalization of lesion SUV to body weight and body background sites was performed. The ratio of lesion SUVmax to body background sites (muscle, aorta, and liver) SUVmax was determined in addition to the ratio of lesion SUVmean to body background sites SUVmean. The second part of the study included the calculations of the body mass index (BMI), body surface area (BSA), and lean body mass (LBM). The normalization of lesion, liver, aorta, and muscle SUV to BMI, BSA, and LBM was calculated and compared to each other. RESULTS After performing the appropriate statistical calculations, the results showed that there is a significant difference in SUV measurements between the three background sites. Lesions normalized to the liver were significantly lower than those normalized to aorta and muscle and the results also showed a higher magnitude of lesions normalized to muscle in comparison to the aorta. The SUVmax and SUVmean normalized to different body weight indices showed the lowest variation with BSA and BMI while being increasingly higher with lean body mass using the two methods James and Janmahasatian, respectively, and then highest with body weight. CONCLUSION The SUVmax and SUVmean showed lower variance in comparison to other background regions. Less variation was also remarkable in SUVmean normalized to BSA and Janma lean mass and also when SUVmax is normalized to James lean body mass. The SUVmax normalized to lean (i.e., James) as well as SUVmean normalized to lean (i.e., Janma) and BSA showed a significant independence with body weight.
Collapse
Affiliation(s)
- Eman M. Badawe
- grid.412093.d0000 0000 9853 2750Department of Physics, Faculty of Science, Helwan University, Cairo, Egypt
| | - Hesham Abdel Gawad
- grid.7776.10000 0004 0639 9286Faculty of Medicine, Kasr Al-Aini Center for Nuclear Medicine and Oncology (NEMROK), Cairo University Hospitals, Cairo University, Giza, Egypt
| | - Mohamed S. El-Nagdy
- grid.412093.d0000 0000 9853 2750Department of Physics, Faculty of Science, Helwan University, Cairo, Egypt
| | - Magdy M. Khalil
- grid.412093.d0000 0000 9853 2750Department of Physics, Faculty of Science, Helwan University, Cairo, Egypt ,grid.507995.70000 0004 6073 8904School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo Egypt
| |
Collapse
|
14
|
Wang Z, Wu Y, Li X, Bai Y, Chen H, Ding J, Shen C, Hu Z, Liang D, Liu X, Zheng H, Yang Y, Zhou Y, Wang M, Sun T. Comparison between a dual-time-window protocol and other simplified protocols for dynamic total-body 18F-FDG PET imaging. EJNMMI Phys 2022; 9:63. [PMID: 36104580 PMCID: PMC9474964 DOI: 10.1186/s40658-022-00492-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 08/29/2022] [Indexed: 11/10/2022] Open
Abstract
PURPOSE Efforts have been made both to avoid invasive blood sampling and to shorten the scan duration for dynamic positron emission tomography (PET) imaging. A total-body scanner, such as the uEXPLORER PET/CT, can relieve these challenges through the following features: First, the whole-body coverage allows for noninvasive input function from the aortic arteries; second, with a dramatic increase in sensitivity, image quality can still be maintained at a high level even with a shorter scan duration than usual. We implemented a dual-time-window (DTW) protocol for a dynamic total-body 18F-FDG PET scan to obtain multiple kinetic parameters. The DTW protocol was then compared to several other simplified quantification methods for total-body FDG imaging that were proposed for conventional setup. METHODS The research included 28 patient scans performed on an uEXPLORER PET/CT. By discarding the corresponding data in the middle of the existing full 60-min dynamic scan, the DTW protocol was simulated. Nonlinear fitting was used to estimate the missing data in the interval. The full input function was obtained from 15 subjects using a hybrid approach with a population-based image-derived input function. Quantification was carried out in three areas: the cerebral cortex, muscle, and tumor lesion. Micro- and macro-kinetic parameters for different scan durations were estimated by assuming an irreversible two-tissue compartment model. The visual performance of parametric images and region of interest-based quantification in several parameters were evaluated. Furthermore, simplified quantification methods (DTW, Patlak, fractional uptake ratio [FUR], and standardized uptake value [SUV]) were compared for similarity to the reference net influx rate Ki. RESULTS Ki and K1 derived from the DTW protocol showed overall good consistency (P < 0.01) with the reference from the 60-min dynamic scan with 10-min early scan and 5-min late scan (Ki correlation: 0.971, 0.990, and 0.990; K1 correlation: 0.820, 0.940, and 0.975 in the cerebral cortex, muscle, and tumor lesion, respectively). Similar correlationss were found for other micro-parameters. The DTW protocol had the lowest bias relative to standard Ki than any of the quantification methods, followed by FUR and Patlak. SUV had the weakest correlation with Ki. The whole-body Ki and K1 images generated by the DTW protocol were consistent with the reference parametric images. CONCLUSIONS Using the DTW protocol, the dynamic total-body FDG scan time can be reduced to 15 min while obtaining accurate Ki and K1 quantification and acceptable visual performance in parametric images. However, the trade-off between quantification accuracy and protocol implementation feasibility must be considered in practice. We recommend that the DTW protocol be used when the clinical task requires reliable visual assessment or quantifying multiple micro-parameters; FUR with a hybrid input function may be a more feasible approach to quantifying regional metabolic rate with a known lesion position or organs of interest.
Collapse
Affiliation(s)
- Zhenguo Wang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Yaping Wu
- Henan Provincial People's Hospital and the People's Hospital of Zhengzhou, University of Zhengzhou, Zhengzhou, People's Republic of China
| | - Xiaochen Li
- Henan Provincial People's Hospital and the People's Hospital of Zhengzhou, University of Zhengzhou, Zhengzhou, People's Republic of China
| | - Yan Bai
- Henan Provincial People's Hospital and the People's Hospital of Zhengzhou, University of Zhengzhou, Zhengzhou, People's Republic of China
| | - Hongzhao Chen
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Jie Ding
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Chushu Shen
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Zhanli Hu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Dong Liang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Yongfeng Yang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Yun Zhou
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, People's Republic of China
- School of Biomedical Engineering, Shanghai Tech University, Shanghai, People's Republic of China
| | - Meiyun Wang
- Henan Provincial People's Hospital and the People's Hospital of Zhengzhou, University of Zhengzhou, Zhengzhou, People's Republic of China.
| | - Tao Sun
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China.
- United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, People's Republic of China.
| |
Collapse
|
15
|
Riauka TA, Baracos VE, Reif R, Juengling FD, Robinson DM, Wieler M, McEwan AJB. Rapid Standardized CT-Based Method to Determine Lean Body Mass SUV for PET-A Significant Improvement Over Prediction Equations. Front Oncol 2022; 12:812777. [PMID: 35875083 PMCID: PMC9302197 DOI: 10.3389/fonc.2022.812777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 05/26/2022] [Indexed: 01/18/2023] Open
Abstract
In 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) studies, maximum standardized uptake value (SUVmax) is the parameter commonly used to provide a measurement of the metabolic activity of a tumor. SUV normalized by body mass is affected by the proportions of body fat and lean tissue, which present high variability in patients with cancer. SUV corrected by lean body mass (LBM), denoted as SUL, is recommended to provide more accurate, consistent, and reproducible SUV results; however, LBM is frequently estimated rather than measured. Given the increasing importance of a quantitative PET parameter, especially when comparing PET studies over time to evaluate disease response clinically, and its use in oncological clinical trials, we set out to evaluate the commonly used equations originally derived by James (1976) and Janmahasatian et al. (2005) against computerized tomography (CT)-derived measures of LBM. Methods Whole-body 18F-FDG PET images of 195 adult patients with cancer were analyzed retrospectively. Representative liver SUVmean was normalized by total body mass. SUL was calculated using a quantitative determination of LBM based on the CT component of the PET/CT study (LBMCT) and compared against the equation-estimated SUL. Bland and Altman plots were generated for SUV-SUL differences. Results This consecutive sample of patients undergoing usual care (men, n = 96; women, n = 99) varied in body mass (38-127 kg) and in Body Mass Index (BMI) (14.7-47.2 kg/m2). LBMCT weakly correlated with body mass (men, r2 = 0.32; women, r2 = 0.22), and thus SUV and SULCT were also weakly correlated (men, r2 = 0.24; women, r2 = 0.11). Equations proved inadequate for the assessment of LBM. LBM estimated by James' equation showed a mean bias (overestimation of LBM compared with LBMCT) in men (+6.13 kg; 95% CI 4.61-7.65) and in women (+6.32 kg; 95% CI 5.26-7.39). Janmahasatian's equation provided similarly poor performance. Conclusions CT-based LBM determinations incorporate the patient's current body composition at the time of a PET/CT study, and the information garnered can provide care teams with information with which to more accurately determine FDG uptake values, allowing comparability over multiple scans and treatment courses and will provide a robust basis for the use of PET Response Criteria in Solid Tumors (PERCIST) in clinical trials.
Collapse
Affiliation(s)
- Terence A. Riauka
- Division of Medical Physics, Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Vickie E. Baracos
- Division of Palliative Care Medicine, Department of Oncology, University of Alberta, Edmonton, AB, Switzerland
| | - Rebecca Reif
- Division of Oncologic Imaging, Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Freimut D. Juengling
- Division of Oncologic Imaging, Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Medical Faculty, University Bern, Bern, Switzerland
| | - Don M. Robinson
- Division of Medical Physics, Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Marguerite Wieler
- Department of Physical Therapy, University of Alberta, Edmonton, AB, Canada
| | - Alexander J. B. McEwan
- Division of Oncologic Imaging, Department of Oncology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
16
|
Jaswal S, Sanders V, Pullarkat P, Teja S, Salter A, Watkins MP, Atagu N, Ludwig DR, Mhlanga J, Mellnick VM, Peterson LR, Bartlett NL, Kahl BS, Fehniger TA, Ghobadi A, Cashen AF, Mehta-Shah N, Ippolito JE. Metabolic Biomarkers Assessed with PET/CT Predict Sex-Specific Longitudinal Outcomes in Patients with Diffuse Large B-Cell Lymphoma. Cancers (Basel) 2022; 14:2932. [PMID: 35740596 PMCID: PMC9221486 DOI: 10.3390/cancers14122932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
In many cancers, including lymphoma, males have higher incidence and mortality than females. Emerging evidence demonstrates that one mechanism underlying this phenomenon is sex differences in metabolism, both with respect to tumor nutrient consumption and systemic alterations in metabolism, i.e., obesity. We wanted to determine if visceral fat and tumor glucose uptake with fluorodeoxyglucose-positron emission tomography/computed tomography (FDG-PET/CT) could predict sex-dependent outcomes in patients with diffuse large B-cell lymphoma (DLBCL). We conducted a retrospective analysis of 160 patients (84 males; 76 females) with DLBCL who had imaging at initial staging and after completion of therapy. CT-based relative visceral fat area (rVFA), PET-based SUVmax normalized to lean body mass (SULmax), and end-of-treatment FDG-PET 5PS score were calculated. Increased rVFA at initial staging was an independent predictor of poor OS only in females. At the end of therapy, increase in visceral fat was a significant predictor of poor survival only in females. Combining the change in rVFA and 5PS scores identified a subgroup of females with visceral fat gain and high 5PS with exceptionally poor outcomes. These data suggest that visceral fat and tumor FDG uptake can predict outcomes in DLBCL patients in a sex-specific fashion.
Collapse
Affiliation(s)
- Shama Jaswal
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.J.); or (V.S.); (D.R.L.); (J.M.); (V.M.M.)
| | - Vanessa Sanders
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.J.); or (V.S.); (D.R.L.); (J.M.); (V.M.M.)
| | - Priyanka Pullarkat
- Washington University School of Medicine, St. Louis, MO 63110, USA; (P.P.); (N.A.)
| | - Stephanie Teja
- Department of Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.T.); (M.P.W.); (N.L.B.); (B.S.K.); (T.A.F.); (A.G.); (A.F.C.)
| | - Amber Salter
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Marcus P. Watkins
- Department of Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.T.); (M.P.W.); (N.L.B.); (B.S.K.); (T.A.F.); (A.G.); (A.F.C.)
| | - Norman Atagu
- Washington University School of Medicine, St. Louis, MO 63110, USA; (P.P.); (N.A.)
| | - Daniel R. Ludwig
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.J.); or (V.S.); (D.R.L.); (J.M.); (V.M.M.)
| | - Joyce Mhlanga
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.J.); or (V.S.); (D.R.L.); (J.M.); (V.M.M.)
| | - Vincent M. Mellnick
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.J.); or (V.S.); (D.R.L.); (J.M.); (V.M.M.)
| | - Linda R. Peterson
- Department of Medicine, Division of Cardiology, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Nancy L. Bartlett
- Department of Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.T.); (M.P.W.); (N.L.B.); (B.S.K.); (T.A.F.); (A.G.); (A.F.C.)
| | - Brad S. Kahl
- Department of Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.T.); (M.P.W.); (N.L.B.); (B.S.K.); (T.A.F.); (A.G.); (A.F.C.)
| | - Todd A. Fehniger
- Department of Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.T.); (M.P.W.); (N.L.B.); (B.S.K.); (T.A.F.); (A.G.); (A.F.C.)
| | - Armin Ghobadi
- Department of Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.T.); (M.P.W.); (N.L.B.); (B.S.K.); (T.A.F.); (A.G.); (A.F.C.)
| | - Amanda F. Cashen
- Department of Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.T.); (M.P.W.); (N.L.B.); (B.S.K.); (T.A.F.); (A.G.); (A.F.C.)
| | - Neha Mehta-Shah
- Department of Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.T.); (M.P.W.); (N.L.B.); (B.S.K.); (T.A.F.); (A.G.); (A.F.C.)
| | - Joseph E. Ippolito
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.J.); or (V.S.); (D.R.L.); (J.M.); (V.M.M.)
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
17
|
Crandall J, Fraum TJ, Wahl RL. Brown adipose tissue: a protective mechanism in "pre-prediabetes"? J Nucl Med 2022; 63:1433-1440. [PMID: 35393347 DOI: 10.2967/jnumed.121.263357] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 03/21/2022] [Indexed: 11/16/2022] Open
Abstract
Brown adipose tissue (BAT) is present in a significant number of adult humans and has been postulated to exert beneficial metabolic effects. Lean, non-diabetic patients undergoing clinical positron emission tomography (PET)/computed tomography (CT) imaging are more likely to exhibit incidental BAT activation. The aim of this study was to assess metabolic changes associated with the cold-activation of BAT and to compare baseline blood metabolites in participants with varying amounts of active BAT. Methods: Serum blood samples were collected from healthy adult volunteers (body mass index 18.0-25.0 and age≤35 years) before and after 2 h cold exposure. 18F-flurodeoxyglucose (FDG) PET/CT imaging was performed immediately following cold exposure. Activated BAT was segmented and fasting glucose, insulin, lipid, and other blood metabolite levels were correlated with volume and intensity of active BAT. Using a median cutoff, subjects were classified as BATHIGH or BATLOW. Results: A higher volume of activated BAT was associated with significantly higher pre-cooling glucose and insulin levels (P<0.001 for each). Pre-cooling thyroid stimulating hormone (TSH) and triglyceride levels were significantly higher in the BATHIGH than in the BATLOW group (P = 0.002 and P<0.001, respectively). Triglyceride levels tended to increase over the cooling period in both BAT groups, but increased significantly more in the BATHIGH group (15.7±13.2 md/dl; P<0.001) than in the BATLOW group (4.5±12.2 mg/dl; P = 0.061). Conclusion: These findings may indicate that BAT is recruited to counteract incipient "pre-prediabetic" states, potentially serving as a first-line protective mechanism against very early metabolic or hormonal variations.
Collapse
|
18
|
Geitenbeek RTJ, Martin E, Graven LH, Broen MPG, Anten MHME, van der Pol JAJ, Verhoef C, Taal W. Diagnostic value of 18F-FDG PET-CT in detecting malignant peripheral nerve sheath tumors among adult and pediatric neurofibromatosis type 1 patients. J Neurooncol 2022; 156:559-567. [PMID: 35025020 PMCID: PMC8860956 DOI: 10.1007/s11060-021-03936-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 12/24/2021] [Indexed: 11/26/2022]
Abstract
Purpose Detecting malignant peripheral nerve sheath tumors (MPNSTs) remains difficult. 18F-FDG PET-CT has been shown helpful, but ideal threshold values of semi-quantitative markers remain unclear, partially because of variation among scanners. Using EU-certified scanners diagnostic accuracy of ideal and commonly used 18F-FDG PET-CT thresholds were investigated and differences between adult and pediatric lesions were evaluated. Methods A retrospective cohort study was performed including patients from two hospitals with a clinical or radiological suspicion of MPNST between 2013 and 2019. Several markers were studied for ideal threshold values and differences among adults and children. A diagnostic algorithm was subsequently developed. Results Sixty patients were included (10 MPNSTs). Ideal threshold values were 5.8 for SUVmax (sensitivity 0.70, specificity 0.92), 5.0 for SUVpeak (sensitivity 0.70, specificity 0.97), 1.7 for TLmax (sensitivity 0.90, specificity 0.86), and 2.3 for TLmean (sensitivity 0.90, specificity 0.79). The standard TLmean threshold value of 2.0 yielded a sensitivity of 0.90 and specificity of 0.74, while the standard SUVmax threshold value of 3.5 yielded a sensitivity of 0.80 and specificity of 0.63. SUVmax and adjusted SUV for lean body mass (SUL) were lower in children, but tumor-to-liver ratios were similar in adult and pediatric lesions. Using TLmean > 2.0 or TLmean < 2.0 and SUVmax > 3.5, a sensitivity and specificity of 1.00 and 0.63 can be achieved. Conclusion 18F-FDG PET-CT offers adequate accuracy to detect MPNSTs. SUV values in pediatric MPNSTs may be lower, but tumor-to-liver ratios are not. By combining TLmean and SUVmax values, a 100% sensitivity can be achieved with acceptable specificity. Supplementary Information The online version contains supplementary material available at 10.1007/s11060-021-03936-y.
Collapse
Affiliation(s)
- Ritch T J Geitenbeek
- Department of Plastic and Reconstructive Surgery G04.126, University Medical Center Utrecht, PO Box 85060, 3508 AB, Utrecht, The Netherlands.,Department of Surgical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
| | - Enrico Martin
- Department of Plastic and Reconstructive Surgery G04.126, University Medical Center Utrecht, PO Box 85060, 3508 AB, Utrecht, The Netherlands. .,Department of Surgical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands.
| | - Laura H Graven
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
| | - Martijn P G Broen
- Department of Neurology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Monique H M E Anten
- Department of Neurology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Jochem A J van der Pol
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
| | - Walter Taal
- Department of Neurology, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
| |
Collapse
|
19
|
Ragab A, Wu J, Ding X, Clark A, Mischen B, Chauhan A, Oates ME, Anthony L, El Khouli R. 68Ga-DOTATATE PET/CT: The Optimum Standardized Uptake Value (SUV) Internal Reference. Acad Radiol 2022; 29:95-106. [PMID: 34756348 DOI: 10.1016/j.acra.2020.08.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/18/2020] [Accepted: 08/18/2020] [Indexed: 01/01/2023]
Abstract
RATIONALE AND OBJECTIVES Standardized Uptake Value (SUV) is an important semiquantitative measurement used in the clinical and research domains to assess radiopharmaceutical concentration in tumors versus normal organs, but is susceptible to many factors beyond the tumor biological environment. So, the aim of this study is to identify the optimum internal reference among organs with physiological uptake in 68Ga-DOTATATE PET/CT (DOTA PET/CT) scans. MATERIALS AND METHODS This HIPAA-compliant, IRB-approved study with waiver of consent included retrospective imaging review of 180 consecutive patients with neuroendocrine tumors presenting for DOTA PET/CT image acquisition: Ga-68 DOTATATE dose was reported as (0.054 mCi/Kg) scans between September 2018 and May 2019. Mean value of body weight normalized SUV (SUVbw) and lean body mass normalized SUV (SUL) of liver and spleen were measured. Information about the patients and scan characteristics were collected. The paired Grambsch test was used to compare variance among the measured SUVs. Spearman's rank correlation coefficient was used to assess correlation between SUVs and potential patient- and scan-specific confounding factors. RESULTS Variance of SUL was significantly lower than variance of SUVbw in both liver and spleen (p-value < 0.0001). Variances of liver SUVbw and SUL were significantly lower than the corresponding spleen SUVs. Liver SUL showed the lowest variance (3.69% ± 1.25%) among all measured SUVs. CONCLUSION SUL is a more reproducible, less variable, and therefore more reliable quantitative measure in DOTA PET/CT scans, compared SUVbw. Among the available organs with physiological uptake, liver SUL is the optimum internal reference given the liver's larger size and uniform SUL values resulting in lower variability and better reproducibility.
Collapse
Affiliation(s)
- Ahmed Ragab
- Yale New Haven Health - Bridgeport Hospital, Bridgeport, Connecticut
| | - Jianrong Wu
- University of Kentucky College of Medicine, Department of Internal Medicine, Division of Cancer Biostatistics, Lexington, Kentucky; University of Kentucky College of Medicine, Markey Cancer Center, Biostatistics and Bioinformatics Shared Resource Facility, Lexington, Kentucky
| | - Xue Ding
- University of Kentucky College of Medicine, Department of Internal Medicine, Division of Cancer Biostatistics, Lexington, Kentucky
| | - Aurela Clark
- University of Kentucky College of Medicine, Department of Radiology, Division of Nuclear Medicine and Molecular imaging, 800 Rose street, Lexington, 40536 KY
| | - Blaine Mischen
- University of Kentucky College of Medicine, Department of Radiology, Division of Nuclear Medicine and Molecular imaging, 800 Rose street, Lexington, 40536 KY
| | - Aman Chauhan
- University of Kentucky College of Medicine, Department of Internal Medicine, Division of Medical Oncology, Lexington, Kentucky; University of Kentucky College of Medicine, Markey Cancer Center, Lexington, Kentucky
| | - M Elizabeth Oates
- University of Kentucky College of Medicine, Department of Radiology, Division of Nuclear Medicine and Molecular imaging, 800 Rose street, Lexington, 40536 KY
| | - Lowell Anthony
- University of Kentucky College of Medicine, Department of Internal Medicine, Division of Medical Oncology, Lexington, Kentucky; University of Kentucky College of Medicine, Markey Cancer Center, Lexington, Kentucky
| | - Riham El Khouli
- University of Kentucky College of Medicine, Department of Radiology, Division of Nuclear Medicine and Molecular imaging, 800 Rose street, Lexington, 40536 KY; University of Kentucky College of Medicine, Markey Cancer Center, Lexington, Kentucky.
| |
Collapse
|
20
|
Moreau N, Rousseau C, Fourcade C, Santini G, Brennan A, Ferrer L, Lacombe M, Guillerminet C, Colombié M, Jézéquel P, Campone M, Normand N, Rubeaux M. Automatic Segmentation of Metastatic Breast Cancer Lesions on 18F-FDG PET/CT Longitudinal Acquisitions for Treatment Response Assessment. Cancers (Basel) 2021; 14:101. [PMID: 35008265 PMCID: PMC8750371 DOI: 10.3390/cancers14010101] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 11/21/2022] Open
Abstract
Metastatic breast cancer patients receive lifelong medication and are regularly monitored for disease progression. The aim of this work was to (1) propose networks to segment breast cancer metastatic lesions on longitudinal whole-body PET/CT and (2) extract imaging biomarkers from the segmentations and evaluate their potential to determine treatment response. Baseline and follow-up PET/CT images of 60 patients from the EPICUREseinmeta study were used to train two deep-learning models to segment breast cancer metastatic lesions: One for baseline images and one for follow-up images. From the automatic segmentations, four imaging biomarkers were computed and evaluated: SULpeak, Total Lesion Glycolysis (TLG), PET Bone Index (PBI) and PET Liver Index (PLI). The first network obtained a mean Dice score of 0.66 on baseline acquisitions. The second network obtained a mean Dice score of 0.58 on follow-up acquisitions. SULpeak, with a 32% decrease between baseline and follow-up, was the biomarker best able to assess patients' response (sensitivity 87%, specificity 87%), followed by TLG (43% decrease, sensitivity 73%, specificity 81%) and PBI (8% decrease, sensitivity 69%, specificity 69%). Our networks constitute promising tools for the automatic segmentation of lesions in patients with metastatic breast cancer allowing treatment response assessment with several biomarkers.
Collapse
Affiliation(s)
- Noémie Moreau
- LS2N, University of Nantes, CNRS, 44000 Nantes, France; (C.F.); (N.N.)
- Keosys Medical Imaging, 13 Imp. Serge Reggiani, 44815 Saint-Herblain, France; (G.S.); (A.B.); (M.R.)
| | - Caroline Rousseau
- CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, 44000 Nantes, France; (C.R.); (P.J.)
- ICO Cancer Center, 49000 Angers, France; (L.F.); (M.L.); (C.G.); (M.C.); (M.C.)
| | - Constance Fourcade
- LS2N, University of Nantes, CNRS, 44000 Nantes, France; (C.F.); (N.N.)
- Keosys Medical Imaging, 13 Imp. Serge Reggiani, 44815 Saint-Herblain, France; (G.S.); (A.B.); (M.R.)
| | - Gianmarco Santini
- Keosys Medical Imaging, 13 Imp. Serge Reggiani, 44815 Saint-Herblain, France; (G.S.); (A.B.); (M.R.)
| | - Aislinn Brennan
- Keosys Medical Imaging, 13 Imp. Serge Reggiani, 44815 Saint-Herblain, France; (G.S.); (A.B.); (M.R.)
| | - Ludovic Ferrer
- ICO Cancer Center, 49000 Angers, France; (L.F.); (M.L.); (C.G.); (M.C.); (M.C.)
- CRCINA, University of Angers, INSERM UMR1232, CNRS-ERL6001, 49000 Angers, France
| | - Marie Lacombe
- ICO Cancer Center, 49000 Angers, France; (L.F.); (M.L.); (C.G.); (M.C.); (M.C.)
| | | | - Mathilde Colombié
- ICO Cancer Center, 49000 Angers, France; (L.F.); (M.L.); (C.G.); (M.C.); (M.C.)
| | - Pascal Jézéquel
- CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, 44000 Nantes, France; (C.R.); (P.J.)
- ICO Cancer Center, 49000 Angers, France; (L.F.); (M.L.); (C.G.); (M.C.); (M.C.)
| | - Mario Campone
- ICO Cancer Center, 49000 Angers, France; (L.F.); (M.L.); (C.G.); (M.C.); (M.C.)
- CRCINA, University of Angers, INSERM UMR1232, CNRS-ERL6001, 49000 Angers, France
| | - Nicolas Normand
- LS2N, University of Nantes, CNRS, 44000 Nantes, France; (C.F.); (N.N.)
| | - Mathieu Rubeaux
- Keosys Medical Imaging, 13 Imp. Serge Reggiani, 44815 Saint-Herblain, France; (G.S.); (A.B.); (M.R.)
| |
Collapse
|
21
|
Matsubara T, Nakajima E, Namikawa H, Ono S, Takada I, Ohira T, Morishita Y, Miyazaki T, Furukawa K, Ikeda N. Investigation of EGFR mutations in non-small cell lung cancer usually undetectable by PCR methods. Mol Clin Oncol 2021; 16:15. [PMID: 34881035 PMCID: PMC8637854 DOI: 10.3892/mco.2021.2447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/10/2021] [Indexed: 12/25/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) mutations are the most significant genomic drivers of non-small cell lung cancer (NSCLC) and determine the efficacy of EGFR tyrosine kinase inhibitor (EGFR-TKI) therapy. PCR methods are used clinically for the detection of EGFR mutations. The Scorpion Amplification Refractory Mutation System (Scorpion-ARMS) and the cobas® EGFR Mutation Test v2 (cobas v2) are widely used PCR methods. However, those PCR methods only selectively detect the common EGFR mutations. The aim of the present study was to reveal the true frequency of EGFR mutations in NSCLC by investigating EGFR mutations usually undetectable by PCR methods by using direct sequencing. A total of 70 Japanese patients who underwent lung resection for NSCLC between September 2016 and March 2019 were included in the present study. Subsequently, PCR methods and direct sequencing were performed. In total, 29 mutations were detected by cobas v2. In total, 41 patients were identified as EGFR wild-type by cobas v2, among whom direct sequencing detected mutations in 3 patients. Subsequent Scorpion-ARMS was performed in the 3 patients in whom direct sequencing detected mutations. In total, one exon 21 L858R + G863D compound mutation was identified as a L858R single mutation, and two other mutations were undetectable. Moreover, 1 patient who was ‘wild-type’ on cobas v2 but ‘EGFR mutation’ on direct sequencing developed recurrence after surgery and responded to EGFR-TKI treatment. In present study, the percentage of undetectable EGFR mutations by cobas v2 was 9.4% in 32 mutations. It was inferred that the cause of the discrepancy in the mutation type (L858R + G863D in exon 21, and L858R in exon 21) between cobas v2 and Scorpion ARMS was due to the different limit of detection between these two PCR methods. In conclusion, the findings of the present study suggested that a selective mutation detection method may decrease the opportunity of patients with NSCLC to receive EGFR-TKI therapy. Thus, the development of a screening test to determine the EGFR status as wild-type or mutant is required for EGFR-TKI therapy.
Collapse
Affiliation(s)
- Taisuke Matsubara
- Department of Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Eiji Nakajima
- Department of Thoracic Surgery, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki 300-0395, Japan
| | - Haruka Namikawa
- Department of Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Shotaro Ono
- Department of Thoracic Surgery, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki 300-0395, Japan
| | - Ikki Takada
- Department of Thoracic Surgery, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki 300-0395, Japan
| | - Tatsuo Ohira
- Department of Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Yukio Morishita
- Department of Diagnostic Pathology, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki 300-0395, Japan
| | - Teruo Miyazaki
- Department of Joint Research Center, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki 300-0395, Japan
| | - Kinya Furukawa
- Department of Thoracic Surgery, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki 300-0395, Japan
| | - Norihiko Ikeda
- Department of Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| |
Collapse
|
22
|
Aksu A, Çapa Kaya G. Is SUV Corrected for Lean Body Mass Superior to SUV of Body Weight in 68Ga-PSMA PET/CT? Mol Imaging Radionucl Ther 2021; 30:144-149. [PMID: 34658229 PMCID: PMC8522520 DOI: 10.4274/mirt.galenos.2021.59254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objectives: This study aimed to investigate the relationship between the standard uptake value (SUV) of body weight and SUV corrected for lean body mass (SUL) parameters obtained from the prostate gland in gallium-68 (68Ga)-prostate-specific membrane antigen (PSMA) positron emission tomography-computed tomography (PET/CT) with Gleason grade (GG) groups, D’Amico risk groups, and presence of metastases. Methods: Patients with prostate adenocarcinoma who underwent 68Ga-PSMA PET/CT for staging at our center between February 2017 and October 2018 were evaluated retrospectively. Maximum SUV (SUVmax), SUVpeak, SULmax, SULpeak, SUVmean, and SULmean values of the prostate tumor were obtained. The difference in these values between GG groups (≥3, <3) and D’Amico risk (low-moderate/high) groups was evaluated with the Mann-Whitney U test. The area under the curve values of SUV and SUL parameters were compared. In addition, SUVmean and SULmean values were obtained from the right liver lobe, and their correlation with body weight was evaluated. Results: A total of 79 patients were included in the study. Significant differences were found in the prostate SUVmax, SULmax, SUVpeak, SULpeak, SUVmean, and SULmean values between the GG (≥3 and <3) groups and between D’Amico risk (low-moderate and high) groups. However, no significant difference was found in the discriminative power of any SUV or SUL parameter when compared with each other. A significant difference in any SUV and SUL parameters was found in patients with and without metastasis. Neither liver SUVmean value nor SULmean value correlated with the body weight. Conclusion: The superiority of SUL values obtained from 68Ga-PSMA PET to SUV was not determined in our study. SUV parameters can also be used for quantitative analysis in 68Ga-PSMA PET.
Collapse
Affiliation(s)
- Ayşegül Aksu
- University of Health Sciences Turkey, Başakşehir Çam and Sakura City Hospital, Clinic of of Nuclear Medicine, İstanbul, Turkey
| | - Gamze Çapa Kaya
- Dokuz Eylül University Faculty of Medicine, Department of Nuclear Medicine, İzmir, Turkey
| |
Collapse
|
23
|
Schierz JH, Sarikaya I, Albatineh AN, Sarikaya A. Assessing the correlation between 68Ga-PSMA-11 renal PET parameters and renal function tests. J Nucl Med Technol 2021; 50:43-48. [PMID: 34330809 DOI: 10.2967/jnmt.121.262462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/07/2021] [Indexed: 11/16/2022] Open
Abstract
Aim: 68Ga -PSMA ligands are used for prostate cancer but also show high renal cortical uptake. In this study, we aimed to assess if there is any correlation between renal PSMA PET parameters and renal function tests using the images of prostate cancer patients. Methods: 68Ga-PSMA-11 PET/CT images of the patients with prostate cancer were retrospectively evaluated. The following PET parameters were obtained: SUVmax, SUVmean, SULmax, SULmean, volume, TLGSUL and counts of both kidneys as well as SUVmean of liver, blood pool and spleen. Total TLGSUL, total volume, kidney to liver and kidney to blood pool ratios were calculated. Patient's creatinine values were obtained and GFR was calculated using the MDRD formula. Statistical analysis was performed to understand if there is a correlation between above parameters and renal function tests. Results: Twenty five patients were included in this study. GFR was significantly/positively correlated and creatinine was significantly/negatively correlated with renal SUV/liver SUV and renal SUV/blood pool SUV ratios. GFR was marginally positively correlated with renal SULmean and creatinine was marginally negatively correlated with total TLGSUL. Total renal parenchymal volume was significantly and directly (positively) associated with GFR and significantly and inversely (negatively) associated with creatinine. Conclusion: Renal 68Ga-PSMA uptake appears to be correlated with renal function tests. Our method of measuring approximate renal parenchymal volume on PET image appears to be reliable.
Collapse
|
24
|
Bos-Liedke A, Cegla P, Matuszewski K, Konstanty E, Piotrowski A, Gross M, Malicki J, Kozak M. Differences among [ 18F]FDG PET-derived parameters in lung cancer produced by three software packages. Sci Rep 2021; 11:13942. [PMID: 34230642 PMCID: PMC8260625 DOI: 10.1038/s41598-021-93436-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Investigation of differences in derived [18F]FDG PET metabolic and volumetric parameters among three different software programs in lung cancer. A retrospective analysis was performed on a group of 98 lung cancer patients who underwent a baseline [18F]FDG PET/CT study. To assess appropriate delineation methods, the NEMA phantom study was first performed using the following software: Philips EBW (Extended Brilliance Workstation), MIM Software and Rover. Based on this study, the best cut-off methods (dependent on tumour size) were selected, extracted and applied for lung cancer delineation. Several semiquantitative [18F]FDG parameters (SUVmax, SUVmean, TLG and MTV) were assessed and compared among the three software programs. The parameters were assessed based on body weight (BW), lean body mass (LBM) and Bq/mL. Statistically significant differences were found in SUVmean (LBM) between MIM Software and Rover (4.62 ± 2.15 vs 4.84 ± 1.20; p < 0.005), in SUVmean (Bq/mL) between Rover and Philips EBW (21,852.30 ± 21,821.23 vs 19,274.81 ± 13,340.28; p < 0.005) and Rover and MIM Software (21,852.30 ± 21,821.23 vs 19,399.40 ± 10,051.30; p < 0.005), and in MTV between MIM Software and Philips EBW (19.87 ± 25.83 vs 78.82 ± 228.00; p = 0.0489). This study showed statistically significant differences in the estimation of semiquantitative parameters using three independent image analysis tools. These findings are important for performing further diagnostic and treatment procedures in lung cancer patients.
Collapse
Affiliation(s)
- Agnieszka Bos-Liedke
- Department of Macromolecular Physics, Adam Mickiewicz University, 61-614, Poznan, Poland
| | - Paulina Cegla
- Department of Nuclear Medicine, Greater Poland Cancer Centre, 61-866, Poznan, Poland.
| | | | - Ewelina Konstanty
- Department of Medical Physics, Greater Poland Cancer Centre, 61-866, Poznan, Poland
| | - Adam Piotrowski
- Department of Macromolecular Physics, Adam Mickiewicz University, 61-614, Poznan, Poland
| | - Magdalena Gross
- Department of Macromolecular Physics, Adam Mickiewicz University, 61-614, Poznan, Poland
| | - Julian Malicki
- Department of Medical Physics, Greater Poland Cancer Centre, 61-866, Poznan, Poland
- Chair, Department of Electroradiology, Poznan University of Medical Science, 61-701, Poznan, Poland
| | - Maciej Kozak
- Department of Macromolecular Physics, Adam Mickiewicz University, 61-614, Poznan, Poland
| |
Collapse
|
25
|
Can the BMI-based dose regimen be used to reduce injection activity and to obtain a constant image quality in oncological patients by 18F-FDG total-body PET/CT imaging? Eur J Nucl Med Mol Imaging 2021; 49:269-278. [PMID: 34185138 DOI: 10.1007/s00259-021-05462-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/10/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE PET image quality is influenced by the patient size according to the current guideline. The study aimed to propose an optimized dose regimen to yield a constant image quality independent of patient habitus to meet the clinical needs. METHODS A first patient cohort of 78 consecutive oncological patients (59.7 ± 13.7 years) who underwent a total-body PET/CT scan were retrospectively enrolled to develop the regimen. The patients were randomly distributed in four body mass index (BMI) groups according to the World Health Organization (WHO) criteria. The liver SNR (signal-to-noise ratio, SNRL) was obtained by manually drawing regions of interest (ROIs) and normalized (SNRnorm) by the product of injected activity and acquisition time. Fits of SNRnorm against different patient-dependent parameters were performed to determine the best correlating parameter and fit method. A qualitative assessment on image quality was performed using a 5-point Likert scale to determine the acceptable threshold of SNRL. Thus, an optimized regimen was proposed and validated by a second patient cohort consisted of prospectively enrolled 38 oncological patients. RESULTS The linear fit showed SNRnorm had the strongest correlation (R2 = 0.69) with the BMI than other patient-dependent parameters and fit method. The qualitative assessment indicated a SNRL value of 14.0 as an acceptable threshold to achieve sufficient image quality. The optimized dose regimen was determined as a quadratic relation with BMI: injected activity (MBq) = 39.2 (MBq)/(- 0.03*BMI + 1.49)2. In the validation study, the SNRL no longer decreased with the increase of BMI. There was no significant difference of the image quality regarding the value of SNRL between different BMI groups (p > 0.05). In addition, the injected activity was reduced by 75.6 ± 2.9%, 72.1 ± 4.0%, 67.1 ± 4.4%, and 64.8 ± 3.5% compared with the first cohort for the four BMI groups, respectively. CONCLUSION The study proposed a quadratic relation between the 18F-FDG injected activity and the patient's BMI for total-body 18F-FDG PET imaging. In this regimen, the image quality can maintain in a constant level independent of patient habitus and meet the clinical requirement with a reduced injected activity.
Collapse
|
26
|
Aujay G, Debordeaux F, Blanc JF, Lapuyade B, Papadopoulos P, Bordenave L, Trillaud H, Pinaquy JB. 18F-choline PET-computed tomography for the prediction of early treatment responses to transarterial radioembolization in patients with hepatocellular carcinoma. Nucl Med Commun 2021; 42:633-638. [PMID: 33660694 DOI: 10.1097/mnm.0000000000001383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Transarterial radioembolization (TARE) is widely used for the treatment of hepatocellular carcinoma (HCC), but early treatment response can be very difficult to assess. The aim was to evaluate 18F-fluorocholine PET/computed tomography (CT) to assess the treatment response in patients with intermediate or locally advanced HCC. METHODS Between March 2019 and July 2020, nine HCC patients treated with TARE, who underwent PET/CT at baseline and 1 month after treatment, were enrolled. The maximum, mean (SUVmean), and peak (SUVpeak) standardized uptake value (SUV), SUV normalized by lean body mass (SUL), and total lesion glycolysis (TLG) were measured. Statistical analysis used the Mann-Whitney test to evaluate the differences in parameters between responders (partial and complete response) and nonresponders (stable or progressive disease) at the 6-month follow-up, according to the modified Response Evaluation Criteria in Solid Tumors. RESULTS Three patients were nonresponders (progressive disease and stable disease) and six were responders. Delta SUVmean, delta SUL, and delta TLG could predict an early response (P = 0.02, P = 0.04, and P = 0.02, respectively). None of the pre-therapeutic parameters were correlated with the response. Post-therapeutic SUL, SUVmean, TLG, and SUVpeak were also predictive of the response. CONCLUSIONS Our preliminary results showed that changes in certain metabolic parameters (from baseline PET to 1-month PET) are predictive of the response to TARE in HCC (Delta SUVmean, delta TLG, and delta SUL). The absence of post-treatment inflammation could lead to a better prediction than MRI evaluation. This study suggests that 1-month 18F-choline PET/CT could modify the clinical management predicting responders.Video Abstract: http://links.lww.com/NMC/A193.
Collapse
|
27
|
Zhao J, Xue Q, Chen X, You Z, Wang Z, Yuan J, Liu H, Hu L. Evaluation of SUVlean consistency in FDG and PSMA PET/MR with Dixon-, James-, and Janma-based lean body mass correction. EJNMMI Phys 2021; 8:17. [PMID: 33598849 PMCID: PMC7889776 DOI: 10.1186/s40658-021-00363-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/04/2021] [Indexed: 01/22/2023] Open
Abstract
PURPOSE To systematically evaluate the consistency of various standardized uptake value (SUV) lean body mass (LBM) normalization methods in a clinical positron emission tomography/magnetic resonance imaging (PET/MR) setting. METHODS SUV of brain, liver, prostate, parotid, blood, and muscle were measured in 90 18F-FDG and 28 18F-PSMA PET/MR scans and corrected for LBM using the James, Janma (short for Janmahasatian), and Dixon approaches. The prospective study was performed from December 2018 to August 2020 at Shanghai East Hospital. Forty dual energy X-ray absorptiometry (DXA) measurements of non-fat mass were used as the reference standard. Agreement between different LBM methods was assessed by linear regression and Bland-Altman statistics. SUV's dependency on BMI was evaluated by means of linear regression and Pearson correlation. RESULTS Compared to DXA, the Dixon approach presented the least bias in LBM/weight% than James and Janma models (bias 0.4±7.3%, - 8.0±9.4%, and - 3.3±8.3% respectively). SUV normalized by body weight (SUVbw) was positively correlated with body mass index (BMI) for both FDG (e.g., liver: r = 0.45, p < 0.001) and PSMA scans (r = 0.20, p = 0.31), while SUV normalized by lean body mass (SUVlean) revealed a decreased dependency on BMI (r = 0.22, 0.08, 0.14, p = 0.04, 0.46, 0.18 for Dixon, James, and Janma models, respectively). The liver SUVbw of obese/overweight patients was significantly larger (p < 0.001) than that of normal patients, whereas the bias was mostly eliminated in SUVlean. One-way ANOVA showed significant difference (p < 0.001) between SUVlean in major organs measured using Dixon method vs James and Janma models. CONCLUSION Significant systematic variation was found using different approaches to calculate SUVlean. A consistent correction method should be applied for serial PET/MR scans. The Dixon method provides the most accurate measure of LBM, yielding the least bias of all approaches when compared to DXA.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Qiaoyi Xue
- Central Research Institute, United Imaging Healthcare Group, Shanghai, China
| | - Xing Chen
- Department of Nuclear Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiwen You
- Department of Nuclear Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhe Wang
- Central Research Institute, United Imaging Healthcare Group, Shanghai, China
| | - Jianmin Yuan
- Central Research Institute, United Imaging Healthcare Group, Shanghai, China
| | - Hui Liu
- Central Research Institute, United Imaging Healthcare Group, Shanghai, China
| | - Lingzhi Hu
- Central Research Institute, United Imaging Healthcare Group, Shanghai, China
| |
Collapse
|
28
|
Shang J, Tan Z, Cheng Y, Tang Y, Guo B, Gong J, Ling X, Wang L, Xu H. A method for evaluation of patient-specific lean body mass from limited-coverage CT images and its application in PERCIST: comparison with predictive equation. EJNMMI Phys 2021; 8:12. [PMID: 33555478 PMCID: PMC7870732 DOI: 10.1186/s40658-021-00358-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 01/28/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Standardized uptake value (SUV) normalized by lean body mass ([LBM] SUL) is recommended as metric by PERCIST 1.0. The James predictive equation (PE) is a frequently used formula for LBM estimation, but may cause substantial error for an individual. The purpose of this study was to introduce a novel and reliable method for estimating LBM by limited-coverage (LC) CT images from PET/CT examinations and test its validity, then to analyse whether SUV normalised by LC-based LBM could change the PERCIST 1.0 response classifications, based on LBM estimated by the James PE. METHODS First, 199 patients who received whole-body PET/CT examinations were retrospectively retrieved. A patient-specific LBM equation was developed based on the relationship between LC fat volumes (FVLC) and whole-body fat mass (FMWB). This equation was cross-validated with an independent sample of 97 patients who also received whole-body PET/CT examinations. Its results were compared with the measurement of LBM from whole-body CT (reference standard) and the results of the James PE. Then, 241 patients with solid tumours who underwent PET/CT examinations before and after treatment were retrospectively retrieved. The treatment responses were evaluated according to the PE-based and LC-based PERCIST 1.0. Concordance between them was assessed using Cohen's κ coefficient and Wilcoxon's signed-ranks test. The impact of differing LBM algorithms on PERCIST 1.0 classification was evaluated. RESULTS The FVLC were significantly correlated with the FMWB (r=0.977). Furthermore, the results of LBM measurement evaluated with LC images were much closer to the reference standard than those obtained by the James PE. The PE-based and LC-based PERCIST 1.0 classifications were discordant in 27 patients (11.2%; κ = 0.823, P=0.837). These discordant patients' percentage changes of peak SUL (SULpeak) were all in the interval above or below 10% from the threshold (±30%), accounting for 43.5% (27/62) of total patients in this region. The degree of variability is related to changes in LBM before and after treatment. CONCLUSIONS LBM algorithm-dependent variability in PERCIST 1.0 classification is a notable issue. SUV normalised by LC-based LBM could change PERCIST 1.0 response classifications based on LBM estimated by the James PE, especially for patients with a percentage variation of SULpeak close to the threshold.
Collapse
Affiliation(s)
- Jingjie Shang
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Road, Guangzhou, 510630, China
| | - Zhiqiang Tan
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Road, Guangzhou, 510630, China
| | - Yong Cheng
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Road, Guangzhou, 510630, China
| | - Yongjin Tang
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Road, Guangzhou, 510630, China
| | - Bin Guo
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Road, Guangzhou, 510630, China
| | - Jian Gong
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Road, Guangzhou, 510630, China
| | - Xueying Ling
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Road, Guangzhou, 510630, China
| | - Lu Wang
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Road, Guangzhou, 510630, China
| | - Hao Xu
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Road, Guangzhou, 510630, China.
| |
Collapse
|
29
|
Multimodality In Vivo Imaging of Perfusion and Glycolysis in a Rat Model of C6 Glioma. Mol Imaging Biol 2021; 23:516-526. [PMID: 33534038 DOI: 10.1007/s11307-021-01585-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/18/2021] [Accepted: 01/25/2021] [Indexed: 12/20/2022]
Abstract
PURPOSE Chemical exchange saturation transfer MRI using an infusion of glucose (glucoCEST) is sensitive to the distribution of glucose in vivo; however, whether glucoCEST is more related to perfusion or glycolysis is still debatable. We compared glucoCEST to computed tomography perfusion (CTP), [18F] fluorodeoxyglucose positron emission tomography (FDG-PET), and hyperpolarized [1-13C] pyruvate magnetic resonance spectroscopy imaging (MRSI) in a C6 rat model of glioma to determine if glucoCEST is more strongly correlated with measurements of perfusion or glycolysis. METHODS 106 C6 glioma cells were implanted in Wistar rat brains (n = 11). CTP (including blood volume, BV; blood flow, BF; and permeability surface area product, PS) and FDG-PET standardized uptake value (SUV) were acquired at 11 to 13 days post-surgery. GlucoCEST measurements (∆CEST) were acquired the following day on a 9.4 T MRI before and after an infusion of glucose solution. This was followed by MRSI on a 3.0 T MRI after the injection of hyperpolarized [1-13C] pyruvate to generate regional maps of the lactate:pyruvate ratio (Lac:Pyr). Pearson's correlations between glucoCEST, CTP, FDG-PET, and Lac:Pyr ratio were evaluated. RESULTS Tumors had significantly higher SUV, BV, and PS than the contralateral brain. Tumor ∆CEST was most strongly correlated with CTP measurements of BV (ρ = 0.74, P = 0.01) and PS (ρ = 0.55, P = 0.04). No significant correlation was found between glycolysis measurements of SUV or Lac:Pyr with tumor ∆CEST. PS significantly correlated with SUV (ρ = 0.58, P = 0.005) and Lac:Pyr (ρ = 0.75, P = 0.005). BV significantly correlated with Lac:Pyr (ρ = 0.57, P = 0.02), and BF significantly correlated with SUV (ρ = 0.49, P = 0.02). CONCLUSION This study determined that glucoCEST is more strongly correlated to measurements of perfusion than glycolysis. GlucoCEST measurements have additional confounds, such as sensitivity to changing pH, that merit additional investigation.
Collapse
|
30
|
Loeliger RC, Maushart CI, Gashi G, Senn JR, Felder M, Becker AS, Müller J, Balaz M, Wolfrum C, Burger IA, Betz MJ. Relation of diet-induced thermogenesis to brown adipose tissue activity in healthy men. Am J Physiol Endocrinol Metab 2021; 320:E93-E101. [PMID: 33225717 DOI: 10.1152/ajpendo.00237.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human brown adipose tissue (BAT) is a thermogenic tissue activated by the sympathetic nervous system in response to cold exposure. It contributes to energy expenditure (EE) and takes up glucose and lipids from the circulation. Studies in rodents suggest that BAT contributes to the transient rise in EE after food intake, so-called diet-induced thermogenesis (DIT). We investigated the relationship between human BAT activity and DIT in response to glucose intake in 17 healthy volunteers. We assessed DIT, cold-induced thermogenesis (CIT), and maximum BAT activity at three separate study visits within 2 wk. DIT was measured by indirect calorimetry during an oral glucose tolerance test. CIT was assessed as the difference in EE after cold exposure of 2-h duration as compared with warm conditions. Maximal activity of BAT was assessed by 18-F-fluoro-deoxyglucose (18F-FDG) 18F-FDG-PET/MRI after cold exposure and concomitant pharmacological stimulation with mirabegron. Seventeen healthy men (mean age = 23.4 yr, mean body mass index = 23.2 kg/m2) participated in the study. EE increased from 1,908 (±181) kcal/24 h to 2,128 (±277) kcal/24 h (P < 0.0001, +11.5%) after mild cold exposure. An oral glucose load increased EE from 1,911 (±165) kcal/24 h to 2,096 (±167) kcal/24 h at 60 min (P < 0.0001, +9.7%). The increase in EE in response to cold was significantly associated with BAT activity (R2 = 0.43, P = 0.004). However, DIT was not associated with BAT activity (R2 = 0.015, P = 0.64). DIT after an oral glucose load was not associated with stimulated 18F-FDG uptake into BAT, suggesting that DIT is independent from BAT activity in humans (Clinicaltrials.gov Registration No. NCT03189511).NEW & NOTEWORTHY Cold-induced thermogenesis (CIT) was related to BAT activity as determined by FDG-PET/MRI after stimulation of BAT. Diet-induced thermogenesis (DIT) was not related to stimulated BAT activity. Supraclavicular skin temperature was related to CIT but not to DIT. DIT in humans is probably not a function of BAT.
Collapse
Affiliation(s)
- Rahel Catherina Loeliger
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Claudia Irene Maushart
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Gani Gashi
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jaël Rut Senn
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Martina Felder
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Anton S Becker
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich/University of Zurich, Zurich, Switzerland
| | - Julian Müller
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich/University of Zurich, Zurich, Switzerland
| | - Miroslav Balaz
- Institute of Food, Nutrition, and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition, and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Irene A Burger
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich/University of Zurich, Zurich, Switzerland
| | - Matthias Johannes Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
31
|
Wahl RL, Hicks RJ. PET Diagnosis and Response Monitoring in Oncology. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00048-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
32
|
The early prediction of pathological response to neoadjuvant chemotherapy and prognosis: comparison of PET Response Criteria in Solid Tumors and European Organization for Research and Treatment of Cancer criteria in breast cancer. Nucl Med Commun 2020; 41:280-287. [PMID: 32032193 DOI: 10.1097/mnm.0000000000001145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To compare the predictive value of European Organization for Research and Treatment of Cancer (EORTC) criteria and PET Response Criteria in Solid Tumors (PERCIST), for the pathological response and prognosis of patients with breast cancer receiving neoadjuvant chemotherapy (NAC). METHODS Consecutive PET/computed tomography scans in 128 operable female patients at baseline and after two courses of NAC were performed. Patients were categorized by complete metabolic response (CMR) and non-CMR groups using 2 PET criteria. CMR and non-CMR were used to predict pathological complete response (pCR) by diagnostic test evaluation, and to predict progression-free survival (PFS) using Kaplan-Meier plots and Cox proportional hazards regression. RESULTS Ninety-two patients were finally analyzed. The sensitivity, specificity, and accuracy for pCR prediction were 69.7, 76.3, and 73.9% with EORTC criteria, and 69.7, 77.9, and 75.0% with PERCIST, respectively. Peak standardized uptake value normalized to lean body mass (SULpeak), maximum standardized uptake value (SUVmax), total lesion glycolysis (TLG), and metabolic tumor volume (MTV) were pCR response with accuracy of 70.7, 60.0, 75.0, and 71.4%, respectively. CMR by the EORTC (P = 0.021) and PERCIST (P = 0.007) was significantly related to a longer PFS. The univariate and multivariate analysis suggested that CMR by PERCIST was an independent predictor of recurrence (P = 0.008). CONCLUSION EORTC criteria and PERCIST had early predictive value to long-term outcome, but moderate value for pCR. Furthermore, PERCIST might show more potential than the EORTC criteria and conventional PET-based parameters to predict prognosis in breast cancer patients following two cycles of neoadjuvant chemotherapy.Video abstract: see http://links.lww.com/NMC/A162.
Collapse
|
33
|
Rohani MFM, Yonan SNM, Tagiling N, Zainon WMNW, Udin Y, Nawi NM. Standardized Uptake Value from Semiquantitative Bone Single-Photon Emission Computed Tomography/Computed Tomography in Normal Thoracic and Lumbar Vertebrae of Breast Cancer Patients. Asian Spine J 2020; 14:629-638. [PMID: 32213791 PMCID: PMC7595810 DOI: 10.31616/asj.2019.0308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 11/28/2022] Open
Abstract
STUDY DESIGN Retrospective study. PURPOSE This study aims to semiquantitatively evaluate the standardized uptake value (SUV) of 99mTc-methylene diphosphonate (MDP) radionuclide tracer in the normal vertebrae of breast cancer patients using an integrated single-photon emission computed tomography (SPECT)/computed tomography (CT) scanner. OVERVIEW OF LITERATURE Molecular imaging techniques using gamma cameras and stand-alone SPECT have traditionally been utilized to evaluate metastatic bone diseases. However, these methods lack quantitative analysis capabilities, impeding accurate uptake characterization. METHODS A total of 30 randomly selected female breast cancer patients were enrolled in this study. The SUV mean (SUVmean) and SUV maximum (SUVmax) values for 286 normal vertebrae at the thoracic and lumbar levels were calculated based on the patients' body weight (BW), body surface area (BSA), and lean body mass (LBM). Additionally, 106 degenerative joint disease (DJD) lesions of the spine were also characterized, and both their BW SUVmean and SUVmax values were obtained. A receiver operating characteristic (ROC) curve analysis was then performed to determine the cutoff value of SUV for differentiating DJD from normal vertebrae. RESULTS The mean±standard deviations for the SUVmean and SUVmax in the normal vertebrae displayed a relatively wide variability: 3.92±0.27 and 6.51±0.72 for BW, 1.05±0.07 and 1.75±0.17 for BSA, and 2.70±0.19 and 4.50±0.44 for LBM, respectively. Generally, the SUVmean had a lower coefficient of variation than the SUVmax. For DJD, the mean±standard deviation for the BW SUVmean and SUVmax was 5.26±3.24 and 7.50±4.34, respectively. Based on the ROC curve, no optimal cutoff value was found to differentiate DJD from normal vertebrae. CONCLUSIONS In this study, the SUV of 99mTc-MDP was successfully determined using SPECT/CT. This research provides an approach that could potentially aid in the clinical quantification of radionuclide uptake in normal vertebrae for the management of breast cancer patients.
Collapse
Affiliation(s)
- Mohd Fazrin Mohd Rohani
- Department of Nuclear Medicine, School of Medical Sciences, Universiti Sains Malaysia (Health Campus), Kubang Kerian, Malaysia
- Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Siti Nurshahirah Mohd Yonan
- Medical Radiation Program, School of Health Sciences, Universiti Sains Malaysia (Health Campus), Kubang Kerian, Malaysia
| | - Nashrulhaq Tagiling
- Department of Nuclear Medicine, School of Medical Sciences, Universiti Sains Malaysia (Health Campus), Kubang Kerian, Malaysia
| | | | - Yusri Udin
- Department of Nuclear Medicine, School of Medical Sciences, Universiti Sains Malaysia (Health Campus), Kubang Kerian, Malaysia
| | - Norazlina Mat Nawi
- Department of Nuclear Medicine, School of Medical Sciences, Universiti Sains Malaysia (Health Campus), Kubang Kerian, Malaysia
- Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
34
|
Rudra S, Fuser D, DeWees TA, Wan L, Gang M, Hui CY, Rao YJ, Siegel BA, Dehdashti F, Mutch DG, Powell MA, Schwarz JK, Grigsby PW, Chen DL, Markovina S. Radiologic Assessment of Groin Lymph Nodes in Pelvic Malignancies. Int J Gynecol Cancer 2020; 30:947-953. [PMID: 32487684 DOI: 10.1136/ijgc-2020-001363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/26/2020] [Accepted: 05/12/2020] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Metastatic involvement of groin nodes can alter radiation therapy planning for pelvic tumors. 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) can identify nodal metastases; however, interpretation of PET/CT-positive nodes can be complicated by non-malignant processes. We evaluated quantitative metrics as methods to identify groin metastases in patients with pelvic tumors by comparison with standard subjective interpretive criteria, with pathology as the reference standard. METHODS We retrospectively identified patients with vulvar, vaginal, or anal cancers who underwent 18F-FDG PET/CT before pathologic evaluation of groin nodes between 2007 and 2017. Because patho-radiologic correlation was not possible for every node, one index node identified on imaging was selected for each groin. For each index node, standardized uptake value measurements, total lesion glycolysis, metabolic tumor volume, CT-based volume, and short and long axes were measured. Multivariate logistic regression was used to identify metrics predictive for pathologically positive groins and generate a probabilistic model. Area under the receiver-operating characteristic curves (AUCs) for the model were compared with clinical interpretation from the diagnostic report via a Wald's χ2 test. RESULTS Of 55 patients identified for analysis, 75 groins had pathologic evaluation resulting in 75 index groin nodes for analysis with 35 groins pathologically positive for malignancy. Logistic regression identified mean standardized-uptake-value (50% threshold) and short-axis length as the most predictive imaging metrics for metastatic nodal involvement. The probabilistic model performed better at predicting pathologic involvement compared with standard clinical interpretation on analysis (AUC 0.91, 95% CI 0.84 to 0.97 vs 0.80, 95% CI 0.71 to 0.89; p<0.01). DISCUSSION Accuracy of 18F-FDG PET/CT for detecting groin nodal metastases in patients with pelvic tumors may be improved with the use of quantitative metrics. Improving prediction of nodal metastases can aid with appropriate selection of patients for pathologic node evaluation and guide radiation volumes and doses.
Collapse
Affiliation(s)
- Soumon Rudra
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Dominique Fuser
- Mallinckrodt Institute of Radiology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Todd A DeWees
- Division of Biomedical Statistics and Informatics, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Leping Wan
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Margery Gang
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Caressa Y Hui
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | - Yuan J Rao
- Department of Radiation Oncology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Barry A Siegel
- Mallinckrodt Institute of Radiology, Washington University in Saint Louis, Saint Louis, Missouri, USA.,Alvin J Siteman Cancer Center, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - Farrokh Dehdashti
- Mallinckrodt Institute of Radiology, Washington University in Saint Louis, Saint Louis, Missouri, USA.,Alvin J Siteman Cancer Center, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - David G Mutch
- Alvin J Siteman Cancer Center, Washington University in Saint Louis, St. Louis, Missouri, USA.,Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - Matthew A Powell
- Alvin J Siteman Cancer Center, Washington University in Saint Louis, St. Louis, Missouri, USA.,Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - Julie K Schwarz
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA.,Alvin J Siteman Cancer Center, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - Perry W Grigsby
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA.,Mallinckrodt Institute of Radiology, Washington University in Saint Louis, Saint Louis, Missouri, USA.,Alvin J Siteman Cancer Center, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - Delphine L Chen
- Mallinckrodt Institute of Radiology, Washington University in Saint Louis, Saint Louis, Missouri, USA.,Department of Radiology, University of Washington, Seattle Cancer Care Alliance, Seattle, Washington, USA
| | - Stephanie Markovina
- Department of Radiation Oncology, Washington University in Saint Louis, Saint Louis, Missouri, USA .,Alvin J Siteman Cancer Center, Washington University in Saint Louis, St. Louis, Missouri, USA
| |
Collapse
|
35
|
Abstract
OBJECTIVE Physiological fluorodeoxyglucose (FDG) uptake of spinal cord needs to be correctly recognized during evaluation of whole-body PET scans, especially for oncological cases. Our aim was to analyze physiological cord FDG uptake and its relation to gender, age, body weight, environmental temperature and time to imaging. MATERIALS AND METHODS PET scans of 254 patients in a single year, one patient for every working day were retrospectively selected. Temperature data were obtained from meteorology recordings. Maximum standard uptake value (SUVmax) of spinal cord at cervical and lower thoracic levels were noted. Spinal canal at L5 level, cerebellum and liver were used for normalization. Correlations with age, body weight, time to imaging and environmental temperature were analyzed. RESULTS Cervical SUV was higher than thoracic SUV (2.5-2.3). Cervical and lower thoracic SUV's were strongly correlated, highest when corrected with L5 level vertebral canal and liver (corr coeff 0.84 and 0.75) and lowest with cerebellum (corr coeff 0.4). Cervical spinal cord FDG uptake was higher for females than males (2.6 to 2.4). Temperature and age did not change spinal cord uptake. There were weak positive correlations with body weight (corr coeff 0.16 and 0.28, cervical and thoracic). There was weak negative correlation of cervical uptake with time to imaging (corr coeff -0.17). CONCLUSION Spinal cord FDG uptake at cervical and lower thoracic levels are strongly correlated. Females have slightly higher cervical SUV. Age and temperature does not change spinal cord FDG uptake in adults. Cord SUV's slightly increased with body weight.
Collapse
|
36
|
Leitner BP, Perry RJ. The Impact of Obesity on Tumor Glucose Uptake in Breast and Lung Cancer. JNCI Cancer Spectr 2020; 4:pkaa007. [PMID: 32368718 PMCID: PMC7190208 DOI: 10.1093/jncics/pkaa007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/16/2019] [Accepted: 02/04/2020] [Indexed: 12/18/2022] Open
Abstract
Obesity confers an increased incidence and poorer clinical prognosis in more than 10 cancer types. Paradoxically, obesity may provide protection from poor outcomes in lung cancer. Mechanisms for the obesity-cancer links are not fully elucidated, with altered glucose metabolism being a promising candidate. Using 18F-fluorodeoxyglucose positron-emission-tomography/computed tomography images from The Cancer Imaging Archive, we explored the relationship between body mass index (BMI) and glucose metabolism in several cancers. In 188 patients (BMI mean [SD] = 27.7 [5.1], range = 17.4-49.3 kg/m2), higher BMI was associated with greater tumor glucose uptake in breast cancer (r = 0.36; P = .02) and with lower tumor glucose uptake in non-small cell lung cancer (r = -0.26; P = .048) using two-sided Pearson correlations. No relationship was observed in soft tissue sarcoma or squamous cell carcinoma. Harnessing the National Cancer Institute's open-access database, we demonstrate altered tumor glucose metabolism as a potential mechanism for the detrimental and protective effects of obesity on breast and lung cancer, respectively.
Collapse
Affiliation(s)
- Brooks P Leitner
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
| | - Rachel J Perry
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
| |
Collapse
|
37
|
Velez EM, Desai B, Ji L, Quinn DI, Colletti PM, Jadvar H. Comparative prognostic implication of treatment response assessments in mCRPC: PERCIST 1.0, RECIST 1.1, and PSA response criteria. Am J Cancer Res 2020; 10:3254-3262. [PMID: 32194866 PMCID: PMC7053201 DOI: 10.7150/thno.39838] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/15/2020] [Indexed: 12/27/2022] Open
Abstract
Accurate appraisal of treatment response in metastatic castrate-resistant prostate cancer (mCRPC) is challenging in view of remarkable tumor heterogeneity and the available choices among many established and novel therapeutic approaches. The purpose of this single-center prospective study was to evaluate the comparative prognostic utility of PERCIST 1.0 in predicting overall survival (OS) in patients with mCRPC compared to RECIST 1.1 and prostate-specific antigen (PSA)-based treatment response assessments. Methods: Patients with mCRPC were prospectively enrolled if they were beginning systemic medical therapy or transitioning to new systemic therapy after not responding to a prior treatment. All patients underwent a baseline 18F-fluorodeoxyglucose (FDG) positron emission tomography/ computed tomography (PET/CT) prior to the initiation of treatment and again 4 months after the start of therapy. Patients' responses to treatment at 4 months compared to baseline were evaluated with RECIST 1.1, PERCIST 1.0 and PSA response criteria. The associations between patients' response categories and OS were evaluated. OS was defined as the duration in time between the date of baseline PET/CT to death from any cause. Patients with different response status were compared with logrank tests. Survival probabilities were calculated using the Kaplan-Meier method. Results: Patients with progressive disease by PSA response criteria at 4 months demonstrated significantly shorter OS (24-month OS probability: 18% ± 11%) compared to patients with stable disease, SD, (44% ± 19%, p=0.03) and complete response, CR, or partial response, PR, (53% ± 11%, p=0.03). RECIST 1.1 response criteria demonstrated a similar trend in OS, however no statistically significant differences were noted between patients with PD (25% ± 15%) compared to SD/non-CR, non-PD (54% ± 13%) and CR/PR (54% ± 14%) (p=0.13). PERCIST 1.0 criteria demonstrated significant differences in OS between responders, CMR/PMR (56% ± 12%), compared to SMD (38% ± 17%, p=0.03) and PMD (21% ± 10%, p=0.01). Patients with progressive disease by both PERICST 1.0 and PSA response criteria demonstrated significantly worse OS (24-month OS: 0%, 12-month OS: 31% ± 14%) compared to patients with progressive disease by either response criteria. Conclusion: PERCIST 1.0 may provide significant prognostic information for patients with mCRPC undergoing systemic chemotherapy, particularly when incorporated with PSA treatment response criteria.
Collapse
|
38
|
18F-FDG-PET/CT in measuring volume and global metabolic activity of thigh muscles. Nucl Med Commun 2020; 41:162-168. [DOI: 10.1097/mnm.0000000000001127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
39
|
Arici S, Karyagar SS, Karyagar S, Geredeli C, Cekin R, Seçmeler Ş, Atci MM, Sakin A, Cihan Ş. The predictive role of metabolic tumor volume on no response to neoadjuvant chemotherapy in patients with breast cancer. J Oncol Pharm Pract 2020; 26:1415-1420. [DOI: 10.1177/1078155219898504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Introduction To evaluate the predictive significance of pretreatment metabolic tumor volume on pathologic response in patients who received neoadjuvant chemotherapy for breast cancer. Methods Seventy patients who received neoadjuvant chemotherapy between 2013 and 2017 years were enrolled in the study. Pathologic responses and 18-fluorodeoxyglucose positron emission tomography/computed tomography metabolic dates of patients were obtained from archive files. Results Forty-six (65.7%) patients were in stage II and 24 (34.3%) patients were in stage III; 25 (35.7%) patients were human epidermal growth factor receptor 2 positive, 46 (65.7%) patients were estrogen receptor-positive, 26 (37.1%) patients were progesterone receptor-positive. According to the Miller-Payne grading system, 24 (34.3%) patients constituted 100% pathological response; patients with 91–99% pathological response were 12 (17.1%), the number of patients with non-pathologic response was 6 (8.6%). Median metabolic tumor volume was 7.3 cm3 (7.1 ± 3.5), 8.8 (11.4 ± 9.4), 7.7 (8.3 ± 4.6) and 22 cm3 (19.8 ± 11.0) in patients with stages IIA, IIB, IIIA, and IIIB, respectively ( p = 0.032). In Miller-Payne grading, the median metabolic tumor volume value was higher in patients with no pathologic response group than 100% response group ( p = 0.003). The cut-off metabolic tumor volume value determining no pathologic response was calculated as higher than 13.62 cm3 (sensitivity 83.3% and specificity 82.8%). Conclusions Our study results suggest that higher pretreatment metabolic tumor volume values are predictive on no pathologic response in patients treated with neoadjuvant chemotherapy for breast cancer.
Collapse
Affiliation(s)
- Serdar Arici
- Department of Medical Oncology, Istanbul Okmeydani Training and Research Hospital, Istanbul, Turkey
| | - Sevda S Karyagar
- Department of Nuclear Medicine, Istanbul Okmeydani Training and Research Hospital, Istanbul, Turkey
| | - Savas Karyagar
- Department of Nuclear Medicine, Istanbul Okmeydani Training and Research Hospital, Istanbul, Turkey
| | - Caglayan Geredeli
- Department of Medical Oncology, Istanbul Okmeydani Training and Research Hospital, Istanbul, Turkey
| | - Ruhper Cekin
- Department of Medical Oncology, Istanbul Okmeydani Training and Research Hospital, Istanbul, Turkey
| | - Şaban Seçmeler
- Department of Medical Oncology, Istanbul Okmeydani Training and Research Hospital, Istanbul, Turkey
| | - Muhammed M Atci
- Department of Medical Oncology, Istanbul Okmeydani Training and Research Hospital, Istanbul, Turkey
| | - Abdullah Sakin
- Faculity of Medicine, Medical Oncology Department, Yuzuncu Yil University, Yuzuncu Yil, Turkey
| | - Şener Cihan
- Department of Medical Oncology, Istanbul Okmeydani Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
40
|
Duarte PS, Sapienza MT. Normalization by bone volume instead of body weight or lean body mass may be better for quantifying skeletal burden in fibrous dysplasia using sodium fluoride PET/CT. Eur J Nucl Med Mol Imaging 2020; 47:1349-1350. [PMID: 31925457 DOI: 10.1007/s00259-020-04688-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 01/06/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Paulo Schiavom Duarte
- Division of Nuclear Medicine, São Paulo Cancer Institute (ICESP), Av. Dr. Arnaldo, 251, 4° SS, Cerqueira César, São Paulo, SP, 01246-000, Brazil.
| | - Marcelo Tatit Sapienza
- Division of Nuclear Medicine, Department of Radiology and Oncology, Medical School of University of São Paulo (FMUSP), São Paulo, Brazil
| |
Collapse
|
41
|
Gafita A, Calais J, Franz C, Rauscher I, Wang H, Roberstson A, Czernin J, Weber WA, Eiber M. Evaluation of SUV normalized by lean body mass (SUL) in 68Ga-PSMA11 PET/CT: a bi-centric analysis. EJNMMI Res 2019; 9:103. [PMID: 31792771 PMCID: PMC6889088 DOI: 10.1186/s13550-019-0572-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 11/04/2019] [Indexed: 12/17/2022] Open
Abstract
Introduction The aim of this analysis was to investigate whether the standardized uptake value (SUV) normalized by lean body mass (SUL) is a more appropriate quantitative parameter compared to the commonly used SUV normalized by patient’s weight in 68Ga-PSMA11 PET/CT. Material and methods 68Ga-PSMA11 PET/CT scans of 121 patients with prostate cancer from two institutions were evaluated. Liver SUV was measured within a 3-cm volume-of-interest (VOI) in the right hepatic lobe and corrected for lean body mass using the Janmahasatian formula. SUV and SUL repeatability between baseline and follow-up scans of the same patients were assessed. Results SUV was significantly positively correlated with body weight (r = 0.35, p = 0.02). In contrast, SUL was not correlated with body weight (r = 0.23, p = 0.07). No significant differences were found between baseline and follow-up scan (p = 0.52). Conclusion The Janmahasatian formula annuls the positive correlations between SUV and body weight, suggesting that SUL is preferable to SUV for quantitative analyses of 68Ga-PSMA11 PET/CT scans.
Collapse
Affiliation(s)
- Andrei Gafita
- Department of Nuclear Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany.
| | - Jeremie Calais
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Charlott Franz
- Department of Nuclear Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Isabel Rauscher
- Department of Nuclear Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Hui Wang
- Department of Nuclear Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Andrew Roberstson
- Department of Nuclear Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Johannes Czernin
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| |
Collapse
|
42
|
de-Lima-Júnior JC, Rodovalho S, Van de Sande-Lee S, Monfort-Pires M, Rachid B, Cintra RM, Ramos CD, Cendes F, Folli F, Velloso LA. Effect of pioglitazone treatment on brown adipose tissue volume and activity and hypothalamic gliosis in patients with type 2 diabetes mellitus: a proof-of-concept study. Acta Diabetol 2019; 56:1333-1339. [PMID: 31506721 DOI: 10.1007/s00592-019-01418-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/29/2019] [Indexed: 02/06/2023]
Abstract
AIMS This study aimed to evaluate the effect of pioglitazone on brown adipose tissue function and hypothalamic gliosis in humans. Brown adipose tissue and the hypothalamus are regarded as important potential pharmacological targets to metabolic diseases, and defining the impact of current therapies on their structure and/or function could provide therapeutic advance in this field. METHODS Six patients with type 2 diabetes were treated for 24 weeks with pioglitazone 30 mg/day as an add-on therapy. Brown adipose tissue glucose uptake and volume were determined using 18F-FDG PET/CT scans; hypothalamic gliosis was determined using MRI scans; blood was collected for hormone and biochemistry measurements. All tests were performed at inclusion and six months after pioglitazone introduction. RESULTS Pioglitazone treatment led to a significant 3% body mass increase. There were neither changes in cold-induced brown adipose tissue glucose uptake and volume nor changes in hypothalamic gliosis. CONCLUSIONS This is a proof-of-concept study that provides clinical evidence for a lack of action of a thiazolidinedione, pioglitazone, to promote homogeneous and measurable changes in brown adipose tissue volume and also in hypothalamic gliosis after 6 months of treatment.
Collapse
Affiliation(s)
- José C de-Lima-Júnior
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Sylka Rodovalho
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Simone Van de Sande-Lee
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Department of Internal Medicine, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Milena Monfort-Pires
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Briana Rachid
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Riobaldo M Cintra
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Celso D Ramos
- Department of Radiology, University of Campinas, Campinas, São Paulo, 13084-970, Brazil
| | - Fernando Cendes
- Neuroimaging Laboratory, Department of Neurology, University of Campinas, Campinas, Brazil
| | - Franco Folli
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
- School of Medicine, Endocrinology and Metabolism Dipartimento di Scienze Della Salute, Universita' degli Studi di Milano, Milan, Italy
- Departmental Unit of Diabetes and Metabolic Disorders, Azienda Socio-Sanitaria Santi Paolo e Carlo, Via A. Di Rudini, 8, 20143, Milan, Italy
| | - Lício A Velloso
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil.
| |
Collapse
|
43
|
Dyke JP, Garfinkel JH, Volpert L, Sanders A, Newcomer M, Dutruel SP, Sofka CM, Ellis SJ, Demetracopoulos CA. Imaging of Bone Perfusion and Metabolism in Subjects Undergoing Total Ankle Arthroplasty Using 18F-Fluoride Positron Emission Tomography. Foot Ankle Int 2019; 40:1351-1357. [PMID: 31597454 DOI: 10.1177/1071100719882717] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Total ankle arthroplasty (TAA) continues to exhibit a relatively high incidence of complications and need for revision surgery compared to knee and hip arthroplasty. One common mode of failure in TAA is talar component subsidence. This may be caused by disruption in the talar blood supply related to the operative technique. The purpose of this study was to quantify changes in talar bone perfusion and turnover before and after TAA with the INBONE II system using 18F-fluoride positron emission tomography / computed tomography (PET/CT). METHODS Nine subjects (5 M/4 F) aged 68.9 ± 8.2 years were enrolled for 18F-fluoride PET/CT imaging before and 3 months after TAA. Regions of interest (ROI) were placed on the postoperative CT images in the body of the talus beneath the talar component and overlaid on the fused static PET images. Standard uptake values (SUVs) along with dynamic K1 (bone blood flow) and ki (bone metabolism or osteoblastic turnover) were calculated. RESULTS The SUV underneath the talar component compared to that measured at baseline before surgery was 1.93 ± 0.29 preoperatively vs 2.47 ± 0.37 postoperatively (P > .05). K1 was 0.84 ± 0.16 mL/min/mL preoperatively vs 1.51 ± 0.23 mL/min/mL postoperatively (P = .026). ki was constant at 0.09 ± 0.03 mL/min/mL preoperatively vs 0.12 ± 0.03 mL/min/mL postoperatively (P > .05). CONCLUSION Our study was the first to link 18F-fluoride PET/CT with pre-post evaluation of total ankle replacements. The study quantified perfusion within the talus beneath the TAA implant supporting the hypothesis that perfusion of the talus remained intact after surgery. LEVEL OF EVIDENCE Level II, prospective cohort study with development of diagnostic criteria.
Collapse
Affiliation(s)
- Jonathan P Dyke
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY, USA
| | | | - Lauren Volpert
- Foot and Ankle Service, Hospital for Special Surgery, New York, NY, USA
| | - Austin Sanders
- Foot and Ankle Service, Hospital for Special Surgery, New York, NY, USA
| | - Meghan Newcomer
- Foot and Ankle Service, Hospital for Special Surgery, New York, NY, USA
| | | | - Carolyn M Sofka
- Department of Radiology, Hospital for Special Surgery, New York, NY, USA
| | - Scott J Ellis
- Foot and Ankle Service, Hospital for Special Surgery, New York, NY, USA
| | | |
Collapse
|
44
|
Sarikaya I, Albatineh AN, Sarikaya A. Revisiting Weight-Normalized SUV and Lean-Body-Mass-Normalized SUV in PET Studies. J Nucl Med Technol 2019; 48:163-167. [PMID: 31604893 DOI: 10.2967/jnmt.119.233353] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/06/2019] [Indexed: 12/31/2022] Open
Abstract
SUV normalized by total body weight is affected by the amount of body fat. The SUV of normal tissues and lesions is higher (overestimated) in obese patients than in patients with a normal body mass index (BMI). SUL (SUV normalized by lean body mass [LBM]) is recommended for more accurate SUV results. Given the importance of a quantitative PET parameter, particularly when comparing PET studies, we aimed to revisit the effect of obesity on SUV, measuring SUL in normal-BMI patients and obese patients and testing the effect of the amount of LBM. Methods: 18F-FDG whole-body images of adult patients were analyzed retrospectively. We measured both SUVmax and SUVmean in the blood pool and liver of patients with a normal BMI (18.5-24.9) and a high BMI (≥30) (obese). In all patients, we calculated LBM via an equation using patient height and weight and corrected all SUVs to SULs. Mean (±SD) SUVs and SULs were compared under various circumstances. Scatterplots were generated for weight and SUV-SUL differences. Results: SUVmean in the liver and blood pool was significantly higher in obese patients (30 patients) than in patients with a normal BMI (20 patients) (4.1 ± 0.7 and 3.0 ± 0.5, respectively, in liver, vs. 3.2 ± 0.6 and 2.4 ± 0.4, respectively, in blood pool; P < 0.001). SULmean was significantly lower in both liver and blood pool in all patients, being approximately 75% of SUVmean in patients with a normal BMI and 55% of SUVmean in obese patients (P < 0.001). SULmean in the liver and blood pool did not significantly differ between obese patients and normal-BMI patients (P > 0.05). The SUV-SUL difference was significantly higher in obese patients than in patients with a normal BMI (P < 0.001). These statistical results were the same when SUVmax and SULmax were compared. Conclusion: SUV overestimates metabolic activity in all patients, and this overestimation is more significant in obese patients than in patients with a normal BMI. SUL is not affected by body weight or the amount of LBM.
Collapse
Affiliation(s)
- Ismet Sarikaya
- Department of Nuclear Medicine, Faculty of Medicine, Kuwait University, and Mubarak Al-Kabeer Hospital, Jabriya, Kuwait
| | - Ahmed N Albatineh
- Department of Community Medicine and Behavioral Sciences, Faculty of Medicine, Kuwait University, Safat, Kuwait; and
| | - Ali Sarikaya
- Department of Nuclear Medicine, Faculty of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
45
|
Das SK, McGurk R, Miften M, Mutic S, Bowsher J, Bayouth J, Erdi Y, Mawlawi O, Boellaard R, Bowen SR, Xing L, Bradley J, Schoder H, Yin FF, Sullivan DC, Kinahan P. Task Group 174 Report: Utilization of [ 18 F]Fluorodeoxyglucose Positron Emission Tomography ([ 18 F]FDG-PET) in Radiation Therapy. Med Phys 2019; 46:e706-e725. [PMID: 31230358 DOI: 10.1002/mp.13676] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 04/30/2019] [Accepted: 06/06/2019] [Indexed: 02/03/2023] Open
Abstract
The use of positron emission tomography (PET) in radiation therapy (RT) is rapidly increasing in the areas of staging, segmentation, treatment planning, and response assessment. The most common radiotracer is 18 F-fluorodeoxyglucose ([18 F]FDG), a glucose analog with demonstrated efficacy in cancer diagnosis and staging. However, diagnosis and RT planning are different endeavors with unique requirements, and very little literature is available for guiding physicists and clinicians in the utilization of [18 F]FDG-PET in RT. The two goals of this report are to educate and provide recommendations. The report provides background and education on current PET imaging systems, PET tracers, intensity quantification, and current utilization in RT (staging, segmentation, image registration, treatment planning, and therapy response assessment). Recommendations are provided on acceptance testing, annual and monthly quality assurance, scanning protocols to ensure consistency between interpatient scans and intrapatient longitudinal scans, reporting of patient and scan parameters in literature, requirements for incorporation of [18 F]FDG-PET in treatment planning systems, and image registration. The recommendations provided here are minimum requirements and are not meant to cover all aspects of the use of [18 F]FDG-PET for RT.
Collapse
Affiliation(s)
- Shiva K Das
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Ross McGurk
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Moyed Miften
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sasa Mutic
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - James Bowsher
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - John Bayouth
- Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Yusuf Erdi
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Osama Mawlawi
- Department of Imaging Physics, University of Texas, M D Anderson Cancer Center, Houston, TX, USA
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Stephen R Bowen
- Department of Radiation Oncology, University of Washington, Seattle, WA, USA
| | - Lei Xing
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey Bradley
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Heiko Schoder
- Molecular Imaging and Therapy Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Fang-Fang Yin
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Daniel C Sullivan
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Paul Kinahan
- Department of Radiology, University of Washington, Seattle, WA, USA
| |
Collapse
|
46
|
Diffusely Decreased Liver Uptake on FDG PET and Cancer-Associated Cachexia With Reduced Survival. Clin Nucl Med 2019; 44:634-642. [PMID: 31274609 DOI: 10.1097/rlu.0000000000002658] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVES We investigated clinical characteristics of patients with extremely increased or decreased physiologic F-FDG uptake of the liver and their prognosis. METHODS One thousand four hundred eighty-seven PET/CT scans of patients with known or suspected malignancy were retrospectively analyzed. A spherical volume of interest (3 cm in diameter) was set on the right lobe of the liver to calculate the SUVmean. Scans with extremely high (SUVmean >97.5th percentile) and low (SUVmean <2.5th percentile) FDG uptake in the liver were evaluated. Physical and laboratory data among a control group (n = 30), the extremely high liver uptake group (HG, n = 36), and the extremely low liver uptake group (LG, n = 36) were compared. Overall survival (OS) of the 3 groups was also compared. RESULTS Body weight and body mass index in the HG (SUVmean ≥3.04) were significantly higher than those in the control group. The LG cases (SUVmean ≤1.78) had anemia, impaired liver function, and systemic inflammation. They were also in a poor nutritional state. The characteristics of LG cases had many things in common with those of cachectic patients. Indeed, 36.1% of LG cases met the diagnostic criteria for cachexia. Moreover, in LG cases with viable and/or recurrent malignant lesions on FDG PET, the proportion of cachexia increased by 52.6%. The OS of LG cases (median, 33 months) was significantly worse than that of controls and HG cases. CONCLUSIONS Our data indicate that cancer patients with extremely decreased liver FDG uptake were likely to have cancer cachexia and a lower OS.
Collapse
|
47
|
Chen W, Zhu L, Yu X, Fu Q, Xu W, Wang P. Quantitative assessment of metabolic tumor burden in molecular subtypes of primary breast cancer with FDG PET/CT. ACTA ACUST UNITED AC 2019; 24:336-341. [PMID: 30406761 DOI: 10.5152/dir.2018.17367] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE We aimed to quantitatively evaluate volumetric metabolic tumor burden including metabolic tumor volume and total lesion glycolysis in different molecular subtypes of breast cancer using 18F-fluorodeoxyglucose (FDG) positron emission tomography/ computed tomography (PET/CT). METHODS This study involved 99 female patients with pathologic diagnosis of primary breast cancer, who underwent 18F-FDG PET/CT before any therapy. Patients were divided into subtypes of luminal A, luminal B, ERBB2+, and basal-like based on the immunohistochemistry results. Metabolic tumor volume (MTV) and total lesion glycolysis (TLG) before and after correction for lean body mass were achieved and compared. Correlations between metabolic tumor burden and Ki-67 were analyzed and diagnostic performances of volumetric metabolic parameters were evaluated. RESULTS TLG values were significantly different between each molecular subtype, while MTV values were not. Values of TLG were significantly reduced after normalizing for lean body mass in each subtype. Both of them showed correlations with Ki-67 and presented high diagnostic ability in identifying patients with basal-like breast cancer from the rest. TLGs before and after normalizing for the lean body mass had similar diagnostic performances in differentiating patients of basal-like subtype from the rest. CONCLUSION Metabolic tumor burden could comprehensively reflect tumor metabolic differences of molecular subtypes of breast cancer, and it can serve to help differentiate patients with basal-like breast cancer.
Collapse
Affiliation(s)
- Wei Chen
- Department of Molecular Imaging and Nuclear Medicine Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| | - Lei Zhu
- Department of Molecular Imaging and Nuclear Medicine Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| | - Xiaozhou Yu
- Department of Molecular Imaging and Nuclear Medicine Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| | - Qiang Fu
- Department of Molecular Imaging and Nuclear Medicine Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| | - Ping Wang
- Department of Molecular Imaging and Nuclear Medicine and Radiation Oncology Tianjin Medical University Cancer Institute and Hospital; Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| |
Collapse
|
48
|
Abstract
Aim To compare weight, lean body mass and body surface area for calculation of standardised uptake value (SUV) in fluorine-18-fluorodeoxyglucose PET/computed tomography, taking sex into account. Patients and methods This was a retrospective study of 161 (97 men) patients. Maximum standardised uptake value (SUVmax) and mean standardised uptake value (SUVmean) were obtained from a 3-cm region of interest over the right lobe of the liver and scaled to weight, scaled to lean body mass (SUL) and scaled to body surface area (SUA). Mean hepatic computed tomography density was used to adjust SUVmean for hepatic fat (SUVFA). Hepatic SUV indices were divided by SUV from left ventricular cavity, thereby, eliminating whole body metric, to obtain a surrogate of blood fluorine-18-fluorodeoxyglucose clearance into liver, and multiplied by blood glucose to give a surrogate of hepatic glucose uptake rate (mSUV). Results SULmax, SUAmax and all scaled to weight indices correlated strongly with weight. SULmean, SULFA, SUAmean and SUAFA, however, correlated weakly or not at all with weight, nor with their corresponding whole body metric in men or women, but correlated strongly when the sexes were combined into one group. This was the result of sex differences in SUL (greater in men) and SUA (greater in women). There was, however, no sex difference in mSUV. Conclusion Weight is unsuitable for calculating SUV. SUL and SUA are also inappropriate as maxima but appropriate as mean and fat-adjusted values. However, SUL is recommended for both sexes because SUA is influenced by both body fat and weight. Sex differences in SUL and SUA give rise to misleading correlations when sexes are combined into one group.
Collapse
|
49
|
de-Lima-Júnior JC, Souza GF, Moura-Assis A, Gaspar RS, Gaspar JM, Rocha AL, Ferrucci DL, Lima TI, Victório SC, Bonfante ILP, Cavaglieri CR, Pareja JC, Brunetto SQ, Ramos CD, Geloneze B, Mori MA, Silveira LR, Segundo GRS, Ropelle ER, Velloso LA. Abnormal brown adipose tissue mitochondrial structure and function in IL10 deficiency. EBioMedicine 2018; 39:436-447. [PMID: 30502051 PMCID: PMC6355943 DOI: 10.1016/j.ebiom.2018.11.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022] Open
Abstract
Background Inflammation is the most relevant mechanism linking obesity with insulin-resistance and metabolic disease. It impacts the structure and function of tissues and organs involved in metabolism, such as the liver, pancreatic islets and the hypothalamus. Brown adipose tissue has emerged as an important component of whole body energy homeostasis, controlling caloric expenditure through the regulation of non-shivering thermogenesis. However, little is known about the impact of systemic inflammation on the structure and function of brown adipose tissue. Methods The relations between IL10 and mitochondria structure/function and also with thermogenesis were evaluated by bioinformatics using human and rodent data. Real-time PCR, immunoblot, fluorescence and transmission electron microscopy were employed to determine the effect of IL10 in the brown adipose tissue of wild type and IL10 knockout mice. Findings IL10 knockout mice, a model of systemic inflammation, present severe structural abnormalities of brown adipose tissue mitochondria, which are round-shaped with loss of cristae structure and increased fragmentation. IL10 deficiency leads to newborn cold intolerance and impaired UCP1-dependent brown adipose tissue mitochondrial respiration. The reduction of systemic inflammation with an anti-TNFα monoclonal antibody partially rescued the structural but not the functional abnormalities of brown adipose tissue mitochondria. Using bioinformatics analyses we show that in both humans and mice, IL10 transcripts correlate with mitochondrial lipid metabolism and caspase gene expression. Interpretation IL10 and systemic inflammation play a central role in the regulation of brown adipose tissue by controlling mitochondrial structure and function. Fund Sao Paulo Research Foundation grant 2013/07607-8.
Collapse
Affiliation(s)
- José C de-Lima-Júnior
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo 13084-970, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil
| | - Gabriela F Souza
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo 13084-970, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil
| | - Alexandre Moura-Assis
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo 13084-970, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil
| | - Rodrigo S Gaspar
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil; CEPECE - Research Center of Sport Sciences, School of Applied Sciences, University of Campinas, Limeira, SP, Brazil(.)
| | - Joana M Gaspar
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo 13084-970, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil
| | - Andréa L Rocha
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP 13083-970, Brazil
| | - Danilo L Ferrucci
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil; National Institute of Photonics Applied to Cell Biology (INFABiC), Campinas, São Paulo, Brazil
| | - Tanes I Lima
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil; Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP 13083-970, Brazil
| | - Sheila C Victório
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo 13084-970, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil
| | - Ivan L P Bonfante
- Laboratory of Exercise Physiology, School of Physical Education, University of Campinas, Campinas, SP 13083-970, Brazil
| | - Claudia R Cavaglieri
- Laboratory of Exercise Physiology, School of Physical Education, University of Campinas, Campinas, SP 13083-970, Brazil
| | - José C Pareja
- Laboratory of Investigation in Metabolism and Diabetes (LIMED)/Gastrocentro, Department of Surgery, University of Campinas (UNICAMP), Campinas, SP 13081-970, Brazil
| | - Sérgio Q Brunetto
- Biomedical Engineering Center, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Celso D Ramos
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil; Department of Radiology, University of Campinas, Campinas, São Paulo 13084-970, Brazil
| | - Bruno Geloneze
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil; Laboratory of Investigation in Metabolism and Diabetes (LIMED)/Gastrocentro, Department of Surgery, University of Campinas (UNICAMP), Campinas, SP 13081-970, Brazil
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP 13083-970, Brazil
| | - Leonardo R Silveira
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil; Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP 13083-970, Brazil
| | - Gesmar R S Segundo
- Department of Pediatrics, Federal University of Uberlandia, Uberlandia, Brazil
| | - Eduardo R Ropelle
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil; CEPECE - Research Center of Sport Sciences, School of Applied Sciences, University of Campinas, Limeira, SP, Brazil(.)
| | - Lício A Velloso
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo 13084-970, Brazil; Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil.
| |
Collapse
|
50
|
Press RH, Shu HKG, Shim H, Mountz JM, Kurland BF, Wahl RL, Jones EF, Hylton NM, Gerstner ER, Nordstrom RJ, Henderson L, Kurdziel KA, Vikram B, Jacobs MA, Holdhoff M, Taylor E, Jaffray DA, Schwartz LH, Mankoff DA, Kinahan PE, Linden HM, Lambin P, Dilling TJ, Rubin DL, Hadjiiski L, Buatti JM. The Use of Quantitative Imaging in Radiation Oncology: A Quantitative Imaging Network (QIN) Perspective. Int J Radiat Oncol Biol Phys 2018; 102:1219-1235. [PMID: 29966725 PMCID: PMC6348006 DOI: 10.1016/j.ijrobp.2018.06.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 05/25/2018] [Accepted: 06/14/2018] [Indexed: 02/07/2023]
Abstract
Modern radiation therapy is delivered with great precision, in part by relying on high-resolution multidimensional anatomic imaging to define targets in space and time. The development of quantitative imaging (QI) modalities capable of monitoring biologic parameters could provide deeper insight into tumor biology and facilitate more personalized clinical decision-making. The Quantitative Imaging Network (QIN) was established by the National Cancer Institute to advance and validate these QI modalities in the context of oncology clinical trials. In particular, the QIN has significant interest in the application of QI to widen the therapeutic window of radiation therapy. QI modalities have great promise in radiation oncology and will help address significant clinical needs, including finer prognostication, more specific target delineation, reduction of normal tissue toxicity, identification of radioresistant disease, and clearer interpretation of treatment response. Patient-specific QI is being incorporated into radiation treatment design in ways such as dose escalation and adaptive replanning, with the intent of improving outcomes while lessening treatment morbidities. This review discusses the current vision of the QIN, current areas of investigation, and how the QIN hopes to enhance the integration of QI into the practice of radiation oncology.
Collapse
Affiliation(s)
- Robert H. Press
- Dept. of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Hui-Kuo G. Shu
- Dept. of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Hyunsuk Shim
- Dept. of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - James M. Mountz
- Dept. of Radiology, University of Pittsburgh, Pittsburgh, PA
| | | | | | - Ella F. Jones
- Dept. of Radiology, University of California, San Francisco, San Francisco, CA
| | - Nola M. Hylton
- Dept. of Radiology, University of California, San Francisco, San Francisco, CA
| | - Elizabeth R. Gerstner
- Dept. of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | | | - Lori Henderson
- Cancer Imaging Program, National Cancer Institute, Bethesda, MD
| | | | - Bhadrasain Vikram
- Radiation Research Program/Division of Cancer Treatment & Diagnosis, National Cancer Institute, Bethesda, MD
| | - Michael A. Jacobs
- Dept. of Radiology and Radiological Science, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore MD
| | - Matthias Holdhoff
- Brain Cancer Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore MD
| | - Edward Taylor
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - David A. Jaffray
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | | | - David A. Mankoff
- Dept. of Radiology, University of Pennsylvania, Philadelphia, PA
| | | | | | - Philippe Lambin
- Dept. of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Thomas J. Dilling
- Dept. of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | | | - John M. Buatti
- Dept. of Radiation Oncology, University of Iowa, Iowa City, IA
| |
Collapse
|