1
|
Ince S, Laforest R, Itani M, Prasad V, Derenoncourt PR, Crandall JP, Ashrafinia S, Smith AM, Wahl RL, Fraum TJ. Quantitative Assessments of Tumor Activity in a General Oncologic PET/CT Population: Which Metric Minimizes Tracer Uptake Time Dependence? J Nucl Med 2024; 65:1349-1356. [PMID: 39142828 PMCID: PMC11372261 DOI: 10.2967/jnumed.123.266469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 07/10/2024] [Indexed: 08/16/2024] Open
Abstract
In oncologic PET, the SUV and standardized uptake ratio (SUR) of a viable tumor generally increase during the postinjection period. In contrast, the net influx rate (Ki ), which is derived from dynamic PET data, should remain relatively constant. Uptake-time-corrected SUV (cSUV) and SUR (cSUR) have been proposed as uptake-time-independent, static alternatives to Ki Our primary aim was to quantify the intrascan repeatability of Ki , SUV, cSUV, SUR, and cSUR among malignant lesions on PET/CT. An exploratory aim was to assess the ability of cSUR to estimate Ki Methods: This prospective, single-center study enrolled adults undergoing standard-of-care oncologic PET/CT. SUV and Ki images were reconstructed from dynamic PET data obtained before (∼35-50 min after injection) and after (∼75-90 min after injection) standard-of-care imaging. Tumors were manually segmented. Quantitative metrics were extracted. cSUVs and cSURs were calculated for a 60-min postinjection reference uptake time. The magnitude of the intrascan test-retest percent change (test-retest |%Δ|) was calculated. Coefficients of determination (R 2) and intraclass correlation coefficients (ICC) were also computed. Differences between metrics were assessed via the Wilcoxon signed-rank test (α, 0.05). Results: This study enrolled 78 subjects; 41 subjects (mean age, 63.8 y; 24 men) with 116 lesions were analyzed. For both tracers, SUVmax and maximum SUR (SURmax) had large early-to-late increases (i.e., poor intrascan repeatability). Among [18F]FDG-avid lesions (n = 93), there were no differences in intrascan repeatability (median test-retest |%Δ|; ICC) between the maximum Ki (Ki ,max) (13%; 0.97) and either the maximum cSUV (cSUVmax) (12%, P = 0.90; 0.96) or the maximum cSUR (cSURmax) (13%, P = 0.67; 0.94). For DOTATATE-avid lesions (n = 23), there were no differences in intrascan repeatability between the Ki ,max (11%; 0.98) and either the cSUVmax (13%, P = 0.41; 0.98) or the cSURmax (11%, P = 0.08; 0.94). The SUVmax, cSUVmax, SURmax, and cSURmax were all strongly correlated with the Ki ,max for both [18F]FDG (R 2, 0.81-0.92) and DOTATATE (R 2, 0.88-0.96), but the cSURmax provided the best agreement with the Ki ,max across early-to-late time points for [18F]FDG (ICC, 0.69-0.75) and DOTATATE (ICC, 0.90-0.91). Conclusion: Ki ,max, cSUVmax, and cSURmax had low uptake time dependence compared with SUVmax and SURmax The Ki ,max can be predicted from cSURmax.
Collapse
Affiliation(s)
- Semra Ince
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Richard Laforest
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Malak Itani
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Vikas Prasad
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | | | - John P Crandall
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Anne M Smith
- Siemens Medical Solutions Inc., Knoxville, Tennessee; and
| | - Richard L Wahl
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Tyler J Fraum
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri;
| |
Collapse
|
2
|
Malaih AA, Kirkwood AA, Johnson P, Radhakrishnan V, Fischer BM, Barrington SF. Healthy tissue metabolism assessed by [ 18F]FDG PET/CT as a marker of prognosis and adverse events in advanced Hodgkin lymphoma patients. Sci Rep 2024; 14:12613. [PMID: 38824206 PMCID: PMC11144227 DOI: 10.1038/s41598-024-63349-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024] Open
Abstract
The aim of the study was to assess healthy tissue metabolism (HTM) using 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) during chemotherapy in Hodgkin lymphoma (HL) and the association of HTM with baseline metabolic tumour volume (MTV), haematological parameters, adverse events (AEs), early response and progression-free survival (PFS). We retrospectively identified 200 patients with advanced HL from the RATHL trial with [18F]FDG-PET/CT before (PET0) and following 2 cycles of chemotherapy (PET2). [18F]FDG-uptake was measured in bone marrow (BM), spleen, liver and mediastinal blood pool (MBP). Deauville score (DS) 1-3 was used to classify responders and DS 4-5, non-responders. [18F]FDG-uptake decreased significantly in BM and spleen and increased in liver and MBP at PET2 (all p < 0.0001), but was not associated with MTV. Higher BM uptake at PET0 was associated with lower baseline haemoglobin and higher absolute neutrophil counts, platelets, and white blood cells. High BM, spleen, and liver uptake at PET0 was associated with neutropenia after cycles 1-2. BM uptake at PET0 was associated with treatment failure at PET2 and non-responders with higher BM uptake at PET2 had significantly inferior PFS (p = 0.023; hazard ratio = 2.31). Based on these results, we concluded that the change in HTM during chemotherapy was most likely a direct impact of chemotherapy rather than a change in MTV. BM uptake has prognostic value in HL.
Collapse
Affiliation(s)
- Afnan A Malaih
- King's College London and Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, Kings Health Partners, London, UK
- Radiologic Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amy A Kirkwood
- Cancer Research UK and UCL Cancer Trials Centre, UCL Cancer Institute, University College London, London, UK
| | - Peter Johnson
- Cancer Research UK Centre, University of Southampton, Southampton, UK
| | | | - Barbara M Fischer
- King's College London and Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, Kings Health Partners, London, UK
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sally F Barrington
- King's College London and Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, Kings Health Partners, London, UK.
| |
Collapse
|
3
|
Rogasch JMM, Michaels L, Baumgärtner GL, Frost N, Rückert JC, Neudecker J, Ochsenreither S, Gerhold M, Schmidt B, Schneider P, Amthauer H, Furth C, Penzkofer T. A machine learning tool to improve prediction of mediastinal lymph node metastases in non-small cell lung cancer using routinely obtainable [ 18F]FDG-PET/CT parameters. Eur J Nucl Med Mol Imaging 2023; 50:2140-2151. [PMID: 36820890 PMCID: PMC10199849 DOI: 10.1007/s00259-023-06145-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/08/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND In patients with non-small cell lung cancer (NSCLC), accuracy of [18F]FDG-PET/CT for pretherapeutic lymph node (LN) staging is limited by false positive findings. Our aim was to evaluate machine learning with routinely obtainable variables to improve accuracy over standard visual image assessment. METHODS Monocentric retrospective analysis of pretherapeutic [18F]FDG-PET/CT in 491 consecutive patients with NSCLC using an analog PET/CT scanner (training + test cohort, n = 385) or digital scanner (validation, n = 106). Forty clinical variables, tumor characteristics, and image variables (e.g., primary tumor and LN SUVmax and size) were collected. Different combinations of machine learning methods for feature selection and classification of N0/1 vs. N2/3 disease were compared. Ten-fold nested cross-validation was used to derive the mean area under the ROC curve of the ten test folds ("test AUC") and AUC in the validation cohort. Reference standard was the final N stage from interdisciplinary consensus (histological results for N2/3 LNs in 96%). RESULTS N2/3 disease was present in 190 patients (39%; training + test, 37%; validation, 46%; p = 0.09). A gradient boosting classifier (GBM) with 10 features was selected as the final model based on test AUC of 0.91 (95% confidence interval, 0.87-0.94). Validation AUC was 0.94 (0.89-0.98). At a target sensitivity of approx. 90%, test/validation accuracy of the GBM was 0.78/0.87. This was significantly higher than the accuracy based on "mediastinal LN uptake > mediastinum" (0.7/0.75; each p < 0.05) or combined PET/CT criteria (PET positive and/or LN short axis diameter > 10 mm; 0.68/0.75; each p < 0.001). Harmonization of PET images between the two scanners affected SUVmax and visual assessment of the LNs but did not diminish the AUC of the GBM. CONCLUSIONS A machine learning model based on routinely available variables from [18F]FDG-PET/CT improved accuracy in mediastinal LN staging compared to established visual assessment criteria. A web application implementing this model was made available.
Collapse
Affiliation(s)
- Julian M M Rogasch
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Liza Michaels
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Georg L Baumgärtner
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Nikolaj Frost
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Jens-Carsten Rückert
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Jens Neudecker
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Sebastian Ochsenreither
- Department of Hematology and Medical Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charité Comprehensive Cancer Center, Berlin, Germany
| | - Manuela Gerhold
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Bernd Schmidt
- Department of Internal Medicine - Pneumology and Sleep Medicine, DRK Kliniken Berlin Mitte, Berlin, Germany
| | - Paul Schneider
- Department of Thoracic Surgery, DRK Kliniken Berlin Mitte, Berlin, Germany
| | - Holger Amthauer
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Tobias Penzkofer
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| |
Collapse
|
4
|
Tamaki N, Hirata K, Kotani T, Nakai Y, Matsushima S, Yamada K. Four-dimensional quantitative analysis using FDG-PET in clinical oncology. Jpn J Radiol 2023:10.1007/s11604-023-01411-4. [PMID: 36947283 PMCID: PMC10366296 DOI: 10.1007/s11604-023-01411-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/02/2023] [Indexed: 03/23/2023]
Abstract
Positron emission tomography (PET) with F-18 fluorodeoxyglucose (FDG) has been commonly used in many oncological areas. High-resolution PET permits a three-dimensional analysis of FDG distributions on various lesions in vivo, which can be applied for tissue characterization, risk analysis, and treatment monitoring after chemoradiotherapy and immunotherapy. Metabolic changes can be assessed using the tumor absolute FDG uptake as standardized uptake value (SUV) and metabolic tumor volume (MTV). In addition, tumor heterogeneity assessment can potentially estimate tumor aggressiveness and resistance to chemoradiotherapy. Attempts have been made to quantify intratumoral heterogeneity using radiomics. Recent reports have indicated the clinical feasibility of a dynamic FDG PET-computed tomography (CT) in pilot cohort studies of oncological cases. Dynamic imaging permits the assessment of temporal changes in FDG uptake after administration, which is particularly useful for differentiating pathological from physiological uptakes with high diagnostic accuracy. In addition, several new parameters have been introduced for the in vivo quantitative analysis of FDG metabolic processes. Thus, a four-dimensional FDG PET-CT is available for precise tissue characterization of various lesions. This review introduces various new techniques for the quantitative analysis of FDG distribution and glucose metabolism using a four-dimensional FDG analysis with PET-CT. This elegant study reveals the important role of tissue characterization and treatment strategies in oncology.
Collapse
Affiliation(s)
- Nagara Tamaki
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Kenji Hirata
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tomoya Kotani
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshitomo Nakai
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigenori Matsushima
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kei Yamada
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
5
|
Ju HM, Kim BC, Lim I, Byun BH, Woo SK. Estimation of an Image Biomarker for Distant Recurrence Prediction in NSCLC Using Proliferation-Related Genes. Int J Mol Sci 2023; 24:ijms24032794. [PMID: 36769108 PMCID: PMC9917349 DOI: 10.3390/ijms24032794] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/22/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
This study aimed to identify a distant-recurrence image biomarker in NSCLC by investigating correlations between heterogeneity functional gene expression and fluorine-18-2-fluoro-2-deoxy-D-glucose positron emission tomography (18F-FDG PET) image features of NSCLC patients. RNA-sequencing data and 18F-FDG PET images of 53 patients with NSCLC (19 with distant recurrence and 34 without recurrence) from The Cancer Imaging Archive and The Cancer Genome Atlas Program databases were used in a combined analysis. Weighted correlation network analysis was performed to identify gene groups related to distant recurrence. Genes were selected for functions related to distant recurrence. In total, 47 image features were extracted from PET images as radiomics. The relationship between gene expression and image features was estimated using a hypergeometric distribution test with the Pearson correlation method. The distant recurrence prediction model was validated by a random forest (RF) algorithm using image texture features and related gene expression. In total, 37 gene modules were identified by gene-expression pattern with weighted gene co-expression network analysis. The gene modules with the highest significance were selected (p-value < 0.05). Nine genes with high protein-protein interaction and area under the curve (AUC) were identified as hub genes involved in the proliferation function, which plays an important role in distant recurrence of cancer. Four image features (GLRLM_SRHGE, GLRLM_HGRE, SUVmean, and GLZLM_GLNU) and six genes were identified to be correlated (p-value < 0.1). AUCs (accuracy: 0.59, AUC: 0.729) from the 47 image texture features and AUCs (accuracy: 0.767, AUC: 0.808) from hub genes were calculated using the RF algorithm. AUCs (accuracy: 0.783, AUC: 0.912) from the four image texture features and six correlated genes and AUCs (accuracy: 0.738, AUC: 0.779) from only the four image texture features were calculated using the RF algorithm. The four image texture features validated by heterogeneity group gene expression were found to be related to cancer heterogeneity. The identification of these image texture features demonstrated that advanced prediction of NSCLC distant recurrence is possible using the image biomarker.
Collapse
Affiliation(s)
- Hye Min Ju
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Republic of Korea
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 07812, Republic of Korea
| | - Byung-Chul Kim
- Department of Nuclear Medicine, Korea Institute of Radiological and Medical Sciences, Seoul 07812, Republic of Korea
| | - Ilhan Lim
- Department of Nuclear Medicine, Korea Institute of Radiological and Medical Sciences, Seoul 07812, Republic of Korea
| | - Byung Hyun Byun
- Department of Nuclear Medicine, Korea Institute of Radiological and Medical Sciences, Seoul 07812, Republic of Korea
| | - Sang-Keun Woo
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Republic of Korea
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 07812, Republic of Korea
- Correspondence: ; Tel.: +82-2-970-1659
| |
Collapse
|
6
|
Shankar LK, Huang E, Litiere S, Hoekstra OS, Schwartz L, Collette S, Boellaard R, Bogaerts J, Seymour L, deVries EGE. Meta-Analysis of the Test-Retest Repeatability of [18F]-Fluorodeoxyglucose Standardized Uptake Values: Implications for Assessment of Tumor Response. Clin Cancer Res 2023; 29:143-153. [PMID: 36302172 DOI: 10.1158/1078-0432.ccr-21-3143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/28/2022] [Accepted: 10/24/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE Currently, guidelines for PET with 18F-fluorodeoxyglucose (FDG-PET) interpretation for assessment of therapy response in oncology primarily involve visual evaluation of FDG-PET/CT scans. However, quantitative measurements of the metabolic activity in tumors may be even more useful in evaluating response to treatment. Guidelines based on such measurements, including the European Organization for Research and Treatment of Cancer Criteria and PET Response Criteria in Solid Tumors, have been proposed. However, more rigorous analysis of response criteria based on FDG-PET measurements is needed to adopt regular use in practice. EXPERIMENTAL DESIGN Well-defined boundaries of repeatability and reproducibility of quantitative measurements to discriminate noise from true signal changes are a needed initial step. An extension of the meta-analysis from de Langen and colleagues (2012) of the test-retest repeatability of quantitative FDG-PET measurements, including mean, maximum, and peak standardized uptake values (SUVmax, SUVmean, and SUVpeak, respectively), was performed. Data from 11 studies in the literature were used to estimate the relationship between the variance in test-retest measurements with uptake level and various study-level, patient-level, and lesion-level characteristics. RESULTS Test-retest repeatability of percentage fluctuations for all three types of SUV measurement (max, mean, and peak) improved with higher FDG uptake levels. Repeatability in all three SUV measurements varied for different lesion locations. Worse repeatability in SUVmean was also associated with higher tumor volumes. CONCLUSIONS On the basis of these results, recommendations regarding SUV measurements for assessing minimal detectable changes based on repeatability and reproducibility are proposed. These should be applied to differentiate between response categories for a future set of FDG-PET-based criteria that assess clinically significant changes in tumor response.
Collapse
Affiliation(s)
| | - Erich Huang
- National Cancer Institute, Bethesda, Maryland
| | - Saskia Litiere
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Otto S Hoekstra
- Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | - Larry Schwartz
- Columbia University Medical Center and New York Presbyterian Hospital, New York, New York
| | | | | | - Jan Bogaerts
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | | | | |
Collapse
|
7
|
van Sluis J, van Snick JH, Brouwers AH, Noordzij W, Dierckx RAJO, Borra RJH, Lammertsma AA, Glaudemans AWJM, Slart RHJA, Yaqub M, Tsoumpas C, Boellaard R. Shortened duration whole body 18F-FDG PET Patlak imaging on the Biograph Vision Quadra PET/CT using a population-averaged input function. EJNMMI Phys 2022; 9:74. [PMID: 36308568 PMCID: PMC9618000 DOI: 10.1186/s40658-022-00504-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/21/2022] [Indexed: 11/13/2022] Open
Abstract
Background Excellent performance characteristics of the Vision Quadra PET/CT, e.g. a substantial increase in sensitivity, allow for precise measurements of image-derived input functions (IDIF) and tissue time activity curves. Previously we have proposed a method for a reduced 30 min (as opposed to 60 min) whole body 18F-FDG Patlak PET imaging procedure using a previously published population-averaged input function (PIF) scaled to IDIF values at 30–60 min post-injection (p.i.). The aim of the present study was to apply this method using the Vision Quadra PET/CT, including the use of a PIF to allow for shortened scan durations. Methods Twelve patients with suspected lung malignancy were included and received a weight-based injection of 18F-FDG. Patients underwent a 65-min dynamic PET acquisition which were reconstructed using European Association of Nuclear Medicine Research Ltd. (EARL) standards 2 reconstruction settings. A volume of interest (VOI) was placed in the ascending aorta (AA) to obtain the IDIF. An external PIF was scaled to IDIF values at 30–60, 40–60, and 50–60 min p.i., respectively, and parametric 18F-FDG influx rate constant (Ki) images were generated using a t* of 30, 40 or 50 min, respectively. Herein, tumour lesions as well as healthy tissues, i.e. liver, muscle tissue, spleen and grey matter, were segmented. Results Good agreement between the IDIF and corresponding PIF scaled to 30–60 min p.i. and 40–60 min p.i. was obtained with 7.38% deviation in Ki. Bland–Altman plots showed excellent agreement in Ki obtained using the PIF scaled to the IDIF at 30–60 min p.i. and at 40–60 min p.i. as all data points were within the limits of agreement (LOA) (− 0.004–0.002, bias: − 0.001); for the 50–60 min p.i. Ki, all except one data point fell in between the LOA (− 0.021–0.012, bias: − 0.005). Conclusions Parametric whole body 18F-FDG Patlak Ki images can be generated non-invasively on a Vision Quadra PET/CT system. In addition, using a scaled PIF allows for a substantial (factor 2 to 3) reduction in scan time without substantial loss of accuracy (7.38% bias) and precision (image quality and noise interference). Supplementary Information The online version contains supplementary material available at 10.1186/s40658-022-00504-9.
Collapse
|
8
|
Pfaehler E, Euba D, Rinscheid A, Hoekstra OS, Zijlstra J, van Sluis J, Brouwers AH, Lapa C, Boellaard R. Convolutional neural networks for automatic image quality control and EARL compliance of PET images. EJNMMI Phys 2022; 9:53. [PMID: 35943622 PMCID: PMC9363539 DOI: 10.1186/s40658-022-00468-w] [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: 11/24/2021] [Accepted: 05/09/2022] [Indexed: 12/02/2022] Open
Abstract
Background Machine learning studies require a large number of images often obtained on different PET scanners. When merging these images, the use of harmonized images following EARL-standards is essential. However, when including retrospective images, EARL accreditation might not have been in place. The aim of this study was to develop a convolutional neural network (CNN) that can identify retrospectively if an image is EARL compliant and if it is meeting older or newer EARL-standards. Materials and methods 96 PET images acquired on three PET/CT systems were included in the study. All images were reconstructed with the locally clinically preferred, EARL1, and EARL2 compliant reconstruction protocols. After image pre-processing, one CNN was trained to separate clinical and EARL compliant reconstructions. A second CNN was optimized to identify EARL1 and EARL2 compliant images. The accuracy of both CNNs was assessed using fivefold cross-validation. The CNNs were validated on 24 images acquired on a PET scanner not included in the training data. To assess the impact of image noise on the CNN decision, the 24 images were reconstructed with different scan durations. Results In the cross-validation, the first CNN classified all images correctly. When identifying EARL1 and EARL2 compliant images, the second CNN identified 100% EARL1 compliant and 85% EARL2 compliant images correctly. The accuracy in the independent dataset was comparable to the cross-validation accuracy. The scan duration had almost no impact on the results. Conclusion The two CNNs trained in this study can be used to retrospectively include images in a multi-center setting by, e.g., adding additional smoothing. This method is especially important for machine learning studies where the harmonization of images from different PET systems is essential.
Supplementary Information The online version contains supplementary material available at 10.1186/s40658-022-00468-w.
Collapse
Affiliation(s)
- Elisabeth Pfaehler
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany.
| | - Daniela Euba
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Andreas Rinscheid
- Medical Physics and Radiation Protection, University Hospital Augsburg, Augsburg, Germany
| | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Josee Zijlstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
9
|
Malaih AA, Dunn JT, Nygård L, Kovacs DG, Andersen FL, Barrington SF, Fischer BM. Test-retest repeatability and interobserver variation of healthy tissue metabolism using 18F-FDG PET/CT of the thorax among lung cancer patients. Nucl Med Commun 2022; 43:549-559. [PMID: 35081091 PMCID: PMC7612596 DOI: 10.1097/mnm.0000000000001537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVES The aim of this study was to assess the test-retest repeatability and interobserver variation in healthy tissue (HT) metabolism using 2-deoxy-2-[18F]fluoro-d-glucose (18F-FDG) PET/computed tomography (PET/CT) of the thorax in lung cancer patients. METHODS A retrospective analysis was conducted in 22 patients with non-small cell lung cancer who had two PET/CT scans of the thorax performed 3 days apart with no interval treatment. The maximum, mean and peak standardized uptake values (SUVs) in different HTs were measured by a single observer for the test-retest analysis and two observers for interobserver variation. Bland-Altman plots were used to assess the repeatability and interobserver variation. Intrasubject variability was evaluated using within-subject coefficients of variation (wCV). RESULTS The wCV of test-retest SUVmean measurements in mediastinal blood pool, bone marrow, skeletal muscles and lungs was less than 20%. The left ventricle (LV) showed higher wCV (>60%) in all SUV parameters with wide limits of repeatability. High interobserver agreement was found with wCV of less than 10% in SUVmean of all HT, but up to 22% was noted in the LV. CONCLUSION HT metabolism is stable in a test-retest scenario and has high interobserver agreement. SUVmean was the most stable metric in organs with low FDG uptake and SUVpeak in HTs with moderate uptake. Test-retest measurements in LV were highly variable irrespective of the SUV parameters used for measurements.
Collapse
Affiliation(s)
- Afnan A Malaih
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, PET Imaging Centre, St Thomas Hospital, King's College London, London, UK
| | - Joel T Dunn
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, PET Imaging Centre, St Thomas Hospital, King's College London, London, UK
| | - Lotte Nygård
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital
| | - David G Kovacs
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Flemming L Andersen
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sally F Barrington
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, PET Imaging Centre, St Thomas Hospital, King's College London, London, UK
| | - Barbara M Fischer
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, PET Imaging Centre, St Thomas Hospital, King's College London, London, UK
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Sui X, Tan H, Yu H, Xiao J, Qi C, Cao Y, Chen S, Zhang Y, Hu P, Shi H. Exploration of the total-body PET/CT reconstruction protocol with ultra-low 18F-FDG activity over a wide range of patient body mass indices. EJNMMI Phys 2022; 9:17. [PMID: 35239037 PMCID: PMC8894532 DOI: 10.1186/s40658-022-00445-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 02/10/2022] [Indexed: 02/02/2023] Open
Abstract
Purpose The purpose of this study was to investigate the image quality and diagnostic performance of different reconstructions over a wide range of patient body mass indices (BMIs) obtained by total-body PET/CT with ultra-low 18F-FDG activity (0.37 MBq/kg). Methods A total of 63 patients who underwent total-body PET/CT with ultra-low activity (0.37 MBq/kg) 18F-FDG were enrolled. Patients were grouped by their BMIs. Images were reconstructed with the following two algorithms: the ordered subset expectation maximization (OSEM) algorithm (2, 3 iterations), both with time of flight (TOF) and point spread function (PSF) corrections (hereinafter referred as OSEM2, OSEM3) and HYPER Iterative algorithm (β-values of 0.3, 0.4, 0.5, 0.6) embedded TOF and PSF technologies (hereinafter referred as HYPER0.3, HYPER0.4, HYPER0.5 and HYPER0.6, respectively). Subjective image quality was assessed by two experienced nuclear medicine physicians according to the Likert quintile, including overall image quality, image noise and lesion conspicuity. The standard deviation (SD) and signal-to-noise ratio (SNR) of the liver, and maximum standard uptake value (SUVmax), peak standard uptake value (SUVpeak), tumour background ratio (T/N) and the largest diameter of lesions were quantitatively analysed by a third reader who did not participate in the subjective image assessment. Results Increased noise was associated with increased BMI in all reconstruction groups. Significant differences occurred in the liver SNR among BMI categories of OSEM reconstructions (P < 0.001) but no difference was seen in the HYPER Iterative reconstructions between any of the BMI categories (P > 0.05). With the increase in BMI, overall image quality and image noise scores decreased significantly in all reconstructions, but there was no statistically significant difference of lesion conspicuity. The overall image quality score of the obese group was not qualified (score = 2.7) in OSEM3, while the others were qualified. The lesion conspicuity scores were significantly higher in HYPER Iterative reconstructions and lower in OSEM2 than in OSEM3 (all P < 0.05). The values of SUVmax, SUVpeak and T/N in HYPER0.3, HYPER0.4 and HYPER0.5 were higher than those in OSEM3. In different reconstructions, there was a correlation between lesion size (median, 1.55 cm; range, 0.7–11.0 cm) and SUVpeak variation rate compared to OSEM3 (r = 0.388, − 0.515, − 0.495, − 0.464, and − 0.423, respectively, and all P < 0.001). Conclusion Considering the image quality and lesion analysis in 18F-FDG total-body PET/CT with ultra-low activity injection, OSEM reconstructions with 3 iterations meet the clinical requirements in patients with BMI < 30. In patients with BMI ≥ 30, it is recommended that the HYPER Iterative algorithm (β-value of 0.3–0.5) be used to ensure consistent visual image quality and quantitative assessment.
Collapse
Affiliation(s)
- Xiuli Sui
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China.,Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Hui Tan
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China.,Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Haojun Yu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China.,Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Jie Xiao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China.,Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Chi Qi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China.,Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Yanyan Cao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China.,Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Shuguang Chen
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China.,Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Yiqiu Zhang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China.,Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Pengcheng Hu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China.,Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China. .,Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China. .,Shanghai Institute of Medical Imaging, Shanghai, 200032, China.
| |
Collapse
|
11
|
Rogasch JMM, Hofheinz F, van Heek L, Voltin CA, Boellaard R, Kobe C. Influences on PET Quantification and Interpretation. Diagnostics (Basel) 2022; 12:451. [PMID: 35204542 PMCID: PMC8871060 DOI: 10.3390/diagnostics12020451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/06/2022] [Accepted: 02/08/2022] [Indexed: 01/21/2023] Open
Abstract
Various factors have been identified that influence quantitative accuracy and image interpretation in positron emission tomography (PET). Through the continuous introduction of new PET technology-both imaging hardware and reconstruction software-into clinical care, we now find ourselves in a transition period in which traditional and new technologies coexist. The effects on the clinical value of PET imaging and its interpretation in routine clinical practice require careful reevaluation. In this review, we provide a comprehensive summary of important factors influencing quantification and interpretation with a focus on recent developments in PET technology. Finally, we discuss the relationship between quantitative accuracy and subjective image interpretation.
Collapse
Affiliation(s)
- Julian M. M. Rogasch
- Department of Nuclear Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany;
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, 10178 Berlin, Germany
| | - Frank Hofheinz
- Institute of Radiopharmaceutical Cancer Research, Helmholtz Center Dresden-Rossendorf, 01328 Dresden, Germany;
| | - Lutz van Heek
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (L.v.H.); (C.-A.V.)
| | - Conrad-Amadeus Voltin
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (L.v.H.); (C.-A.V.)
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam (CCA), Amsterdam University Medical Center, Free University Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Carsten Kobe
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (L.v.H.); (C.-A.V.)
| |
Collapse
|
12
|
Kolinger GD, Vállez García D, Kramer GM, Frings V, Zwezerijnen GJC, Smit EF, De Langen AJ, Buvat I, Boellaard R. Effects of tracer uptake time in non-small cell lung cancer 18F-FDG PET radiomics. J Nucl Med 2021; 63:919-924. [PMID: 34933890 DOI: 10.2967/jnumed.121.262660] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/21/2021] [Indexed: 11/16/2022] Open
Abstract
Positron emission tomography (PET) radiomics applied to oncology allows the measurement of intra-tumoral heterogeneity. This quantification can be affected by image protocols hence there is an increased interest in understanding how radiomic expression on PET images is affected by different imaging conditions. To address that, this study explores how radiomic features are affected by changes in 18F-FDG uptake time, image reconstruction, lesion delineation, and radiomics binning settings. Methods: Ten non-small cell lung cancer (NSCLC) patients underwent 18F-FDG PET scans on two consecutive days. On each day, scans were obtained at 60min and 90min post-injection and reconstructed following EARL version 1 (EARL1) and with point-spread-function resolution modelling (PSF-EARL2). Lesions were delineated using thresholds at SUV=4.0, 40% of SUVmax, and with a contrast-based isocontour. PET image intensity was discretized with both fixed bin width (FBW) and fixed bin number (FBN) before the calculation of the radiomic features. Repeatability of features was measured with intraclass correlation (ICC), and the change in feature value over time was calculated as a function of its repeatability. Features were then classified on use-case scenarios based on their repeatability and susceptibility to tracer uptake time. Results: With PSF-EARL2 reconstruction, 40% of SUVmax lesion delineation, and FBW intensity discretization, most features (94%) were repeatable at both uptake times (ICC>0.9), 39% being classified for dual-time-point use-case for being sensitive to changes in uptake time, 39% were classified for cross-sectional studies with unclear dependency on time, 20% classified for cross-sectional use while being robust to tracer uptake time changes, and 6% were discarded for poor repeatability. EARL1 images had one less repeatable feature than PSF-EARL2 (Neighborhood Gray-Level Different Matrix Coarseness), the contrast-based delineation had the poorest repeatability of the delineation methods with 45% features being discarded, and FBN resulted in lower repeatability than FBW (45% and 6% features were discarded, respectively). Conclusion: Repeatability was maximized with PSF-EARL2 reconstruction, lesion delineation at 40% of SUVmax, and FBW intensity discretization. Based on their susceptibility to tracer uptake time, radiomic features were classified into specific NSCLC PET radiomics use-cases.
Collapse
Affiliation(s)
| | - David Vállez García
- Medical Imaging Center, University Medical Center Groningen, University of Groningen, Netherlands
| | - Gerbrand Maria Kramer
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VU Medical Center, Netherlands
| | - Virginie Frings
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VU Medical Center, Netherlands
| | | | - Egbert F Smit
- Department of Pulmonology, Amsterdam University Medical Center, location VU Medical Center, Netherlands
| | | | - Irène Buvat
- Laboratoire d'Imagerie Translationnelle en Oncologie, INSERM, Institut Curie, Université Paris-Saclay, France
| | - Ronald Boellaard
- Medical Imaging Center, University Medical Center Groningen, University of Groningen, Netherlands
| |
Collapse
|
13
|
Yamane T, Takahashi M, Matsusaka Y, Fukushima K, Seto A, Kuji I, Matsunari I. Satisfied quantitative value can be acquired by short-time bone SPECT/CT using a whole-body cadmium-zinc-telluride gamma camera. Sci Rep 2021; 11:24320. [PMID: 34934145 PMCID: PMC8692318 DOI: 10.1038/s41598-021-03853-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/10/2021] [Indexed: 11/15/2022] Open
Abstract
The aim of this study was to evaluate the quantitative values of short-time scan (STS) of metastatic lesions compared with a standard scan (SS) when acquired by whole-body bone SPECT/CT with cadmium-zinc-telluride (CZT) detectors. We retrospectively reviewed 13 patients with bone metastases from prostate cancer, who underwent SPECT/CT performed on whole-body CZT gamma cameras. STSs were obtained using 75, 50, 25, 10, and 5% of the list-mode data for SS, respectively. Regions of interest (ROIs) were set on the increased uptake areas diagnosed as metastases. Intraclass correlation coefficients (ICCs) of standardized uptake values (SUVs) for the ROIs were calculated between the SS and each STS, and ICC ≥ 0.8 was set as a perfect correlation. Moreover, the repeatability coefficient (RC) was calculated, and RC ≤ 20% was defined as acceptable. A total of 152 metastatic lesions were included in the analysis. The ICCs between the SS vs. 75%-STS, 50%-STS, 25%-STS, 10%-STS, and 5%-STS were 0.999, 0.997, 0.994, 0.983, and 0.955, respectively. The RCs of the SS vs. 75%-STS, 50%-STS, 25%-STS, 10%-STS, and 5%-STS were 7.9, 12.4, 19.8, 30.8, and 41.3%, respectively. When evaluating the quality of CZT bone SPECT/CT acquired by a standard protocol, 25%-STS may provide adequate quantitative values.
Collapse
Affiliation(s)
- Tomohiko Yamane
- Division of Nuclear Medicine, Department of Radiology, Saitama Medical University Hospital, 38 Moro-Hongo, Moroyama, 350-0495, Japan.
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-0491, Japan.
| | - Masafumi Takahashi
- Department of Central Radiological Technology, Saitama Medical University Hospital, 38 Moro-Hongo, Moroyama, 350-0495, Japan
| | - Yohji Matsusaka
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-0491, Japan
| | - Kenji Fukushima
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-0491, Japan
| | - Akira Seto
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-0491, Japan
| | - Ichiei Kuji
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-0491, Japan
| | - Ichiro Matsunari
- Division of Nuclear Medicine, Department of Radiology, Saitama Medical University Hospital, 38 Moro-Hongo, Moroyama, 350-0495, Japan
| |
Collapse
|
14
|
Zuo M, Yao L, Wen L, Shen J, Zhang N, Bai T, Huang Q. The expression of miRNA-216b is negatively correlated with 18F-FDG uptake in non-small cell lung cancer. World J Surg Oncol 2021; 19:262. [PMID: 34470640 PMCID: PMC8411519 DOI: 10.1186/s12957-021-02376-2] [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: 03/09/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background This study aimed to investigate the correlation between miRNA-216b expression in patients with non-small cell lung cancer (NSCLC) and 18F-fluorodeoxyglucose (FDG) uptake by PET/CT and to explore the clinical application value of 18F-FDG PET/CT in miRNA-216b based on therapy for NSCLC. Methods Eighty patients with NSCLC and 40 healthy subjects were enrolled in our study. The SUVmax of the lesion area by PET/CT imaging was calculated. SUVmax represented the highest concentration of 18F-FDG in the lesion. The expression of miRNA-216b in the plasma and fiber bronchoscopic puncture of NSCLC patients was detected by RT qPCR. Then Pearson correlation analysis was used to analyze the correlation between miRNA-216b expression and 18F-FDG uptake in patients with different types of NSCLC. Results Compared with healthy subjects, SUVmax of early adenocarcinoma and advanced adenocarcinoma were increased. Compared with healthy subjects, SUVmax of early squamous and advanced squamous were increased. And the SUVmax content of advanced adenocarcinoma and squamous cell carcinoma was higher than that of early adenocarcinoma and squamous cell carcinoma. Compared with healthy subjects, the expression of miRNA-216b in the plasma of patients with early and advanced adenocarcinoma was reduced, and the expression of miRNA-216b in the plasma of patients with early and advanced squamous cell carcinoma was reduced. Compared with adjacent tissues, the expression of miRNA-216b in early adenocarcinoma tissues and advanced adenocarcinoma tissues was reduced, and the expression in early squamous cell carcinoma and advanced squamous cell carcinoma was reduced. Pearson correlation analysis showed a negative correlation between SUVmax and miRNA-216b (plasma and tissue) in patients with four types of NSCLC. Conclusion miRNA-216b expression was negatively correlated with 18F-FDG uptake in NSCLC. miRNA-216b could be used for the classification and staging of non-small cell lung cancer. 18F-FDG PET/CT may be used to evaluate the therapeutic response in application of miRNA-216b-based cancer treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-021-02376-2.
Collapse
Affiliation(s)
- Mingfei Zuo
- Imaging Center, The Third Affiliated Hospital of Qiqihar Medical University, No. 27 Taishun Street, Tiefeng District, Qiqihar, 161002, Heilongjiang, China
| | - Lan Yao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161002, Heilongjiang, China
| | - Lijuan Wen
- Imaging Center, The Third Affiliated Hospital of Qiqihar Medical University, No. 27 Taishun Street, Tiefeng District, Qiqihar, 161002, Heilongjiang, China
| | - Jianfei Shen
- Department of Nuclear Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161002, Heilongjiang, China
| | - Na Zhang
- Imaging Center, The Third Affiliated Hospital of Qiqihar Medical University, No. 27 Taishun Street, Tiefeng District, Qiqihar, 161002, Heilongjiang, China
| | - Tian Bai
- Imaging Center, The Third Affiliated Hospital of Qiqihar Medical University, No. 27 Taishun Street, Tiefeng District, Qiqihar, 161002, Heilongjiang, China
| | - Qicheng Huang
- Imaging Center, The Third Affiliated Hospital of Qiqihar Medical University, No. 27 Taishun Street, Tiefeng District, Qiqihar, 161002, Heilongjiang, China.
| |
Collapse
|
15
|
Wibmer AG, Morris MJ, Gonen M, Zheng J, Hricak H, Larson S, Scher HI, Vargas HA. Quantification of Metastatic Prostate Cancer Whole-Body Tumor Burden with 18F-FDG PET Parameters and Associations with Overall Survival After First-Line Abiraterone or Enzalutamide: A Single-Center Retrospective Cohort Study. J Nucl Med 2021; 62:1050-1056. [PMID: 33419944 PMCID: PMC8833874 DOI: 10.2967/jnumed.120.256602] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022] Open
Abstract
New biomarkers for metastatic prostate cancer are needed. The aim of this study was to evaluate the prognostic value of 18F-FDG PET whole-body tumor burden parameters in patients with metastatic prostate cancer who received first-line abiraterone or enzalutamide therapy. Methods: This was a retrospective study of patients with metastatic castration-sensitive prostate cancer (mCSPC, n = 25) and metastatic castration-resistant prostate cancer (mCRPC, n = 71) who underwent 18F-FDG PET/CT within 90 d before first-line treatment with abiraterone or enzalutamide at a tertiary-care academic cancer center. Whole-body tumor burden on PET/CT was quantified as metabolic tumor volume (MTV) and total lesion glycolysis (TLG) and correlated with overall survival (OS) probabilities using Kaplan-Meier curves and Cox models. Results: The median follow-up in survivors was 56.3 mo (interquartile range, 37.7-66.8 mo); the median OSs for patients with mCRPC and mCSPC were 27.8 and 76.1 mo, respectively (P < 0.001). On univariate analysis, the OS probability of mCRPC patients was significantly associated with plasma levels of alkaline phosphatase (hazard ratio [HR], 1.90; P < 0.001), plasma levels of lactate dehydrogenase (HR, 1.01; P < 0.001), hemoglobin levels (HR, 0.80; P = 0.013), whole-body SUVmax (HR, 1.14; P < 0.001), the number of 18F-FDG-avid metastases (HR, 1.08; P < 0.001), whole-body metabolic tumor volume (HR, 1.86; P < 0.001), and TLG (HR, 1.84; P < 0.001). On multivariable analysis with stepwise variable selection, hemoglobin levels (HR, 0.81; P = 0.013) and whole-body TLG (HR, 1.88; P < 0.001) were independently associated with OS. In mCSPC patients, no significant association was observed between these variables and OS. Conclusion: In patients with mCRPC receiving first-line treatment with abiraterone or enzalutamide, 18F-FDG PET WB TLG is independently associated with OS and might be used as a quantitative prognostic imaging biomarker.
Collapse
Affiliation(s)
- Andreas G Wibmer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
| | - Michael J Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Junting Zheng
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hedvig Hricak
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Howard I Scher
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | | |
Collapse
|
16
|
Rijnsdorp S, Roef MJ, Arends AJ. Impact of the Noise Penalty Factor on Quantification in Bayesian Penalized Likelihood (Q.Clear) Reconstructions of 68Ga-PSMA PET/CT Scans. Diagnostics (Basel) 2021; 11:diagnostics11050847. [PMID: 34066854 PMCID: PMC8150604 DOI: 10.3390/diagnostics11050847] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 01/04/2023] Open
Abstract
Functional imaging with 68Ga prostate-specific membrane antigen (PSMA) and positron emission tomography (PET) can fulfill an important role in treatment selection and adjustment in prostate cancer. This article focusses on quantitative assessment of 68Ga-PSMA-PET. The effect of various parameters on standardized uptake values (SUVs) is explored, and an optimal Bayesian penalized likelihood (BPL) reconstruction is suggested. PET acquisitions of two phantoms consisting of a background compartment and spheres with diameter 4 mm to 37 mm, both filled with solutions of 68Ga in water, were performed with a GE Discovery 710 PET/CT scanner. Recovery coefficients (RCs) in multiple reconstructions with varying noise penalty factors and acquisition times were determined and analyzed. Apparent recovery coefficients of spheres with a diameter smaller than 17 mm were significantly lower than those of spheres with a diameter of 17 mm and bigger (p < 0.001) for a tumor-to-background (T/B) ratio of 10:1 and a scan time of 10 min per bed position. With a T/B ratio of 10:1, the four largest spheres exhibit significantly higher RCs than those with a T/B ratio of 20:1 (p < 0.0001). For spheres with a diameter of 8 mm and less, alignment with the voxel grid potentially affects the RC. Evaluation of PET/CT scans using (semi-)quantitative measures such as SUVs should be performed with great caution, as SUVs are influenced by scanning and reconstruction parameters. Based on the evaluation of multiple reconstructions with different β of phantom scans, an intermediate β (600) is suggested as the optimal value for the reconstruction of clinical 68Ga-PSMA PET/CT scans, considering that both detectability and reproducibility are relevant.
Collapse
Affiliation(s)
- Sjoerd Rijnsdorp
- Department of Medical Physics, Catharina Hospital Eindhoven, Michelangelolaan 2, 5623 EJ Eindhoven, The Netherlands;
- Correspondence:
| | - Mark J. Roef
- Department of Nuclear Medicine, Catharina Hospital Eindhoven, Michelangelolaan 2, 5623 EJ Eindhoven, The Netherlands;
| | - Albert J. Arends
- Department of Medical Physics, Catharina Hospital Eindhoven, Michelangelolaan 2, 5623 EJ Eindhoven, The Netherlands;
| |
Collapse
|
17
|
Kim JS, Park CR, Yoon SH, Lee JA, Kim TY, Yang HJ. Improvement of image quality using amplitude-based respiratory gating in PET-computed tomography scanning. Nucl Med Commun 2021; 42:553-565. [PMID: 33625179 DOI: 10.1097/mnm.0000000000001368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES This study sought to provide data supporting the expanded clinical use of respiratory gating by assessing the diagnostic accuracy of breathing motion correction using amplitude-based respiratory gating. METHODS A respiratory movement tracking device was attached to a PET-computed tomography scanner, and images were obtained in respiratory gating mode using a motion phantom that was capable of sensing vertical motion. Specifically, after setting amplitude changes and intervals according to the movement cycle using a total of nine combinations of three waveforms and three amplitude ranges, respiratory motion-corrected images were reconstructed using the filtered back projection method. After defining areas of interest in the acquired images in the same image planes, statistical analyses were performed to compare differences in standardized uptake value (SUV), lesion volume, full width at half maximum (FWHM), signal-to-noise ratio (SNR) and contrast-to-noise ratio (CNR). RESULTS SUVmax increased by 89.9%, and lesion volume decreased by 27.9%. Full width at half maximum decreased by 53.9%, signal-to-noise ratio increased by 11% and contrast-to-noise ratio increased by 16.3%. Optimal results were obtained when using a rest waveform and 35% duty cycle, in which the change in amplitude in the respiratory phase signal was low, and a constant level of long breaths was maintained. CONCLUSIONS These results demonstrate that respiratory-gated PET-CT imaging can be used to accurately correct for SUV changes and image distortion caused by respiratory motion, thereby providing excellent imaging information and quality.
Collapse
Affiliation(s)
- Jung-Soo Kim
- Department of Radiological Technology, Dongnam Health University, Suwon
- Department of Biomedical Science, The Korea University, Sejong
| | - Chan-Rok Park
- Department of Biomedical Science, The Korea University, Sejong
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul
| | - Seok-Hwan Yoon
- Department of Biomedical Science, The Korea University, Sejong
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul
| | - Joo-Ah Lee
- Department of Biomedical Science, The Korea University, Sejong
- Department of Radiation Oncology, Catholic University Incheon St. Mary's Hospital, Incheon
| | - Tae-Yoon Kim
- Department of Radiation Oncology, Catholic University Incheon St. Mary's Hospital, Incheon
- Department of Radiation Oncology, National Cancer Center, Goyang
| | - Hyung-Jin Yang
- Department of Radiation Oncology, Catholic University Incheon St. Mary's Hospital, Incheon
- Department of Physics, The Korea University, Sejong, Korea
| |
Collapse
|
18
|
Yamane T, Fukushima K, Shirotake S, Nishimoto K, Okabe T, Oyama M, Seto A, Kuji I. Test-retest repeatability of quantitative bone SPECT/CT. Ann Nucl Med 2021; 35:338-346. [PMID: 33400148 DOI: 10.1007/s12149-020-01568-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/11/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Technological innovations in single-photon emission computed tomography (SPECT) have enabled a more accurate quantitative evaluation of the uptake, and the standardized uptake value (SUV) can be measured as a semi-quantitative value, as in positron emission tomography. Nevertheless, the reliability of the SUV of bone SPECT has not been well established. The purpose of this study is to evaluate the test-retest repeatability of the SUV of bone SPECT/CT in clinical settings. METHODS This prospective study recruited patients with prostate cancer planning to receive bone SPECT/CT for the evaluation of bone abnormality between August 2017 and September 2019. Bone images were acquired twice by an integrated SPECT/CT scanner (Symbia Intevo, Siemens) within a 4- to 10-day interval. The maximum SUV (SUVmax) and peak SUV (SUVpeak) were calculated for the volumes of interests on the normal bone areas, degeneration/fracture lesions, and metastatic lesions. To determine repeatability, we calculated statistical indicators, including intraclass correlation coefficient (ICC), repeatability coefficient (RC), and mean absolute percentage difference (MAPD). For the ICC, the 95% confidential interval (CI) was also calculated, and an ICC of ≥ 0.8 was defined as an almost perfect correlation. RESULTS Twelve male patients were enrolled in the study (58-86 years; median, 71 years), and a total of 229 volumes of the interest were included in the analyses. The ICCs were 0.968 [95% CI (0.959, 0.975)] for SUVmax and 0.976 [95% CI (0.969, 0.981)] for SUVpeak. The RCs of the relative difference were 30.7% for SUVmax and 27.6% for SUVpeak, and the MAPDs (± standardized deviation) of all lesions were 12.3 ± 9.9% for SUVmax and 11.5 ± 8.3% for SUVpeak. The RCs and the MAPDs showed comparable value with the previous report regarding repeatability studies on PET. CONCLUSION An almost perfect correlation was demonstrated by repeated SUVmax and SUVpeak measured by quantitative integrated SPECT/CT. The quantitative values could be reliable indicators in patient management.
Collapse
Affiliation(s)
- Tomohiko Yamane
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1298, Japan.
| | - Kenji Fukushima
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1298, Japan
| | - Suguru Shirotake
- Department of Uro-Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1298, Japan
| | - Koshiro Nishimoto
- Department of Uro-Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1298, Japan
| | - Takashi Okabe
- Department of Uro-Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1298, Japan
| | - Masafumi Oyama
- Department of Uro-Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1298, Japan
| | - Akira Seto
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1298, Japan
| | - Ichiei Kuji
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1298, Japan
| |
Collapse
|
19
|
Pfaehler E, Mesotten L, Zhovannik I, Pieplenbosch S, Thomeer M, Vanhove K, Adriaensens P, Boellaard R. Plausibility and redundancy analysis to select FDG-PET textural features in non-small cell lung cancer. Med Phys 2021; 48:1226-1238. [PMID: 33368399 PMCID: PMC7985880 DOI: 10.1002/mp.14684] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 01/06/2023] Open
Abstract
Background Radiomics refers to the extraction of a large number of image biomarker describing the tumor phenotype displayed in a medical image. Extracted from positron emission tomography (PET) images, radiomics showed diagnostic and prognostic value for several cancer types. However, a large number of radiomic features are nonreproducible or highly correlated with conventional PET metrics. Moreover, radiomic features used in the clinic should yield relevant information about tumor texture. In this study, we propose a framework to identify technical and clinical meaningful features and exemplify our results using a PET non‐small cell lung cancer (NSCLC) dataset. Materials and methods The proposed selection procedure consists of several steps. A priori, we only include features that were found to be reproducible in a multicenter setting. Next, we apply a voxel randomization step to identify features that reflect actual textural information, that is, that yield in 90% of the patient scans a value significantly different from random texture. Finally, the remaining features were correlated with standard PET metrics to further remove redundancy with common PET metrics. The selection procedure was performed for different volume ranges, that is, excluding lesions with smaller volumes in order to assess the effect of tumor size on the results. To exemplify our procedure, the selected features were used to predict 1‐yr survival in a dataset of 150 NSCLC patients. A predictive model was built using volume as predictive factor for smaller, and one of the selected features as predictive factor for bigger lesions. The prediction accuracy of the both models were compared with the prediction accuracy of volume. Results The number of selected features depended on the lesion size included in the analysis. When including the whole dataset, from 19 features reflecting actual texture only two were found to be not strongly correlated with conventional PET metrics. When excluding lesions smaller than 11.49 and 33.10 mL (25 and 50 percentile of the dataset), four out of 27 features and 13 out of 29 features remained after eliminating features highly correlated with standard PET metrics. When excluding lesions smaller than 103.9 mL (75 percentile), 33 out of 53 features remained. For larger lesions, some of these features outperformed volume in terms of classification accuracy (increase of 4–10%). The combination of using volume as predictor for smaller and one of the selected features for larger lesions also improved the accuracy when compared with volume only (increase from 72% to 76%). Conclusion When performing radiomic analysis for smaller lesions, it should be first carefully investigated if a textural feature reflects actual heterogeneity information. Next, verification of the absence of correlation with all conventional PET metrics is essential in order to assess the additional value of radiomic features. Radiomic analysis with lesions larger than 11.4 mL might give additional information to conventional metrics while at the same time reflecting actual tumor texture. Using a combination of volume and one of the selected features for prediction yields promise to increase accuracy and reliability of a radiomic model.
Collapse
Affiliation(s)
- Elisabeth Pfaehler
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Liesbet Mesotten
- Faculty of Medicine and Life Sciences, Hasselt University, Agoralaan building D, Diepenbeek, B-3590, Belgium.,Department of Nuclear Medicine, Ziekenhuis Oost Limburg, Schiepse Bos 6, Genk, B-3600, Belgium
| | - Ivan Zhovannik
- Department of Radiation Oncology, Radboudumc, Nijmegen, The Netherlands.,Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht, The Netherlands
| | - Simone Pieplenbosch
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Michiel Thomeer
- Faculty of Medicine and Life Sciences, Hasselt University, Agoralaan building D, Diepenbeek, B-3590, Belgium.,Department of Nuclear Medicine, Ziekenhuis Oost Limburg, Schiepse Bos 6, Genk, B-3600, Belgium
| | - Karolien Vanhove
- Faculty of Medicine and Life Sciences, Hasselt University, Agoralaan building D, Diepenbeek, B-3590, Belgium.,Department of Respiratory Medicine, Ziekenhuis Oost Limburg, Schiepse Bos 6, Genk, B-3600, Belgium
| | - Peter Adriaensens
- Hasselt University, Institute for Materials Research (IMO) - Division Chemistry, Agoralaan Building D, Diepenbeek, B 3590, Belgium
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
20
|
van Sluis J, Yaqub M, Brouwers AH, Dierckx RAJO, Noordzij W, Boellaard R. Use of population input functions for reduced scan duration whole-body Patlak 18F-FDG PET imaging. EJNMMI Phys 2021; 8:11. [PMID: 33547518 PMCID: PMC7865035 DOI: 10.1186/s40658-021-00357-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/22/2021] [Indexed: 11/10/2022] Open
Abstract
Abstract Whole-body Patlak images can be obtained from an acquisition of first 6 min of dynamic imaging over the heart to obtain the arterial input function (IF), followed by multiple whole-body sweeps up to 60 min pi. The use of a population-averaged IF (PIF) could exclude the first dynamic scan and minimize whole-body sweeps to 30–60 min pi. Here, the effects of (incorrect) PIFs on the accuracy of the proposed Patlak method were assessed. In addition, the extent of mitigating these biases through rescaling of the PIF to image-derived IF values at 30–60 min pi was evaluated. Methods Using a representative IF and rate constants from the literature, various tumour time-activity curves (TACs) were simulated. Variations included multiplication of the IF with a positive and negative gradual linear bias over 60 min of 5, 10, 15, 20, and 25% (generating TACs using an IF different from the PIF); use of rate constants (K1, k3, and both K1 and k2) multiplied by 2, 1.5, and 0.75; and addition of noise (μ = 0 and σ = 5, 10 and 15%). Subsequent Patlak analysis using the original IF (representing the PIF) was used to obtain the influx constant (Ki) for the differently simulated TACs. Next, the PIF was scaled towards the (simulated) IF value using the 30–60-min pi time interval, simulating scaling of the PIF to image-derived values. Influence of variabilities in IF and rate constants, and rescaling the PIF on bias in Ki was evaluated. Results Percentage bias in Ki observed using simulated modified IFs varied from − 16 to 16% depending on the simulated amplitude and direction of the IF modifications. Subsequent scaling of the PIF reduced these Ki biases in most cases (287 out of 290) to < 5%. Conclusions Simulations suggest that scaling of a (possibly incorrect) PIF to IF values seen in whole-body dynamic imaging from 30 to 60 min pi can provide accurate Ki estimates. Consequently, dynamic Patlak imaging protocols may be performed for 30–60 min pi making whole-body Patlak imaging clinically feasible.
Collapse
Affiliation(s)
- Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Walter Noordzij
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Predicting tumor response and outcome of second-look surgery with 18F-FDG PET/CT: insights from the GINECO CHIVA phase II trial of neoadjuvant chemotherapy plus nintedanib in stage IIIc-IV FIGO ovarian cancer. Eur J Nucl Med Mol Imaging 2020; 48:1998-2008. [PMID: 33221969 PMCID: PMC8113167 DOI: 10.1007/s00259-020-05092-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Abstract
Background This ancillary study aimed to evaluate 18F-FDG PET parameter changes after one cycle of treatment compared to baseline in patients receiving first-line neoadjuvant anti-angiogenic nintedanib combined to paclitaxel-carboplatin chemotherapy or chemotherapy plus placebo and to evaluate the ability of 18F-FDG PET parameters to predict progression-free survival (PFS), overall survival (OS), and success of second-look surgery. Materials and methods Central review was performed by two readers blinded to the received treatment and to the patients’ outcome, in consensus, by computing percentage change in PET metrics within a volume of interest encompassing the entire tumor burden. EORTC and PERCIST criteria were applied to classify patients as responders (partial metabolic response and complete metabolic response) or non-responders (stable metabolic disease and progressive metabolic disease). Also analyzed was the percentage change in metabolic active tumor volume (MATV) and total lesion glycolysis (TLG). Results Twenty-four patients were included in this ancillary study: 10 received chemotherapy + placebo and 14 chemotherapy + nintedanib. PERCIST and EORTC criteria showed similar discriminative power in predicting PSF and OS. Variation in MATV/TLG did not predict PFS or OS, and no optimal threshold could be found for MATV/TLG for predicting survival. Complete cytoreductive surgery (no residual disease versus residual disease < 0.25 cm/0.25–2.5 cm/> 2.5 cm) was more frequent in responders versus non-responders (P = 0.002 for PERCIST and P = 0.02 for EORTC criteria). No correlation was observed between the variation of PET data and the variation of CA-125 blood level between baseline sample and that performed contemporary to the interim PET, but a statistically significant correlation was observed between ΔSULpeak and ΔCA-125 between baseline sample and that performed after the second cycle. Conclusion 18F-FDG PET using EORTC or PERCIST criteria appeared to be a useful tool in ovarian cancer trials to analyze early tumor response, and predict second-look surgery outcome and survival. An advantage of PERCIST is the correlation of ΔSULpeak and ΔCA-125, PET response preceding tumor markers response by 1 month. Neither MATV nor TLG was useful in predicting survival. Trial registration NCT01583322 ARCAGY/ GINECO GROUP GINECO-OV119, 24 April 2012 Supplementary Information The online version contains supplementary material available at 10.1007/s00259-020-05092-3.
Collapse
|
22
|
Rogasch JMM, Furth C, Bluemel S, Radojewski P, Amthauer H, Hofheinz F. Asphericity of tumor FDG uptake in non-small cell lung cancer: reproducibility and implications for harmonization in multicenter studies. EJNMMI Res 2020; 10:134. [PMID: 33140213 PMCID: PMC7606415 DOI: 10.1186/s13550-020-00725-y] [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: 09/16/2020] [Accepted: 10/21/2020] [Indexed: 11/15/2022] Open
Abstract
Background Asphericity (ASP) of the primary tumor’s metabolic tumor volume (MTV) in FDG-PET/CT is independently predictive for survival in patients with non-small cell lung cancer (NSCLC). However, comparability between PET systems may be limited. Therefore, reproducibility of ASP was evaluated at varying image reconstruction and acquisition times to assess feasibility of ASP assessment in multicenter studies.
Methods This is a retrospective study of 50 patients with NSCLC (female 20; median age 69 years) undergoing pretherapeutic FDG-PET/CT (median 3.7 MBq/kg; 180 s/bed position). Reconstruction used OSEM with TOF4/16 (iterations 4; subsets 16; in-plane filter 2.0, 6.4 or 9.5 mm), TOF4/8 (4 it; 8 ss; filter 2.0/6.0/9.5 mm), PSF + TOF2/17 (2 it; 17 ss; filter 2.0/7.0/10.0 mm) or Bayesian-penalized likelihood (Q.Clear; beta, 600/1750/4000). Resulting reconstructed spatial resolution (FWHM) was determined from hot sphere inserts of a NEMA IEC phantom. Data with approx. 5-mm FWHM were retrospectively smoothed to achieve 7-mm FWHM. List mode data were rebinned for acquisition times of 120/90/60 s. Threshold-based delineation of primary tumor MTV was followed by evaluation of relative ASP/SUVmax/MTV differences between datasets and resulting proportions of discordantly classified cases.
Results Reconstructed resolution for narrow/medium/wide in-plane filter (or low/medium/high beta) was approx. 5/7/9 mm FWHM. Comparing different pairs of reconstructed resolution between TOF4/8, PSF + TOF2/17, Q.Clear and the reference algorithm TOF4/16, ASP differences was lowest at FWHM of 7 versus 7 mm. Proportions of discordant cases (ASP > 19.5% vs. ≤ 19.5%) were also lowest at 7 mm (TOF4/8, 2%; PSF + TOF2/17, 4%; Q.Clear, 10%). Smoothing of 5-mm data to 7-mm FWHM significantly reduced discordant cases (TOF4/8, 38% reduced to 2%; PSF + TOF2/17, 12% to 4%; Q.Clear, 10% to 6%), resulting in proportions comparable to original 7-mm data. Shorter acquisition time only increased proportions of discordant cases at < 90 s. Conclusions ASP differences were mainly determined by reconstructed spatial resolution, and multicenter studies should aim at comparable FWHM (e.g., 7 mm; determined by in-plane filter width). This reduces discordant cases (high vs. low ASP) to an acceptable proportion for TOF and PSF + TOF of < 5% (Q.Clear: 10%). Data with better resolution (i.e., lower FWHM) could be retrospectively smoothed to the desired FWHM, resulting in a comparable number of discordant cases.
Collapse
Affiliation(s)
- Julian M M Rogasch
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Christian Furth
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Stephanie Bluemel
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Piotr Radojewski
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Holger Amthauer
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Frank Hofheinz
- Institute for Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| |
Collapse
|
23
|
Boers J, de Vries EFJ, Glaudemans AWJM, Hospers GAP, Schröder CP. Application of PET Tracers in Molecular Imaging for Breast Cancer. Curr Oncol Rep 2020; 22:85. [PMID: 32627087 PMCID: PMC7335757 DOI: 10.1007/s11912-020-00940-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Molecular imaging with positron emission tomography (PET) is a powerful tool to visualize breast cancer characteristics. Nonetheless, implementation of PET imaging into cancer care is challenging, and essential steps have been outlined in the international "imaging biomarker roadmap." In this review, we identify hurdles and provide recommendations for implementation of PET biomarkers in breast cancer care, focusing on the PET tracers 2-[18F]-fluoro-2-deoxyglucose ([18F]-FDG), sodium [18F]-fluoride ([18F]-NaF), 16α-[18F]-fluoroestradiol ([18F]-FES), and [89Zr]-trastuzumab. RECENT FINDINGS Technical validity of [18F]-FDG, [18F]-NaF, and [18F]-FES is established and supported by international guidelines. However, support for clinical validity and utility is still pending for these PET tracers in breast cancer, due to variable endpoints and procedures in clinical studies. Assessment of clinical validity and utility is essential towards implementation; however, these steps are still lacking for PET biomarkers in breast cancer. This could be solved by adding PET biomarkers to randomized trials, development of imaging data warehouses, and harmonization of endpoints and procedures.
Collapse
Affiliation(s)
- Jorianne Boers
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Erik F J de Vries
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andor W J M Glaudemans
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Geke A P Hospers
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Carolina P Schröder
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
24
|
Kwiecinski J, Cadet S, Daghem M, Lassen ML, Dey D, Dweck MR, Berman DS, Newby DE, Slomka PJ. Whole-vessel coronary 18F-sodium fluoride PET for assessment of the global coronary microcalcification burden. Eur J Nucl Med Mol Imaging 2020; 47:1736-1745. [PMID: 31897586 PMCID: PMC7271818 DOI: 10.1007/s00259-019-04667-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 12/20/2019] [Indexed: 01/12/2023]
Abstract
PURPOSE 18F-sodium fluoride (18F-NaF) has shown promise in assessing disease activity in coronary arteries, but currently used measures of activity - such as maximum target to background ratio (TBRmax) - are defined by single pixel count values. We aimed to develop a novel coronary-specific measure of 18F-NaF PET reflecting activity throughout the entire coronary vasculature (coronary microcalcification activity [CMA]). METHODS Patients with recent myocardial infarction and multi-vessel coronary artery disease underwent 18F-NaF PET and coronary CT angiography. We assessed the association between coronary 18F-NaF uptake (both TBRmax and CMA) and coronary artery calcium scores (CACS) as well as low attenuation plaque (LAP, attenuation < 30 Hounsfield units) volume. RESULTS In 50 patients (64% males, 63 ± 7 years), CMA and TBRmax were higher in vessels with LAP compared to those without LAP (1.09 [0.02, 2.34] versus 0.0 [0.0, 0.0], p < 0.001 and 1.23 [1.16, 1.37] versus 1.04 [0.93, 1.11], p < 0.001). Compared to a TBRmax threshold of 1.25, CMA > 0 had a higher diagnostic accuracy for detection of LAP: sensitivity of 93.1 (83.3-98.1)% versus 58.6 (44.9-71.4)% and a specificity of 95.7 (88.0-99.1)% versus 80.0 (68.7-88.6)% (both p < 0.001). 18F-NaF uptake assessed by CMA correlated more closely with LAP (r = 0.86, p < 0.001) than the CT calcium score (r = 0.39, p < 0.001), with these associations outperforming those observed for TBRmax values (LAP r = 0.63, p < 0.001; CT calcium score r = 0.30, p < 0.001). CONCLUSIONS Automated assessment of disease activity across the entire coronary vasculature is feasible using 18F-NaF CMA, providing a single measurement that has closer agreement with CT markers of plaque vulnerability than more traditional measures of plaque activity.
Collapse
Affiliation(s)
- Jacek Kwiecinski
- Departments of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Interventional Cardiology and Angiology, Institute of Cardiology, Alpejska 42, 04-628, Warsaw, Poland
| | - Sebastien Cadet
- Departments of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Marwa Daghem
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Martin L Lassen
- Departments of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Damini Dey
- Departments of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Marc R Dweck
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Daniel S Berman
- Departments of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - David E Newby
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Piotr J Slomka
- Departments of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
25
|
Pfaehler E, Burggraaff C, Kramer G, Zijlstra J, Hoekstra OS, Jalving M, Noordzij W, Brouwers AH, Stevenson MG, de Jong J, Boellaard R. PET segmentation of bulky tumors: Strategies and workflows to improve inter-observer variability. PLoS One 2020; 15:e0230901. [PMID: 32226030 PMCID: PMC7105134 DOI: 10.1371/journal.pone.0230901] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 03/11/2020] [Indexed: 12/26/2022] Open
Abstract
Background PET-based tumor delineation is an error prone and labor intensive part of image analysis. Especially for patients with advanced disease showing bulky tumor FDG load, segmentations are challenging. Reducing the amount of user-interaction in the segmentation might help to facilitate segmentation tasks especially when labeling bulky and complex tumors. Therefore, this study reports on segmentation workflows/strategies that may reduce the inter-observer variability for large tumors with complex shapes with different levels of user-interaction. Methods Twenty PET images of bulky tumors were delineated independently by six observers using four strategies: (I) manual, (II) interactive threshold-based, (III) interactive threshold-based segmentation with the additional presentation of the PET-gradient image and (IV) the selection of the most reasonable result out of four established semi-automatic segmentation algorithms (Select-the-best approach). The segmentations were compared using Jaccard coefficients (JC) and percentage volume differences. To obtain a reference standard, a majority vote (MV) segmentation was calculated including all segmentations of experienced observers. Performed and MV segmentations were compared regarding positive predictive value (PPV), sensitivity (SE), and percentage volume differences. Results The results show that with decreasing user-interaction the inter-observer variability decreases. JC values and percentage volume differences of Select-the-best and a workflow including gradient information were significantly better than the measurements of the other segmentation strategies (p-value<0.01). Interactive threshold-based and manual segmentations also result in significant lower and more variable PPV/SE values when compared with the MV segmentation. Conclusions FDG PET segmentations of bulky tumors using strategies with lower user-interaction showed less inter-observer variability. None of the methods led to good results in all cases, but use of either the gradient or the Select-the-best workflow did outperform the other strategies tested and may be a good candidate for fast and reliable labeling of bulky and heterogeneous tumors.
Collapse
Affiliation(s)
- Elisabeth Pfaehler
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- * E-mail:
| | - Coreline Burggraaff
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Gem Kramer
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Josée Zijlstra
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Otto S. Hoekstra
- Department of Oncology Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mathilde Jalving
- Department of Oncology Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Walter Noordzij
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Adrienne H. Brouwers
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marc G. Stevenson
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johan de Jong
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ronald Boellaard
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Koopman D, van Dalen JA, Stevens H, Slump CH, Knollema S, Jager PL. Performance of Digital PET Compared with High-Resolution Conventional PET in Patients with Cancer. J Nucl Med 2020; 61:1448-1454. [PMID: 32060217 DOI: 10.2967/jnumed.119.238105] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/03/2020] [Indexed: 01/02/2023] Open
Abstract
Recently introduced PET systems using silicon photomultipliers with digital readout (dPET) have an improved timing and spatial resolution, aiming at a better image quality than conventional PET (cPET) systems. We prospectively evaluated the performance of a dPET system in patients with cancer, as compared with high-resolution (HR) cPET imaging. Methods: After a single 18F-FDG injection, 66 patients underwent dPET and cPET imaging in randomized order. We used HR reconstructions (2 × 2 × 2 mm voxels) for both scanners and determined SUVmax, SUVmean, lesion-to-background ratio (LBR), metabolic tumor volume (MTV), and lesion diameter in up to 5 18F-FDG-positive lesions per patient. Furthermore, we counted the number of visible and measurable lesions on each PET scan. Two nuclear medicine specialists determined, in a masked manner, the TNM score from both image sets in 30 patients referred for initial staging. For all 66 patients, these specialists separately evaluated image quality (4-point scale) and determined the scan preference. Results: We included 238 lesions that were visible and measurable on both PET scans. For 27 patients, we found 37 additional lesions on dPET (41%) that were unmeasurable (n = 14) or invisible (n = 23) on cPET. Mean (±SD) SUVmean, SUVmax, LBR, and MTV on cPET were 5.2 ± 3.9, 6.9 ± 5.6, 5.0 ± 3.6, and 2,991 ± 13,251 mm3, respectively. On dPET, SUVmean, SUVmax, and LBR increased by 24%, 23%, and 27%, respectively (P < 0.001) whereas MTV decreased by 13% (P < 0.001), compared with cPET. Visual analysis showed TNM upstaging with dPET in 13% of the patients (4/30). dPET images also had higher scores for quality (P = 0.003) and were visually preferred in most cases (65%). Conclusion: dPET improved the detection of small lesions, upstaged the disease, and produced images that were visually preferred to those from HR cPET. More studies are necessary to confirm the superior diagnostic performance of dPET.Keywords: digital PET; conventional PET; FDG PET; lesion detection; cancer imaging.
Collapse
Affiliation(s)
- Daniëlle Koopman
- Department of Nuclear Medicine, Isala, Zwolle, The Netherlands .,Technical Medicine Centre, University of Twente, Enschede, The Netherlands; and
| | | | - Henk Stevens
- Department of Nuclear Medicine, Isala, Zwolle, The Netherlands
| | - Cornelis H Slump
- Technical Medicine Centre, University of Twente, Enschede, The Netherlands; and
| | - Siert Knollema
- Department of Nuclear Medicine, Isala, Zwolle, The Netherlands
| | - Pieter L Jager
- Department of Nuclear Medicine, Isala, Zwolle, The Netherlands
| |
Collapse
|
27
|
de Boer SA, Spoor DS, Slart RHJA, Mulder DJ, Reijrink M, Borra RJH, Kramer GM, Hoekstra OS, Boellaard R, Greuter MJ. Performance Evaluation of a Semi-automated Method for [ 18F]FDG Uptake in Abdominal Visceral Adipose Tissue. Mol Imaging Biol 2019; 21:159-167. [PMID: 29789994 DOI: 10.1007/s11307-018-1211-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE Severity of abdominal obesity and possibly levels of metabolic activity of abdominal visceral adipose tissue (VAT) are associated with an increased risk for cardiovascular disease (CVD). In this context, the purpose of the current study was to evaluate the reproducibility and repeatability of a semi-automated method for assessment of the metabolic activity of VAT using 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET)/x-ray computed tomography (CT). PROCEDURES Ten patients with lung cancer who underwent two baseline whole-body [18F]FDG PET/low-dose (LD) CT scans within 1 week were included. Abdominal VAT was automatically segmented using CT between levels L1-L5. The initial CT-based segmentation was further optimized using PET data with a standardized uptake value (SUV) threshold approach (range 1.0-2.5) and morphological erosion (range 0-5 pixels). The [18F]FDG uptake in SUV that was measured by the automated method was compared with manual analysis. The reproducibility and repeatability were quantified using intraclass correlation coefficients (ICCs). RESULTS The metabolic assessment of VAT on [18F]FDG PET/LDCT scans expressed as SUVmean, using an automated method showed high inter and intra observer (all ICCs > 0.99) and overall repeatability (ICC = 0.98). The manual method showed reproducible inter observer (all ICCs > 0.92), but less intra observer (ICC = 0.57) and less overall repeatability (ICC = 0.78) compared with the automated method. CONCLUSIONS Our proposed semi-automated method provided reproducible and repeatable quantitative analysis of [18F]FDG uptake in VAT. We expect this method to aid future research regarding the role of VAT in development of CVD.
Collapse
Affiliation(s)
- Stefanie A de Boer
- Department of Vascular Medicine, University Medical Center Groningen, University of Groningen, HP AA41, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - Daan S Spoor
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Riemer H J A Slart
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Biomedical Technology and Technical Medicine (MIRA), University of Twente, Enschede, The Netherlands
| | - Douwe J Mulder
- Department of Vascular Medicine, University Medical Center Groningen, University of Groningen, HP AA41, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Melanie Reijrink
- Department of Vascular Medicine, University Medical Center Groningen, University of Groningen, HP AA41, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Ronald J H Borra
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Medical Imaging Center of Southwest Finland, Department of Diagnostic Radiology, Turku University Hospital, University of Turku, Turku, Finland.,Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerbrand M Kramer
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Marcel J Greuter
- Biomedical Technology and Technical Medicine (MIRA), University of Twente, Enschede, The Netherlands.,Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
28
|
Botman E, Raijmakers PGHM, Yaqub M, Teunissen B, Netelenbos C, Lubbers W, Schwarte LA, Micha D, Bravenboer N, Schoenmaker T, de Vries TJ, Pals G, Smit JM, Koolwijk P, Trotter DG, Lammertsma AA, Eekhoff EMW. Evolution of heterotopic bone in fibrodysplasia ossificans progressiva: An [ 18F]NaF PET/CT study. Bone 2019; 124:1-6. [PMID: 30858149 DOI: 10.1016/j.bone.2019.03.009] [Citation(s) in RCA: 23] [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] [Received: 11/01/2018] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 11/30/2022]
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare, autosomal dominant disorder characterized by heterotopic ossification (HO) in muscles, ligaments and tendons. Flare-ups often precede the formation of HO, resulting in immobilization of joints. Due to progression of the disease without signs of a flare-up, co-existence of a chronic progression of HO has been postulated, but conclusive evidence is lacking. Recently, it has been shown that [18F]NaF PET/CT is able to identify early ossifying disease activity during flare-ups. Therefore, the purpose of the present study was to assess whether [18F]NaF PET/CT might also be able to identify the possible presence of chronic progressive HO in FOP. A total of thirteen [18F]NaF PET/CT scans from five FOP patients were analysed. Scans were acquired over a period of 0.5 to 2 years. Volumes of HO and standardized uptake values (SUV) were obtained based on manual segmentation of CT images. SUVpeak values, defined as the average SUV value of a 1 mL sphere containing the hottest voxel pixels, were obtained. Two out of five patients experienced ≥1 active clinical flare-ups at the time of the [18F]NaF PET/CT scan. In addition, in four out of five patients, serial scans showed radiological progression of HO (3 to 8 cm3), as assessed by CT volume, in the absence of a clinical flare-up. This volumetric increase was present in 6/47 (12.8%) of identified HO structures and, in all cases, was accompanied by increased [18F]NaF uptake, with SUVpeak ranging from 8.4 to 17.9. In conclusion, HO may progress without signs of a flare-up. [18F]NaF PET/CT is able to identify these asymptomatic, but progressive HO lesions, thereby demonstrating the presence of chronic activity in FOP. Consequently, future drugs should not only target new HO formation, but also this chronic HO progression.
Collapse
Affiliation(s)
- Esmée Botman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Internal Medicine Section Endocrinology, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands
| | - Pieter G H M Raijmakers
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, the Netherlands
| | - Maqsood Yaqub
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, the Netherlands
| | - Bernd Teunissen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, the Netherlands
| | - Coen Netelenbos
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Internal Medicine Section Endocrinology, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands
| | - Wouter Lubbers
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anaesthesiology, the Netherlands
| | - Lothar A Schwarte
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anaesthesiology, the Netherlands
| | - Dimitra Micha
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Clinical Genetics, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands
| | - Nathalie Bravenboer
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Clinical Chemistry, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands
| | - Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit
| | - Teun J de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit
| | - Gerard Pals
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Clinical Genetics, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands
| | - Jan Maerten Smit
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Bone Center, the Netherlands
| | - Pieter Koolwijk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Physiology, the Netherlands
| | | | - Adriaan A Lammertsma
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, the Netherlands
| | - E Marelise W Eekhoff
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Internal Medicine Section Endocrinology, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands.
| |
Collapse
|
29
|
Repeatability of brown adipose tissue measurements on FDG PET/CT following a simple cooling procedure for BAT activation. PLoS One 2019; 14:e0214765. [PMID: 30995248 PMCID: PMC6469763 DOI: 10.1371/journal.pone.0214765] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
Brown Adipose Tissue (BAT) is present in a significant number of adult humans and can be activated by exposure to cold. Measurement of active BAT presence, activity, and volume are desirable for determining the efficacy of potential treatments intended to activate BAT. The repeatability of 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) measurements of BAT presence, activity, and volume under controlled conditions has not been extensively studied. Eleven female volunteers underwent double baseline FDG PET imaging performed following a simple, regional cold intervention intended to activate brown fat. The cold intervention involved the lightly-clothed participants intermittently placing their feet on a block of ice while sitting in a cooled room. A repeat study was performed under the same conditions within a target of two weeks. FDG scans were obtained and maximum standardized uptake value adjusted for lean body mass (SULmax), CT Hounsfield units (HU), BAT metabolic volume (BMV), and total BAT glycolysis (TBG) were determined according to the Brown Adipose Reporting Criteria in Imaging STudies (BARCIST) 1.0. A Lin’s concordance correlation (CCC) of 0.80 was found for BMV between test and retest imaging. Intersession BAT SULmax was significantly correlated (r = 0.54; p < 0.05). The session #1 mean SULmax of 4.92 ± 4.49 g/mL was not significantly different from that of session #2 with a mean SULmax of 7.19 ± 7.34 g/mL (p = 0.16). BAT SULmax was highly correlated with BMV in test and retest studies (r ≥ 0.96, p < 0.001). Using a simplified ice-block cooling method, BAT was activated in the majority (9/11) of a group of young, lean female participants. Quantitative assessments of BAT SUL and BMV were not substantially different between test and retest imaging, but individual BMV could vary considerably. Intrasession BMV and SULmax were strongly correlated. The variability in estimates of BAT activity and volume on test-retest with FDG should inform sample size choice in studies quantifying BAT physiology and support the dynamic metabolic characteristics of this tissue. A more sophisticated cooling method potentially may reduce variations in test-retest BAT studies.
Collapse
|
30
|
Jauw YWS, Heijtel DF, Zijlstra JM, Hoekstra OS, de Vet HCW, Vugts DJ, Verheul HM, Boellaard R, Zweegman S, van Dongen GAMS, der Houven van Oordt CWMV, Lammertsma AA, Huisman MC. Noise-Induced Variability of Immuno-PET with Zirconium-89-Labeled Antibodies: an Analysis Based on Count-Reduced Clinical Images. Mol Imaging Biol 2019; 20:1025-1034. [PMID: 29713958 PMCID: PMC6244539 DOI: 10.1007/s11307-018-1200-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Purpose Positron emission tomography (PET) with Zirconium-89 (Zr-89)-labeled antibodies can be used for in vivo quantification of antibody uptake. Knowledge about measurement variability is required to ensure correct interpretation. However, no clinical studies have been reported on measurement variability of Zr-89 immuno-PET. As variability due to low signal-to-noise is part of the total measurement variability, the aim of this study was to assess noise-induced variability of Zr-89 -immuno-PET using count-reduced clinical images. Procedures Data were acquired from three previously reported clinical studies with [89Zr]antiCD20 (74 MBq, n = 7), [89Zr]antiEGFR (37 MBq, n = 7), and [89Zr]antiCD44 (37 MBq, n = 13), with imaging obtained 1 to 6 days post injection (D0–D6). Volumes of interest (VOIs) were manually delineated for liver, spleen, kidney, lung, brain, and tumor. For blood pool and bone marrow, fixed-size VOIs were used. Original PET list mode data were split and reconstructed, resulting in two count-reduced images at 50 % of the original injected dose (e.g., 37 MBq74inj). Repeatability coefficients (RC) were obtained from Bland-Altman analysis on standardized uptake values (SUV) derived from VOIs applied to these images. Results The RC for the combined manually delineated organs for [89Zr] antiCD20 (37 MBq74inj) increased from D0 to D6 and was less than 6 % at all time points. Blood pool and bone marrow had higher RC, up to 43 % for 37 MBq74inj at D6. For tumor, the RC was up to 42 % for [89Zr]antiCD20 (37 MBq74inj). For [89Zr]antiCD20, (18 MBq74inj), [89Zr]antiEGFR (18 MBq37inj), and [89Zr]antiCD44 (18 MBq37inj), measurement variability was independent of the investigated antibody. Conclusions Based on this study, noise-induced variability results in a RC for Zr-89-immuno-PET (37 MBq) around 6 % for manually delineated organs combined, increasing up to 43 % at D6 for blood pool and bone marrow, assuming similar biodistribution of antibodies. The signal-to-noise ratio leads to tumor RC up to 42 %. Electronic supplementary material The online version of this article (10.1007/s11307-018-1200-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yvonne W S Jauw
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands. .,Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.
| | | | - Josée M Zijlstra
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Otto S Hoekstra
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Henrica C W de Vet
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Danielle J Vugts
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Henk M Verheul
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Philips Healthcare, Best, the Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Guus A M S van Dongen
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Marc C Huisman
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
31
|
Kramer GM, Yaqub M, Vargas HA, Schuit RC, Windhorst AD, van den Eertwegh AJM, van der Veldt AAM, Bergman AM, Burnazi EM, Lewis JS, Chua S, Staton KD, Beattie BJ, Humm JL, Davis ID, Weickhardt AJ, Scott AM, Morris MJ, Hoekstra OS, Lammertsma AA. Assessment of Simplified Methods for Quantification of 18F-FDHT Uptake in Patients with Metastatic Castration-Resistant Prostate Cancer. J Nucl Med 2019; 60:1221-1227. [PMID: 30850488 DOI: 10.2967/jnumed.118.220111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 02/06/2019] [Indexed: 11/16/2022] Open
Abstract
18F-fluorodihydrotestosterone (18F-FDHT) PET/CT potentially provides a noninvasive method for assessment of androgen receptor expression in patients with metastatic castration-resistant prostate cancer (mCRPC). The objective of this study was to assess simplified methods for quantifying 18F-FDHT uptake in mCRPC patients and to assess effects of tumor perfusion on these 18F-FDHT uptake metrics. Methods: Seventeen mCRPC patients were included in this prospective observational multicenter study. Test and retest 30-min dynamic 18F-FDHT PET/CT scans with venous blood sampling were performed in 14 patients. In addition, arterial blood sampling and dynamic 15O-H2O scans were obtained in a subset of 6 patients. Several simplified methods were assessed: Patlak plots; SUV normalized to body weight (SUVBW), lean body mass (SUVLBM), whole blood (SUVWB), parent plasma activity concentration (SUVPP), area under the parent plasma curve (SUVAUC,PP), and area under the whole-blood input curve (SUVAUC,WB); and SUVBW corrected for sex hormone-binding globulin levels (SUVSHBG). Results were correlated with parameters derived from full pharmacokinetic 18F-FDHT and 15O-H2O. Finally, the repeatability of individual quantitative uptake metrics was assessed. Results: Eighty-seven 18F-FDHT-avid lesions were evaluated. 18F-FDHT uptake was best described by an irreversible 2-tissue-compartment model. Replacing the continuous metabolite-corrected arterial plasma input function with an image-derived input function in combination with venous sample data provided similar K i results (R 2 = 0.98). Patlak K i and SUVAUC,PP showed an excellent correlation (R 2 > 0.9). SUVBW showed a moderate correlation to K i (R 2 = 0.70, presumably due to fast 18F-FDHT metabolism. When calculating SUVSHBG, correlation to K i improved (R 2 = 0.88). The repeatability of full kinetic modeling parameters was inferior to that of simplified methods (repeatability coefficients > 36% vs. < 28%, respectively). 18F-FDHT uptake showed minimal blood flow dependency. Conclusion: 18F-FDHT kinetics in mCRPC patients are best described by an irreversible 2-tissue-compartment model with blood volume parameter. SUVAUC,PP showed a near-perfect correlation with the irreversible 2-tissue-compartment model analysis and can be used for accurate quantification of 18F-FDHT uptake in whole-body PET/CT scans. In addition, SUVSHBG could potentially be used as an even simpler method to quantify 18F-FDHT uptake when less complex scanning protocols and accuracy are required.
Collapse
Affiliation(s)
- Gerbrand M Kramer
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Herbert A Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert C Schuit
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Astrid A M van der Veldt
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Departments of Medical Oncology, Radiology, and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andries M Bergman
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Eva M Burnazi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Sua Chua
- Department of Nuclear Medicine, Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Kevin D Staton
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brad J Beattie
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ian D Davis
- Monash University and Eastern Health, Eastern Health Clinical School, Box Hill, Australia
| | - Andrew J Weickhardt
- Department of Medical Oncology, Olivia Newton-John Cancer Research Institute, Austin Hospital, Melbourne, Victoria, Australia
| | - Andrew M Scott
- Department of Medical Oncology, Olivia Newton-John Cancer Research Institute, Austin Hospital, Melbourne, Victoria, Australia.,Department of Molecular Imaging and Therapy, University of Melbourne, Heidelberg, Victoria, Australia
| | - Michael J Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; and.,Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Maebatake A, Morita K, Akamatsu G, Tsutsui Y, Himuro K, Baba S, Sasaki M. The Influence of Minimal Misalignment on the Repeatability of PET Images Examined by the Repositioning of Point Sources. J Nucl Med Technol 2019; 47:55-59. [DOI: 10.2967/jnmt.118.208835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 08/23/2018] [Indexed: 11/16/2022] Open
|
33
|
Kolinger GD, Vállez García D, Kramer GM, Frings V, Smit EF, de Langen AJ, Dierckx RAJO, Hoekstra OS, Boellaard R. Repeatability of [ 18F]FDG PET/CT total metabolic active tumour volume and total tumour burden in NSCLC patients. EJNMMI Res 2019; 9:14. [PMID: 30734113 PMCID: PMC6367490 DOI: 10.1186/s13550-019-0481-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 01/25/2019] [Indexed: 12/15/2022] Open
Abstract
Background Total metabolic active tumour volume (TMATV) and total tumour burden (TTB) are increasingly studied as prognostic and predictive factors in non-small cell lung cancer (NSCLC) patients. In this study, we investigated the repeatability of TMATV and TTB as function of uptake interval, positron emission tomography/computed tomography (PET/CT) image reconstruction settings, and lesion delineation method. We used six lesion delineation methods, four direct PET image-derived delineations and two based on a majority vote approach, i.e. intersection between two or more delineations (MV2) and between three or more delineations (MV3). To evaluate the accuracy of those methods, they were compared with a reference delineation obtained from the consensus of the segmentations performed by three experienced observers. Ten NSCLC patients underwent two baseline whole-body [18F]2-Fluoro-2-deoxy-2-D-glucose ([18F]FDG) PET/CT studies on separate days, within 3 days. Two scans were obtained on each day at 60 and 90 min post-injection to assess the influence of tracer uptake interval. PET/CT images were reconstructed following the European Association of Nuclear Medicine Research Ltd. (EARL) compliant settings and with point-spread-function (PSF) modelling. Repeatability between the measurements of each day was determined and the influence of uptake interval, reconstruction settings, and lesion delineation method was assessed using the generalized estimating equations model. Results Based on the Jaccard index with the reference delineation, the MV2 lesion delineation method was the most successful method for automated lesion segmentation. The best overall repeatability (lowest repeatability coefficient, RC) was found for TTB from 90 min of tracer uptake scans reconstructed with EARL compliant settings and delineated with 41% of lesion’s maximum SUV method (RC = 11%). In most cases, TMATV and TTB repeatability were not significantly affected by changes in tracer uptake time or reconstruction settings. However, some lesion delineation methods had significantly different repeatability when applied to the same images. Conclusions This study suggests that under some circumstances TMATV and TTB repeatability are significantly affected by the lesion delineation method used. Performing the delineation with a majority vote approach improves reliability and does not hamper repeatability, regardless of acquisition and reconstruction settings. It is therefore concluded that by using a majority vote based tumour segmentation approach, TMATV and TTB in NSCLC patients can be measured with high reliability and precision. Electronic supplementary material The online version of this article (10.1186/s13550-019-0481-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guilherme D Kolinger
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - David Vállez García
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Gerbrand M Kramer
- Amsterdam University Medical Centers, location VU Medical Center, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Virginie Frings
- Amsterdam University Medical Centers, location VU Medical Center, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Egbert F Smit
- Amsterdam University Medical Centers, location VU Medical Center, Department of Pulmonary Disease, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,Netherlands Cancer Institute, Department of Thoracic Oncology, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Adrianus J de Langen
- Amsterdam University Medical Centers, location VU Medical Center, Department of Pulmonary Disease, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,Netherlands Cancer Institute, Department of Thoracic Oncology, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Otto S Hoekstra
- Amsterdam University Medical Centers, location VU Medical Center, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Ronald Boellaard
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands. .,Amsterdam University Medical Centers, location VU Medical Center, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
34
|
Fahey F, Christian P, Zukotynski K, Sexton-Stallone B, Kiss C, Clarke B, Onar-Thomas A, Poussaint TY. Use of a Qualification Phantom for PET Brain Imaging in a Multicenter Consortium: A Collaboration Between the Pediatric Brain Tumor Consortium and the SNMMI Clinical Trials Network. J Nucl Med 2018; 60:677-682. [PMID: 30530829 DOI: 10.2967/jnumed.118.219998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/14/2018] [Indexed: 01/13/2023] Open
Abstract
The purpose of this study was to assess image quality and quantitative brain PET across a multicenter consortium. Methods: All academic centers and children's hospitals in the Pediatric Brain Tumor Consortium (PBTC) scanned a phantom developed by the Society of Nuclear Medicine and Molecular Imaging Clinical Trials Network (SNMMI CTN) for the validation of brain PET studies associated with clinical trials. The phantom comprises 2 separate, fillable sections: a resolution/uniformity section and a clinical simulation section. The resolution/uniformity section is a cylinder 12.7 cm long and 20 cm in diameter; spatial resolution is evaluated subjectively with 2 sets of rods (hot and cold) of varying diameter (4.0, 5.0, 6.25, 7.81, 9.67, and 12.2 mm) and spacing (twice the rod diameter). The clinical simulation section simulates a transverse section of midbrain with ventricles and gray and white matter compartments. If properly filled, hot rods have a 4:1 target-to-background ratio, and gray-to-white matter sections have a 4:1 ratio. Uniformity and image quality were evaluated using the SUV in a small volume of interest as well as subjectively by 2 independent observers using a 4-point scale. Results: Eleven PBTC sites scanned the phantom on 13 PET scanners. The phantom's complexity led to suboptimal filling, particularly of the hot rod section, in 5 sites. The SUV in the uniformity section was within 10% of unity on only 5 of 13 scanners, although 12 of 13 were subjectively judged to have very good to excellent uniformity. Four of 6 hot rods were discernable by all 13 scanners, whereas 3 of 6 cold rods were discernable by only 5 scanners. Four of 13 scanners had a gray-to-white matter ratio between 3.0 and 5.0 (4.0 is truth); however, 11 of 13 scanners were subjectively judged to have very good or excellent image quality. Conclusion: Eleven sites were able to image a powerful phantom developed by the SNMMI CTN that evaluated image uniformity, spatial resolution, and image quality of brain PET. There was considerable variation in PET data across the PBTC sites, possibly resulting from variations in scanning across the sites due to challenges in filling the phantom.
Collapse
Affiliation(s)
- Frederic Fahey
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Boston Children's Hospital, Boston, Massachusetts .,Department of Radiology, Harvard Medical School, Boston, Massachusetts
| | - Paul Christian
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Katherine Zukotynski
- Department of Medicine and Radiology, McMaster University, Hamilton, Ontario, Canada
| | - Briana Sexton-Stallone
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Boston Children's Hospital, Boston, Massachusetts
| | - Christina Kiss
- Clinical Trials Network, Society of Nuclear Medicine and Molecular Imaging, Reston Virginia
| | - Bonnie Clarke
- Clinical Trials Network, Society of Nuclear Medicine and Molecular Imaging, Reston Virginia
| | | | - Tina Young Poussaint
- Department of Radiology, Harvard Medical School, Boston, Massachusetts.,Division of Neuroradiology, Department of Radiology, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
35
|
Fledelius J. Editorial on "What FDG-PET response-assessment method best predicts survival after curative-intent chemoradiation in non-small cell lung cancer (NSCLC): EORTC, PERCIST, Peter Mac or Deauville criteria?". J Thorac Dis 2018; 10:S4076-S4079. [PMID: 30631559 DOI: 10.21037/jtd.2018.09.136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Joan Fledelius
- Department of Nuclear Medicine, Herning Hospital, Herning, Denmark
| |
Collapse
|
36
|
Stevenson MG, Been LB, Hoekstra HJ, Suurmeijer AJH, Boellaard R, Brouwers AH. Volume of interest delineation techniques for 18F-FDG PET-CT scans during neoadjuvant extremity soft tissue sarcoma treatment in adults: a feasibility study. EJNMMI Res 2018; 8:42. [PMID: 29881881 PMCID: PMC5992109 DOI: 10.1186/s13550-018-0397-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/08/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND This study explores various volume of interest (VOI) delineation techniques for fluorine-18-fluorodeoxyglucose positron emission tomography with computed tomography (18F-FDG PET-CT) scans during neoadjuvant extremity soft tissue sarcoma (ESTS) treatment. RESULTS During neoadjuvant treatment, hyperthermic isolated limb perfusion (HILP) and preoperative external beam radiotherapy (EBRT), 11 patients underwent three 18F-FDG PET-CT scans. The first scan was made prior to the HILP, the second after the HILP but prior to the start of the EBRT, and the third prior to surgical resection. An automatically drawn VOIauto, a manually drawn VOIman, and two gradient-based semi-automatically drawn VOIs (VOIgrad and VOIgrad+) were obtained. Maximum standardized uptake value (SUVmax), SUVpeak, SUVmean, metabolically active tumor volume (MATV), and total lesion glycolysis (TLG) were calculated from each VOI. The correlation and level of agreement between VOI delineation techniques was explored. Lastly, the changes in metabolic tumor activity were related to the histopathologic response. The strongest correlation and an acceptable level of agreement was found between the VOIman and the VOIgrad+ delineation techniques. A decline (VOIman) in SUVmax, SUVpeak, SUVmean, TLG, and MATV (all p < 0.05) was found between the three scans. A > 75% decline in TLG between scan 1 and scan 3 possibly identifies histopathologic response. CONCLUSIONS The VOIgrad+ delineation technique was identified as most reliable considering reproducibility when compared with the other VOI delineation techniques during the multimodality neoadjuvant treatment of locally advanced ESTS. A significant decline in metabolic tumor activity during the treatment was found. TLG deserves further exploration as predictor for histopathologic response after multimodality ESTS treatment.
Collapse
Affiliation(s)
- Marc G. Stevenson
- Department of Surgical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Lukas B. Been
- Department of Surgical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Harald J. Hoekstra
- Department of Surgical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Albert J. H. Suurmeijer
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB Groningen, The Netherlands
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Adrienne H. Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
37
|
Kramer GM, Liu Y, de Langen AJ, Jansma EP, Trigonis I, Asselin MC, Jackson A, Kenny L, Aboagye EO, Hoekstra OS, Boellaard R. Repeatability of quantitative 18F-FLT uptake measurements in solid tumors: an individual patient data multi-center meta-analysis. Eur J Nucl Med Mol Imaging 2018; 45:951-961. [PMID: 29362858 PMCID: PMC5915500 DOI: 10.1007/s00259-017-3923-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/26/2017] [Indexed: 12/26/2022]
Abstract
INTRODUCTION 3'-deoxy-3'-[18F]fluorothymidine (18F-FLT) positron emission tomography (PET) provides a non-invasive method to assess cellular proliferation and response to antitumor therapy. Quantitative 18F-FLT uptake metrics are being used for evaluation of proliferative response in investigational setting, however multi-center repeatability needs to be established. The aim of this study was to determine the repeatability of 18F-FLT tumor uptake metrics by re-analyzing individual patient data from previously published reports using the same tumor segmentation method and repeatability metrics across cohorts. METHODS A systematic search in PubMed, EMBASE.com and the Cochrane Library from inception-October 2016 yielded five 18F-FLT repeatability cohorts in solid tumors. 18F-FLT avid lesions were delineated using a 50% isocontour adapted for local background on test and retest scans. SUVmax, SUVmean, SUVpeak, proliferative volume and total lesion uptake (TLU) were calculated. Repeatability was assessed using the repeatability coefficient (RC = 1.96 × SD of test-retest differences), linear regression analysis, and the intra-class correlation coefficient (ICC). The impact of different lesion selection criteria was also evaluated. RESULTS Images from four cohorts containing 30 patients with 52 lesions were obtained and analyzed (ten in breast cancer, nine in head and neck squamous cell carcinoma, and 33 in non-small cell lung cancer patients). A good correlation was found between test-retest data for all 18F-FLT uptake metrics (R2 ≥ 0.93; ICC ≥ 0.96). Best repeatability was found for SUVpeak (RC: 23.1%), without significant differences in RC between different SUV metrics. Repeatability of proliferative volume (RC: 36.0%) and TLU (RC: 36.4%) was worse than SUV. Lesion selection methods based on SUVmax ≥ 4.0 improved the repeatability of volumetric metrics (RC: 26-28%), but did not affect the repeatability of SUV metrics. CONCLUSIONS In multi-center studies, differences ≥ 25% in 18F-FLT SUV metrics likely represent a true change in tumor uptake. Larger differences are required for FLT metrics comprising volume estimates when no lesion selection criteria are applied.
Collapse
Affiliation(s)
- G M Kramer
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, Netherlands.
| | - Y Liu
- European Organisation for Research and Treatment for Cancer (EORTC), Headquarters, Brussels, Belgium
| | - A J de Langen
- Department of Pulmonology, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, Netherlands
| | - E P Jansma
- Medical Library, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - I Trigonis
- Division of Informatics, Imaging and Data Sciences Institute of Population Health, Wolfson Molecular Imaging Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, UK
| | - M-C Asselin
- Division of Informatics, Imaging and Data Sciences Institute of Population Health, Wolfson Molecular Imaging Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, UK
| | - A Jackson
- Division of Informatics, Imaging and Data Sciences Institute of Population Health, Wolfson Molecular Imaging Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, UK
| | - L Kenny
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, London, UK
| | - E O Aboagye
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, London, UK
| | - O S Hoekstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, Netherlands
| | - R Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, Netherlands
| |
Collapse
|
38
|
Comparison Between Different PET and CT-Based Imaging Interpretation Criteria at Interim Imaging in Patients With Diffuse Large B-Cell Lymphoma. Clin Nucl Med 2018; 43:1-8. [PMID: 29076913 DOI: 10.1097/rlu.0000000000001880] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate the predictive value of interim PET (iPET) in diffuse large B-cell lymphoma (DLBCL) using 5 different imaging interpretation criteria: Deauville 5-point scale criteria, International Harmonization Project (IHP) criteria, Response Evaluation Criteria In Solid Tumors (RECIST) 1.1, European Organization for Research and Treatment of Cancer, and PET Response Criteria in Solid Tumors (PERCIST) 1.0. METHODS We retrospectively reviewed records from 38 patients with DLBCL who underwent baseline and iPET at our institution. Imaging was interpreted according to the previously mentioned criteria. Results were correlated with end-of-treatment response, based on reports at the end of treatment radiological examinations, overall survival (OS), and progression-free survival (PFS) to assess and compare the predictive value of iPET according to each criterion. We also evaluated the concordance between different criteria. RESULTS The Deauville and PERCIST criteria were the most reliable for predicting end-of-treatment response, reporting an accuracy of 81.6%. They also correlated with OS and PFS (P = 0.0004 and P = 0.0001, and P = 0.0007 and P = 0.0002, for Deauville and PERCIST, respectively). Interim PET according to European Organization for Research and Treatment of Cancer also predicted the end-of-treatment response with an accuracy of 73.7% and had a significant correlation with OS (P = 0.007) and PFS (P = 0.007). In contrast, the IHP criteria and RECIST did not predict outcomes: the accuracy for end-of-treatment response was 34.2% and 36.8%, respectively, with no significant correlation with OS or PFS (P = 0.182 and P = 0.357, and P = 0.341 and P = 0.215, for OS and PFS, respectively). CONCLUSIONS The predictive value of iPET in DLBCL patients is most reliable using the Deauville and PERCIST criteria. Criteria that rely on anatomical characteristics, namely, RECIST and IHP criteria, are less accurate in predicting patient outcomes in DLBCL.
Collapse
|
39
|
Kasalak Ö, Overbosch J, Glaudemans AWJM, Boellaard R, Jutte PC, Kwee TC. Primary tumor volume measurements in Ewing sarcoma: MRI inter- and intraobserver variability and comparison with FDG-PET. Acta Oncol 2018; 57:534-540. [PMID: 29117758 DOI: 10.1080/0284186x.2017.1398411] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Primary tumor volume is as an important and independent prognostic factor in Ewing sarcoma. However, the observer variability of magnetic resonance imaging (MRI)-based primary tumor volume measurements in newly diagnosed Ewing sarcoma has never been investigated. Furthermore, it is unclear how MRI-based volume measurements compare to 18F-fluoro-2-deoxy-D-glucose (FDG) positron emission tomography (PET)-based volume measurements. This study aimed to determine the observer variability of simplified MRI-based primary tumor volume measurements in newly diagnosed treatment-naive Ewing sarcoma and to compare them to the actual primary tumor volume at MRI and the FDG-PET-based metabolically active tumor volume (MATV). MATERIAL AND METHODS Twenty-nine newly diagnosed Ewing sarcoma patients with pretreatment MRI (of whom 11 also underwent FDG-PET) were included. Both exact and dichotomized (according to the proposed threshold of 200 mL) primary tumor volume measurements were analyzed. RESULTS Mean inter- and intraobserver differences of MRI-based simplified tumor volume ± limits of agreement varied between 15-42 ± 155-204 mL and between 9-16 ± 64-250 mL, respectively. Inter- and intraobserver agreements of dichotomized MRI-based simplified tumor volume measurements was very good (κ = 0.827-1.000). Mean difference between simplified and actual tumor volumes at MRI ± limits of agreement was 60 ± 381 mL. Agreement between dichotomized simplified and actual tumor volumes at MRI was very good (κ = 0.839). Mean difference between MRI-based simplified tumor volume and MATV ± limits of agreement was 181 ± 549 mL and almost significantly different (p = .0581). Agreement between dichotomized MRI-based simplified tumor volume and MATV was moderate (κ = 0.560). CONCLUSIONS Exact MRI-based simplified primary tumor volume measurements in Ewing sarcoma suffer from considerable observer variability, but observer agreement of dichotomized measurements (≤200 mL vs. >200 mL) is very good and generally matches MRI-based actual volume measurements. MRI-based primary tumor volume measurements poorly-moderately agree with and tend to be higher than the MATV.
Collapse
Affiliation(s)
- Ömer Kasalak
- Department of Radiology, Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jelle Overbosch
- Department of Radiology, Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Andor W. J. M. Glaudemans
- Department of Radiology, Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ronald Boellaard
- Department of Radiology, Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Paul C. Jutte
- Departments of Orthopedics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Thomas C. Kwee
- Department of Radiology, Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
40
|
Kaalep A, Sera T, Rijnsdorp S, Yaqub M, Talsma A, Lodge MA, Boellaard R. Feasibility of state of the art PET/CT systems performance harmonisation. Eur J Nucl Med Mol Imaging 2018; 45:1344-1361. [PMID: 29500480 PMCID: PMC5993859 DOI: 10.1007/s00259-018-3977-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 02/12/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE The objective of this study was to explore the feasibility of harmonising performance for PET/CT systems equipped with time-of-flight (ToF) and resolution modelling/point spread function (PSF) technologies. A second aim was producing a working prototype of new harmonising criteria with higher contrast recoveries than current EARL standards using various SUV metrics. METHODS Four PET/CT systems with both ToF and PSF capabilities from three major vendors were used to acquire and reconstruct images of the NEMA NU2-2007 body phantom filled conforming EANM EARL guidelines. A total of 15 reconstruction parameter sets of varying pixel size, post filtering and reconstruction type, with three different acquisition durations were used to compare the quantitative performance of the systems. A target range for recovery curves was established such that it would accommodate the highest matching recoveries from all investigated systems. These updated criteria were validated on 18 additional scanners from 16 sites in order to demonstrate the scanners' ability to meet the new target range. RESULTS Each of the four systems was found to be capable of producing harmonising reconstructions with similar recovery curves. The five reconstruction parameter sets producing harmonising results significantly increased SUVmean (25%) and SUVmax (26%) contrast recoveries compared with current EARL specifications. Additional prospective validation performed on 18 scanners from 16 EARL accredited sites demonstrated the feasibility of updated harmonising specifications. SUVpeak was found to significantly reduce the variability in quantitative results while producing lower recoveries in smaller (≤17 mm diameter) sphere sizes. CONCLUSIONS Harmonising PET/CT systems with ToF and PSF technologies from different vendors was found to be feasible. The harmonisation of such systems would require an update to the current multicentre accreditation program EARL in order to accommodate higher recoveries. SUVpeak should be further investigated as a noise resistant alternative quantitative metric to SUVmax.
Collapse
Affiliation(s)
- Andres Kaalep
- Department of Medical Technology, North Estonia Medical Centre Foundation, J. Sutiste Str 19, 13419, Tallinn, Estonia.
| | - Terez Sera
- Department of Nuclear Medicine, University of Szeged, Szeged, Hungary
- On behalf of EANM Research Limited (EARL), Vienna, Austria
| | - Sjoerd Rijnsdorp
- Department of Medical Physics, Catharina Hospital, Eindhoven, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Anne Talsma
- Department of Radiology, Martini Hospital, Groningen, Netherlands
| | - Martin A Lodge
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Ronald Boellaard
- On behalf of EANM Research Limited (EARL), Vienna, Austria.
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, Groningen, the Netherlands.
| |
Collapse
|
41
|
Measurement uncertainty of lesion and reference mediastinum standardized uptake value in lung cancer. Nucl Med Commun 2017; 38:509-514. [PMID: 28489690 DOI: 10.1097/mnm.0000000000000677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To assess standardized uptake value (SUV) measurement uncertainty (MU) of lung cancer lesions with uptake greater than mediastinum but less than or equal to the liver and that of the mediastinum blood pool, and to compare lesion SUV with mediastinum SUV by assessing MU of their ratio. PATIENTS AND METHODS Dynamic PET data involving 10 frames were retrospectively analyzed in 10 patients, yielding maximal SUV of 25 lesions (Lesion-SUVmax), 10 mediastinum SUV, either maximal or mean (Med-SUVmax, Med-SUVmean), 25 Rmax ratios (=Lesion-SUVmax/Med-SUVmax), and 25 Rmean ratios (=Lesion-SUVmax/Med-SUVmean). A mean coefficient of variation was calculated for each parameter, leading to relative measurement uncertainty (MUr), respectively. RESULTS MU of Rmax was found to involve both Lesion-SUVmax and Med-SUVmax MU: MUr=33.3-23.3-21.9%, respectively (95% confidence level). No significant difference in MUr was found between Med-SUVmax and Med-SUVmean and between Rmax and Rmean. CONCLUSION Comparison between target lesion SUV and reference mediastinum SUV must take into account SUV MU of both. Therefore, no MU reduction can be expected from using the lesion/mediastinum SUVmax ratio instead of Lesion-SUVmax. Moreover, no MU reduction can be expected from using the mean mediastinum SUV instead of the maximal one.
Collapse
|
42
|
Mansor S, Pfaehler E, Heijtel D, Lodge MA, Boellaard R, Yaqub M. Impact of PET/CT system, reconstruction protocol, data analysis method, and repositioning on PET/CT precision: An experimental evaluation using an oncology and brain phantom. Med Phys 2017; 44:6413-6424. [PMID: 28994465 PMCID: PMC5734628 DOI: 10.1002/mp.12623] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 12/02/2022] Open
Abstract
Purpose In longitudinal oncological and brain PET/CT studies, it is important to understand the repeatability of quantitative PET metrics in order to assess change in tracer uptake. The present studies were performed in order to assess precision as function of PET/CT system, reconstruction protocol, analysis method, scan duration (or image noise), and repositioning in the field of view. Methods Multiple (repeated) scans have been performed using a NEMA image quality (IQ) phantom and a 3D Hoffman brain phantom filled with 18F solutions on two systems. Studies were performed with and without randomly (< 2 cm) repositioning the phantom and all scans (12 replicates for IQ phantom and 10 replicates for Hoffman brain phantom) were performed at equal count statistics. For the NEMA IQ phantom, we studied the recovery coefficients (RC) of the maximum (SUVmax), peak (SUVpeak), and mean (SUVmean) uptake in each sphere as a function of experimental conditions (noise level, reconstruction settings, and phantom repositioning). For the 3D Hoffman phantom, the mean activity concentration was determined within several volumes of interest and activity recovery and its precision was studied as function of experimental conditions. Results The impact of phantom repositioning on RC precision was mainly seen on the Philips Ingenuity PET/CT, especially in the case of smaller spheres (< 17 mm diameter, P < 0.05). This effect was much smaller for the Siemens Biograph system. When exploring SUVmax, SUVpeak, or SUVmean of the spheres in the NEMA IQ phantom, it was observed that precision depended on phantom repositioning, reconstruction algorithm, and scan duration, with SUVmax being most and SUVpeak least sensitive to phantom repositioning. For the brain phantom, regional averaged SUVs were only minimally affected by phantom repositioning (< 2 cm). Conclusion The precision of quantitative PET metrics depends on the combination of reconstruction protocol, data analysis methods and scan duration (scan statistics). Moreover, precision was also affected by phantom repositioning but its impact depended on the data analysis method in combination with the reconstructed voxel size (tissue fraction effect). This study suggests that for oncological PET studies the use of SUVpeak may be preferred over SUVmax because SUVpeak is less sensitive to patient repositioning/tumor sampling.
Collapse
Affiliation(s)
- Syahir Mansor
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Elisabeth Pfaehler
- Faculty of Medical Sciences, Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Dennis Heijtel
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Martin A Lodge
- PET Center, Johns Hopkins Hospital, Nelson B1125, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,Faculty of Medical Sciences, Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Kramer GM, Hoekstra OS, Boellaard R. Reply: Repeatability of Quantitative Whole-Body 18F-FDG PET/CT Uptake Measures in Patients with Non-Small Cell Lung Cancer: Dynamic Versus Test-Retest Design. J Nucl Med 2017; 58:1528-1529. [PMID: 28619733 DOI: 10.2967/jnumed.117.195461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Gerbrand Maria Kramer
- VU University Medical Center P.O. Box 7057 1007 MB Amsterdam, The Netherlands E-mail:
| | | | | |
Collapse
|
44
|
Fledelius J, Winther-Larsen A, Khalil AA, Bylov CM, Hjorthaug K, Bertelsen A, Frøkiær J, Meldgaard P. 18F-FDG PET/CT for Very Early Response Evaluation Predicts CT Response in Erlotinib-Treated Non–Small Cell Lung Cancer Patients: A Comparison of Assessment Methods. J Nucl Med 2017; 58:1931-1937. [DOI: 10.2967/jnumed.117.193003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 04/25/2017] [Indexed: 01/01/2023] Open
|
45
|
Hofheinz F, Apostolova I, Oehme L, Kotzerke J, van den Hoff J. Test-Retest Variability in Lesion SUV and Lesion SUR in 18F-FDG PET: An Analysis of Data from Two Prospective Multicenter Trials. J Nucl Med 2017; 58:1770-1775. [PMID: 28473598 DOI: 10.2967/jnumed.117.190736] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/14/2017] [Indexed: 01/20/2023] Open
Abstract
Quantitative assessment of radio- and chemotherapy response with 18F-FDG whole-body PET has attracted increasing interest in recent years. In most published work, SUV has been used for this purpose. In the context of therapy response assessment, the reliability of lesion SUVs, notably their test-retest stability, thus becomes crucial. However, a recent study demonstrated substantial test-retest variability (TRV) in SUVs. The purpose of the present study was to investigate whether the tumor-to-blood SUV ratio (SUR) can improve TRV in tracer uptake. Methods: 73 patients with advanced non-small cell lung cancer from the prospective multicenter trials ACRIN 6678 (n = 34) and MK-0646-008 (n = 39) were included in this study. All patients underwent two 18F-FDG PET/CT investigations on two different days (time difference, 3.6 ± 2.1 d; range, 1-7 d) before therapy. For each patient, up to 7 tumor lesions were evaluated. For each lesion, SUVmax and SUVpeak were determined. Blood SUV was determined as the mean value of a 3-dimensional aortic region of interest that was delineated on the attenuation CT image and transferred to the PET image. SURs were computed as the ratio of tumor SUV to blood SUV and were uptake time-corrected to 75 min after injection. TRV was quantified as 1.96 multiplied by the root-mean-square deviation of the fractional paired differences in SUV and SUR. The combined effect of blood normalization and uptake time correction was inspected by considering RTRV (TRVSUR/TRVSUV), a ratio reflecting the reduction in the TRV in SUR relative to SUV. RTRV was correlated with the group-averaged-value difference (δ) in CFmean (δCFmean) of the quantity δCF = |CF - 1|, where CF is the numeric factor that converts individual ratios of paired SUVs into corresponding SURs. This correlation analysis was performed by successively increasing a threshold value δCFmin and computing δCFmean and RTRV for the remaining subgroup of patients/lesions with δCF ≥ δCFminResults: The group-averaged TRVSUV and TRVSUR were 32.1 and 29.0, respectively, which correspond to a reduction of variability in SUR by an RTRV factor of 0.9 in comparison to SUV. This rather marginal improvement can be understood to be a consequence of the atypically low intrasubject variability in blood SUV and uptake time and the accordingly small δCF values in the investigated prospective study groups. In fact, subgroup analysis with increasing δCFmin thresholds revealed a pronounced negative correlation (Spearman ρ = -0.99, P < 0.001) between RTRV and δCFmean, where RTRV ≈ 0.4 in the δCFmin = 20% subgroup, corresponding to a more than 2-fold reduction of TRVSUR compared with TRVSUVConclusion: Variability in blood SUV and uptake time has been identified as a causal factor in the TRV in lesion SUV. Therefore, TRV in lesion uptake measurements can be reduced by replacing SUV with SUR as the uptake measure. The improvement becomes substantial for the level of variability in blood SUV and uptake time typically observed in the clinical context.
Collapse
Affiliation(s)
- Frank Hofheinz
- Helmholtz-Zentrum Dresden-Rossendorf, PET Center, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Ivayla Apostolova
- Klinik für Radiologie Nuklearmedizin, Universitätsklinikum Magdeburg A.ö.R., Magdeburg, Germany; and
| | - Liane Oehme
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jörg Kotzerke
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jörg van den Hoff
- Helmholtz-Zentrum Dresden-Rossendorf, PET Center, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
46
|
Lodge MA. Repeatability of SUV in Oncologic 18F-FDG PET. J Nucl Med 2017; 58:523-532. [PMID: 28232605 DOI: 10.2967/jnumed.116.186353] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/21/2017] [Indexed: 11/16/2022] Open
Abstract
Quantitative analysis can potentially improve the accuracy and consistency of 18F-FDG PET, particularly for the assessment of tumor response to treatment. Although not without limitations, SUV has emerged as the predominant metric for tumor quantification with 18F-FDG PET. Growing literature suggests that the difference between SUVs measured before and after treatment can be used to predict tumor response at an early stage. SUV is, however, associated with multiple sources of variability, and to best use SUV for response assessment, an understanding of the repeatability of the technique is required. Test-retest studies involve repeated scanning of the same patient on the same scanner using the same protocol no more than a few days apart and provide basic information on the repeatability of the technique. Multiple test-retest studies have been performed to assess SUV repeatability, although a comparison of reports is complicated by the use of different methodologies and statistical metrics. This article reviews the available data, addressing issues such as different repeatability metrics, relative units, log transformation, and asymmetric limits of repeatability. When acquired with careful attention to protocol, tumor SUV has a within-subject coefficient of variation of approximately 10%. In a response assessment setting, SUV reductions of more than 25% and increases of more than 33% are unlikely to be due to measurement variability. Broader margins may be required for sites with less rigorous protocol compliance, but in general, SUV is a highly repeatable imaging biomarker that is ideally suited to monitoring tumor response to treatment in individual patients.
Collapse
Affiliation(s)
- Martin A Lodge
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
47
|
Thie JA. Need for Glucose Correction for 18F-FDG PET Influenced by Glucose Sensitivities to Types of Tissue and Random Factors. J Nucl Med 2017; 58:1527. [PMID: 28126886 DOI: 10.2967/jnumed.116.188185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Joseph A Thie
- University of Tennessee 12334 Bluff Shore Dr. Knoxville, TN 37922 E-mail:
| |
Collapse
|
48
|
Laffon E, Marthan R. Repeatability of Quantitative Whole-Body 18F-FDG PET/CT Uptake Measures in Patients with Non-Small Cell Lung Cancer: Dynamic Versus Test-Retest Design. J Nucl Med 2016; 58:1528. [PMID: 27932561 DOI: 10.2967/jnumed.116.186676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Eric Laffon
- Hôpital du Haut-Lévèque avenue de Magellan 33604 Pessac, France E-mail:
| | | |
Collapse
|
49
|
Meijer TWH, de Geus-Oei LF, Visser EP, Oyen WJG, Looijen-Salamon MG, Visvikis D, Verhagen AFTM, Bussink J, Vriens D. Tumor Delineation and Quantitative Assessment of Glucose Metabolic Rate within Histologic Subtypes of Non-Small Cell Lung Cancer by Using Dynamic 18F Fluorodeoxyglucose PET. Radiology 2016; 283:547-559. [PMID: 27846378 DOI: 10.1148/radiol.2016160329] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Purpose To assess whether dynamic fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography (PET) has added value over static 18F-FDG PET for tumor delineation in non-small cell lung cancer (NSCLC) radiation therapy planning by using pathology volumes as the reference standard and to compare pharmacokinetic rate constants of 18F-FDG metabolism, including regional variation, between NSCLC histologic subtypes. Materials and Methods The study was approved by the institutional review board. Patients gave written informed consent. In this prospective observational study, 1-hour dynamic 18F-FDG PET/computed tomographic examinations were performed in 35 patients (36 resectable NSCLCs) between 2009 and 2014. Static and parametric images of glucose metabolic rate were obtained to determine lesion volumes by using three delineation strategies. Pathology volume was calculated from three orthogonal dimensions (n = 32). Whole tumor and regional rate constants and blood volume fraction (VB) were computed by using compartment modeling. Results Pathology volumes were larger than PET volumes (median difference, 8.7-25.2 cm3; Wilcoxon signed rank test, P < .001). Static fuzzy locally adaptive Bayesian (FLAB) volumes corresponded best with pathology volumes (intraclass correlation coefficient, 0.72; P < .001). Bland-Altman analyses showed the highest precision and accuracy for static FLAB volumes. Glucose metabolic rate and 18F-FDG phosphorylation rate were higher in squamous cell carcinoma (SCC) than in adenocarcinoma (AC), whereas VB was lower (Mann-Whitney U test or t test, P = .003, P = .036, and P = .019, respectively). Glucose metabolic rate, 18F-FDG phosphorylation rate, and VB were less heterogeneous in AC than in SCC (Friedman analysis of variance). Conclusion Parametric images are not superior to static images for NSCLC delineation. FLAB-based segmentation on static 18F-FDG PET images is in best agreement with pathology volume and could be useful for NSCLC autocontouring. Differences in glycolytic rate and VB between SCC and AC are relevant for research in targeting agents and radiation therapy dose escalation. © RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Tineke W H Meijer
- From the Departments of Radiation Oncology (T.W.H.M., J.B.), Radiology and Nuclear Medicine (L.F.d.G.O., E.P.V., W.J.G.O.), Pathology (M.G.L.S.), and Cardiothoracic Surgery (A.F.T.M.V.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands (L.F.d.G.O., D. Vriens); Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, the Netherlands (L.F.d.G.O.); Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, England (W.J.G.O.); and INSERM, UMR 1101, LaTIM, Université de Bretagne Occidentale, Brest, France (D. Visvikis)
| | - Lioe-Fee de Geus-Oei
- From the Departments of Radiation Oncology (T.W.H.M., J.B.), Radiology and Nuclear Medicine (L.F.d.G.O., E.P.V., W.J.G.O.), Pathology (M.G.L.S.), and Cardiothoracic Surgery (A.F.T.M.V.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands (L.F.d.G.O., D. Vriens); Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, the Netherlands (L.F.d.G.O.); Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, England (W.J.G.O.); and INSERM, UMR 1101, LaTIM, Université de Bretagne Occidentale, Brest, France (D. Visvikis)
| | - Eric P Visser
- From the Departments of Radiation Oncology (T.W.H.M., J.B.), Radiology and Nuclear Medicine (L.F.d.G.O., E.P.V., W.J.G.O.), Pathology (M.G.L.S.), and Cardiothoracic Surgery (A.F.T.M.V.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands (L.F.d.G.O., D. Vriens); Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, the Netherlands (L.F.d.G.O.); Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, England (W.J.G.O.); and INSERM, UMR 1101, LaTIM, Université de Bretagne Occidentale, Brest, France (D. Visvikis)
| | - Wim J G Oyen
- From the Departments of Radiation Oncology (T.W.H.M., J.B.), Radiology and Nuclear Medicine (L.F.d.G.O., E.P.V., W.J.G.O.), Pathology (M.G.L.S.), and Cardiothoracic Surgery (A.F.T.M.V.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands (L.F.d.G.O., D. Vriens); Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, the Netherlands (L.F.d.G.O.); Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, England (W.J.G.O.); and INSERM, UMR 1101, LaTIM, Université de Bretagne Occidentale, Brest, France (D. Visvikis)
| | - Monika G Looijen-Salamon
- From the Departments of Radiation Oncology (T.W.H.M., J.B.), Radiology and Nuclear Medicine (L.F.d.G.O., E.P.V., W.J.G.O.), Pathology (M.G.L.S.), and Cardiothoracic Surgery (A.F.T.M.V.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands (L.F.d.G.O., D. Vriens); Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, the Netherlands (L.F.d.G.O.); Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, England (W.J.G.O.); and INSERM, UMR 1101, LaTIM, Université de Bretagne Occidentale, Brest, France (D. Visvikis)
| | - Dimitris Visvikis
- From the Departments of Radiation Oncology (T.W.H.M., J.B.), Radiology and Nuclear Medicine (L.F.d.G.O., E.P.V., W.J.G.O.), Pathology (M.G.L.S.), and Cardiothoracic Surgery (A.F.T.M.V.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands (L.F.d.G.O., D. Vriens); Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, the Netherlands (L.F.d.G.O.); Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, England (W.J.G.O.); and INSERM, UMR 1101, LaTIM, Université de Bretagne Occidentale, Brest, France (D. Visvikis)
| | - Ad F T M Verhagen
- From the Departments of Radiation Oncology (T.W.H.M., J.B.), Radiology and Nuclear Medicine (L.F.d.G.O., E.P.V., W.J.G.O.), Pathology (M.G.L.S.), and Cardiothoracic Surgery (A.F.T.M.V.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands (L.F.d.G.O., D. Vriens); Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, the Netherlands (L.F.d.G.O.); Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, England (W.J.G.O.); and INSERM, UMR 1101, LaTIM, Université de Bretagne Occidentale, Brest, France (D. Visvikis)
| | - Johan Bussink
- From the Departments of Radiation Oncology (T.W.H.M., J.B.), Radiology and Nuclear Medicine (L.F.d.G.O., E.P.V., W.J.G.O.), Pathology (M.G.L.S.), and Cardiothoracic Surgery (A.F.T.M.V.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands (L.F.d.G.O., D. Vriens); Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, the Netherlands (L.F.d.G.O.); Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, England (W.J.G.O.); and INSERM, UMR 1101, LaTIM, Université de Bretagne Occidentale, Brest, France (D. Visvikis)
| | - Dennis Vriens
- From the Departments of Radiation Oncology (T.W.H.M., J.B.), Radiology and Nuclear Medicine (L.F.d.G.O., E.P.V., W.J.G.O.), Pathology (M.G.L.S.), and Cardiothoracic Surgery (A.F.T.M.V.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands (L.F.d.G.O., D. Vriens); Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, the Netherlands (L.F.d.G.O.); Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, England (W.J.G.O.); and INSERM, UMR 1101, LaTIM, Université de Bretagne Occidentale, Brest, France (D. Visvikis)
| |
Collapse
|
50
|
Comparative evaluation of SUV, tumor-to-blood standard uptake ratio (SUR), and dual time point measurements for assessment of the metabolic uptake rate in FDG PET. EJNMMI Res 2016; 6:53. [PMID: 27334609 PMCID: PMC4917506 DOI: 10.1186/s13550-016-0208-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/14/2016] [Indexed: 11/26/2022] Open
Abstract
Background We have demonstrated recently that the tumor-to-blood standard uptake ratio (SUR) is superior to tumor standardized uptake value (SUV) as a surrogate of the metabolic uptake rate Km of fluorodeoxyglucose (FDG), overcoming several of the known shortcomings of the SUV approach: excellent linear correlation of SUR and Km from Patlak analysis was found using dynamic imaging of liver metastases. However, due to the perfectly standardized uptake period used for SUR determination and the comparatively short uptake period, these results are not automatically valid and applicable for clinical whole-body examinations in which the uptake periods (T) are distinctly longer and can vary considerably. Therefore, the aim of this work was to investigate the correlation between SUR derived from clinical static whole-body scans and Km-surrogate derived from dual time point (DTP) measurements. Methods DTP 18F-FDG PET/CT was performed in 90 consecutive patients with histologically proven non-small cell lung cancer (NSCLC). In the PET images, the primary tumor was delineated with an adaptive threshold method. For determination of the blood SUV, an aorta region of interest (ROI) was delineated manually in the attenuation CT and transferred to the PET image. Blood SUV was computed as the mean value of the aorta ROI. SUR values were computed as ratio of tumor SUV and blood SUV. SUR values from the early time point of each DTP measurement were scan time corrected to 75 min postinjection (SURtc). As surrogate of Km, we used the SUR(T) slope, Kslope, derived from DTP measurements since it is proportional to the latter under the given circumstances. The correlation of SUV and SURtc with Kslope was investigated. The prognostic value of SUV, SURtc, and Kslope for overall survival (OS) and progression-free survival (PFS) was investigated with univariate Cox regression in a homogeneous subgroup (N=31) treated with primary chemoradiation. Results Correlation analysis revealed for both, SUV and SURtc, a clear linear correlation with Kslope (P<0.001). Correlation SUR vs. Kslope was considerably stronger than correlation SUV vs. Kslope (R2=0.92 and R2=0.69, respectively, P<0.001). Univariate Cox regression revealed SURtc and Kslope as significant prognostic factors for PFS (hazard ratio (HR) =3.4/ P=0.017 and HR =4.3/ P=0.020, respectively). For SUV, no significant effect was found. None of the investigated parameters was prognostic for OS. Conclusions Scan-time-corrected SUR is a significantly better surrogate of tumor FDG metabolism in clinical whole-body PET compared to SUV. The very high linear correlation of SUR and DTP-derived Kslope (which is proportional to actual Km) implies that for histologically proven malignant lesions, FDG-DTP does not provide added value in comparison to the SUR approach in NSCLC.
Collapse
|