1
|
Baier JM, Funck KL, Dons-Jensen A, Munk OL, Tolbod LP, Laugesen E, Poulsen PL, Gormsen LC, Dias AH. Test-retest repeatability of quantitative organ and tissue uptake using 20-minute dynamic multiparametric whole-body [ 18F]FDG PET/CT in patients with type 2 diabetes. EJNMMI Res 2025; 15:56. [PMID: 40354003 PMCID: PMC12069784 DOI: 10.1186/s13550-025-01249-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/25/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Recently developed dynamic whole-body PET/CT (D-WB PET/CT) protocols allow for measurements of potentially more precise metabolic parameters than the commonly used semiquantitative SUV. Most notable is the metabolic rate of FDG uptake (MRFDG), which reflects quantitative glucose uptake into tissues and organs. However, data on the reproducibility of MRFDG measurements are scarce, particularly in patients with perturbed glucose homeostasis such as type 2 diabetes. We therefore aimed to evaluate the test-retest repeatability of both MRFDG and SUV in these patients. RESULTS Fifteen participants (mean age 71 ± 7 years; 2 females) with type 2 diabetes underwent a short 20-minute [18F]FDG D-WB PET/CT after 6 h fasting on two consecutive days. Both SUV and MRFDG images were reconstructed from D-WB PET/CT data obtained 60-80 min post-injection of [18F]FDG. MRFDG and SUV data were measured in organs and tissues, and repeatability was assessed with Bland-Altman analysis, intraclass correlation coefficients (ICC), repeatability coefficients (RPC) and coefficients of variation (wCV). There was high repeatability of both SUVmean and MRFDG-mean in all measured organs (ICC range: 0.65-0.95 for SUVmean and 0.66-0.94 for MRFDG-mean). SUVmean generally demonstrated higher reliability (ICC) and lower variability (%RPC and %wCV) when compared to MRFDG-mean. However, MRFDG test-retest variation was < 19% in most analysed tissues, demonstrating that MRFDG may be used as a precise marker of treatment response. CONCLUSION This study demonstrates that MRFDG calculated from D-WB PET/CT exhibit high repeatability, comparable to SUVs across most organs in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Jonathan M Baier
- Dept. of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Kristian L Funck
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Dept. of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Anna Dons-Jensen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Ole L Munk
- Dept. of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, Aarhus N, DK-8200, Denmark
| | - Lars P Tolbod
- Dept. of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, Aarhus N, DK-8200, Denmark
| | - Esben Laugesen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Diagnostic Centre, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Per L Poulsen
- Dept. of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Lars C Gormsen
- Dept. of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, Aarhus N, DK-8200, Denmark
| | - André H Dias
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, Aarhus N, DK-8200, Denmark.
| |
Collapse
|
2
|
Mohymen AA, Farag HI, Reda SM, Monem AS, Ali SA. Optimization of Reconstruction Parameters for Discovery 710 Positron Emission Tomography/Computed Tomography. J Med Phys 2025; 50:118-130. [PMID: 40256189 PMCID: PMC12005667 DOI: 10.4103/jmp.jmp_167_24] [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: 10/03/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 04/22/2025] Open
Abstract
Aim This study aimed to optimize the quantitative aspects of (18F) fluorodeoxyglucose (FDG) positron emission tomography (PET)/computed tomography (CT) imaging by investigating the impact of various reconstruction parameters on the recovery coefficients (RCs) using the NEMA image quality phantom. Specifically, the study aims to assess how different matrix sizes, iterations, subsets, and Gaussian postfilters affect the accuracy of standardized uptake value (SUV) quantification in (18F) FDG PET/CT imaging. Materials and Methods The study utilized the "Vue Point FX + Sharp IR" algorithm for PET image reconstruction, incorporating 3D-ordered subset expectation maximization (3D-OSEM), time-of-flight, and point spread function technologies. Various reconstruction parameters were explored, including two distinct matrix sizes, multiple iterations, subsets, and a wide range of Gaussian postfilters. The investigation focused on the impact of these parameters on RCs using the NEMA image quality phantom. Results The results of the study indicated that for accurate SUV quantification in spheres ≥17 mm, the 256 × 256 matrix size and mean SUV should be employed. Conversely, for spheres ≤13 mm, maximum SUV was found to be more suitable. The choice of postfiltering value was shown to have a significant impact on SUV quantification accuracy, particularly for small-sized spheres. In addition, a larger matrix size was found to partially mitigate the effects of Gibbs artifact and slightly enhance SUV quantification for the spheres of various sizes. Conclusion This study highlights the critical importance of optimizing PET reconstruction parameters in accordance with the guidelines set by European Association of Nuclear Medicine/EARL. By optimizing these parameters, the accuracy and reliability of SUV quantification in (18F) FDG PET imaging can be significantly enhanced, especially for small-sized spheres. This underscores the necessity of carefully considering reconstruction parameters to ensure precise and reliable quantitative measurements in PET/CT imaging.
Collapse
Affiliation(s)
- Ahmed Abdel Mohymen
- Department of Nuclear Medicine and Radiation Therapy, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Hamed Ibrahim Farag
- Department of Nuclear Medicine and Radiation Therapy, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Sameh M. Reda
- Department of Radiometry, National Institute of Standards, Giza, Egypt
| | - Ahmed Soltan Monem
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | | |
Collapse
|
3
|
Metrard G, Cohen C, Bailly M. Comprehensive literature review of oral and intravenous contrast-enhanced PET/CT: a step forward? Front Med (Lausanne) 2024; 11:1373260. [PMID: 38566921 PMCID: PMC10985176 DOI: 10.3389/fmed.2024.1373260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
The integration of diagnostic CT scans into PET/CT facilitates a comprehensive single examination, presenting potential advantages for patients seeking a thorough one-shot check-up. The introduction of iodinated contrast media during PET scanning raises theoretical concerns about potential interference with uptake quantification, due to the modification of tissue density on CT. Nevertheless, this impact appears generally insignificant for clinical use, compared to the intrinsic variability of standardized uptake values. On the other hand, with the growing indications of PET, especially 18F-FDG PET, contrast enhancement increases the diagnostic performances of the exam, and provides additional information. This improvement in performance achieved through contrast-enhanced PET/CT must be carefully evaluated considering the associated risks and side-effects stemming from the administration of iodinated contrast media. Within this article, we present a comprehensive literature review of contrast enhanced PET/CT, examining the potential impact of iodinated contrast media on quantification, additional side-effects and the pivotal clinically demonstrated benefits of an all-encompassing examination for patients. In conclusion, the clinical benefits of iodinated contrast media are mainly validated by the large diffusion in PET protocols. Contrary to positive oral contrast, which does not appear to offer any major advantage in patient management, intravenous iodine contrast media provides clinical benefits without significant artifact on images or quantification. However, studies on the benefit-risk balance for patients are still lacking.
Collapse
Affiliation(s)
- Gilles Metrard
- Nuclear Medicine Department, Orléans University Hospital, Orléans, France
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d’Orléans, Orléans, France
| | - Clara Cohen
- Radiology Department, Orléans University Hospital, Orléans, France
| | - Matthieu Bailly
- Nuclear Medicine Department, Orléans University Hospital, Orléans, France
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d’Orléans, Orléans, France
| |
Collapse
|
4
|
Wyatt JJ, Kaushik S, Cozzini C, Pearson RA, Petrides G, Wiesinger F, McCallum HM, Maxwell RJ. Evaluating a radiotherapy deep learning synthetic CT algorithm for PET-MR attenuation correction in the pelvis. EJNMMI Phys 2024; 11:10. [PMID: 38282050 PMCID: PMC11266329 DOI: 10.1186/s40658-024-00617-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/15/2024] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Positron emission tomography-magnetic resonance (PET-MR) attenuation correction is challenging because the MR signal does not represent tissue density and conventional MR sequences cannot image bone. A novel zero echo time (ZTE) MR sequence has been previously developed which generates signal from cortical bone with images acquired in 65 s. This has been combined with a deep learning model to generate a synthetic computed tomography (sCT) for MR-only radiotherapy. This study aimed to evaluate this algorithm for PET-MR attenuation correction in the pelvis. METHODS Ten patients being treated with ano-rectal radiotherapy received a [Formula: see text]F-FDG-PET-MR in the radiotherapy position. Attenuation maps were generated from ZTE-based sCT (sCTAC) and the standard vendor-supplied MRAC. The radiotherapy planning CT scan was rigidly registered and cropped to generate a gold standard attenuation map (CTAC). PET images were reconstructed using each attenuation map and compared for standard uptake value (SUV) measurement, automatic thresholded gross tumour volume (GTV) delineation and GTV metabolic parameter measurement. The last was assessed for clinical equivalence to CTAC using two one-sided paired t tests with a significance level corrected for multiple testing of [Formula: see text]. Equivalence margins of [Formula: see text] were used. RESULTS Mean whole-image SUV differences were -0.02% (sCTAC) compared to -3.0% (MRAC), with larger differences in the bone regions (-0.5% to -16.3%). There was no difference in thresholded GTVs, with Dice similarity coefficients [Formula: see text]. However, there were larger differences in GTV metabolic parameters. Mean differences to CTAC in [Formula: see text] were [Formula: see text] (± standard error, sCTAC) and [Formula: see text] (MRAC), and [Formula: see text] (sCTAC) and [Formula: see text] (MRAC) in [Formula: see text]. The sCTAC was statistically equivalent to CTAC within a [Formula: see text] equivalence margin for [Formula: see text] and [Formula: see text] ([Formula: see text] and [Formula: see text]), whereas the MRAC was not ([Formula: see text] and [Formula: see text]). CONCLUSION Attenuation correction using this radiotherapy ZTE-based sCT algorithm was substantially more accurate than current MRAC methods with only a 40 s increase in MR acquisition time. This did not impact tumour delineation but did significantly improve the accuracy of whole-image and tumour SUV measurements, which were clinically equivalent to CTAC. This suggests PET images reconstructed with sCTAC would enable accurate quantitative PET images to be acquired on a PET-MR scanner.
Collapse
Affiliation(s)
- Jonathan J Wyatt
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
- Northern Centre for Cancer Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| | - Sandeep Kaushik
- GE Healthcare, Munich, Germany
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | | | - Rachel A Pearson
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Northern Centre for Cancer Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - George Petrides
- Nuclear Medicine Department, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Hazel M McCallum
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Northern Centre for Cancer Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Ross J Maxwell
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
5
|
So H, Lee H, Hyun SH, Cho YS, Moon SH, Choi JY, Lee KH. Metabolic bulk volume from FDG PET as an independent predictor of progression-free survival in follicular lymphoma. Front Oncol 2023; 13:1283582. [PMID: 38023238 PMCID: PMC10655116 DOI: 10.3389/fonc.2023.1283582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Background Total metabolic tumor volume (TMTV) in 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) predicts patient outcome in follicular lymphoma (FL); however, it requires laborious segmentation of all lesions. We investigated the prognostic value of the metabolic bulk volume (MBV) obtained from the single largest lesion. Methods Pretreatment FDG PET/computed tomography (CT) scans of 201 patients were analyzed for TMTV and MBV using a 41% maximum standardized uptake value (SUVmax) threshold. Results During a median follow-up of 3.2 years, 54 events, including 14 deaths, occurred. Optimal cut-offs were 121.1 cm3 for TMTV and 24.8 cm3 for MBV. Univariable predictors of progression-free survival (PFS) included a high Follicular Lymphoma International Prognostic Index 2 (FLIPI2) score, TMTV, and MBV. In the multivariable analysis, high TMTV and MBV were independent predictors of worse PFS (P =0.015 and 0.033). Furthermore, in a sub-group with FLIP2 scores of 0-2 (n = 132), high MBV could identify patients with worse PFS (P = 0.007). . Conclusion Readily measurable MBV is useful for stratifying risk in FL patients.
Collapse
Affiliation(s)
- Heejune So
- School of Medicine, University of Galway, Galway, Ireland
| | - Hyunjong Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Hyup Hyun
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Seok Cho
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
6
|
Natarajan A, Khan S, Liang X, Nguyen H, Das N, Anders D, Malik N, Oderinde OM, Chin FT, Rosenthal E, Pratx G. Preclinical Evaluation of 89Zr-Panitumumab for Biology-Guided Radiation Therapy. Int J Radiat Oncol Biol Phys 2023; 116:927-934. [PMID: 36669541 PMCID: PMC11290461 DOI: 10.1016/j.ijrobp.2023.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/27/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023]
Abstract
PURPOSE Biology-guided radiation therapy (BgRT) uses real-time line-of-response data from on-board positron emission tomography (PET) detectors to guide beamlet delivery during therapeutic radiation. The current workflow requires 18F-fluorodeoxyglucose (FDG) administration daily before each treatment fraction. However, there are advantages to reducing the number of tracer injections by using a PET tracer with a longer decay time. In this context, we investigated 89Zr-panitumumab (89Zr-Pan), an antibody PET tracer with a half-life of 78 hours that can be imaged for up to 9 days using PET. METHODS AND MATERIALS The BgRT workflow was evaluated preclinically in mouse colorectal cancer xenografts (HCT116) using small-animal positron emission tomography/computed tomography (PET/CT) for imaging and image-guided kilovoltage conformal irradiation for therapy. Mice (n = 5 per group) received 7 MBq of 89Zr-Pan as a single dose 2 weeks after tumor induction, with or without fractionated radiation therapy (RT; 6 × 6.6 Gy) to the tumor region. The mice were imaged longitudinally to assess the kinetics of the tracer over 9 days. PET images were then analyzed to determine the stability of the PET signal in irradiated tumors over time. RESULTS Mice in the treatment group experienced complete tumor regression, whereas those in the control group were killed because of tumor burden. PET imaging of 89Zr-Pan showed well-delineated tumors with minimal background in both groups. On day 9 postinjection, tumor uptake of 89Zr-Pan was 7.2 ± 1.7 in the control group versus 5.2 ± 0.5 in the treatment group (mean percentage of injected dose per gram of tissue [%ID/g] ± SD; P = .07), both significantly higher than FDG uptake (1.1 ± 0.5 %ID/g) 1 hour postinjection. To assess BgRT feasibility, the clinical eligibility criteria was computed using human-equivalent uptake values that were extrapolated from preclinical PET data. Based on this semiquantitative analysis, BgRT may be feasible for 5 consecutive days after a single 740-MBq injection of 89Zr-Pan. CONCLUSIONS This study indicates the potential of long-lived antibody-based PET tracers for guiding clinical BgRT.
Collapse
Affiliation(s)
| | - Syamantak Khan
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Xuanwei Liang
- Department of Physics, Foothill College, Los Altos, California
| | - Hieu Nguyen
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Neeladrisingha Das
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - David Anders
- Department of Radiology, Stanford University, Stanford, California
| | - Noeen Malik
- Department of Radiology, Stanford University, Stanford, California
| | | | - Frederick T Chin
- Department of Radiology, Stanford University, Stanford, California
| | - Eben Rosenthal
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Guillem Pratx
- Department of Radiation Oncology, Stanford University, Stanford, California.
| |
Collapse
|
7
|
AlRasheedi M, Han S, Thygesen H, Neilson M, Hendry F, Alkarn A, Maclay JD, Leung HY. A Comparative Evaluation of Mediastinal Nodal SUVmax and Derived Ratios from 18F-FDG PET/CT Imaging to Predict Nodal Metastases in Non-Small Cell Lung Cancer. Diagnostics (Basel) 2023; 13:1209. [PMID: 37046427 PMCID: PMC10093125 DOI: 10.3390/diagnostics13071209] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 04/14/2023] Open
Abstract
18F-FDG positron emission tomography with computed tomography (PET/CT) is a standard imaging modality for the nodal staging of non-small cell lung cancer (NSCLC). To improve the accuracy of pre-operative staging, we compare the staging accuracy of mediastinal lymph node (LN) standard uptake values (SUV) with four derived SUV ratios based on the SUV values of primary tumours (TR), the mediastinal blood pool (MR), liver (LR), and nodal size (SR). In 2015-2017, 53 patients (29 women and 24 men, mean age 67.4 years, range 53-87) receiving surgical resection have pre-operative evidence of mediastinal nodal involvement (cN2). Among these, 114 mediastinal nodes are resected and available for correlative PET/CT analysis. cN2 status accuracy is low, with only 32.5% of the cN2 cases confirmed pathologically. Using receiver operating characteristic (ROC) curve analyses, a SUVmax of N2 LN performs well in predicting the presence of N2 disease (AUC, 0.822). Based on the respective selected thresholds for each ROC curve, normalisation of LN SUVmax to that for mediastinum, liver and tumour improved sensitivities of LN SUVmax from 68% to 81.1-89.2% while maintaining acceptable specificity (68-70.1%). In conclusion, normalised SUV ratios (particularly LR) improve current pre-operative staging performance in detecting mediastinal nodal involvement.
Collapse
Affiliation(s)
- Maha AlRasheedi
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (M.A.)
- West of Scotland PET Centre, Gartnavel General Hospital, NHS Greater Glasgow and Clyde, Glasgow G12 0YN, UK
| | - Sai Han
- West of Scotland PET Centre, Gartnavel General Hospital, NHS Greater Glasgow and Clyde, Glasgow G12 0YN, UK
| | - Helene Thygesen
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (M.A.)
| | - Matt Neilson
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Fraser Hendry
- West of Scotland PET Centre, Gartnavel General Hospital, NHS Greater Glasgow and Clyde, Glasgow G12 0YN, UK
| | - Ahmed Alkarn
- Department of Respiratory Medicine, Glasgow Royal Infirmary, NHS Greater Glasgow and Clyde, Glasgow G4 0SF, UK
| | - John D. Maclay
- Department of Respiratory Medicine, Glasgow Royal Infirmary, NHS Greater Glasgow and Clyde, Glasgow G4 0SF, UK
| | - Hing Y. Leung
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; (M.A.)
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
8
|
Fragoso Costa P, Jentzen W, SÜßELBECK F, Fendler WP, Rischpler C, Herrmann K, Conti M, Kersting D, Weber M. Reduction of emission time for [68Ga]Ga-PSMA PET/CT using the digital biograph vision: a phantom study. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2023; 67:57-68. [PMID: 34309334 DOI: 10.23736/s1824-4785.21.03300-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The aim of this phantom study was to optimize the [68Ga]Ga-PSMA PET/CT examination in terms of scan time duration and image reconstruction parameters, in combination with PSF and TOF modelling, in a digital Biograph Vision PET/CT scanner. METHODS Three types of phantoms were used: 1) soft-tissue tumor phantom consisting of six spheres mounted in a torso phantom; 2) bone-lung tumor phantom; 3) resolution phantom. Phantom inserts were filled with activity concentrations (ACs) that were derived from clinical data. Phantom data were acquired in list-mode at one bed position. Images with emission data ranging from 30 to 210 s in 30-s increments were reconstructed from a reference image acquired with 3.5-min emission. Iterative image reconstruction (OSEM), point-spread-function (PSF) and time-of-flight (TOF) options were applied using different iterations, Gaussian filters, and voxel sizes. The criteria for image quality was lesion detectability and lesion quantification, evaluated as contrast-to-noise ratio (CNR) and maximum AC (peak AC), respectively. A threshold value of CNR above 6 and percentage maximum AC (peak AC) deviation range of ±20% of the reference image were considered acceptable. The proposed single-bed scan time reduction was projected to a whole-body examination (patient validation scan) using the continuous-bed-motion mode. RESULTS Sphere and background ACs of 20 kBq/mL and 1 kBq/mL were selected, respectively. The optimized single-bed scan time was approximately 60 s using OSEM-TOF or OSEM-TOF+PSF (four iterations, 4.0-mm Gaussian filter and almost isotropic voxel size of 3.0-mm side length), resulting in a PET spatial resolution of 6.3 mm for OSEM-TOF and 5.5 mm for OSEM-TOF+PSF. In the patient validation, the maximum percentage difference in lesion quantification between standard and optimized protocol (whole-body scan time of 15 vs. 5 min) was below 19%. CONCLUSIONS A reduction of single-bed and whole-body scan time for [68Ga]Ga-PSMA PET/CT compared to current recommended clinical acquisition protocols is postulated. Clinical studies are warranted to validate the applicability of this protocol.
Collapse
Affiliation(s)
- Pedro Fragoso Costa
- Department of Nuclear Medicine, Essen University Hospital, Duisburg-Essen University, Essen, Germany -
| | - Walter Jentzen
- Department of Nuclear Medicine, Essen University Hospital, Duisburg-Essen University, Essen, Germany
| | - Finja SÜßELBECK
- Department of Nuclear Medicine, Essen University Hospital, Duisburg-Essen University, Essen, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, Essen University Hospital, Duisburg-Essen University, Essen, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, Essen University Hospital, Duisburg-Essen University, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, Essen University Hospital, Duisburg-Essen University, Essen, Germany
| | | | - David Kersting
- Department of Nuclear Medicine, Essen University Hospital, Duisburg-Essen University, Essen, Germany
| | - Manuel Weber
- Department of Nuclear Medicine, Essen University Hospital, Duisburg-Essen University, Essen, Germany
| |
Collapse
|
9
|
Ricard F, Barrington S, Korn R, Brueggenwerth G, Trotman J, Cheson B, Salles G, Schwartz L, Goldmacher G, Jarecha R, Narang J, Broussais F, Galette P, Liu M, Bajpai S, Perlman E, Gillis J, Smalberg I, Terve P, Zahlmann G, Schmid A. Application of the Lugano Classification for Initial Evaluation, Staging, and Response Assessment of Hodgkin and Non-Hodgkin Lymphoma: The PRoLoG Consensus Initiative (Part 2-Technical). J Nucl Med 2023; 64:239-243. [PMID: 35835581 PMCID: PMC9902846 DOI: 10.2967/jnumed.122.264124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
The aim of this initiative was to provide consensus recommendations from a consortium of academic and industry experts in the field of lymphoma and imaging for the consistent application of imaging assessment with the Lugano classification. Methods: Consensus was obtained through a series of meetings from July 2019 to October 2021 sponsored by the PINTaD (Pharma Imaging Network for Therapeutics and Diagnostics) as part of the ProLoG (PINTaD RespOnse criteria in Lymphoma wOrking Group) consensus initiative. Results: Consensus recommendations encompass all technical imaging aspects of the Lugano classification. Some technical considerations for PET/CT and diagnostic CT are clarified with regards to required imaging series and scan visits, as well as acquisition and reconstruction of PET images and influence of lesion size and background activity. Recommendations are given on the role of imaging and clinical reviewers as well as on training and monitoring. Finally, an example template of an imaging case report form is provided to support efficient collection of data with Lugano Classification. Conclusion: Consensus recommendations are made to comprehensively address technical and imaging areas of inconsistency and ambiguity in the classification encountered by end users. Such guidance should be used to support standardized acquisition and evaluation with the Lugano 2014.
Collapse
Affiliation(s)
| | - Sally Barrington
- King's College London and Guy's and St. Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, London, United Kingdom
| | - Ron Korn
- Adjunct Faculty TGEN/City of Hope and Imaging Endpoints Core Lab, Scottsdale, Arizona
| | | | - Judith Trotman
- Concord Repatriation General Hospital, University of Sydney, Concord, Australia
| | - Bruce Cheson
- Lymphoma Research Foundation, New York, New York
| | - Gilles Salles
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weil Cornell Medicine, New York, New York
| | - Larry Schwartz
- Department of Radiology, Columbia University College of Physicians and Surgeons and Radiologist-in-Chief, The New York Presbyterian Hospital, Columbia, New York
| | | | | | - Jayant Narang
- Takeda Pharmaceutical Company Ltd, Cambridge, Massachusetts
| | - Florence Broussais
- Lymphoma Study Association Research Center LYSARC, Pierre Benite, France Calyx International, Billerica, Massachusetts
| | | | - Min Liu
- Autolus Therapeutics, London, United Kingdom
| | | | - Eric Perlman
- Perlman Advisory Group LLC, Boynton Beach, Florida
| | | | - Ira Smalberg
- Saint John's Cancer Institute and Tower Imaging Medical Group, Sherman Oaks, California
| | - Pierre Terve
- KEOSYS Medical Imaging, Saint Herblain, France; and
| | - Gudrun Zahlmann
- QIBA/RSNA, Radiological Society of North America, Oak Brook, Illinois
| | - Annette Schmid
- Takeda Pharmaceutical Company Ltd, Cambridge, Massachusetts
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Fragoso Costa P, Jentzen W, Brahmer A, Mavroeidi IA, Zarrad F, Umutlu L, Fendler WP, Rischpler C, Herrmann K, Conti M, Seifert R, Sraieb M, Weber M, Kersting D. Phantom-based acquisition time and image reconstruction parameter optimisation for oncologic FDG PET/CT examinations using a digital system. BMC Cancer 2022; 22:899. [PMID: 35978274 PMCID: PMC9387080 DOI: 10.1186/s12885-022-09993-4] [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: 12/05/2021] [Accepted: 08/08/2022] [Indexed: 11/18/2022] Open
Abstract
Background New-generation silicon-photomultiplier (SiPM)-based PET/CT systems exhibit an improved lesion detectability and image quality due to a higher detector sensitivity. Consequently, the acquisition time can be reduced while maintaining diagnostic quality. The aim of this study was to determine the lowest 18F-FDG PET acquisition time without loss of diagnostic information and to optimise image reconstruction parameters (image reconstruction algorithm, number of iterations, voxel size, Gaussian filter) by phantom imaging. Moreover, patient data are evaluated to confirm the phantom results. Methods Three phantoms were used: a soft-tissue tumour phantom, a bone-lung tumour phantom, and a resolution phantom. Phantom conditions (lesion sizes from 6.5 mm to 28.8 mm in diameter, lesion activity concentration of 15 kBq/mL, and signal-to-background ratio of 5:1) were derived from patient data. PET data were acquired on an SiPM-based Biograph Vision PET/CT system for 10 min in list-mode format and resampled into time frames from 30 to 300 s in 30-s increments to simulate different acquisition times. Different image reconstructions with varying iterations, voxel sizes, and Gaussian filters were probed. Contrast-to-noise-ratio (CNR), maximum, and peak signal were evaluated using the 10-min acquisition time image as reference. A threshold CNR value ≥ 5 and a maximum (peak) deviation of ± 20% were considered acceptable. 20 patient data sets were evaluated regarding lesion quantification as well as agreement and correlation between reduced and full acquisition time standard uptake values (assessed by Pearson correlation coefficient, intraclass correlation coefficient, Bland–Altman analyses, and Krippendorff’s alpha). Results An acquisition time of 60 s per bed position yielded acceptable detectability and quantification results for clinically relevant phantom lesions ≥ 9.7 mm in diameter using OSEM-TOF or OSEM-TOF+PSF image reconstruction, a 4-mm Gaussian filter, and a 1.65 × 1.65 x 2.00-mm3 or 3.30 × 3.30 x 3.00-mm3 voxel size. Correlation and agreement of patient lesion quantification between full and reduced acquisition times were excellent. Conclusion A threefold reduction in acquisition time is possible. Patients might benefit from more comfortable examinations or reduced radiation exposure, if instead of the acquisition time the applied activity is reduced. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09993-4.
Collapse
Affiliation(s)
- Pedro Fragoso Costa
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Walter Jentzen
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Alissa Brahmer
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Ilektra-Antonia Mavroeidi
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.,Department of Medical Oncology, University Hospital Essen, West German Cancer Center (WTZ), University Duisburg-Essen, 45147, Essen, Germany
| | - Fadi Zarrad
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Lale Umutlu
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.,Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University Duisburg-Essen, 45147, Essen, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | | | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Miriam Sraieb
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Manuel Weber
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - David Kersting
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany. .,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.
| |
Collapse
|
13
|
Harmonization based on quantitative analysis of standardized uptake value variations across PET/CT scanners: a multicenter phantom study. Nucl Med Commun 2022; 43:1004-1014. [PMID: 35836388 DOI: 10.1097/mnm.0000000000001598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES This study aimed to measure standardized uptake value (SUV) variations across different PET/computed tomography (CT) scanners to harmonize quantification across systems. METHODS We acquired images using the National Electrical Manufacturers Association International Electrotechnical Commission phantom from three PET/CT scanners operated using routine imaging protocols at each site. The SUVs of lesions were assessed in the presence of reference values by a digital reference object (DRO) and recommendations by the European Association of Nuclear Medicine (EANM/EARL) to measure inter-site variations. For harmonization, Gaussian filters with tuned full width at half maximum (FWHM) values were applied to images to minimize differences in SUVs between reference and images. Inter-site variation of SUVs was evaluated in both pre- and postharmonization situations. Test-retest analysis was also carried out to evaluate repeatability. RESULTS SUVs from different scanners became significantly more consistent, and inter-site differences decreased for SUVmean, SUVmax and SUVpeak from 17.3, 20.7, and 15.5% to 4.8, 4.7, and 2.7%, respectively, by harmonization (P values <0.05 for all). The values for contrast-to-noise ratio in the smallest lesion of the phantom verified preservation of image quality following harmonization (>2.8%). CONCLUSIONS Harmonization significantly lowered variations in SUV measurements across different PET/CT scanners, improving reproducibility while preserving image quality.
Collapse
|
14
|
Chen W, Liu L, Li Y, Li S, Li Z, Zhang W, Zhang X, Wu R, Hu D, Sun H, Zhou Y, Fan W, Zhao Y, Zhang Y, Hu Y. Evaluation of pediatric malignancies using total-body PET/CT with half-dose [ 18F]-FDG. Eur J Nucl Med Mol Imaging 2022; 49:4145-4155. [PMID: 35788704 DOI: 10.1007/s00259-022-05893-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/25/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE To explore the impact of a true half dose of [18F]-FDG on image quality in pediatric oncological patients undergoing total-body PET/CT and investigate short acquisition times with half-dose injected activity. METHODS One hundred pediatric oncological patients who underwent total-body PET/CT using the uEXPLORER scanner after receiving a true half dose of [18F]-FDG (1.85 MBq/kg) were retrospectively enrolled. The PET images were first reconstructed using complete 600-s data and then split into 300-s, 180-s, 60-s, 40-s, and 20-s duration groups (G600 to G20). The subjective analysis was performed using 5-point Likert scales. Objective quantitative metrics included the maximum standard uptake value (SUVmax), SUVmean, standard deviation (SD), signal-to-noise ratio (SNR), and SNRnorm of the background. The variabilities in lesion SUVmean, SUVmax, and tumor-to-background ratio (TBR) were also calculated. RESULTS The overall image quality scores in the G600, G300, G180, and G60 groups were 4.9 ± 0.2, 4.9 ± 0.3, 4.4 ± 0.5, and 3.5 ± 0.5 points, respectively. All the lesions identified in the half-dose images were localized in the G60 images, while 56% of the lesions could be clearly identified in the G20 images. With reduced acquisition time, the SUVmax and SD of the backgrounds were gradually increased, while the TBR values showed no statistically significant differences among the groups (all p > 0.1). Using the half-dose images as a reference, the variability in the lesion SUVmax gradually increased from the G180 to G20 images, while the lesion SUVmean remained stable across all age groups. SNRnorm was highly negatively correlated with age. CONCLUSION Total-body PET/CT with a half dose of [18F]-FDG (1.85 MBq/kg, estimated whole-body effective dose: 1.76-2.57 mSv) achieved good performance in pediatric patients, with sufficient image quality and good lesion conspicuity. Sufficient image quality and lesion conspicuity could be maintained at a fast scanning time of 60 s with half-dose activity.
Collapse
Affiliation(s)
- Wanqi Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfengdong Road, Guangzhou, 510060, Guangdong, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Lei Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfengdong Road, Guangzhou, 510060, Guangdong, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yinghe Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfengdong Road, Guangzhou, 510060, Guangdong, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shatong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfengdong Road, Guangzhou, 510060, Guangdong, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhijian Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfengdong Road, Guangzhou, 510060, Guangdong, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Weiguang Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfengdong Road, Guangzhou, 510060, Guangdong, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xu Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfengdong Road, Guangzhou, 510060, Guangdong, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Runze Wu
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - Debin Hu
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - Hongyan Sun
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - Yun Zhou
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - Wei Fan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfengdong Road, Guangzhou, 510060, Guangdong, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yumo Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfengdong Road, Guangzhou, 510060, Guangdong, China.
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Yizhuo Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfengdong Road, Guangzhou, 510060, Guangdong, China.
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Yingying Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfengdong Road, Guangzhou, 510060, Guangdong, China.
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
15
|
Tanaka A, Sekine T, Ter Voert EEGW, Zeimpekis KG, Delso G, de Galiza Barbosa F, Warnock G, Kumita SI, Veit Haibach P, Huellner M. Reproducibility of Standardized Uptake Values Including Volume Metrics Between TOF-PET-MR and TOF-PET-CT. Front Med (Lausanne) 2022; 9:796085. [PMID: 35308500 PMCID: PMC8924656 DOI: 10.3389/fmed.2022.796085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/07/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose To investigate the reproducibility of tracer uptake measurements, including volume metrics, such as metabolic tumor volume (MTV) and tumor lesion glycolysis (TLG) obtained by TOF-PET-CT and TOF-PET-MR. Materials and Methods Eighty consecutive patients with different oncologic diagnoses underwent TOF-PET-CT (Discovery 690; GE Healthcare) and TOF-PET-MR (SIGNA PET-MR; GE Healthcare) on the same day with single dose−18F-FDG injection. The scan order, PET-CT following or followed by PET-MR, was randomly assigned. A spherical volume of interest (VOI) of 30 mm was placed on the liver in accordance with the PERCIST criteria. For liver, the maximum and mean standard uptake value for body weight (SUV) and lean body mass (SUL) were obtained. For tumor delineation, VOI with a threshold of 40 and 50% of SUVmax was used (VOI40 and VOI50). The SUVmax, SUVmean, SUVpeak, MTV and TLG were calculated. The measurements were compared between the two scanners. Results In total, 80 tumor lesions from 35 patients were evaluated. There was no statistical difference observed in liver regions, whereas in tumor lesions, SUVmax, SUV mean, and SUVpeak of PET-MR were significantly underestimated (p < 0.001) in both VOI40 and VOI50. Among volume metrics, there was no statistical difference observed except TLG on VOI50 (p = 0.03). Correlation between PET-CT and PET-MR of each metrics were calculated. There was a moderate correlation of the liver SUV and SUL metrics (r = 0.63–0.78). In tumor lesions, SUVmax and SUVmean had a stronger correlation with underestimation in PET-MR on VOI 40 (SUVmax and SUVmean; r = 0.92 and 0.91 with slope = 0.71 and 0.72, respectively). In the evaluation of MTV and TLG, the stronger correlations were observed both on VOI40 (MTV and TLG; r = 0.75 and 0.92) and VOI50 (MTV and TLG; r = 0.88 and 0.95) between PET-CT and PET-MR. Conclusion PET metrics on TOF-PET-MR showed a good correlation with that of TOF-PET-CT. SUVmax and SUVpeak of tumor lesions were underestimated by 16% on PET-MRI. MTV with % threshold can be regarded as identical volumetric markers for both TOF-PET-CT and TOF-PET-MR.
Collapse
Affiliation(s)
- Aruki Tanaka
- Department of Radiology, Nippon Medical School Hospital, Tokyo, Japan
| | - Tetsuro Sekine
- Department of Radiology, Nippon Medical School Hospital, Tokyo, Japan.,Department of Radiology, Nippon Medical School Musashi Kosugi Hospital, Kanagawa, Japan.,Departments of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.,University of Zurich, Zurich, Switzerland
| | - Edwin E G W Ter Voert
- Departments of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.,University of Zurich, Zurich, Switzerland
| | - Konstantinos G Zeimpekis
- Departments of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.,University of Zurich, Zurich, Switzerland.,Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Felipe de Galiza Barbosa
- Departments of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.,University of Zurich, Zurich, Switzerland
| | - Geoffrey Warnock
- Departments of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.,University of Zurich, Zurich, Switzerland.,PMOD Technologies Ltd., Zurich, Switzerland
| | | | - Patrick Veit Haibach
- Departments of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.,University of Zurich, Zurich, Switzerland.,Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, Toronto, ON, Canada.,Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Martin Huellner
- Departments of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.,University of Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
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
|
17
|
Fournier L, de Geus-Oei LF, Regge D, Oprea-Lager DE, D’Anastasi M, Bidaut L, Bäuerle T, Lopci E, Cappello G, Lecouvet F, Mayerhoefer M, Kunz WG, Verhoeff JJC, Caruso D, Smits M, Hoffmann RT, Gourtsoyianni S, Beets-Tan R, Neri E, deSouza NM, Deroose CM, Caramella C. Twenty Years On: RECIST as a Biomarker of Response in Solid Tumours an EORTC Imaging Group - ESOI Joint Paper. Front Oncol 2022; 11:800547. [PMID: 35083155 PMCID: PMC8784734 DOI: 10.3389/fonc.2021.800547] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Response evaluation criteria in solid tumours (RECIST) v1.1 are currently the reference standard for evaluating efficacy of therapies in patients with solid tumours who are included in clinical trials, and they are widely used and accepted by regulatory agencies. This expert statement discusses the principles underlying RECIST, as well as their reproducibility and limitations. While the RECIST framework may not be perfect, the scientific bases for the anticancer drugs that have been approved using a RECIST-based surrogate endpoint remain valid. Importantly, changes in measurement have to meet thresholds defined by RECIST for response classification within thus partly circumventing the problems of measurement variability. The RECIST framework also applies to clinical patients in individual settings even though the relationship between tumour size changes and outcome from cohort studies is not necessarily translatable to individual cases. As reproducibility of RECIST measurements is impacted by reader experience, choice of target lesions and detection/interpretation of new lesions, it can result in patients changing response categories when measurements are near threshold values or if new lesions are missed or incorrectly interpreted. There are several situations where RECIST will fail to evaluate treatment-induced changes correctly; knowledge and understanding of these is crucial for correct interpretation. Also, some patterns of response/progression cannot be correctly documented by RECIST, particularly in relation to organ-site (e.g. bone without associated soft-tissue lesion) and treatment type (e.g. focal therapies). These require specialist reader experience and communication with oncologists to determine the actual impact of the therapy and best evaluation strategy. In such situations, alternative imaging markers for tumour response may be used but the sources of variability of individual imaging techniques need to be known and accounted for. Communication between imaging experts and oncologists regarding the level of confidence in a biomarker is essential for the correct interpretation of a biomarker and its application to clinical decision-making. Though measurement automation is desirable and potentially reduces the variability of results, associated technical difficulties must be overcome, and human adjudications may be required.
Collapse
Affiliation(s)
- Laure Fournier
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Université de Paris, Assistance Publique–Hôpitaux de Paris (AP-HP), Hopital europeen Georges Pompidou, Department of Radiology, Paris Cardiovascular Research Center (PARCC) Unité Mixte de Recherche (UMRS) 970, Institut national de la santé et de la recherche médicale (INSERM), Paris, France
| | - Lioe-Fee de Geus-Oei
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Daniele Regge
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Surgical Sciences, University of Turin, Turin, Italy
- Radiology Unit, Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-Istituto Di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Turin, Italy
| | - Daniela-Elena Oprea-Lager
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology & Nuclear Medicine, Cancer Centre Amsterdam, Amsterdam University Medical Centers [Vrije Universiteit (VU) University], Amsterdam, Netherlands
| | - Melvin D’Anastasi
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Medical Imaging Department, Mater Dei Hospital, University of Malta, Msida, Malta
| | - Luc Bidaut
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- College of Science, University of Lincoln, Lincoln, United Kingdom
| | - Tobias Bäuerle
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Egesta Lopci
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine Unit, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) – Humanitas Research Hospital, Milan, Italy
| | - Giovanni Cappello
- Department of Surgical Sciences, University of Turin, Turin, Italy
- Radiology Unit, Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-Istituto Di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Turin, Italy
| | - Frederic Lecouvet
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Marius Mayerhoefer
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang G. Kunz
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Joost J. C. Verhoeff
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Damiano Caruso
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Marion Smits
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
- Brain Tumour Centre, Erasmus Medical Centre (MC) Cancer Institute, Rotterdam, Netherlands
| | - Ralf-Thorsten Hoffmann
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Institute and Policlinic for Diagnostic and Interventional Radiology, University Hospital, Carl-Gustav-Carus Technical University Dresden, Dresden, Germany
| | - Sofia Gourtsoyianni
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, School of Medicine, National and Kapodistrian University of Athens, Areteion Hospital, Athens, Greece
| | - Regina Beets-Tan
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- School For Oncology and Developmental Biology (GROW) School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Emanuele Neri
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Diagnostic and Interventional Radiology, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Nandita M. deSouza
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Quantitative Imaging Biomarkers Alliance, Radiological Society of North America, Oak Brook, IL, United States
| | - Christophe M. Deroose
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Caroline Caramella
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Radiology Department, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph Centre International des Cancers Thoraciques, Université Paris-Saclay, Le Plessis-Robinson, France
| |
Collapse
|
18
|
Wang D, Li Y, Chen X, Li P. Prognostic significance of volume-based 18F-FDG PET/CT parameters and correlation with PD-L1 expression in patients with surgically resected lung adenocarcinoma. Medicine (Baltimore) 2021; 100:e27100. [PMID: 34477147 PMCID: PMC8415941 DOI: 10.1097/md.0000000000027100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 08/11/2021] [Indexed: 12/09/2022] Open
Abstract
The aim of this study was to retrospectively analyze 18F-FDG positron emission tomography/computed tomography (18F-FDG PET/CT) metabolic variables, programmed death-ligand 1 (PD-L1) and phosphorylated signal transducer and activator of transcription 3 (p-STAT3) tumor expression, and other factors as predictors of disease-free survival (DFS) in patients with lung adenocarcinoma (LUAD) (stage IA-IIIA) who underwent surgical resection. We still lack predictor of immune checkpoint (programmed cell death-1 [PD-1]/PD-L1) inhibitors. Herein, we investigated the correlation between metabolic parameters from 18F-FDG PET/CT and PD-L1 expression in patients with surgically resected LUAD.Seventy-four patients who underwent 18F-FDG PET/CT prior to treatment were consecutively enrolled. The main 18F-FDG PET/CT-derived variables were primary tumor maximum standardized uptake value (SUVmax), metabolic tumor volume (MTV), and total lesion glycolysis (TLG). Surgical tumor specimens were analyzed for PD-L1 and p-STAT3 expression using immunohistochemistry. Correlations between immunohistochemistry results and 18F-FDG PET/CT-derived variables were compared. Associations of PD-L1 and p-STAT3 tumor expression, 18F-FDG PET/CT-derived variables, and other factors with DFS in resected LUAD were evaluated.All tumors were FDG-avid. The cutoff values of low and high SUVmax, MTV, and TLG were 12.60, 14.87, and 90.85, respectively. The results indicated that TNM stage, PD-L1 positivity, and high 18F-FDG PET/CT metabolic volume parameters (TLG ≥90.85 or MTV ≥14.87) were independent predictors of worse DFS in resected LUAD. No 18F-FDG metabolic parameters associated with PD-L1 expression were observed (chi-square test), but we found that patients with positive PD-L1 expression have significantly higher SUVmax (P = .01), MTV (P = .00), and TLG (P = .00) than patients with negative PD-L1 expression.18F-FDG PET/CT metabolic volume parameters (TLG ≥90.85 or MTV ≥14.87) were more helpful in prognostication than the conventional parameter (SUVmax), PD-L1 expression was an independent predictor of DFS in patients with resected LUAD. Metabolic parameters on 18F-FDG PET/CT have a potential role for 18F-FDG PET/CT in selecting candidate LUAD for treatment with checkpoint inhibitors.
Collapse
Affiliation(s)
- Dalong Wang
- Department of PET/CT, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yingci Li
- Department of PET/CT, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Xiaolin Chen
- Department of PET/CT, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ping Li
- Department of PET/CT, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
19
|
Evaluation of Quantitative Ga-68 PSMA PET/CT Repeatability of Recurrent Prostate Cancer Lesions Using Both OSEM and Bayesian Penalized Likelihood Reconstruction Algorithms. Diagnostics (Basel) 2021; 11:diagnostics11061100. [PMID: 34208531 PMCID: PMC8233885 DOI: 10.3390/diagnostics11061100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 01/04/2023] Open
Abstract
Rationale: To formally determine the repeatability of Ga-68 PSMA lesion uptake in both relapsing and metastatic tumor. In addition, it was hypothesized that the BPL algorithm Q. Clear has the ability to lower SUV signal variability in the small lesions typically encountered in Ga-68 PSMA PET imaging of prostate cancer. Methods: Patients with biochemical recurrence of prostate cancer were prospectively enrolled in this single center pilot test-retest study and underwent two Ga-68 PSMA PET/CT scans within 7.9 days on average. Lesions were classified as suspected local recurrence, lymph node metastases or bone metastases. Two datasets were generated: one standard PSF + OSEM and one with PSF + BPL reconstruction algorithm. For tumor lesions, SUVmax was determined. Repeatability was formally assessed using Bland–Altman analysis for both BPL and standard reconstruction. Results: A total number of 65 PSMA-positive tumor lesions were found in 23 patients (range 1 to 12 lesions a patient). Overall repeatability in the 65 lesions was −1.5% ± 22.7% (SD) on standard reconstructions and −2.1% ± 29.1% (SD) on BPL reconstructions. Ga-68 PSMA SUVmax had upper and lower limits of agreement of +42.9% and −45.9% for standard reconstructions and +55.0% and −59.1% for BPL reconstructions, respectively (NS). Tumor SUVmax repeatability was dependent on lesion area, with smaller lesions exhibiting poorer repeatability on both standard and BPL reconstructions (F-test, p < 0.0001). Conclusion: A minimum response of 50% seems appropriate in this clinical situation. This is more than the recommended 30% for other radiotracers and clinical situations (PERCIST response criteria). BPL does not seem to lower signal variability in these cases.
Collapse
|
20
|
Sugita S, Yamato M, Hatabu T, Kataoka Y. Involvement of cancer-derived EMT cells in the accumulation of 18F-fluorodeoxyglucose in the hypoxic cancer microenvironment. Sci Rep 2021; 11:9668. [PMID: 33994540 PMCID: PMC8126561 DOI: 10.1038/s41598-021-88414-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
A high rate of glycolysis, one of the most common features of cancer, is used in positron emission tomography (PET) imaging to visualize tumor tissues using 18F-fluorodeoxyglucose (18F-FDG). Heterogeneous intratumoral distribution of 18F-FDG in tissues has been established in some types of cancer, and the maximum standardized uptake value (SUVmax) has been correlated with poor prognosis. However, the phenotype of cells that show high 18F-FDG accumulation in tumors remains unknown. Here, we combined quantitative micro-autoradiography with fluorescence immunohistochemistry to simultaneously visualize 18F-FDG distribution, the expression of multiple proteins, and hypoxic regions in the cancer microenvironment of a human A431 xenograft tumor in C.B-17/Icr-scid/scid mice. We found that the highest 18F-FDG accumulation was in cancer-derived cells undergoing epithelial-mesenchymal transition (EMT) in hypoxic regions, implicating these regions as a major contributor to increased glucose metabolism, as measured by 18F-FDG-PET.
Collapse
Affiliation(s)
- Sachi Sugita
- Laboratory of Animal Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama, Okayama, 700-8530, Japan.,Laboratory for Cellular Function Imaging, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Masanori Yamato
- Laboratory for Cellular Function Imaging, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.,Multi-Modal Microstructure Analysis Unit, RIKEN-JEOL Collaboration Center, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Toshimitsu Hatabu
- Laboratory of Animal Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama, Okayama, 700-8530, Japan
| | - Yosky Kataoka
- Laboratory for Cellular Function Imaging, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan. .,Multi-Modal Microstructure Analysis Unit, RIKEN-JEOL Collaboration Center, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| |
Collapse
|
21
|
Ferjančič P, Ebert MA, Francis R, Nowak AK, Jeraj R. Repeatability of Quantitative 18F-FET PET in Glioblastoma. Biomed Phys Eng Express 2021; 7. [PMID: 33887712 DOI: 10.1088/2057-1976/abfae9] [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: 12/03/2020] [Accepted: 04/22/2021] [Indexed: 12/16/2022]
Abstract
Purpose: O-(2-[18F]fluoroethyl)-L-tyrosine (FET), a PET radiotracer of amino acid uptake, has shown potential for diagnosis and treatment planning in patients with glioblastoma (GBM). To improve quantitative assessment of FET PET imaging, we evaluated the repeatability of uptake of this tracer in patients with GBM.Methods: Test-retest FET PET imaging was performed on 8 patients with histologically confirmed GBM, who previously underwent surgical resection of the tumour. Data were acquired according to the protocol of a prospective clinical trial validating FET PET as a clinical tool in GBM. SUVmean, SUVmaxand SUV98%metrics were extracted for both test and retest images and used to calculate 95% Bland-Altman limits of agreement (LoA) on lesion-level, as well as on volumes of varying sizes. Impact of healthy brain normalization on repeatability of lesion SUV metrics was evaluated.Results: Tumour LoA were [0.72, 1.46] for SUVmeanand SUVtotal, [0.79,1.23] for SUVmax, and [0.80,1.18] for SUV98%. Healthy brain LoA were [0.80,1.25] for SUVmean, [0.80,1.25] for SUVmax, and [0.81,1.23] for SUV98%. Voxel-level SUV LoA were [0.76, 1.32] for tumour volumes and [0.80, 1.25] for healthy brain. When sampled over maximum volume, SUV LoA were [0.90,1.12] for tumour and [0.92,1.08] for healthy brain. Normalization of uptake using healthy brain volumes was found to improve repeatability, but not after normalization volume size of about 15 cm3.Conclusions Advances in Knowledge and Implications for Patient Care: Repeatability of FET PET is comparable to existing tracers such as FDG and FLT. Healthy brain uptake is slightly more repeatable than uptake of tumour volumes. Repeatability was found to increase with sampled volume. SUV normalization between scans using healthy brain uptake should be performed using volumes at least 15 cm3in size to ensure best imaging repeatability.
Collapse
Affiliation(s)
- Peter Ferjančič
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Martin A Ebert
- Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, University of Western Australia, Crawley, Western Australia, Australia.,5D Clinics, Perth, Western Australia, Australia
| | - Roslyn Francis
- Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, University of Western Australia, Crawley, Western Australia, Australia
| | - Anna K Nowak
- Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, University of Western Australia, Crawley, Western Australia, Australia
| | - Robert Jeraj
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States of America.,Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Cho SY, Huff DT, Jeraj R, Albertini MR. FDG PET/CT for Assessment of Immune Therapy: Opportunities and Understanding Pitfalls. Semin Nucl Med 2020; 50:518-531. [PMID: 33059821 PMCID: PMC8201415 DOI: 10.1053/j.semnuclmed.2020.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immune checkpoint blockade has demonstrated the ability to modulate the immune system to produce durable responses in a wide range of cancers and has significantly impacted the standard of care. However, many cancer patients still do not respond to immune checkpoint blockade or have a limited duration of antitumor responses. Moreover, immune-related adverse events caused by immune checkpoint blockade can be severe and debilitating for some patients, limiting continuation of therapy and resulting in severe autoimmune conditions. Standard-of-care conventional anatomic imaging modalities and tumor response criteria have limitations to adequately assess tumor responses, especially early in the course of therapy, for risk-adapted clinical management to inform care of patients treated with immunotherapy. Molecular imaging with position emission tomography (PET) provides a noninvasive functional biomarker of tumor response, and of immune activation, for patients on immune-based therapies to help address these needs. 18F-FDG (FDG) PET/CT is readily available clinically and a number of studies have evaluated the use of this agent for assessment of prognosis, treatment response and immune activation for patients treated with immune checkpoint blockade. In this review paper, we discuss the current oncologic applications and imaging needs of cancer immunotherapy, recent studies applying FDG PET/CT for tumor response assessment, and evaluation of immune-related adverse events for improving clinical management. We largely focus on metastatic melanoma; however, we generalize where applicable to immunotherapy in other tumor types. We also briefly discuss PET imaging and quantitation as well as emerging non-FDG PET imaging radiotracers for cancer immunotherapy imaging.
Collapse
Affiliation(s)
- Steve Y Cho
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| | - Daniel T Huff
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Robert Jeraj
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Mark R Albertini
- University of Wisconsin Carbone Cancer Center, Madison, WI; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI; Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
| |
Collapse
|
25
|
Duan C, Chaovalitwongse WA, Bai F, Hippe DS, Wang S, Thammasorn P, Pierce LA, Liu X, You J, Miyaoka RS, Vesselle HJ, Kinahan PE, Rengan R, Zeng J, Bowen SR. Sensitivity analysis of FDG PET tumor voxel cluster radiomics and dosimetry for predicting mid-chemoradiation regional response of locally advanced lung cancer. Phys Med Biol 2020; 65:205007. [PMID: 33027064 PMCID: PMC7593986 DOI: 10.1088/1361-6560/abb0c7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We investigated the sensitivity of regional tumor response prediction to variability in voxel clustering techniques, imaging features, and machine learning algorithms in 25 patients with locally advanced non-small cell lung cancer (LA-NSCLC) enrolled on the FLARE-RT clinical trial. Metabolic tumor volumes (MTV) from pre-chemoradiation (PETpre) and mid-chemoradiation fluorodeoxyglucose-positron emission tomography (FDG PET) images (PETmid) were subdivided into K-means or hierarchical voxel clusters by standardized uptake values (SUV) and 3D-positions. MTV cluster separability was evaluated by CH index, and morphologic changes were captured by Dice similarity and centroid Euclidean distance. PETpre conventional features included SUVmean, MTV/MTV cluster size, and mean radiation dose. PETpre radiomics consisted of 41 intensity histogram and 3D texture features (PET Oncology Radiomics Test Suite) extracted from MTV or MTV clusters. Machine learning models (multiple linear regression, support vector regression, logistic regression, support vector machines) of conventional features or radiomic features were constructed to predict PETmid response. Leave-one-out-cross-validated root-mean-squared-error (RMSE) for continuous response regression (ΔSUVmean) and area-under-receiver-operating-characteristic-curve (AUC) for binary response classification were calculated. K-means MTV 2-clusters (MTVhi, MTVlo) achieved maximum CH index separability (Friedman p < 0.001). Between PETpre and PETmid, MTV cluster pairs overlapped (Dice 0.70-0.87) and migrated 0.6-1.1 cm. PETmid ΔSUVmean response prediction was superior in MTV and MTVlo (RMSE = 0.17-0.21) compared to MTVhi (RMSE = 0.42-0.52, Friedman p < 0.001). PETmid ΔSUVmean response class prediction performance trended higher in MTVlo (AUC = 0.83-0.88) compared to MTVhi (AUC = 0.44-0.58, Friedman p = 0.052). Models were more sensitive to MTV/MTV cluster regions (Friedman p = 0.026) than feature sets/algorithms (Wilcoxon signed-rank p = 0.36). Top-ranked radiomic features included GLZSM-LZHGE (large-zone-high-SUV), GTSDM-CP (cluster-prominence), GTSDM-CS (cluster-shade) and NGTDM-CNT (contrast). Top-ranked features were consistent between MTVhi and MTVlo cluster pairs but varied between MTVhi-MTVlo clusters, reflecting distinct regional radiomic phenotypes. Variability in tumor voxel cluster response prediction can inform robust radiomic target definition for risk-adaptive chemoradiation in patients with LA-NSCLC. FLARE-RT trial: NCT02773238.
Collapse
Affiliation(s)
- Chunyan Duan
- Department of Mechanical Engineering, Tongji University School of Mechanical Engineering, Shanghai China
- Department of Industrial Engineering, University of Arkansas College of Engineering, Fayetteville AR
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle WA
| | - W. Art Chaovalitwongse
- Department of Industrial Engineering, University of Arkansas College of Engineering, Fayetteville AR
| | - Fangyun Bai
- Department of Management Science and Engineering, Tongji University School of Economics and Management, Shanghai China
- Department of Industrial, Manufacturing, & Systems Engineering, University of Texas at Arlington College of Engineering, Arlington, TX
| | - Daniel S. Hippe
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| | - Shouyi Wang
- Department of Industrial, Manufacturing, & Systems Engineering, University of Texas at Arlington College of Engineering, Arlington, TX
| | - Phawis Thammasorn
- Department of Industrial Engineering, University of Arkansas College of Engineering, Fayetteville AR
| | - Larry A. Pierce
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| | - Xiao Liu
- Department of Industrial Engineering, University of Arkansas College of Engineering, Fayetteville AR
| | - Jianxin You
- Department of Management Science and Engineering, Tongji University School of Economics and Management, Shanghai China
| | - Robert S. Miyaoka
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| | - Hubert J. Vesselle
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| | - Paul E. Kinahan
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| | - Ramesh Rengan
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle WA
| | - Jing Zeng
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle WA
| | - Stephen R. Bowen
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle WA
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| |
Collapse
|
26
|
Derlin T, Gatidis S, Krause BJ, Antoch G, Kotzerke J, Pinto Dos Santos D, Eiber M, Weber MA, Giesel F, Pfannenberg C, Schlemmer HP, Persigehl T, Herrmann K, Umutlu L. [Structured reporting in oncologic hybrid imaging: a consensus recommendation]. Nuklearmedizin 2020; 59:288-293. [PMID: 32544954 DOI: 10.1055/a-1176-0275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Since the clinical introduction of PET/CT in the year of 2001 and PET/MRI in the year of 2010, hybrid imaging-guided precision medicine has become an important component of diagnostic algorithms in oncology. The written report represents the primary mode of communication between the referring physician and both the nuclear medicine physician and the radiologist. Reports have considerable impact on patient management and patient outcome, and serve as a legal documentation of the services provided and the expert impression of the interpreting physician. A high-quality hybrid imaging study should result in a likewise high-quality, structured written report which satisfactorily answers the clinical question of the referring physician. In this manuscript, consensus recommendations for structure and content of oncologic hybrid imaging reports and conclusive impressions are provided. Moreover, exemplary structured reports are provided. The recommendations for structured reporting provided in this document should foster further standardization and harmonization of oncologic reports in the context of hybrid imaging. They should also simplify communication with referring physicians and support both acceptance and appreciation of the clinical value of oncologic hybrid imaging. CITATION FORMAT: · Derlin T, Gatidis S, Krause BJ et al. Konsensusempfehlung zur strukturierten Befunderstellung onkologischer PET-Hybridbildgebung. Nuklearmedizin 2020; 59: DOI:10.1055/a-1176-0275.
Collapse
Affiliation(s)
- Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Sergios Gatidis
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany
| | - Bernd Joachim Krause
- Department of Nuclear Medicine, University Medical Center Rostock, Rostock, Germany
| | - Gerald Antoch
- Department of Diagnostic and Interventional Radiology, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Jörg Kotzerke
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Daniel Pinto Dos Santos
- Institute of Diagnostic and Interventional Radiology, University Hospital of Cologne, Cologne, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - Marc-André Weber
- Institute of Diagnostic and Interventional Radiology, Pediatric Radiology and Neuroradiology, University Medical Center Rostock, Rostock, Germany
| | - Frederik Giesel
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Christina Pfannenberg
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany
| | | | - Thorsten Persigehl
- Institute of Diagnostic and Interventional Radiology, University Hospital of Cologne, Cologne, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Lale Umutlu
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| |
Collapse
|
27
|
Hong IK, Lee JM, Hwang IK, Paik SS, Kim C, Lee SH. Diagnostic and Predictive Values of 18F-FDG PET/CT Metabolic Parameters in EGFR-Mutated Advanced Lung Adenocarcinoma. Cancer Manag Res 2020; 12:6453-6465. [PMID: 32801885 PMCID: PMC7396957 DOI: 10.2147/cmar.s259055] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/16/2020] [Indexed: 12/23/2022] Open
Abstract
Purpose The clinical implications of the metabolic parameters of 18F-fluorodeoxyglucose positron emission tomography-computed tomography (18F-FDG PET/CT) in epidermal growth factor receptor (EGFR)-mutated lung cancer are not fully understood. The aim of this study was to evaluate the diagnostic and prognostic utility of the parameters in EGFR-mutated lung cancer patients. Patients and Methods We retrospectively enrolled 134 patients with advanced lung adenocarcinoma (72 EGFR-negative and 62 EGFR-positive). We evaluated the correlation between EGFR mutational status and the maximum standardized uptake value (SUVmax), as well as the associations between treatment outcomes in EGFR-mutated patients and various metabolic parameters of primary tumors. For the best predictive parameters, we calculated the metabolic tumor volume (MTV) and total lesion glycolysis (TLG) using two SUV cutoffs: 1.5 (MTV1.5, TLG1.5) and 2.5 (MTV2.5, TLG2.5). Results Mean SUVmax was lower for EGFR-mutated tumors compared with EGFR wild-type (6.11 vs 10.41, p < 0.001) tumors. Low SUVmax was significantly associated with positive EGFR mutation (odds ratio = 1.74). Multivariate analysis for survival demonstrated that high MTV1.5, TLG1.5, MTV2.5, and TLG2.5 were independently associated with shorter progression-free survival (PFS) and overall survival (OS), and the highest hazard ratios were found in TLG1.5 (3.26 for PFS and 4.62 for OS). Conclusion SUVmax may be predictive for EGFR mutational status, and MTV and TLG of primary tumors may be promising prognostic parameters; 18F-FDG PET/CT has potential utility for the risk stratification of EGFR-mutated patients treated with targeted therapy.
Collapse
Affiliation(s)
- Il Ki Hong
- Department of Nuclear Medicine, Kyung Hee University Medical Center, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Jeong Mi Lee
- Department of Internal Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - In Kyoung Hwang
- Department of Internal Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Seung Sook Paik
- Department of Internal Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Chanwoo Kim
- Department of Nuclear Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Seung Hyeun Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Kyung Hee University Medical Center, Kyung Hee University School of Medicine, Seoul, South Korea
| |
Collapse
|
28
|
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
|
29
|
Schmall JP, Surti S, Otero HJ, Servaes S, Karp JS, States LJ. Investigating Low-Dose Image Quality in Whole-Body Pediatric 18F-FDG Scans Using Time-of-Flight PET/MRI. J Nucl Med 2020; 62:123-130. [DOI: 10.2967/jnumed.119.240127] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022] Open
|
30
|
Korbin S, Salerno M, Achonu JU, Huang M, Vaska P, Pawlak A, Komatsu DE, Paci JM. PET/MRI reveals ongoing metabolic activity in ACL grafts one year post-ACL reconstruction. J Exp Orthop 2020; 7:40. [PMID: 32483664 PMCID: PMC7264087 DOI: 10.1186/s40634-020-00258-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/25/2020] [Indexed: 12/29/2022] Open
Abstract
Purpose To use serial PET/MRI imaging to radiographically evaluate the metabolic activity of the ACL graft over the first post-operative year. Methods Six patients undergoing primary ACL reconstruction were recruited in this prospective study in an inpatient university hospital. All patients underwent femoral and tibial suspensory cortical fixation with quadrupled semitendinosus autograft hamstring ACL reconstruction by an orthopaedic surgeon. Simultaneous 18F-FDG PET and MRI of both the operative and non-operative knee was performed at three, six, and 12 months post-operatively. Quantification of the mean standardized uptake value (SUV) within the whole-knee, as well as tibial tunnel, femoral tunnel, and intra-articular graft regions of interest (ROIs). Results PET whole-knee activity was increased at all time-points post-operatively compared to the control, non-operative knee. Activity decreased over time, yet considerable generalized activity remained 1 year post-operatively, with relative intensity 34% percent higher than control. When the operative knee was divided into three whole-regions, there was greater activity in the tibia at three than 12 months, the femur at six than 12 months, and in the tibia compared to the intra-articular region at 3 months. When they were separated into sub-regions, results demonstrated greater activity closer to the joint surface. Conclusions PET/MRI evaluation of ACL graft reconstructions demonstrates evolving biologic activity within the graft and both tunnels. Focal areas of increased activity within the tunnels may indicate of ligamento-osseous morphologic changes. These data suggest that graft incorporation continues well beyond 1 year post-operatively. Level of evidence Level IV.
Collapse
Affiliation(s)
- Seth Korbin
- Department of Orthopaedics, Stony Brook University Hospital, Stony Brook, New York, USA
| | - Michael Salerno
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Justice U Achonu
- Department of Orthopaedics, Stony Brook University Hospital, Stony Brook, New York, USA
| | - Mingqian Huang
- Department of Radiology, Stony Brook University Hospital, Stony Brook, New York, USA
| | - Paul Vaska
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Amanda Pawlak
- Department of Orthopaedics, Stony Brook University Hospital, Stony Brook, New York, USA
| | - David E Komatsu
- Department of Orthopaedics, Stony Brook University Hospital, Stony Brook, New York, USA.
| | - James M Paci
- Orlin & Cohen Orthopedic Group, Zucker School of Medicine at Hofstra/Northwell School of Medicine, Hempstead, New York, USA
| |
Collapse
|
31
|
Wangaryattawanich P, Branstetter BF, Ly JD, Duvvuri U, Heron DE, Rath TJ. Positive Predictive Value of Neck Imaging Reporting and Data System Categories 3 and 4 Posttreatment FDG-PET/CT in Head and Neck Squamous Cell Carcinoma. AJNR Am J Neuroradiol 2020; 41:1070-1075. [PMID: 32467187 DOI: 10.3174/ajnr.a6589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/04/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE The Neck Imaging Reporting and Data System is a standardized reporting system intended to risk stratify patients treated for head and neck squamous cell carcinoma. The purpose of this study is to investigate the positive predictive value of the Neck Imaging Reporting and Data System categories 3 and 4 on posttreatment PET/CT in patients treated definitively for head and neck squamous cell carcinoma. MATERIALS AND METHODS We retrospectively identified patients treated definitively for head and neck squamous cell carcinoma between 2006 and 2018. Patients whose posttreatment PET/CT scans were interpreted as Neck Imaging Reporting and Data System 3 (suspicious) or 4 (definitive recurrence) at the primary site, regional nodes, or at distant sites were included. The reference standard was histopathology or unequivocal imaging or clinical evidence of treatment failure. The positive predictive values of Neck Imaging Reporting and Data System 3 and 4 posttreatment PET/CT were calculated. RESULTS Seventy-two of 128 patients with posttreatment PET/CT interpreted as Neck Imaging Reporting and Data System 3 at the primary site, regional nodes, or distant sites were proved to have treatment failure at the suspicious sites, yielding an overall positive predictive value of 56% (95% CI, 48%-65%). The positive predictive values of Neck Imaging Reporting and Data System 3 by subsite were as follows: primary site, 56% (44/79); regional nodes, 65% (34/52); and distant sites, 79% (42/53). All 69 patients with posttreatment PET/CT interpreted as Neck Imaging Reporting and Data System 4 had true treatment failure, yielding a positive predictive value of 100% (95% CI, 96%-100%): primary site, 100% (28/28); regional nodes, 100% (32/32); and distant sites, 100% (29/29). CONCLUSIONS The positive predictive value of Neck Imaging Reporting and Data System 3 on posttreatment PET/CT is relatively low. Thus, Neck Imaging Reporting and Data System 3 findings should be confirmed with tissue sampling before instituting new salvage treatment regimens to avoid unnecessary overtreatment and its associated toxicities. Neck Imaging Reporting and Data System 4 reliably indicates recurrent disease.
Collapse
Affiliation(s)
| | - B F Branstetter
- From the Departments of Radiology (P.W., B.F.B., J.D.L.).,Otolaryngology (B.F.B., U.D., D.E.H.).,Biomedical Informatics (B.F.B.)
| | - J D Ly
- From the Departments of Radiology (P.W., B.F.B., J.D.L.)
| | - U Duvvuri
- Otolaryngology (B.F.B., U.D., D.E.H.)
| | - D E Heron
- Otolaryngology (B.F.B., U.D., D.E.H.).,Radiation Oncology (D.E.H.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - T J Rath
- Neuroradiology Section, Department of Radiology (T.J.R.), Mayo Clinic Hospital, Phoenix, Arizona.
| |
Collapse
|
32
|
Kinahan PE, Perlman ES, Sunderland JJ, Subramaniam R, Wollenweber SD, Turkington TG, Lodge MA, Boellaard R, Obuchowski NA, Wahl RL. The QIBA Profile for FDG PET/CT as an Imaging Biomarker Measuring Response to Cancer Therapy. Radiology 2020; 294:647-657. [PMID: 31909700 PMCID: PMC7053216 DOI: 10.1148/radiol.2019191882] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/15/2019] [Accepted: 11/04/2019] [Indexed: 01/22/2023]
Abstract
The Quantitative Imaging Biomarkers Alliance (QIBA) Profile for fluorodeoxyglucose (FDG) PET/CT imaging was created by QIBA to both characterize and reduce the variability of standardized uptake values (SUVs). The Profile provides two complementary claims on the precision of SUV measurements. First, tumor glycolytic activity as reflected by the maximum SUV (SUVmax) is measurable from FDG PET/CT with a within-subject coefficient of variation of 10%-12%. Second, a measured increase in SUVmax of 39% or more, or a decrease of 28% or more, indicates that a true change has occurred with 95% confidence. Two applicable use cases are clinical trials and following individual patients in clinical practice. Other components of the Profile address the protocols and conformance standards considered necessary to achieve the performance claim. The Profile is intended for use by a broad audience; applications can range from discovery science through clinical trials to clinical practice. The goal of this report is to provide a rationale and overview of the FDG PET/CT Profile claims as well as its context, and to outline future needs and potential developments.
Collapse
Affiliation(s)
- Paul E. Kinahan
- From the Department of Radiology, University of Washington, 1959 NE
Pacific St, RR215, Box 357115, Seattle, WA 98195-7117 (P.E.K.); Perlman Advisory
Group, LLC, Hillsdale, NY (E.S.P.); Department of Radiology, University of Iowa,
Iowa City, Iowa (J.J.S.); Department of Radiology, University of Texas
Southwestern, Dallas, Tex (R.S.); GE Healthcare, Waukesha, Wis (S.D.W.);
Department of Radiology, Duke University Medical Center, Durham, NC (T.G.T.);
The Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University, Baltimore, Md (M.A.L.); Department of Radiology and Nuclear
Medicine, Amsterdam, the Netherlands (R.B.); Quantitative Health Sciences,
Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Mallinckrodt
Institute of Radiology, Washington University School of Medicine, St Louis, Mo
(R.L.W.)
| | - Eric S. Perlman
- From the Department of Radiology, University of Washington, 1959 NE
Pacific St, RR215, Box 357115, Seattle, WA 98195-7117 (P.E.K.); Perlman Advisory
Group, LLC, Hillsdale, NY (E.S.P.); Department of Radiology, University of Iowa,
Iowa City, Iowa (J.J.S.); Department of Radiology, University of Texas
Southwestern, Dallas, Tex (R.S.); GE Healthcare, Waukesha, Wis (S.D.W.);
Department of Radiology, Duke University Medical Center, Durham, NC (T.G.T.);
The Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University, Baltimore, Md (M.A.L.); Department of Radiology and Nuclear
Medicine, Amsterdam, the Netherlands (R.B.); Quantitative Health Sciences,
Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Mallinckrodt
Institute of Radiology, Washington University School of Medicine, St Louis, Mo
(R.L.W.)
| | - John J. Sunderland
- From the Department of Radiology, University of Washington, 1959 NE
Pacific St, RR215, Box 357115, Seattle, WA 98195-7117 (P.E.K.); Perlman Advisory
Group, LLC, Hillsdale, NY (E.S.P.); Department of Radiology, University of Iowa,
Iowa City, Iowa (J.J.S.); Department of Radiology, University of Texas
Southwestern, Dallas, Tex (R.S.); GE Healthcare, Waukesha, Wis (S.D.W.);
Department of Radiology, Duke University Medical Center, Durham, NC (T.G.T.);
The Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University, Baltimore, Md (M.A.L.); Department of Radiology and Nuclear
Medicine, Amsterdam, the Netherlands (R.B.); Quantitative Health Sciences,
Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Mallinckrodt
Institute of Radiology, Washington University School of Medicine, St Louis, Mo
(R.L.W.)
| | - Rathan Subramaniam
- From the Department of Radiology, University of Washington, 1959 NE
Pacific St, RR215, Box 357115, Seattle, WA 98195-7117 (P.E.K.); Perlman Advisory
Group, LLC, Hillsdale, NY (E.S.P.); Department of Radiology, University of Iowa,
Iowa City, Iowa (J.J.S.); Department of Radiology, University of Texas
Southwestern, Dallas, Tex (R.S.); GE Healthcare, Waukesha, Wis (S.D.W.);
Department of Radiology, Duke University Medical Center, Durham, NC (T.G.T.);
The Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University, Baltimore, Md (M.A.L.); Department of Radiology and Nuclear
Medicine, Amsterdam, the Netherlands (R.B.); Quantitative Health Sciences,
Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Mallinckrodt
Institute of Radiology, Washington University School of Medicine, St Louis, Mo
(R.L.W.)
| | - Scott D. Wollenweber
- From the Department of Radiology, University of Washington, 1959 NE
Pacific St, RR215, Box 357115, Seattle, WA 98195-7117 (P.E.K.); Perlman Advisory
Group, LLC, Hillsdale, NY (E.S.P.); Department of Radiology, University of Iowa,
Iowa City, Iowa (J.J.S.); Department of Radiology, University of Texas
Southwestern, Dallas, Tex (R.S.); GE Healthcare, Waukesha, Wis (S.D.W.);
Department of Radiology, Duke University Medical Center, Durham, NC (T.G.T.);
The Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University, Baltimore, Md (M.A.L.); Department of Radiology and Nuclear
Medicine, Amsterdam, the Netherlands (R.B.); Quantitative Health Sciences,
Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Mallinckrodt
Institute of Radiology, Washington University School of Medicine, St Louis, Mo
(R.L.W.)
| | - Timothy G. Turkington
- From the Department of Radiology, University of Washington, 1959 NE
Pacific St, RR215, Box 357115, Seattle, WA 98195-7117 (P.E.K.); Perlman Advisory
Group, LLC, Hillsdale, NY (E.S.P.); Department of Radiology, University of Iowa,
Iowa City, Iowa (J.J.S.); Department of Radiology, University of Texas
Southwestern, Dallas, Tex (R.S.); GE Healthcare, Waukesha, Wis (S.D.W.);
Department of Radiology, Duke University Medical Center, Durham, NC (T.G.T.);
The Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University, Baltimore, Md (M.A.L.); Department of Radiology and Nuclear
Medicine, Amsterdam, the Netherlands (R.B.); Quantitative Health Sciences,
Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Mallinckrodt
Institute of Radiology, Washington University School of Medicine, St Louis, Mo
(R.L.W.)
| | - Martin A. Lodge
- From the Department of Radiology, University of Washington, 1959 NE
Pacific St, RR215, Box 357115, Seattle, WA 98195-7117 (P.E.K.); Perlman Advisory
Group, LLC, Hillsdale, NY (E.S.P.); Department of Radiology, University of Iowa,
Iowa City, Iowa (J.J.S.); Department of Radiology, University of Texas
Southwestern, Dallas, Tex (R.S.); GE Healthcare, Waukesha, Wis (S.D.W.);
Department of Radiology, Duke University Medical Center, Durham, NC (T.G.T.);
The Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University, Baltimore, Md (M.A.L.); Department of Radiology and Nuclear
Medicine, Amsterdam, the Netherlands (R.B.); Quantitative Health Sciences,
Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Mallinckrodt
Institute of Radiology, Washington University School of Medicine, St Louis, Mo
(R.L.W.)
| | - Ronald Boellaard
- From the Department of Radiology, University of Washington, 1959 NE
Pacific St, RR215, Box 357115, Seattle, WA 98195-7117 (P.E.K.); Perlman Advisory
Group, LLC, Hillsdale, NY (E.S.P.); Department of Radiology, University of Iowa,
Iowa City, Iowa (J.J.S.); Department of Radiology, University of Texas
Southwestern, Dallas, Tex (R.S.); GE Healthcare, Waukesha, Wis (S.D.W.);
Department of Radiology, Duke University Medical Center, Durham, NC (T.G.T.);
The Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University, Baltimore, Md (M.A.L.); Department of Radiology and Nuclear
Medicine, Amsterdam, the Netherlands (R.B.); Quantitative Health Sciences,
Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Mallinckrodt
Institute of Radiology, Washington University School of Medicine, St Louis, Mo
(R.L.W.)
| | - Nancy A. Obuchowski
- From the Department of Radiology, University of Washington, 1959 NE
Pacific St, RR215, Box 357115, Seattle, WA 98195-7117 (P.E.K.); Perlman Advisory
Group, LLC, Hillsdale, NY (E.S.P.); Department of Radiology, University of Iowa,
Iowa City, Iowa (J.J.S.); Department of Radiology, University of Texas
Southwestern, Dallas, Tex (R.S.); GE Healthcare, Waukesha, Wis (S.D.W.);
Department of Radiology, Duke University Medical Center, Durham, NC (T.G.T.);
The Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University, Baltimore, Md (M.A.L.); Department of Radiology and Nuclear
Medicine, Amsterdam, the Netherlands (R.B.); Quantitative Health Sciences,
Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Mallinckrodt
Institute of Radiology, Washington University School of Medicine, St Louis, Mo
(R.L.W.)
| | - Richard L. Wahl
- From the Department of Radiology, University of Washington, 1959 NE
Pacific St, RR215, Box 357115, Seattle, WA 98195-7117 (P.E.K.); Perlman Advisory
Group, LLC, Hillsdale, NY (E.S.P.); Department of Radiology, University of Iowa,
Iowa City, Iowa (J.J.S.); Department of Radiology, University of Texas
Southwestern, Dallas, Tex (R.S.); GE Healthcare, Waukesha, Wis (S.D.W.);
Department of Radiology, Duke University Medical Center, Durham, NC (T.G.T.);
The Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University, Baltimore, Md (M.A.L.); Department of Radiology and Nuclear
Medicine, Amsterdam, the Netherlands (R.B.); Quantitative Health Sciences,
Cleveland Clinic Foundation, Cleveland, Ohio (N.A.O.); and Mallinckrodt
Institute of Radiology, Washington University School of Medicine, St Louis, Mo
(R.L.W.)
| |
Collapse
|
33
|
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
|
34
|
Qualitative and quantitative reproducibility of [18]fluoromethycholine PET/computed tomography in prostate cancer. Nucl Med Commun 2020; 41:147-154. [PMID: 31939917 DOI: 10.1097/mnm.0000000000001129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Recurrence occurs in more than 50% of prostate cancer. To be effective, treatments require precise localization of tumor cells. [F]fluoromethylcholine ([18F]FCH) PET/computed tomography (CT) is currently used to restage disease in cases of biochemical relapse. To be used for therapy response as has been suggested, repeatability limits of PET derived indices need to be established. OBJECTIVE The aim of our study was to prospectively assess the qualitative and quantitative reproducibility [18F]FCH PET/CT in prostate cancer. METHODS Patients with histologically proven prostate cancer referred for initial staging or restaging were prospectively included. All patients underwent two [18F]FCH PET/CTs in the same conditions within a maximum of 3 weeks' time. We studied the repeatability of the visual report and the repeatability of SUVmax and its evolution over the acquisition time in lesions, liver and vascular background. Statistical analysis was performed using the Bland-Altman approach. RESULTS Twenty-one patients were included. Reporting repeatability was excellent with 97.8% of concordance. Mean repeatability of SUVmax considering all times and all lesions was 2.2% ± 20. Evolution of SUVmax was unpredictable, either increasing or decreasing over the acquisition time, both for lesions and for physiological activity. CONCLUSION Our study demonstrated that visual report of [18F]FCH PET/CT was very reproducible and that the repeatability limits of SUVmax was similar to those of other PET radiotracers. An SUVmax difference of more than 40% should be considered as representing a treatment response effect. Change of SUVmax during the acquisition time varied and should not be considered as an interpretation criterion.
Collapse
|
35
|
Laffon E, Marthan R. Prognostic value of 18F-FDG-PET in patients with mantle cell lymphoma: results from the LyMa-PET Project. Haematologica 2020; 105:e41. [PMID: 31894098 DOI: 10.3324/haematol.2019.236869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Eric Laffon
- CHU de Bordeaux, Bordeaux .,Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux.,INSERM U-1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France
| | - Roger Marthan
- CHU de Bordeaux, Bordeaux.,Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux.,INSERM U-1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France
| |
Collapse
|
36
|
Koopman D, Jager PL, Slump CH, Knollema S, van Dalen JA. SUV variability in EARL-accredited conventional and digital PET. EJNMMI Res 2019; 9:106. [PMID: 31823097 PMCID: PMC6904705 DOI: 10.1186/s13550-019-0569-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 10/16/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND A high SUV-reproducibility is crucial when different PET scanners are in use. We evaluated the SUV variability in whole-body FDG-PET scans of patients with suspected or proven cancer using an EARL-accredited conventional and digital PET scanner. In a head-to-head comparison we studied images of 50 patients acquired on a conventional scanner (cPET, Ingenuity TF PET/CT, Philips) and compared them with images acquired on a digital scanner (dPET, Vereos PET/CT, Philips). The PET scanning order was randomised and EARL-compatible reconstructions were applied. We measured SUVmean, SUVpeak, SUVmax and lesion diameter in up to 5 FDG-positive lesions per patient. The relative difference ΔSUV between cPET and dPET was calculated for each SUV-parameter. Furthermore, we calculated repeatability coefficients, reflecting the 95% confidence interval of ΔSUV. RESULTS We included 128 lesions with an average size of 19 ± 14 mm. Average ΔSUVs were 6-8% with dPET values being higher for all three SUV-parameters (p < 0.001). ΔSUVmax was significantly higher than ΔSUVmean (8% vs. 6%, p = 0.002) and than ΔSUVpeak (8% vs. 7%, p = 0.03). Repeatability coefficients across individual lesions were 27% (ΔSUVmean and ΔSUVpeak) and 33% (ΔSUVmax) (p < 0.001). CONCLUSIONS With EARL-accredited conventional and digital PET, we found a limited SUV variability with average differences up to 8%. Furthermore, only a limited number of lesions showed a SUV difference of more than 30%. These findings indicate that EARL standardisation works. TRIAL REGISTRATION This prospective study was registered on the 31th of October 2017 at ClinicalTrials.cov. URL: https://clinicaltrials.gov/ct2/show/NCT03457506?id=03457506&rank=1.
Collapse
Affiliation(s)
- Daniëlle Koopman
- Department of Nuclear Medicine, Isala, Dokter van Heesweg 2, 8025, AB, Zwolle, the Netherlands. .,Technical Medicine Center, University of Twente, Enschede, the Netherlands.
| | - Pieter L Jager
- Department of Nuclear Medicine, Isala, Dokter van Heesweg 2, 8025, AB, Zwolle, the Netherlands
| | - Cornelis H Slump
- Technical Medicine Center, University of Twente, Enschede, the Netherlands
| | - Siert Knollema
- Department of Nuclear Medicine, Isala, Dokter van Heesweg 2, 8025, AB, Zwolle, the Netherlands
| | | |
Collapse
|
37
|
Mendoza DP, Digumarthy SR. The added value of quantitative 18F-FDG-PET/CT parameters in the assessment of pulmonary lymphangitic carcinomatosis in lung cancer. J Thorac Dis 2019; 11:E239-E242. [PMID: 31903292 DOI: 10.21037/jtd.2019.10.10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Dexter P Mendoza
- Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Subba R Digumarthy
- Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
38
|
Saadani H, van der Hiel B, Aalbersberg EA, Zavrakidis I, Haanen JBAG, Hoekstra OS, Boellaard R, Stokkel MPM. Metabolic Biomarker-Based BRAFV600 Mutation Association and Prediction in Melanoma. J Nucl Med 2019; 60:1545-1552. [PMID: 31481581 DOI: 10.2967/jnumed.119.228312] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022] Open
Abstract
The aim of this study was to associate and predict B-rapidly accelerated fibrosarcoma valine 600 (BRAFV600) mutation status with both conventional and radiomics 18F-FDG PET/CT features, while exploring several methods of feature selection in melanoma radiomics. Methods: Seventy unresectable stage III-IV melanoma patients who underwent a baseline 18F-FDG PET/CT scan were identified. Patients were assigned to the BRAFV600 group or BRAF wild-type group according to mutational status. 18F-FDG uptake quantification was performed by semiautomatic lesion delineation. Four hundred eighty radiomics features and 4 conventional PET features (SUVmax, SUVmean, SUVpeak, and total lesion glycolysis) were extracted per lesion. Six different methods of feature selection were implemented, and 10-fold cross-validated predictive models were built for each. Model performances were evaluated with areas under the curve (AUCs) for the receiver operating characteristic curves. Results: Thirty-five BRAFV600 mutated patients (100 lesions) and 35 BRAF wild-type patients (79 lesions) were analyzed. AUCs predicting the BRAFV600 mutation varied from 0.54 to 0.62 and were susceptible to feature selection method. The best AUCs were achieved by feature selection based on literature, a penalized binary logistic regression model, and random forest model. No significant difference was found between the BRAFV600 and BRAF wild-type group in conventional PET features or predictive value. Conclusion: BRAFV600 mutation status is not associated with, nor can it be predicted with, conventional PET features, whereas radiomics features were of low predictive value (AUC = 0.62). We showed feature selection methods to influence predictive model performance, describing and evaluating 6 unique methods. Detecting BRAFV600 status in melanoma based on 18F-FDG PET/CT alone does not yet provide clinically relevant knowledge.
Collapse
Affiliation(s)
- Hanna Saadani
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bernies van der Hiel
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Else A Aalbersberg
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ioannis Zavrakidis
- Department of Epidemiology and Biostatistics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands; and
| | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Marcel P M Stokkel
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
39
|
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
|
40
|
Lu T, Zhan C, Huang Y, Zhao M, Yang X, Ge D, Shi Y, Wang Q. Small pulmonary granuloma is often misdiagnosed as lung cancer by positron emission tomography/computer tomography in diabetic patients. Interact Cardiovasc Thorac Surg 2019; 28:394-398. [PMID: 30165660 DOI: 10.1093/icvts/ivy263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/21/2018] [Accepted: 07/28/2018] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES A small pulmonary granuloma (SPG) is often misdiagnosed as lung cancer in diabetic patients by positron emission tomography/computed tomography (PET/CT). The present study was conducted to investigate whether diabetes is the influencing factor and to determine other related factors that have an impact on the diagnostic results following PET/CT examination. METHODS All clinical, imaging and pathological data of patients diagnosed with pulmonary nodules by PET/CT from January 2004 to December 2017 in our department were collected. Patients with an SPG who were wrongly diagnosed with lung cancer by PET/CT were enrolled (n = 79). The propensity score matching method was used to create a comparable control adenocarcinoma group (n = 395). Maximum standard uptake values, diabetes and fasting blood-glucose (FBG) were determined and analysed. RESULTS The average maximum standard uptake values in the 2 groups were comparable (P = 0.801). Maximum standard uptake values in 5 subsections were not significantly different between the 2 groups (P = 0.135). The odds ratio (OR) of 3.326 [95% confidence interval (CI) 1.671-6.623] for diabetes favoured misdiagnosis and was statistically significant (P < 0.001). Furthermore, in patients with high FBG levels (≥7.0 mmol/l), the risk of misdiagnosis of SPG increased significantly compared with normal FBG level (OR 2.601, 95% CI 1.174-5.761; P = 0.015). CONCLUSIONS Diabetes and high FBG level were the influencing factors in the false-positive results of lung cancer by PET/CT examination.
Collapse
Affiliation(s)
- Tao Lu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaodong Yang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Di Ge
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Letellier A, Johnson AC, Kit NH, Savigny JF, Batalla A, Parienti JJ, Aide N. Uptake of Radium-223 Dichloride and Early [ 18F]NaF PET Response Are Driven by Baseline [ 18F]NaF Parameters: a Pilot Study in Castration-Resistant Prostate Cancer Patients. Mol Imaging Biol 2019; 20:482-491. [PMID: 29027074 DOI: 10.1007/s11307-017-1132-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE The purpose of this study is to identify predictive factors on baseline [18F]NaF positron emission tomography (PET)/computed tomography (CT) of early response to radium-223 dichloride after 3 cycles of treatment in metastatic castration-resistant prostate cancer patients. PROCEDURES Analysis of 152 metastases was performed in six consecutive patients who underwent [18F]NaF PET/CT at baseline and for early monitoring after 3 cycles of radium-223 dichloride. All metastases depicted on whole-body [18F]NaF PET/CT were contoured and CT (density in Hounsfield units, sclerotic, mixed, or lytic appearance) as well as [18F]NaF [maximum standardized uptake value (SUVmax), SUVmean, and lesion volume (V18F-NaF)] patterns were recorded. Tumor response was defined as percentage change in SUVmax and SUVmean between baseline and post-treatment PET. Bone lesions were defined as stable, responsive, or progressive, according to thresholds derived from a recent multicentre test-retest study in [18F]NaF PET/CT. Total [18F]NaF uptake in metastases, defined as MATV × SUVmean, was correlated to uptake of radium-223 on biodistribution scintigraphy performed 7 days after the first cycle of treatment. RESULTS Among metastases, 116 involved the axial skeleton and 36 the appendicular skeleton. Lesions were sclerotic in 126 cases and mixed in 26 cases. No lytic lesion was depicted. ROC analysis showed that SUVmax and SUVmean were better predictors of lesion response than V18F-NaF and density on CT (P < 0.0001 and P = 0.001, respectively). SUVmax and SUVmean were predictors of individual tumor response in separate multivariate models (P = 0.01 and P = 0.02, respectively). CT pattern (mixed versus sclerotic) and lesion density were independent predictors only when assessing response with delta SUVmax (P = 0.002 and 0.007, respectively). A good correlation between total [18F]NaF uptake within metastases and their relative radium-223 uptake assessed by two observers 7 days after treatment (r = 0.72 and 0.77, P < 0.0001) was found. CONCLUSIONS SUVmax and SUVmean on baseline [18F]NaF PET/CT are independent predictors of bone lesions' response to 3 cycles of radium-223 dichloride, supporting the use of NaF to select patients more likely to respond to treatment.
Collapse
Affiliation(s)
- Arthur Letellier
- Nuclear Medicine Department, Caen University Hospital, 14000, Caen, France.,Radiology Department, University Hospital, Caen, France
| | | | - Nicolas How Kit
- Nuclear Medicine Department, Caen University Hospital, 14000, Caen, France.,Radiology Department, University Hospital, Caen, France
| | | | - Alain Batalla
- Medical Physics, François Baclesse Cancer Centre, Caen, France
| | - Jean-Jacques Parienti
- Clinical Research Unit, University Hospital, Caen, France.,EA2656 (GRAM 2.0), Normandie University, Caen, France
| | - Nicolas Aide
- Nuclear Medicine Department, Caen University Hospital, 14000, Caen, France. .,INSERM 1199 ANTICIPE, Normandie University, Caen, France.
| |
Collapse
|
42
|
Reinert CP, Schuhmann MU, Bender B, Gugel I, la Fougère C, Schäfer J, Gatidis S. Comprehensive anatomical and functional imaging in patients with type I neurofibromatosis using simultaneous FDG-PET/MRI. Eur J Nucl Med Mol Imaging 2018; 46:776-787. [DOI: 10.1007/s00259-018-4227-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/25/2018] [Indexed: 12/31/2022]
|
43
|
Ferretti A, Chondrogiannis S, Rampin L, Bellan E, Marzola MC, Grassetto G, Gusella S, Maffione AM, Gava M, Rubello D. How to harmonize SUVs obtained by hybrid PET/CT scanners with and without point spread function correction. Phys Med Biol 2018; 63:235010. [PMID: 30474620 DOI: 10.1088/1361-6560/aaee27] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
State of the art point-spread function (PSF) corrections implemented in positron emission tomography/computed tomography (PET/CT) reconstruction improved image quality and diagnostic performance but caused an increase in the standardized uptake value (SUV) compared to a conventional OSEM reconstruction system. The EANM suggested one produce two reconstructions, one optimised for maximum lesion detection and one for semi-quantitative analysis. In this work we investigated an alternative methodology, using a single reconstruction data set together with a post-reconstruction algorithm for SUV harmonization. Data acquisition was performed on a Siemens Biograph mCT system equipped with lutetium oxyorthosilicat crystals, PSF and time-of-flight algorithms and on a General Electric Discovery STE system equipped with BGO crystals. Both a EANM double reconstruction method and a dedicated post-reconstruction algorithm (marketed as EQ-filter) were tested to harmonize the quantitative values of the two PET/CT scanners. For phantom measurements we used a NEMA IQ phantom and a Jaszczak cylindrical phantom equipped with small spheres (lesion to background ratios of 8:1 and 4:1). Several different reconstruction settings were tested in order to provide a general methodology. Data obtained by phantom measurements were validated on seven oncologic patients who performed a one-bed extra acquisition on a different scanner. The evaluation regarded 39 small lesions (diameters: 0.3-2.6 cm) and was performed by two experienced nuclear medicine physicians. The SUV recoveries measured with the PSF reconstruction exceeded those obtained by the OSEM reconstruction with deviations ranging from 16% to 150%. These discrepancies resulted below 7% applying the optimized value of the EQ.filter or the double-reconstruction methods. For each reconstruction setting the optimal value of the EQ.filter was identified in order to minimize these discrepancies. Patient data, analyzed by Wilcoxon statistical test, confirmed and validated phantom measurements. EQ.filter can harmonize SUV values between different PET/CT scanners using a single reconstruction optimized to maximum lesion detectability. In this way, the second reconstruction proposed by EANM/EARL is avoided.
Collapse
Affiliation(s)
- Alice Ferretti
- Medical Physics Unit, Santa Maria della Misericordia Hospital, Rovigo, Italy. Author to whom any correspondence should be addressed. These authors equally contributed to the preparation of the study
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Kurland BF, Peterson LM, Shields AT, Lee JH, Byrd DW, Novakova-Jiresova A, Muzi M, Specht JM, Mankoff DA, Linden HM, Kinahan PE. Test-Retest Reproducibility of 18F-FDG PET/CT Uptake in Cancer Patients Within a Qualified and Calibrated Local Network. J Nucl Med 2018; 60:608-614. [PMID: 30361381 DOI: 10.2967/jnumed.118.209544] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 10/01/2018] [Indexed: 11/16/2022] Open
Abstract
Calibration and reproducibility of quantitative 18F-FDG PET measures are essential for adopting integral 18F-FDG PET/CT biomarkers and response measures in multicenter clinical trials. We implemented a multicenter qualification process using National Institute of Standards and Technology-traceable reference sources for scanners and dose calibrators, and similar patient and imaging protocols. We then assessed SUV in patient test-retest studies. Methods: Five 18F-FDG PET/CT scanners from 4 institutions (2 in a National Cancer Institute-designated Comprehensive Cancer Center, 3 in a community-based network) were qualified for study use. Patients were scanned twice within 15 d, on the same scanner (n = 10); different but same model scanners within an institution (n = 2); or different model scanners at different institutions (n = 11). SUVmax was recorded for lesions, and SUVmean for normal liver uptake. Linear mixed models with random intercept were fitted to evaluate test-retest differences in multiple lesions per patient and to estimate the concordance correlation coefficient. Bland-Altman plots and repeatability coefficients were also produced. Results: In total, 162 lesions (82 bone, 80 soft tissue) were assessed in patients with breast cancer (n = 17) or other cancers (n = 6). Repeat scans within the same institution, using the same scanner or 2 scanners of the same model, had an average difference in SUVmax of 8% (95% confidence interval, 6%-10%). For test-retest on different scanners at different sites, the average difference in lesion SUVmax was 18% (95% confidence interval, 13%-24%). Normal liver uptake (SUVmean) showed an average difference of 5% (95% confidence interval, 3%-10%) for the same scanner model or institution and 6% (95% confidence interval, 3%-11%) for different scanners from different institutions. Protocol adherence was good; the median difference in injection-to-acquisition time was 2 min (range, 0-11 min). Test-retest SUVmax variability was not explained by available information on protocol deviations or patient or lesion characteristics. Conclusion: 18F-FDG PET/CT scanner qualification and calibration can yield highly reproducible test-retest tumor SUV measurements. Our data support use of different qualified scanners of the same model for serial studies. Test-retest differences from different scanner models were greater; more resolution-dependent harmonization of scanner protocols and reconstruction algorithms may be capable of reducing these differences to values closer to same-scanner results.
Collapse
Affiliation(s)
- Brenda F Kurland
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lanell M Peterson
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - Andrew T Shields
- Department of Radiology, University of Washington, Seattle, Washington; and
| | - Jean H Lee
- Department of Radiology, University of Washington, Seattle, Washington; and
| | - Darrin W Byrd
- Department of Radiology, University of Washington, Seattle, Washington; and
| | - Alena Novakova-Jiresova
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - Mark Muzi
- Department of Radiology, University of Washington, Seattle, Washington; and
| | - Jennifer M Specht
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - David A Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hannah M Linden
- Division of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, Seattle, Washington
| | - Paul E Kinahan
- Department of Radiology, University of Washington, Seattle, Washington; and
| |
Collapse
|
46
|
Wangaryattawanich P, Branstetter BF, Hughes M, Clump DA, Heron DE, Rath TJ. Negative Predictive Value of NI-RADS Category 2 in the First Posttreatment FDG-PET/CT in Head and Neck Squamous Cell Carcinoma. AJNR Am J Neuroradiol 2018; 39:1884-1888. [PMID: 30166429 DOI: 10.3174/ajnr.a5767] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/28/2018] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND PURPOSE FDG PET/CT has a high negative predictive value in patients with head and neck squamous cell carcinoma who responds completely to non-operative therapy. However, the treatment failure rate in patients with a partial but incomplete response is unclear. Our aim was to investigate the negative predictive value of the first posttreatment FDG-PET/CT in patients with head and neck squamous cell carcinoma with incomplete response interpreted as Neck Imaging Reporting and Data System (NI-RADS) category 2. MATERIALS AND METHODS We retrospectively identified patients with head and neck squamous cell carcinoma treated with chemoradiation or radiation therapy with curative intent in our institution between 2008 and 2016. We included patients whose first posttreatment FDG-PET/CT was interpreted as showing marked improvement of disease but who had a mild residual mass or FDG avidity in either the primary tumor bed or lymph nodes (NI-RADS 2). The negative predictive value of FDG-PET/CT was calculated, including the 95% CI, using the Newcombe method. Two-year disease-free survival was the reference standard. RESULTS Seventeen of 110 patients (15%) experienced locoregional treatment failure within 2 years of completing treatment, yielding a negative predictive value of 85% (95% Cl, 77%-90%). The most common location of tumor recurrence was the cervical lymph nodes (59%). The median time interval between completion of therapy and treatment failure was 10 months (range, 5-24 months). CONCLUSIONS In patients with an incomplete response after treatment of head and neck squamous cell carcinoma, the negative predictive value of the first posttreatment FDG-PET/CT was 85%, which is lower than the 91% negative predictive value of FDG-PET/CT in patients with an initial complete response. Patients with an incomplete response (NI-RADS 2) should undergo more frequent clinical and imaging surveillance than patients with an initial complete response (NI-RADS 1).
Collapse
Affiliation(s)
| | - B F Branstetter
- From the Departments of Radiology (P.W, B.F.B., M.H., T.J.R.).,Otolaryngology (B.F.B., M.H., T.J.R.).,Biomedical Informatics (B.F.B.)
| | - M Hughes
- From the Departments of Radiology (P.W, B.F.B., M.H., T.J.R.).,Otolaryngology (B.F.B., M.H., T.J.R.)
| | - D A Clump
- Radiation Oncology (D.A.C., D.E.H.), University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - D E Heron
- Radiation Oncology (D.A.C., D.E.H.), University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - T J Rath
- From the Departments of Radiology (P.W, B.F.B., M.H., T.J.R.) .,Otolaryngology (B.F.B., M.H., T.J.R.)
| |
Collapse
|
47
|
Abstract
Although visual assessment using the Deauville criteria is strongly recommended by guidelines for treatment response monitoring in all FDG-avid lymphoma histologies, the high rate of false-positives and concerns about interobserver variability have motivated the development of quantitative tools to facilitate objective measurement of tumor response in both routine and clinical trial settings. Imaging studies using functional quantitative measures play a significant role in profiling oncologic processes. These quantitative metrics allow for objective end points in multicenter clinical trials. However, the standardization of imaging procedures including image acquisition parameters, reconstruction and analytic measures, and validation of these methods are essential to enable an individualized treatment approach. A robust quality control program associated with the inclusion of proper scanner calibration, cross-calibration with dose calibrators and across other scanners is required for accurate quantitative measurements. In this section, we will review the technical and methodological considerations related to PET-derived quantitative metrics and the relevant published data to emphasize the potential value of these metrics in the prediction of patient prognosis in lymphoma.
Collapse
Affiliation(s)
- Lale Kostakoglu
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Stéphane Chauvie
- Department of Medical Physics, 'Santa Croce e Carle' Hospital, Cuneo, Italy
| |
Collapse
|
48
|
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
|
49
|
Stafford WC, Peng X, Olofsson MH, Zhang X, Luci DK, Lu L, Cheng Q, Trésaugues L, Dexheimer TS, Coussens NP, Augsten M, Ahlzén HSM, Orwar O, Östman A, Stone-Elander S, Maloney DJ, Jadhav A, Simeonov A, Linder S, Arnér ESJ. Irreversible inhibition of cytosolic thioredoxin reductase 1 as a mechanistic basis for anticancer therapy. Sci Transl Med 2018; 10:eaaf7444. [PMID: 29444979 PMCID: PMC7059553 DOI: 10.1126/scitranslmed.aaf7444] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 02/01/2017] [Accepted: 12/14/2017] [Indexed: 12/25/2022]
Abstract
Cancer cells adapt to their inherently increased oxidative stress through activation of the glutathione (GSH) and thioredoxin (TXN) systems. Inhibition of both of these systems effectively kills cancer cells, but such broad inhibition of antioxidant activity also kills normal cells, which is highly unwanted in a clinical setting. We therefore evaluated targeting of the TXN pathway alone and, more specifically, selective inhibition of the cytosolic selenocysteine-containing enzyme TXN reductase 1 (TXNRD1). TXNRD1 inhibitors were discovered in a large screening effort and displayed increased specificity compared to pan-TXNRD inhibitors, such as auranofin, that also inhibit the mitochondrial enzyme TXNRD2 and additional targets. For our lead compounds, TXNRD1 inhibition correlated with cancer cell cytotoxicity, and inhibitor-triggered conversion of TXNRD1 from an antioxidant to a pro-oxidant enzyme correlated with corresponding increases in cellular production of H2O2 In mice, the most specific TXNRD1 inhibitor, here described as TXNRD1 inhibitor 1 (TRi-1), impaired growth and viability of human tumor xenografts and syngeneic mouse tumors while having little mitochondrial toxicity and being better tolerated than auranofin. These results display the therapeutic anticancer potential of irreversibly targeting cytosolic TXNRD1 using small molecules and present potent and selective TXNRD1 inhibitors. Given the pronounced up-regulation of TXNRD1 in several metastatic malignancies, it seems worthwhile to further explore the potential benefit of specific irreversible TXNRD1 inhibitors for anticancer therapy.
Collapse
Affiliation(s)
- William C Stafford
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77 Stockholm, Sweden
- Oblique Therapeutics AB, SE 413 46 Gothenburg, Sweden
| | - Xiaoxiao Peng
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Maria Hägg Olofsson
- Department of Oncology-Pathology, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Xiaonan Zhang
- Department of Oncology-Pathology, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Diane K Luci
- NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-4874, USA
| | - Li Lu
- Karolinska Experimental Research and Imaging Center, Karolinska University Hospital, SE 171 76 Stockholm, Sweden
| | - Qing Cheng
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Lionel Trésaugues
- Division of Biophysics, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Thomas S Dexheimer
- NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-4874, USA
| | - Nathan P Coussens
- NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-4874, USA
| | - Martin Augsten
- Department of Oncology-Pathology, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Hanna-Stina Martinsson Ahlzén
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Owe Orwar
- Oblique Therapeutics AB, SE 413 46 Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, SE 171 77 Stockholm, Sweden
- University of Bergen, Postboks 7804, N-5020 Bergen, Norway
| | - Sharon Stone-Elander
- Department of Neuroradiology, Positron Emission Tomography Radiochemistry, Karolinska University Hospital, SE 171 76 Stockholm, Sweden
- Department of Clinical Neurosciences, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - David J Maloney
- NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-4874, USA
| | - Ajit Jadhav
- NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-4874, USA
| | - Anton Simeonov
- NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-4874, USA
| | - Stig Linder
- Department of Oncology-Pathology, Karolinska Institutet, SE 171 77 Stockholm, Sweden
- Division of Drug Research, Department of Medicine and Health, Linköping University, SE 581 83 Linköping, Sweden
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77 Stockholm, Sweden.
| |
Collapse
|
50
|
Crespo-Jara A, Redal-Peña MC, Martinez-Navarro EM, Sureda M, Fernandez-Morejon FJ, Garcia-Cases FJ, Manzano RG, Brugarolas A. A novel genomic signature predicting FDG uptake in diverse metastatic tumors. EJNMMI Res 2018; 8:4. [PMID: 29349517 PMCID: PMC5773462 DOI: 10.1186/s13550-017-0355-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/27/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Building a universal genomic signature predicting the intensity of FDG uptake in diverse metastatic tumors may allow us to understand better the biological processes underlying this phenomenon and their requirements of glucose uptake. METHODS A balanced training set (n = 71) of metastatic tumors including some of the most frequent histologies, with matched PET/CT quantification measurements and whole human genome gene expression microarrays, was used to build the signature. Selection of microarray features was carried out exclusively on the basis of their strong association with FDG uptake (as measured by SUVmean35) by means of univariate linear regression. A thorough bioinformatics study of these genes was performed, and multivariable models were built by fitting several state of the art regression techniques to the training set for comparison. RESULTS The 909 probes with the strongest association with the SUVmean35 (comprising 742 identifiable genes and 62 probes not matched to a symbol) were used to build the signature. Partial least squares using three components (PLS-3) was the best performing model in the training dataset cross-validation (root mean square error, RMSE = 0.443) and was validated further in an independent validation dataset (n = 13) obtaining a performance within the 95% CI of that obtained in the training dataset (RMSE = 0.645). Significantly overrepresented biological processes correlating with the SUVmean35 were identified beyond glycolysis, such as ribosome biogenesis and DNA replication (correlating with a higher SUVmean35) and cytoskeleton reorganization and autophagy (correlating with a lower SUVmean35). CONCLUSIONS PLS-3 is a signature predicting accurately the intensity of FDG uptake in diverse metastatic tumors. FDG-PET might help in the design of specific targeted therapies directed to counteract the identified malignant biological processes more likely activated in a tumor as inferred from the SUVmean35 and also from its variations in response to antineoplastic treatments.
Collapse
Affiliation(s)
- Aurora Crespo-Jara
- Plataforma de Oncologia, Hospital Quironsalud Torrevieja, Pda. La Loma s/n, 03184, Torrevieja, Alicante, Spain.,Catedra Oncologia Multidisciplinar, Universidad Catolica de Murcia, Murcia, Spain
| | - Maria Carmen Redal-Peña
- Plataforma de Oncologia, Hospital Quironsalud Torrevieja, Pda. La Loma s/n, 03184, Torrevieja, Alicante, Spain.,Catedra Oncologia Multidisciplinar, Universidad Catolica de Murcia, Murcia, Spain
| | - Elena Maria Martinez-Navarro
- Plataforma de Oncologia, Hospital Quironsalud Torrevieja, Pda. La Loma s/n, 03184, Torrevieja, Alicante, Spain.,Catedra Oncologia Multidisciplinar, Universidad Catolica de Murcia, Murcia, Spain
| | - Manuel Sureda
- Plataforma de Oncologia, Hospital Quironsalud Torrevieja, Pda. La Loma s/n, 03184, Torrevieja, Alicante, Spain.,Catedra Oncologia Multidisciplinar, Universidad Catolica de Murcia, Murcia, Spain
| | - Francisco Jose Fernandez-Morejon
- Plataforma de Oncologia, Hospital Quironsalud Torrevieja, Pda. La Loma s/n, 03184, Torrevieja, Alicante, Spain.,Catedra Oncologia Multidisciplinar, Universidad Catolica de Murcia, Murcia, Spain
| | - Francisco J Garcia-Cases
- Plataforma de Oncologia, Hospital Quironsalud Torrevieja, Pda. La Loma s/n, 03184, Torrevieja, Alicante, Spain.,Catedra Oncologia Multidisciplinar, Universidad Catolica de Murcia, Murcia, Spain
| | - Ramon Gonzalez Manzano
- Plataforma de Oncologia, Hospital Quironsalud Torrevieja, Pda. La Loma s/n, 03184, Torrevieja, Alicante, Spain. .,Catedra Oncologia Multidisciplinar, Universidad Catolica de Murcia, Murcia, Spain.
| | - Antonio Brugarolas
- Plataforma de Oncologia, Hospital Quironsalud Torrevieja, Pda. La Loma s/n, 03184, Torrevieja, Alicante, Spain.,Catedra Oncologia Multidisciplinar, Universidad Catolica de Murcia, Murcia, Spain
| |
Collapse
|