1
|
Gutsche R, Lowis C, Ziemons K, Kocher M, Ceccon G, Régio Brambilla C, Shah NJ, Langen KJ, Galldiks N, Isensee F, Lohmann P. Automated Brain Tumor Detection and Segmentation for Treatment Response Assessment Using Amino Acid PET. J Nucl Med 2023; 64:1594-1602. [PMID: 37562802 DOI: 10.2967/jnumed.123.265725] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/31/2023] [Indexed: 08/12/2023] Open
Abstract
Evaluation of metabolic tumor volume (MTV) changes using amino acid PET has become an important tool for response assessment in brain tumor patients. MTV is usually determined by manual or semiautomatic delineation, which is laborious and may be prone to intra- and interobserver variability. The goal of our study was to develop a method for automated MTV segmentation and to evaluate its performance for response assessment in patients with gliomas. Methods: In total, 699 amino acid PET scans using the tracer O-(2-[18F]fluoroethyl)-l-tyrosine (18F-FET) from 555 brain tumor patients at initial diagnosis or during follow-up were retrospectively evaluated (mainly glioma patients, 76%). 18F-FET PET MTVs were segmented semiautomatically by experienced readers. An artificial neural network (no new U-Net) was configured on 476 scans from 399 patients, and the network performance was evaluated on a test dataset including 223 scans from 156 patients. Surface and volumetric Dice similarity coefficients (DSCs) were used to evaluate segmentation quality. Finally, the network was applied to a recently published 18F-FET PET study on response assessment in glioblastoma patients treated with adjuvant temozolomide chemotherapy for a fully automated response assessment in comparison to an experienced physician. Results: In the test dataset, 92% of lesions with increased uptake (n = 189) and 85% of lesions with iso- or hypometabolic uptake (n = 33) were correctly identified (F1 score, 92%). Single lesions with a contiguous uptake had the highest DSC, followed by lesions with heterogeneous, noncontiguous uptake and multifocal lesions (surface DSC: 0.96, 0.93, and 0.81 respectively; volume DSC: 0.83, 0.77, and 0.67, respectively). Change in MTV, as detected by the automated segmentation, was a significant determinant of disease-free and overall survival, in agreement with the physician's assessment. Conclusion: Our deep learning-based 18F-FET PET segmentation allows reliable, robust, and fully automated evaluation of MTV in brain tumor patients and demonstrates clinical value for automated response assessment.
Collapse
Affiliation(s)
- Robin Gutsche
- Institute of Neuroscience and Medicine, Forschungszentrum Juelich GmbH, Juelich, Germany
- RWTH Aachen University, Aachen, Germany
| | - Carsten Lowis
- Institute of Neuroscience and Medicine, Forschungszentrum Juelich GmbH, Juelich, Germany
| | - Karl Ziemons
- Medical Engineering and Technomathematics, FH Aachen University of Applied Sciences, Juelich, Germany
| | - Martin Kocher
- Institute of Neuroscience and Medicine, Forschungszentrum Juelich GmbH, Juelich, Germany
- Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Garry Ceccon
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Cláudia Régio Brambilla
- Institute of Neuroscience and Medicine, Forschungszentrum Juelich GmbH, Juelich, Germany
- JARA-BRAIN-Translational Medicine, Aachen, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine, Forschungszentrum Juelich GmbH, Juelich, Germany
- JARA-BRAIN-Translational Medicine, Aachen, Germany
- Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Forschungszentrum Juelich GmbH, Juelich, Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine, Forschungszentrum Juelich GmbH, Juelich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
| | - Fabian Isensee
- Applied Computer Vision Lab, Helmholtz Imaging, Heidelberg, Germany; and
- Division of Medical Image Computing, German Cancer Research Center, Heidelberg, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine, Forschungszentrum Juelich GmbH, Juelich, Germany;
| |
Collapse
|
2
|
Ladefoged CN, Andersen FL, Andersen TL, Anderberg L, Engkebølle C, Madsen K, Højgaard L, Henriksen OM, Law I. DeepDixon synthetic CT for [ 18F]FET PET/MRI attenuation correction of post-surgery glioma patients with metal implants. Front Neurosci 2023; 17:1142383. [PMID: 37090806 PMCID: PMC10115992 DOI: 10.3389/fnins.2023.1142383] [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: 01/11/2023] [Accepted: 03/08/2023] [Indexed: 04/25/2023] Open
Abstract
Purpose Conventional magnetic resonance imaging (MRI) can for glioma assessment be supplemented by positron emission tomography (PET) imaging with radiolabeled amino acids such as O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET), which provides additional information on metabolic properties. In neuro-oncology, patients often undergo brain and skull altering treatment, which is known to challenge MRI-based attenuation correction (MR-AC) methods and thereby impact the simplified semi-quantitative measures such as tumor-to-brain ratio (TBR) used in clinical routine. The aim of the present study was to examine the applicability of our deep learning method, DeepDixon, for MR-AC in [18F]FET PET/MRI scans of a post-surgery glioma cohort with metal implants. Methods The MR-AC maps were assessed for all 194 included post-surgery glioma patients (318 studies). The subgroup of 147 patients (222 studies, 200 MBq [18F]FET PET/MRI) with tracer uptake above 1 ml were subsequently reconstructed with DeepDixon, vendor-default atlas-based method, and a low-dose computed tomography (CT) used as reference. The biological tumor volume (BTV) was delineated on each patient by isocontouring tracer uptake above a TBR threshold of 1.6. We evaluated the MR-AC methods using the recommended clinical metrics BTV and mean and maximum TBR on a patient-by-patient basis against the reference with CT-AC. Results Ninety-seven percent of the studies (310/318) did not have any major artifacts using DeepDixon, which resulted in a Dice coefficient of 0.89/0.83 for tissue/bone, respectively, compared to 0.84/0.57 when using atlas. The average difference between DeepDixon and CT-AC was within 0.2% across all clinical metrics, and no statistically significant difference was found. When using DeepDixon, only 3 out of 222 studies (1%) exceeded our acceptance criteria compared to 72 of the 222 studies (32%) with the atlas method. Conclusion We evaluated the performance of a state-of-the-art MR-AC method on the largest post-surgical glioma patient cohort to date. We found that DeepDixon could overcome most of the issues arising from irregular anatomy and metal artifacts present in the cohort resulting in clinical metrics within acceptable limits of the reference CT-AC in almost all cases. This is a significant improvement over the vendor-provided atlas method and of particular importance in response assessment.
Collapse
|
3
|
Li Z, Holzgreve A, Unterrainer LM, Ruf VC, Quach S, Bartos LM, Suchorska B, Niyazi M, Wenter V, Herms J, Bartenstein P, Tonn JC, Unterrainer M, Albert NL, Kaiser L. Combination of pre-treatment dynamic [ 18F]FET PET radiomics and conventional clinical parameters for the survival stratification in patients with IDH-wildtype glioblastoma. Eur J Nucl Med Mol Imaging 2023; 50:535-545. [PMID: 36227357 PMCID: PMC9816231 DOI: 10.1007/s00259-022-05988-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 10/03/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE The aim of this study was to build and evaluate a prediction model which incorporates clinical parameters and radiomic features extracted from static as well as dynamic [18F]FET PET for the survival stratification in patients with newly diagnosed IDH-wildtype glioblastoma. METHODS A total of 141 patients with newly diagnosed IDH-wildtype glioblastoma and dynamic [18F]FET PET prior to surgical intervention were included. Patients with a survival time ≤ 12 months were classified as short-term survivors. First order, shape, and texture radiomic features were extracted from pre-treatment static (tumor-to-background ratio; TBR) and dynamic (time-to-peak; TTP) images, respectively, and randomly divided into a training (n = 99) and a testing cohort (n = 42). After feature normalization, recursive feature elimination was applied for feature selection using 5-fold cross-validation on the training cohort, and a machine learning model was constructed to compare radiomic models and combined clinical-radiomic models with selected radiomic features and clinical parameters. The area under the ROC curve (AUC), accuracy, sensitivity, specificity, and positive and negative predictive values were calculated to assess the predictive performance for identifying short-term survivors in both the training and testing cohort. RESULTS A combined clinical-radiomic model comprising six clinical parameters and six selected dynamic radiomic features achieved highest predictability of short-term survival with an AUC of 0.74 (95% confidence interval, 0.60-0.88) in the independent testing cohort. CONCLUSIONS This study successfully built and evaluated prediction models using [18F]FET PET-based radiomic features and clinical parameters for the individualized assessment of short-term survival in patients with a newly diagnosed IDH-wildtype glioblastoma. The combination of both clinical parameters and dynamic [18F]FET PET-based radiomic features reached highest accuracy in identifying patients at risk. Although the achieved accuracy level remained moderate, our data shows that the integration of dynamic [18F]FET PET radiomic data into clinical prediction models may improve patient stratification beyond established prognostic markers.
Collapse
Affiliation(s)
- Zhicong Li
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Lena M Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Viktoria C Ruf
- Center for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Laura M Bartos
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Bogdana Suchorska
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
- Department of Neurosurgery, Sana Hospital, Duisburg, Germany
| | - Maximilian Niyazi
- Department of Radiotherapy, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Vera Wenter
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Joerg-Christian Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcus Unterrainer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|
4
|
Dynamic 11C-Methionine PET-CT: Prognostic Factors for Disease Progression and Survival in Patients with Suspected Glioma Recurrence. Cancers (Basel) 2021; 13:cancers13194777. [PMID: 34638262 PMCID: PMC8508090 DOI: 10.3390/cancers13194777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/19/2021] [Accepted: 09/10/2021] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Recurrence after initial treatments is an expected event in glioma patients, particularly for high-grade glioma, with a median progression-free survival of 8–11 weeks. The prognostic evaluation of disease is a crucial step in the planning of therapeutic strategies, in both the primary and recurrence stages of disease. The aim of our retrospective study was to assess the prognostic value of 11C-methionine PET-CT dynamic and semiquantitative parameters in patients with suspected glioma recurrence at MR, in terms of progression-free survival and overall survival. In a population of sixty-seven consecutive patients, both static and kinetic analyses provided parameters (i.e., tumour-to-background ratio and SUVmax associated with time-to-peak, respectively) able to predict both progression-free and overall survival in the whole population and in the high-grade glioma subgroup of patients. Dynamic 11C-methionine PET-CT can be a useful diagnostic tool, in patients with suspicion of glioma recurrence, able to produce significant prognostic indices. Abstract Purpose: The prognostic evaluation of glioma recurrence patients is important in the therapeutic management. We investigated the prognostic value of 11C-methionine PET-CT (MET-PET) dynamic and semiquantitative parameters in patients with suspected glioma recurrence. Methods: Sixty-seven consecutive patients who underwent MET-PET for suspected glioma recurrence at MR were retrospectively included. Twenty-one patients underwent static MET-PET; 46/67 underwent dynamic MET-PET. In all patients, SUVmax, SUVmean and tumour-to-background ratio (T/B) were calculated. From dynamic acquisition, the shape and slope of time-activity curves, time-to-peak and its SUVmax (SUVmaxTTP) were extrapolated. The prognostic value of PET parameters on progression-free (PFS) and overall survival (OS) was evaluated using Kaplan–Meier survival estimates and Cox regression. Results: The overall median follow-up was 19 months from MET-PET. Recurrence patients (38/67) had higher SUVmax (p = 0.001), SUVmean (p = 0.002) and T/B (p < 0.001); deceased patients (16/67) showed higher SUVmax (p = 0.03), SUVmean (p = 0.03) and T/B (p = 0.006). All static parameters were associated with PFS (all p < 0.001); T/B was associated with OS (p = 0.031). Regarding kinetic analyses, recurrence (27/46) and deceased (14/46) patients had higher SUVmaxTTP (p = 0.02, p = 0.01, respectively). SUVmaxTTP was the only dynamic parameter associated with PFS (p = 0.02) and OS (p = 0.006). At univariate analysis, SUVmax, SUVmean, T/B and SUVmaxTTP were predictive for PFS (all p < 0.05); SUVmaxTTP was predictive for OS (p = 0.02). At multivariate analysis, SUVmaxTTP remained significant for PFS (p = 0.03). Conclusion: Semiquantitative parameters and SUVmaxTTP were associated with clinical outcomes in patients with suspected glioma recurrence. Dynamic PET-CT acquisition, with static and kinetic parameters, can be a valuable non-invasive prognostic marker, identifying patients with worse prognosis who require personalised therapy.
Collapse
|
5
|
Li Z, Kaiser L, Holzgreve A, Ruf VC, Suchorska B, Wenter V, Quach S, Herms J, Bartenstein P, Tonn JC, Unterrainer M, Albert NL. Prediction of TERTp-mutation status in IDH-wildtype high-grade gliomas using pre-treatment dynamic [ 18F]FET PET radiomics. Eur J Nucl Med Mol Imaging 2021; 48:4415-4425. [PMID: 34490493 PMCID: PMC8566644 DOI: 10.1007/s00259-021-05526-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/05/2021] [Indexed: 12/22/2022]
Abstract
Purpose To evaluate radiomic features extracted from standard static images (20–40 min p.i.), early summation images (5–15 min p.i.), and dynamic [18F]FET PET images for the prediction of TERTp-mutation status in patients with IDH-wildtype high-grade glioma. Methods A total of 159 patients (median age 60.2 years, range 19–82 years) with newly diagnosed IDH-wildtype diffuse astrocytic glioma (WHO grade III or IV) and dynamic [18F]FET PET prior to surgical intervention were enrolled and divided into a training (n = 112) and a testing cohort (n = 47) randomly. First-order, shape, and texture radiomic features were extracted from standard static (20–40 min summation images; TBR20–40), early static (5–15 min summation images; TBR5–15), and dynamic (time-to-peak; TTP) images, respectively. Recursive feature elimination was used for feature selection by 10-fold cross-validation in the training cohort after normalization, and logistic regression models were generated using the radiomic features extracted from each image to differentiate TERTp-mutation status. The areas under the ROC curve (AUC), accuracy, sensitivity, specificity, and positive and negative predictive value were calculated to illustrate diagnostic power in both the training and testing cohort. Results The TTP model comprised nine selected features and achieved highest predictability of TERTp-mutation with an AUC of 0.82 (95% confidence interval 0.71–0.92) and sensitivity of 92.1% in the independent testing cohort. Weak predictive capability was obtained in the TBR5–15 model, with an AUC of 0.61 (95% CI 0.42–0.80) in the testing cohort, while no predictive power was observed in the TBR20–40 model. Conclusions Radiomics based on TTP images extracted from dynamic [18F]FET PET can predict the TERTp-mutation status of IDH-wildtype diffuse astrocytic high-grade gliomas with high accuracy preoperatively. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05526-6.
Collapse
Affiliation(s)
- Zhicong Li
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Viktoria C Ruf
- Center for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Bogdana Suchorska
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
- Department of Neurosurgery, Sana Hospital, Duisburg, Germany
| | - Vera Wenter
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jörg-Christian Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcus Unterrainer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
6
|
Di Giorgio E, Cuocolo A, Mansi L, Sicignano M, Squame F, Gaudieri V, Giordano P, Giugliano FM, Mazzaferro MP, Negro A, Villa A, Spadafora M. Assessment of therapy response to Regorafenib by 18F-DOPA-PET/CT in patients with recurrent high-grade gliomas: a case series. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00416-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
7
|
Gutsche R, Scheins J, Kocher M, Bousabarah K, Fink GR, Shah NJ, Langen KJ, Galldiks N, Lohmann P. Evaluation of FET PET Radiomics Feature Repeatability in Glioma Patients. Cancers (Basel) 2021; 13:cancers13040647. [PMID: 33562803 PMCID: PMC7915742 DOI: 10.3390/cancers13040647] [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: 12/22/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Initial studies suggested the additional diagnostic value of amino acid positron emission tomography (PET) radiomics using the tracer O-(2-[18F]fluoroethyl)-L-tyrosine (FET) in brain tumor patient management. However, to ensure the reliable performance of the generated FET PET radiomics models for clinical diagnostics, repeatability of radiomics features is essential. Hence, we assessed the impact of brain tumor volumes and key molecular alterations such as an isocitrate dehydrogenase (IDH) mutation on the repeatability of FET PET radiomics features in 50 newly diagnosed glioma patients. In a test–retest approach based on routinely acquired FET PET scans, we identified 297 repeatable features. The IDH genotype did not affect feature repeatability. Moreover, these robust features were able to differentiate patients with IDH-wildtype glioma from those with an IDH mutation. Our results suggest that robust radiomics features can be obtained from routinely acquired FET PET scans, which are valuable for further standardization of radiomics analyses in neurooncology. Abstract Amino acid PET using the tracer O-(2-[18F]fluoroethyl)-L-tyrosine (FET) has attracted considerable interest in neurooncology. Furthermore, initial studies suggested the additional diagnostic value of FET PET radiomics in brain tumor patient management. However, the conclusiveness of radiomics models strongly depends on feature generalizability. We here evaluated the repeatability of feature-based FET PET radiomics. A test–retest analysis based on equivalent but statistically independent subsamples of FET PET images was performed in 50 newly diagnosed and histomolecularly characterized glioma patients. A total of 1,302 radiomics features were calculated from semi-automatically segmented tumor volumes-of-interest (VOIs). Furthermore, to investigate the influence of the spatial resolution of PET on repeatability, spherical VOIs of different sizes were positioned in the tumor and healthy brain tissue. Feature repeatability was assessed by calculating the intraclass correlation coefficient (ICC). To further investigate the influence of the isocitrate dehydrogenase (IDH) genotype on feature repeatability, a hierarchical cluster analysis was performed. For tumor VOIs, 73% of first-order features and 71% of features extracted from the gray level co-occurrence matrix showed high repeatability (ICC 95% confidence interval, 0.91–1.00). In the largest spherical tumor VOIs, 67% of features showed high repeatability, significantly decreasing towards smaller VOIs. The IDH genotype did not affect feature repeatability. Based on 297 repeatable features, two clusters were identified separating patients with IDH-wildtype glioma from those with an IDH mutation. Our results suggest that robust features can be obtained from routinely acquired FET PET scans, which are valuable for further standardization of radiomics analyses in neurooncology.
Collapse
Affiliation(s)
- Robin Gutsche
- Research Center Juelich, Institute of Neuroscience and Medicine (INM-3, -4, -11), 52425 Juelich, Germany; (R.G.); (J.S.); (M.K.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- RWTH Aachen University, 52062 Aachen, Germany
| | - Jürgen Scheins
- Research Center Juelich, Institute of Neuroscience and Medicine (INM-3, -4, -11), 52425 Juelich, Germany; (R.G.); (J.S.); (M.K.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
| | - Martin Kocher
- Research Center Juelich, Institute of Neuroscience and Medicine (INM-3, -4, -11), 52425 Juelich, Germany; (R.G.); (J.S.); (M.K.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
- Center for Integrated Oncology (CIO), Universities Aachen, Bonn, Duesseldorf and Cologne, 50937 Cologne, Germany
| | - Khaled Bousabarah
- Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
| | - Gereon R. Fink
- Research Center Juelich, Institute of Neuroscience and Medicine (INM-3, -4, -11), 52425 Juelich, Germany; (R.G.); (J.S.); (M.K.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Nadim J. Shah
- Research Center Juelich, Institute of Neuroscience and Medicine (INM-3, -4, -11), 52425 Juelich, Germany; (R.G.); (J.S.); (M.K.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Neurology, University Hospital RWTH Aachen, 52074 Aachen, Germany
- JARA-BRAIN-Translational Medicine, 52074 Aachen, Germany
| | - Karl-Josef Langen
- Research Center Juelich, Institute of Neuroscience and Medicine (INM-3, -4, -11), 52425 Juelich, Germany; (R.G.); (J.S.); (M.K.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Nuclear Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany
- Center for Integrated Oncology (CIO), Universities Aachen, Bonn, Duesseldorf and Cologne, 52074 Aachen, Germany
| | - Norbert Galldiks
- Research Center Juelich, Institute of Neuroscience and Medicine (INM-3, -4, -11), 52425 Juelich, Germany; (R.G.); (J.S.); (M.K.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Center for Integrated Oncology (CIO), Universities Aachen, Bonn, Duesseldorf and Cologne, 50937 Cologne, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Philipp Lohmann
- Research Center Juelich, Institute of Neuroscience and Medicine (INM-3, -4, -11), 52425 Juelich, Germany; (R.G.); (J.S.); (M.K.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
- Correspondence:
| |
Collapse
|
8
|
Shymanskaya A, Worthoff WA, Stoffels G, Lindemeyer J, Neumaier B, Lohmann P, Galldiks N, Langen KJ, Shah NJ. Comparison of [ 18F]Fluoroethyltyrosine PET and Sodium MRI in Cerebral Gliomas: a Pilot Study. Mol Imaging Biol 2021; 22:198-207. [PMID: 30989437 DOI: 10.1007/s11307-019-01349-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE Positron emission tomography (PET) using O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) improves the diagnostics of cerebral gliomas compared with conventional magnetic resonance imaging (MRI). Sodium MRI is an evolving method to assess tumor metabolism. In this pilot study, we explored the relationship of [18F]FET-PET and sodium MRI in patients with cerebral gliomas in relation to the mutational status of the enzyme isocitrate dehydrogenase (IDH). PROCEDURES Ten patients with untreated cerebral gliomas and one patient with a recurrent glioblastoma (GBM) were investigated by dynamic [18F]FET-PET and sodium MRI using an enhanced simultaneous single-quantum- and triple-quantum-filtered imaging of 23Na (SISTINA) sequence to estimate total (NaT), weighted non-restricted (NaNR, mainly extracellular), and restricted (NaR, mainly intracellular) sodium in tumors and normal brain tissue. [18F]FET uptake and sodium parameters in tumors with a different IDH mutational status were compared. After biopsy or resection, histology and the IDH mutational status were determined neuropathologically. RESULTS NaT (p = 0.05), tumor-to-brain ratios (TBR) of NaT (p = 0.02), NaNR (p = 0.003), and the ratio of NaT/NaR (p < 0.001) were significantly higher in IDH-mutated than in IDH-wild-type gliomas (n = 5 patients each) while NaR was significantly lower in IDH-mutated gliomas (p = 0.01). [18F]FET parameters (TBR, time-to-peak) were not predictive of IDH status in this small cohort of patients. There was no obvious relationship between sodium distribution and [18F]FET uptake. The patient with a recurrent GBM exhibited an additional radiation injury with strong abnormalities in sodium MRI. CONCLUSIONS Sodium MRI appears to be more strongly related to the IDH mutational status than are [18F]FET-PET parameters. A further evaluation of the combination of the two methods in a larger group of high- and low-grade gliomas seems promising.
Collapse
Affiliation(s)
- Aliaksandra Shymanskaya
- Institute of Neuroscience and Medicine (3, 4, 5, 11), Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - Wieland A Worthoff
- Institute of Neuroscience and Medicine (3, 4, 5, 11), Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (3, 4, 5, 11), Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - Johannes Lindemeyer
- Institute of Neuroscience and Medicine (3, 4, 5, 11), Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine (3, 4, 5, 11), Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (3, 4, 5, 11), Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (3, 4, 5, 11), Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany.,Center of Integrated Oncology (CIO), Universities of Bonn and Cologne, Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (3, 4, 5, 11), Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.,Department of Nuclear Medicine, RWTH Aachen University, Aachen, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Aachen, Germany
| | - N Jon Shah
- Institute of Neuroscience and Medicine (3, 4, 5, 11), Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Aachen, Germany.,Department of Neurology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
9
|
Lohmann P, Elahmadawy MA, Gutsche R, Werner JM, Bauer EK, Ceccon G, Kocher M, Lerche CW, Rapp M, Fink GR, Shah NJ, Langen KJ, Galldiks N. FET PET Radiomics for Differentiating Pseudoprogression from Early Tumor Progression in Glioma Patients Post-Chemoradiation. Cancers (Basel) 2020; 12:cancers12123835. [PMID: 33353180 PMCID: PMC7766151 DOI: 10.3390/cancers12123835] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 11/19/2022] Open
Abstract
Simple Summary Following chemoradiation with alkylating agents in glioma patients, structural magnetic resonance imaging (MRI) may suggest tumor progression which subsequently improves during the course of the disease without any treatment change. This phenomenon has been termed pseudoprogression. Despite advances in medical imaging, a reliable diagnosis of pseudoprogression remains a challenging task. Radiomics is a subdiscipline of artificial intelligence and allows the identification and extraction of imaging features from various routine imaging modalities. These features can be used for the generation of mathematical models to improve diagnostics in patients with brain tumors. The present study highlights the potential of radiomics obtained from amino acid positron emission tomography (PET) for the diagnosis of pseudoprogression. In 34 patients with suspicious MRI early after chemoradiation completion, our radiomics model correctly identified all patients with pseudoprogression. Abstract Currently, a reliable diagnostic test for differentiating pseudoprogression from early tumor progression is lacking. We explored the potential of O-(2-[18F]fluoroethyl)-L-tyrosine (FET) positron emission tomography (PET) radiomics for this clinically important task. Thirty-four patients (isocitrate dehydrogenase (IDH)-wildtype glioblastoma, 94%) with progressive magnetic resonance imaging (MRI) changes according to the Response Assessment in Neuro-Oncology (RANO) criteria within the first 12 weeks after completing temozolomide chemoradiation underwent a dynamic FET PET scan. Static and dynamic FET PET parameters were calculated. For radiomics analysis, the number of datasets was increased to 102 using data augmentation. After randomly assigning patients to a training and test dataset, 944 features were calculated on unfiltered and filtered images. The number of features for model generation was limited to four to avoid data overfitting. Eighteen patients were diagnosed with early tumor progression, and 16 patients had pseudoprogression. The FET PET radiomics model correctly diagnosed pseudoprogression in all test cohort patients (sensitivity, 100%; negative predictive value, 100%). In contrast, the diagnostic performance of the best FET PET parameter (TBRmax) was lower (sensitivity, 81%; negative predictive value, 80%). The results suggest that FET PET radiomics helps diagnose patients with pseudoprogression with a high diagnostic performance. Given the clinical significance, further studies are warranted.
Collapse
Affiliation(s)
- Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4, -11), Research Center Juelich, 52425 Juelich, Germany; (M.A.E.); (R.G.); (M.K.); (C.W.L.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
- Correspondence:
| | - Mai A. Elahmadawy
- Institute of Neuroscience and Medicine (INM-3, -4, -11), Research Center Juelich, 52425 Juelich, Germany; (M.A.E.); (R.G.); (M.K.); (C.W.L.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Nuclear Medicine, National Cancer Institute (NCI), Cairo University, 11796 Cairo, Egypt
| | - Robin Gutsche
- Institute of Neuroscience and Medicine (INM-3, -4, -11), Research Center Juelich, 52425 Juelich, Germany; (M.A.E.); (R.G.); (M.K.); (C.W.L.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- RWTH Aachen University, 52062 Aachen, Germany
| | - Jan-Michael Werner
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (J.-M.W.); (E.K.B.); (G.C.)
| | - Elena K. Bauer
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (J.-M.W.); (E.K.B.); (G.C.)
| | - Garry Ceccon
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (J.-M.W.); (E.K.B.); (G.C.)
| | - Martin Kocher
- Institute of Neuroscience and Medicine (INM-3, -4, -11), Research Center Juelich, 52425 Juelich, Germany; (M.A.E.); (R.G.); (M.K.); (C.W.L.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
- Center for Integrated Oncology (CIO), Universities Aachen, Bonn, Duesseldorf and Cologne, 50937 Cologne, Germany
| | - Christoph W. Lerche
- Institute of Neuroscience and Medicine (INM-3, -4, -11), Research Center Juelich, 52425 Juelich, Germany; (M.A.E.); (R.G.); (M.K.); (C.W.L.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
| | - Marion Rapp
- Department of Neurosurgery, University of Duesseldorf, 40255 Duesseldorf, Germany;
| | - Gereon R. Fink
- Institute of Neuroscience and Medicine (INM-3, -4, -11), Research Center Juelich, 52425 Juelich, Germany; (M.A.E.); (R.G.); (M.K.); (C.W.L.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (J.-M.W.); (E.K.B.); (G.C.)
| | - Nadim J. Shah
- Institute of Neuroscience and Medicine (INM-3, -4, -11), Research Center Juelich, 52425 Juelich, Germany; (M.A.E.); (R.G.); (M.K.); (C.W.L.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Neurology, University Hospital RWTH Aachen, 52074 Aachen, Germany
- JARA-BRAIN-Translational Medicine, 52074 Aachen, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4, -11), Research Center Juelich, 52425 Juelich, Germany; (M.A.E.); (R.G.); (M.K.); (C.W.L.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Nuclear Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany
- Center for Integrated Oncology (CIO), Universities Aachen, Bonn, Duesseldorf and Cologne, 52074 Aachen, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, -4, -11), Research Center Juelich, 52425 Juelich, Germany; (M.A.E.); (R.G.); (M.K.); (C.W.L.); (G.R.F.); (N.J.S.); (K.-J.L.); (N.G.)
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (J.-M.W.); (E.K.B.); (G.C.)
- Center for Integrated Oncology (CIO), Universities Aachen, Bonn, Duesseldorf and Cologne, 50937 Cologne, Germany
| |
Collapse
|
10
|
Non-Invasive Prediction of IDH Mutation in Patients with Glioma WHO II/III/IV Based on F-18-FET PET-Guided In Vivo 1H-Magnetic Resonance Spectroscopy and Machine Learning. Cancers (Basel) 2020; 12:cancers12113406. [PMID: 33212941 PMCID: PMC7698334 DOI: 10.3390/cancers12113406] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/08/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Approximately 75–80% of according to the classification of world health organization (WHO) grade II and III gliomas are characterized by a mutation of the isocitrate dehydrogenase (IDH) enzymes, which are very important in glioma cell metabolism. Patients with IDH mutated glioma have a significantly better prognosis than patients with IDH wildtype status, typically seen in glioblastoma WHO grade IV. Here we used a prospective O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET) positron emission tomography guided single-voxel 1H-magnetic resonance spectroscopy approach to predict the IDH status before surgery. Finally, 34 patients were included in this neuroimaging study, of whom eight had additionally tissue analysis. Using a machine learning technique, we predicted IDH status with an accuracy of 88.2%, a sensitivity of 95.5% and a specificity of 75.0%. It was newly recognized, that two metabolites (myo-inositol and glycine) have a particularly important role in the determination of the IDH status. Abstract Isocitrate dehydrogenase (IDH)-1 mutation is an important prognostic factor and a potential therapeutic target in glioma. Immunohistological and molecular diagnosis of IDH mutation status is invasive. To avoid tumor biopsy, dedicated spectroscopic techniques have been proposed to detect D-2-hydroxyglutarate (2-HG), the main metabolite of IDH, directly in vivo. However, these methods are technically challenging and not broadly available. Therefore, we explored the use of machine learning for the non-invasive, inexpensive and fast diagnosis of IDH status in standard 1H-magnetic resonance spectroscopy (1H-MRS). To this end, 30 of 34 consecutive patients with known or suspected glioma WHO grade II-IV were subjected to metabolic positron emission tomography (PET) imaging with O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET) for optimized voxel placement in 1H-MRS. Routine 1H-magnetic resonance (1H-MR) spectra of tumor and contralateral healthy brain regions were acquired on a 3 Tesla magnetic resonance (3T-MR) scanner, prior to surgical tumor resection and molecular analysis of IDH status. Since 2-HG spectral signals were too overlapped for reliable discrimination of IDH mutated (IDHmut) and IDH wild-type (IDHwt) glioma, we used a nested cross-validation approach, whereby we trained a linear support vector machine (SVM) on the complete spectral information of the 1H-MRS data to predict IDH status. Using this approach, we predicted IDH status with an accuracy of 88.2%, a sensitivity of 95.5% (95% CI, 77.2–99.9%) and a specificity of 75.0% (95% CI, 42.9–94.5%), respectively. The area under the curve (AUC) amounted to 0.83. Subsequent ex vivo 1H-nuclear magnetic resonance (1H-NMR) measurements performed on metabolite extracts of resected tumor material (eight specimens) revealed myo-inositol (M-ins) and glycine (Gly) to be the major discriminators of IDH status. We conclude that our approach allows a reliable, non-invasive, fast and cost-effective prediction of IDH status in a standard clinical setting.
Collapse
|
11
|
Galldiks N, Abdulla DSY, Scheffler M, Wolpert F, Werner JM, Hüllner M, Stoffels G, Schweinsberg V, Schlaak M, Kreuzberg N, Landsberg J, Lohmann P, Ceccon G, Baues C, Trommer M, Celik E, Ruge MI, Kocher M, Marnitz S, Fink GR, Tonn JC, Weller M, Langen KJ, Wolf J, Mauch C. Treatment Monitoring of Immunotherapy and Targeted Therapy Using 18F-FET PET in Patients with Melanoma and Lung Cancer Brain Metastases: Initial Experiences. J Nucl Med 2020; 62:464-470. [PMID: 32887757 DOI: 10.2967/jnumed.120.248278] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/30/2020] [Indexed: 12/15/2022] Open
Abstract
We investigated the value of O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) PET for treatment monitoring of immune checkpoint inhibition (ICI) or targeted therapy (TT) alone or in combination with radiotherapy in patients with brain metastasis (BM) since contrast-enhanced MRI often remains inconclusive. Methods: We retrospectively identified 40 patients with 107 BMs secondary to melanoma (n = 29 with 75 BMs) or non-small cell lung cancer (n = 11 with 32 BMs) treated with ICI or TT who had 18F-FET PET (n = 60 scans) for treatment monitoring from 2015 to 2019. Most patients (n = 37; 92.5%) had radiotherapy during the course of the disease. In 27 patients, 18F-FET PET was used to differentiate treatment-related changes from BM relapse after ICI or TT. In 13 patients, 18F-FET PET was performed for response assessment to ICI or TT using baseline and follow-up scans (median time between scans, 4.2 mo). In all lesions, static and dynamic 18F-FET PET parameters were obtained (i.e., mean tumor-to-brain ratios [TBR], time-to-peak values). Diagnostic accuracies of PET parameters were evaluated by receiver-operating-characteristic analyses using the clinical follow-up or neuropathologic findings as a reference. Results: A TBR threshold of 1.95 differentiated BM relapse from treatment-related changes with an accuracy of 85% (P = 0.003). Metabolic responders to ICI or TT on 18F-FET PET had a significantly longer stable follow-up (threshold of TBR reduction relative to baseline, ≥10%; accuracy, 82%; P = 0.004). Furthermore, at follow-up, time to peak in metabolic responders increased significantly (P = 0.019). Conclusion: 18F-FET PET may add valuable information for treatment monitoring in BM patients treated with ICI or TT.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany .,Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Diana S Y Abdulla
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Lung Cancer Group, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Matthias Scheffler
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Lung Cancer Group, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Fabian Wolpert
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Jan-Michael Werner
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Martin Hüllner
- Department of Nuclear Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Viola Schweinsberg
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Max Schlaak
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nicole Kreuzberg
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jennifer Landsberg
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Dermatology, University Hospital Bonn, Bonn, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Garry Ceccon
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Christian Baues
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Maike Trommer
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Eren Celik
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Maximilian I Ruge
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Martin Kocher
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Simone Marnitz
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Jörg-Christian Tonn
- Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany; and
| | - Michael Weller
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany
| | - Jürgen Wolf
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Lung Cancer Group, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Cornelia Mauch
- Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
12
|
Grant R, Dowswell T, Tomlinson E, Brennan PM, Walter FM, Ben-Shlomo Y, Hunt DW, Bulbeck H, Kernohan A, Robinson T, Lawrie TA. Interventions to reduce the time to diagnosis of brain tumours. Cochrane Database Syst Rev 2020; 9:CD013564. [PMID: 32901926 PMCID: PMC8082957 DOI: 10.1002/14651858.cd013564.pub2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Brain tumours are recognised as one of the most difficult cancers to diagnose because presenting symptoms, such as headache, cognitive symptoms, and seizures, may be more commonly attributable to other, more benign conditions. Interventions to reduce the time to diagnosis of brain tumours include national awareness initiatives, expedited pathways, and protocols to diagnose brain tumours, based on a person's presenting symptoms and signs; and interventions to reduce waiting times for brain imaging pathways. If such interventions reduce the time to diagnosis, it may make it less likely that people experience clinical deterioration, and different treatment options may be available. OBJECTIVES To systematically evaluate evidence on the effectiveness of interventions that may influence: symptomatic participants to present early (shortening the patient interval), thresholds for primary care referral (shortening the primary care interval), and time to imaging diagnosis (shortening the secondary care interval and diagnostic interval). To produce a brief economic commentary, summarising the economic evaluations relevant to these interventions. SEARCH METHODS For evidence on effectiveness, we searched CENTRAL, MEDLINE, and Embase from January 2000 to January 2020; Clinicaltrials.gov to May 2020, and conference proceedings from 2014 to 2018. For economic evidence, we searched the UK National Health Services Economic Evaluation Database from 2000 to December 2014. SELECTION CRITERIA We planned to include studies evaluating any active intervention that may influence the diagnostic pathway, e.g. clinical guidelines, direct access imaging, public health campaigns, educational initiatives, and other interventions that might lead to early identification of primary brain tumours. We planned to include randomised and non-randomised comparative studies. Included studies would include people of any age, with a presentation that might suggest a brain tumour. DATA COLLECTION AND ANALYSIS Two review authors independently assessed titles identified by the search strategy, and the full texts of potentially eligible studies. We resolved discrepancies through discussion or, if required, by consulting another review author. MAIN RESULTS We did not identify any studies for inclusion in this review. We excluded 115 studies. The main reason for exclusion of potentially eligible intervention studies was their study design, due to a lack of control groups. We found no economic evidence to inform a brief economic commentary on this topic. AUTHORS' CONCLUSIONS In this version of the review, we did not identify any studies that met the review inclusion criteria for either effectiveness or cost-effectiveness. Therefore, there is no evidence from good quality studies on the best strategies to reduce the time to diagnosis of brain tumours, despite the prioritisation of research on early diagnosis by the James Lind Alliance in 2015. This review highlights the need for research in this area.
Collapse
Affiliation(s)
- Robin Grant
- Edinburgh Centre for Neuro-Oncology (ECNO), Western General Hospital, Edinburgh, UK
| | - Therese Dowswell
- C/o Cochrane Pregnancy and Childbirth Group, Department of Women's and Children's Health, The University of Liverpool, Liverpool, UK
| | - Eve Tomlinson
- Cochrane Gynaecological, Neuro-oncology and Orphan Cancers, 1st Floor Education Centre, Royal United Hospital, Bath, UK
| | - Paul M Brennan
- Translational Neurosurgery Department, Western General Hospital, Edinburgh, UK
| | - Fiona M Walter
- Public Health & Primary Care, University of Cambridge, Cambridge, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, Bristol, UK
| | - David William Hunt
- Foundation School/Dept of Clinical and Experimental Medicine, Royal Surrey County Hospital/University of Surrey, Guildford, UK
| | | | - Ashleigh Kernohan
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Tomos Robinson
- Institute of Health & Society, Newcastle University, Newcastle upon Tyne, UK
| | | |
Collapse
|
13
|
Galldiks N, Unterrainer M, Judov N, Stoffels G, Rapp M, Lohmann P, Vettermann F, Dunkl V, Suchorska B, Tonn JC, Kreth FW, Fink GR, Bartenstein P, Langen KJ, Albert NL. Photopenic defects on O-(2-[18F]-fluoroethyl)-L-tyrosine PET: clinical relevance in glioma patients. Neuro Oncol 2020; 21:1331-1338. [PMID: 31077276 DOI: 10.1093/neuonc/noz083] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND O-(2-[18F]-fluoroethyl)-L-tyrosine (FET) PET has a sensitivity of more than 90% to detect gliomas. In the remaining small fraction of gliomas without increased tracer uptake, some tumors even show photopenic defects whose clinical significance is unclear. METHODS Glioma patients with a negative FET PET scan prior to neuropathological confirmation were identified retrospectively. Gliomas were rated visually as (i) having indifferent FET uptake or (ii) photopenic, if FET uptake was below background activity. FET uptake in the area of signal hyperintensity on the T2/fluid attenuated inversion recovery-weighted MRI was evaluated by mean standardized uptake value (SUV) and mean tumor-to-brain ratio (TBR). The progression-free survival (PFS) of photopenic gliomas was compared with that of gliomas with indifferent FET uptake. RESULTS Of 100 FET-negative gliomas, 40 cases with photopenic defects were identified. Fifteen of these 40 cases (38%) had World Health Organization (WHO) grades III and IV gliomas. FET uptake in photopenic gliomas was significantly decreased compared with both the healthy-appearing brain tissue (SUV, 0.89 ± 0.26 vs 1.08 ± 0.23; P < 0.001) and gliomas with indifferent FET uptake (TBR, 0.82 ± 0.09 vs 0.96 ± 0.13; P < 0.001). Irrespective of the applied treatment, isocitrate dehydrogenase (IDH)-mutated WHO grade II diffuse astrocytoma patients with indifferent FET uptake (n = 25) had a significantly longer PFS than patients with IDH-mutated diffuse astrocytomas (WHO grade II) with photopenic defects (n = 11) (51 vs 24 mo; P = 0.027). The multivariate survival analysis indicated that photopenic defects predict an unfavorable PFS (P = 0.009). CONCLUSION Photopenic gliomas in negative FET PET scans should be managed more actively, as they seem to have a higher risk of harboring a higher-grade glioma and an unfavorable outcome.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany.,Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Düsseldorf, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich (LMU), Munich, Germany
| | - Natalie Judov
- Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany
| | - Marion Rapp
- Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany
| | - Franziska Vettermann
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich (LMU), Munich, Germany
| | - Veronika Dunkl
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | | | - Jörg C Tonn
- Department of Neurosurgery, LMU, Munich, Germany
| | | | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich (LMU), Munich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich (LMU), Munich, Germany
| |
Collapse
|
14
|
Kertels O, Kessler AF, Mihovilovic MI, Stolzenburg A, Linsenmann T, Samnick S, Brändlein S, Monoranu CM, Ernestus RI, Buck AK, Löhr M, Lapa C. Prognostic Value of O-(2-[ 18F]Fluoroethyl)-L-Tyrosine PET/CT in Newly Diagnosed WHO 2016 Grade II and III Glioma. Mol Imaging Biol 2020; 21:1174-1181. [PMID: 30977078 DOI: 10.1007/s11307-019-01357-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE The use of [18F]fluoroethyl)-L-tyrosine ([18F]FET) positron emission tomography/computed tomography (PET/CT) has proven valuable in brain tumor management. This study aimed to investigate the prognostic value of radiotracer uptake in newly diagnosed grade II or III gliomas according to the current 2016 World Health Organization (WHO) classification. PROCEDURES A total of 35 treatment-naive patients (mean age, 48 ± 17 years) with histologically proven WHO grade II or III gliomas as defined by the current 2016 WHO classification were included. Static PET/CT imaging was performed 20 min after intravenous [18F]FET injection. Images were assessed visually and semi-quantitatively using regions of interest for both tumor (SUVmax, SUVmean) and background (BKGmean) to calculate tumor-to-background (TBR) ratios. The association among histological results, molecular markers (including isocitrate dehydrogenase enzyme and methylguanine-DNA methyltransferase status), clinical features (age), and PET findings was tested and compared with outcome (progression-free [PFS] and overall survival [OS]). RESULTS Fourteen patients presented with grade II (diffuse astrocytoma n = 10, oligodendroglioma n = 4) and 21 patients with grade III glioma (anaplastic astrocytoma n = 15, anaplastic oligodendroglioma n = 6). Twenty-seven out of the 35 patients were PET-positive (grade II n = 8/14, grade III n = 19/21), with grade III tumors exhibiting significantly higher amino acid uptake (TBRmean and TBRmax; p = 0.03 and p = 0.02, respectively). PET-negative lesions demonstrated significantly prolonged PFS (p = 0.003) as compared to PET-positive gliomas. PET-positive disease had a complementary value in prognostication in addition to patient age, glioma grade, and molecular markers. CONCLUSIONS Amino acid uptake as assessed by [18F]FET-PET/CT imaging is useful as non-invasive read-out for tumor biology and prognosis in newly diagnosed, treatment-naive gliomas according to the 2016 WHO classification.
Collapse
Affiliation(s)
- Olivia Kertels
- Institute of Diagnostic Radiology, University Hospital Würzburg, Wurzburg, Germany
| | - Almuth F Kessler
- Department of Neurosurgery, University Hospital Würzburg, Wurzburg, Germany
| | - Milena I Mihovilovic
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Wurzburg, Germany
| | - Antje Stolzenburg
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Wurzburg, Germany
| | - Thomas Linsenmann
- Department of Neurosurgery, University Hospital Würzburg, Wurzburg, Germany
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Wurzburg, Germany
| | - Stephanie Brändlein
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Wurzburg, Germany
| | - Camelia Maria Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Wurzburg, Germany
| | - Ralf-Ingo Ernestus
- Department of Neurosurgery, University Hospital Würzburg, Wurzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Wurzburg, Germany
| | - Mario Löhr
- Department of Neurosurgery, University Hospital Würzburg, Wurzburg, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Wurzburg, Germany.
| |
Collapse
|
15
|
Bauer EK, Stoffels G, Blau T, Reifenberger G, Felsberg J, Werner JM, Lohmann P, Rosen J, Ceccon G, Tscherpel C, Rapp M, Sabel M, Filss CP, Shah NJ, Neumaier B, Fink GR, Langen KJ, Galldiks N. Prediction of survival in patients with IDH-wildtype astrocytic gliomas using dynamic O-(2-[ 18F]-fluoroethyl)-L-tyrosine PET. Eur J Nucl Med Mol Imaging 2020; 47:1486-1495. [PMID: 32034446 PMCID: PMC7188701 DOI: 10.1007/s00259-020-04695-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/12/2020] [Indexed: 11/27/2022]
Abstract
PURPOSE Integrated histomolecular diagnostics of gliomas according to the World Health Organization (WHO) classification of 2016 has refined diagnostic accuracy and prediction of prognosis. This study aimed at exploring the prognostic value of dynamic O-(2-[18F]-fluoroethyl)-L-tyrosine (FET) PET in newly diagnosed, histomolecularly classified astrocytic gliomas of WHO grades III or IV. METHODS Before initiation of treatment, dynamic FET PET imaging was performed in patients with newly diagnosed glioblastoma (GBM) and anaplastic astrocytoma (AA). Static FET PET parameters such as maximum and mean tumour/brain ratios (TBRmax/mean), the metabolic tumour volume (MTV) as well as the dynamic FET PET parameters time-to-peak (TTP) and slope, were obtained. The predictive ability of FET PET parameters was evaluated concerning the progression-free and overall survival (PFS, OS). Using ROC analyses, threshold values for FET PET parameters were obtained. Subsequently, univariate Kaplan-Meier and multivariate Cox regression survival analyses were performed to assess the predictive power of these parameters for survival. RESULTS Sixty patients (45 GBM and 15 AA patients) of two university centres were retrospectively identified. Patients with isocitrate dehydrogenase (IDH)-mutant or O6-methylguanine-DNA-methyltransferase (MGMT) promoter-methylated tumours had a significantly longer PFS and OS (both P < 0.001). Furthermore, ROC analysis of IDH-wildtype glioma patients (n = 45) revealed that a TTP > 25 min (AUC, 0.90; sensitivity, 90%; specificity, 87%; P < 0.001) was highly prognostic for longer PFS (13 vs. 7 months; P = 0.005) and OS (29 vs. 12 months; P < 0.001). In contrast, at a lower level of significance, TBRmax, TBRmean, and MTV were only prognostic for longer OS (P = 0.004, P = 0.038, and P = 0.048, respectively). Besides complete resection and a methylated MGMT promoter, TTP remained significant in multivariate survival analysis (all P ≤ 0.02), indicating an independent predictor for OS. CONCLUSIONS Our data suggest that dynamic FET PET allows the identification of patients with longer OS among patients with newly diagnosed IDH-wildtype GBM and AA.
Collapse
Affiliation(s)
- Elena K Bauer
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937, Cologne, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Research Centre Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Tobias Blau
- Department of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute of Neuropathology, University Hospital Essen, Essen, Germany
| | - Guido Reifenberger
- Institute of Neuropathology, Heinrich Heine University, Duesseldorf, Germany.,Center of Integrated Oncology (CIO), University of Duesseldorf, Duesseldorf, Germany
| | - Jörg Felsberg
- Institute of Neuropathology, Heinrich Heine University, Duesseldorf, Germany.,Center of Integrated Oncology (CIO), University of Duesseldorf, Duesseldorf, Germany
| | - Jan M Werner
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937, Cologne, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Research Centre Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Jurij Rosen
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937, Cologne, Germany
| | - Garry Ceccon
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937, Cologne, Germany
| | - Caroline Tscherpel
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937, Cologne, Germany
| | - Marion Rapp
- Center of Integrated Oncology (CIO), University of Duesseldorf, Duesseldorf, Germany.,Department of Neurosurgery, Heinrich Heine University, Duesseldorf, Germany
| | - Michael Sabel
- Center of Integrated Oncology (CIO), University of Duesseldorf, Duesseldorf, Germany.,Department of Neurosurgery, Heinrich Heine University, Duesseldorf, Germany
| | - Christian P Filss
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Research Centre Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany.,Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Research Centre Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany.,Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Research Centre Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4, -5), Research Centre Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Research Centre Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany.,Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany.,Center of Integrated Oncology (CIO), University of Aachen, Aachen, Germany
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937, Cologne, Germany. .,Institute of Neuroscience and Medicine (INM-3, -4, -5), Research Centre Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany. .,Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany.
| |
Collapse
|
16
|
Lohmeier J, Bohner G, Siebert E, Brenner W, Hamm B, Makowski MR. Quantitative biparametric analysis of hybrid 18F-FET PET/MR-neuroimaging for differentiation between treatment response and recurrent glioma. Sci Rep 2019; 9:14603. [PMID: 31601829 PMCID: PMC6787240 DOI: 10.1038/s41598-019-50182-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 09/04/2019] [Indexed: 11/09/2022] Open
Abstract
We investigated the diagnostic potential of simultaneous 18F-FET PET/MR-imaging for differentiation between recurrent glioma and post-treatment related effects (PTRE) using quantitative volumetric (3D-VOI) lesion analysis. In this retrospective study, a total of 42 patients including 32 patients with histologically proven glioma relapse and 10 patients with PTRE (histopathologic follow-up, n = 4, serial imaging follow-up, n = 6) were evaluated regarding recurrence. PET/MR-imaging was semi-automatically analysed based on FET tracer uptake using conservative SUV thresholding (isocontour 80%) with emphasis on the metabolically most active regions. Mean (relative) apparent diffusion coefficient (ADCmean, rADCmean), standardised-uptake-value (SUV) including target-to-background (TBR) ratio were determined. Glioma relapse presented higher ADCmean (MD ± SE, 284 ± 91, p = 0.003) and TBRmax (MD ± SE, 1.10 ± 0.45, p = 0.02) values than treatment-related changes. Both ADCmean (AUC ± SE = 0.82 ± 0.07, p-value < 0.001) and TBRmax (AUC ± SE = 0.81 ± 0.08, p-value < 0.001) achieved reliable diagnostic performance in differentiating glioma recurrence from PTRE. Bivariate analysis based on a combination of ADCmean and TBRmax demonstrated highest diagnostic accuracy (AUC ± SE = 0.90 ± 0.05, p-value < 0.001), improving clinical (false negative and false positive) classification. In conclusion, biparametric analysis using DWI and FET PET, both providing distinct information regarding the underlying pathophysiology, presented best diagnostic accuracy and clinical benefit in differentiating recurrent glioma from treatment-related changes.
Collapse
Affiliation(s)
- Johannes Lohmeier
- Charité Universitätsmedizin Berlin, Department of Radiology, Campus Charité Mitte (CCM), Charitéplatz 1, 10117, Berlin, Germany.
| | - Georg Bohner
- Charité Universitätsmedizin Berlin, Department of Neuroradiology, Campus Charité Mitte (CCM), Charitéplatz 1, 10117, Berlin, Germany
| | - Eberhard Siebert
- Charité Universitätsmedizin Berlin, Department of Neuroradiology, Campus Charité Mitte (CCM), Charitéplatz 1, 10117, Berlin, Germany
| | - Winfried Brenner
- Charité Universitätsmedizin Berlin, Department of Nuclear Medicine, Campus Virchow-Klinikum (CVK), Augustenburger Platz 1, 13353, Berlin, Germany
| | - Bernd Hamm
- Charité Universitätsmedizin Berlin, Department of Radiology, Campus Charité Mitte (CCM), Charitéplatz 1, 10117, Berlin, Germany
| | - Marcus R Makowski
- Charité Universitätsmedizin Berlin, Department of Radiology, Campus Charité Mitte (CCM), Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
17
|
Influence of Dexamethasone on O-(2-[ 18F]-Fluoroethyl)-L-Tyrosine Uptake in the Human Brain and Quantification of Tumor Uptake. Mol Imaging Biol 2019; 21:168-174. [PMID: 29845426 DOI: 10.1007/s11307-018-1221-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
PURPOSE O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) is an established positron emission tomography (PET) tracer for brain tumor imaging. This study explores the influence of dexamethasone therapy on [18F]FET uptake in the normal brain and its influence on the maximum and mean tumor-to-brain ratio (TBR). PROCEDURES [18F]FET PET scans of 160 brain tumor patients were evaluated (80 dexamethasone treated, 80 untreated; each group with 40 men/40 women). The standardized uptake value of [18F]FET uptake in the normal brain (SUVbrain) in the different groups was compared. Nine patients were examined repeatedly with and without dexamethasone therapy. RESULTS SUVbrain of [18F]FET uptake was significantly higher in dexamethasone-treated patients than in untreated patients (SUVbrain 1.33 ± 0.1 versus 1.06 ± 0.16 in male and 1.45 ± 0.25 versus 1.31 ± 0.28 in female patients). Similar results were observed in patients with serial PET scans. Furthermore, compared to men, a significantly higher SUVbrain was found in women, both with and without dexamethasone treatment. There were no significant differences between the different groups for TBRmax and TBRmean, which could have been masked by the high standard deviation. In a patient with a stable brain metastasis investigated twice with and without dexamethasone, the TBRmax and the biological tumor volume (BTV) decreased considerably after dexamethasone due to an increased SUVbrain. CONCLUSION Dexamethasone treatment appears to increase the [18F]FET uptake in the normal brain. An effect on TBRmax, TBRmean, and BTV cannot be excluded which should be considered especially for treatment monitoring and the estimation of BTV using [18F]FET PET.
Collapse
|
18
|
Investigation of cis-4-[ 18F]Fluoro-D-Proline Uptake in Human Brain Tumors After Multimodal Treatment. Mol Imaging Biol 2019; 20:1035-1043. [PMID: 29687323 DOI: 10.1007/s11307-018-1197-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
PURPOSE Cis-4-[18F]fluoro-D-proline (D-cis-[18F]FPro) has been shown to pass the intact blood-brain barrier and to accumulate in areas of secondary neurodegeneration and necrosis in the rat brain while uptake in experimental brain tumors is low. This pilot study explores the uptake behavior of D-cis-[18F]FPro in human brain tumors after multimodal treatment. PROCEDURES In a prospective study, 27 patients with suspected recurrent brain tumor after treatment with surgery, radiotherapy, and/or chemotherapy (SRC) were investigated by dynamic positron emission tomography (PET) using D-cis-[18F]FPro (22 high-grade gliomas, one unspecified glioma, and 4 metastases). Furthermore, two patients with untreated lesions were included (one glioblastoma, one reactive astrogliosis). Data were compared with the results of PET using O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) which detects viable tumor tissue. Tracer distribution, mean and maximum lesion-to-brain ratios (LBRmean, LBRmax), and time-to-peak (TTP) of the time activity curve (TAC) of tracer uptake were evaluated. Final diagnosis was determined by histology (n = 9), clinical follow-up (n = 10), or by [18F]FET PET (n = 10). RESULTS D-cis-[18F]FPro showed high uptake in both recurrent brain tumors (n = 11) and lesions classified as treatment-related changes (TRC) only (n = 16) (LBRmean 2.2 ± 0.7 and 2.1 ± 0.6, n.s.; LBRmax 3.4 ± 1.2 and 3.2 ± 1.3, n.s.). The untreated glioblastoma and the lesion showing reactive astrogliosis exhibited low D-cis-[18F]FPro uptake. Distribution of [18F]FET and D-cis-[18F]FPro uptake was discordant in 21/29 cases indicating that the uptake mechanisms are different. CONCLUSION The high accumulation of D-cis-[18F]FPro in pretreated brain tumors and TRC supports the hypothesis that tracer uptake is related to cell death. Further studies before and after therapy are needed to assess the potential of D-cis-[18F]FPro for treatment monitoring.
Collapse
|
19
|
Peeken JC, Goldberg T, Pyka T, Bernhofer M, Wiestler B, Kessel KA, Tafti PD, Nüsslin F, Braun AE, Zimmer C, Rost B, Combs SE. Combining multimodal imaging and treatment features improves machine learning-based prognostic assessment in patients with glioblastoma multiforme. Cancer Med 2018; 8:128-136. [PMID: 30561851 PMCID: PMC6346243 DOI: 10.1002/cam4.1908] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 12/22/2022] Open
Abstract
Background For Glioblastoma (GBM), various prognostic nomograms have been proposed. This study aims to evaluate machine learning models to predict patients' overall survival (OS) and progression‐free survival (PFS) on the basis of clinical, pathological, semantic MRI‐based, and FET‐PET/CT‐derived information. Finally, the value of adding treatment features was evaluated. Methods One hundred and eighty‐nine patients were retrospectively analyzed. We assessed clinical, pathological, and treatment information. The VASARI set of semantic imaging features was determined on MRIs. Metabolic information was retained from preoperative FET‐PET/CT images. We generated multiple random survival forest prediction models on a patient training set and performed internal validation. Single feature class models were created including "clinical," "pathological," "MRI‐based," and "FET‐PET/CT‐based" models, as well as combinations. Treatment features were combined with all other features. Results Of all single feature class models, the MRI‐based model had the highest prediction performance on the validation set for OS (C‐index: 0.61 [95% confidence interval: 0.51‐0.72]) and PFS (C‐index: 0.61 [0.50‐0.72]). The combination of all features did increase performance above all single feature class models up to C‐indices of 0.70 (0.59‐0.84) and 0.68 (0.57‐0.78) for OS and PFS, respectively. Adding treatment information further increased prognostic performance up to C‐indices of 0.73 (0.62‐0.84) and 0.71 (0.60‐0.81) on the validation set for OS and PFS, respectively, allowing significant stratification of patient groups for OS. Conclusions MRI‐based features were the most relevant feature class for prognostic assessment. Combining clinical, pathological, and imaging information increased predictive power for OS and PFS. A further increase was achieved by adding treatment features.
Collapse
Affiliation(s)
- Jan C Peeken
- Department of Radiation Oncology, Klinikum rechts der Isar der Technischem Universität München (TUM), München, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany.,Department of Radiation Sciences (DRS), Institute of Innovative Radiotherapy (iRT), Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Thomas Pyka
- Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München (TUM), Munich, Germany
| | - Michael Bernhofer
- Department for Bioinformatics and Computational Biology, Technical University of Munich (TUM), Garching, Germany
| | - Benedikt Wiestler
- Department of Neuroradiology, Klinikum rechts der Isar der Technischen Universität, Munich (TUM), München, Germany
| | - Kerstin A Kessel
- Department of Radiation Oncology, Klinikum rechts der Isar der Technischem Universität München (TUM), München, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany.,Department of Radiation Sciences (DRS), Institute of Innovative Radiotherapy (iRT), Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Fridtjof Nüsslin
- Department of Radiation Oncology, Klinikum rechts der Isar der Technischem Universität München (TUM), München, Germany
| | | | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar der Technischen Universität, Munich (TUM), München, Germany
| | - Burkhard Rost
- Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München (TUM), Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar der Technischem Universität München (TUM), München, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany.,Department of Radiation Sciences (DRS), Institute of Innovative Radiotherapy (iRT), Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
20
|
Law I, Albert NL, Arbizu J, Boellaard R, Drzezga A, Galldiks N, la Fougère C, Langen KJ, Lopci E, Lowe V, McConathy J, Quick HH, Sattler B, Schuster DM, Tonn JC, Weller M. Joint EANM/EANO/RANO practice guidelines/SNMMI procedure standards for imaging of gliomas using PET with radiolabelled amino acids and [ 18F]FDG: version 1.0. Eur J Nucl Med Mol Imaging 2018; 46:540-557. [PMID: 30519867 PMCID: PMC6351513 DOI: 10.1007/s00259-018-4207-9] [Citation(s) in RCA: 361] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 10/29/2018] [Indexed: 01/12/2023]
Abstract
These joint practice guidelines, or procedure standards, were developed collaboratively by the European Association of Nuclear Medicine (EANM), the Society of Nuclear Medicine and Molecular Imaging (SNMMI), the European Association of Neurooncology (EANO), and the working group for Response Assessment in Neurooncology with PET (PET-RANO). Brain PET imaging is being increasingly used to supplement MRI in the clinical management of glioma. The aim of these standards/guidelines is to assist nuclear medicine practitioners in recommending, performing, interpreting and reporting the results of brain PET imaging in patients with glioma to achieve a high-quality imaging standard for PET using FDG and the radiolabelled amino acids MET, FET and FDOPA. This will help promote the appropriate use of PET imaging and contribute to evidence-based medicine that may improve the diagnostic impact of this technique in neurooncological practice. The present document replaces a former version of the guidelines published in 2006 (Vander Borght et al. Eur J Nucl Med Mol Imaging. 33:1374–80, 2006), and supplements a recent evidence-based recommendation by the PET-RANO working group and EANO on the clinical use of PET imaging in patients with glioma (Albert et al. Neuro Oncol. 18:1199–208, 2016). The information provided should be taken in the context of local conditions and regulations.
Collapse
Affiliation(s)
- Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, 9, Blegdamsvej, 2100-DK, Copenhagen Ø, Denmark.
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Javier Arbizu
- Department of Nuclear Medicine, Clínica Universidad de Navarra, University of Navarre, Pamplona, Spain
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands.,Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Norbert Galldiks
- Department of Neurology, University Hospital Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Julich, Julich, Germany
| | - Christian la Fougère
- Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, University of Tübingen, Tübingen, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Julich, Julich, Germany.,Department of Nuclear Medicine, RWTH University Aachen, Aachen, Germany
| | - Egesta Lopci
- Department of Nuclear Medicine, Humanitas Clinical and Research Hospital, Rozzano, Italy
| | - Val Lowe
- Department of Radiology, Nuclear Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jonathan McConathy
- Division of Molecular Imaging and Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Harald H Quick
- High-Field and Hybrid MR Imaging, University Hospital Essen, Essen, Germany
| | - Bernhard Sattler
- Department for Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - David M Schuster
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Jörg-Christian Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michael Weller
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
Lohmann P, Stavrinou P, Lipke K, Bauer EK, Ceccon G, Werner JM, Neumaier B, Fink GR, Shah NJ, Langen KJ, Galldiks N. FET PET reveals considerable spatial differences in tumour burden compared to conventional MRI in newly diagnosed glioblastoma. Eur J Nucl Med Mol Imaging 2018; 46:591-602. [PMID: 30327856 DOI: 10.1007/s00259-018-4188-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/03/2018] [Indexed: 01/02/2023]
Abstract
PURPOSE Areas of contrast enhancement (CE) on MRI are usually the target for resection or radiotherapy target volume definition in glioblastomas. However, the solid tumour mass may extend beyond areas of CE. Amino acid PET can detect parts of the tumour that show no CE. We systematically investigated tumour volumes delineated by amino acid PET and MRI in patients with newly diagnosed, untreated glioblastoma. METHODS Preoperatively, 50 patients with neuropathologically confirmed glioblastoma underwent O-(2-[18F]-fluoroethyl)-L-tyrosine (FET) PET, and fluid-attenuated inversion recovery (FLAIR) and contrast-enhanced MRI. Areas of CE were manually segmented. FET PET tumour volumes were segmented using a tumour-to-brain ratio of ≥1.6. The percentage overlap volumes, and Dice and Jaccard spatial similarity coefficients (DSC, JSC) were calculated. FLAIR images were evaluated visually. RESULTS In 43 patients (86%), the FET tumour volume was significantly larger than the CE volume (21.5 ± 14.3 mL vs. 9.4 ± 11.3 mL; P < 0.001). Forty patients (80%) showed both increased uptake of FET and CE. In these 40 patients, the spatial similarity between FET uptake and CE was low (mean DSC 0.39 ± 0.21, mean JSC 0.26 ± 0.16). Ten patients (20%) showed no CE, and one of these patients showed no FET uptake. In five patients (10%), increased FET uptake was present outside areas of FLAIR hyperintensity. CONCLUSION Our results show that the metabolically active tumour volume delineated by FET PET is significantly larger than tumour volume delineated by CE. Furthermore, the results strongly suggest that the information derived from both imaging modalities should be integrated into the management of patients with newly diagnosed glioblastoma.
Collapse
Affiliation(s)
- Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, 52425, Juelich, Germany.
| | | | - Katharina Lipke
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, 52425, Juelich, Germany
| | - Elena K Bauer
- Department of Neurology, University of Cologne, Cologne, Germany
| | - Garry Ceccon
- Department of Neurology, University of Cologne, Cologne, Germany
| | | | - Bernd Neumaier
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, 52425, Juelich, Germany
| | - Gereon R Fink
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, 52425, Juelich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, 52425, Juelich, Germany.,Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, 52425, Juelich, Germany.,Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, 52425, Juelich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany.,Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany
| |
Collapse
|
22
|
Voxel-wise analysis of dynamic 18F-FET PET: a novel approach for non-invasive glioma characterisation. EJNMMI Res 2018; 8:91. [PMID: 30203138 PMCID: PMC6131687 DOI: 10.1186/s13550-018-0444-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/26/2018] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Glioma grading with dynamic 18F-FET PET (0-40 min p.i.) is typically performed by analysing the mean time-activity curve of the entire tumour or a suspicious area within a heterogeneous tumour. This work aimed to ensure a reader-independent glioma characterisation and identification of aggressive sub-volumes by performing a voxel-based analysis with diagnostically relevant kinetic and static 18F-FET PET parameters. One hundred sixty-two patients with a newly diagnosed glioma classified according to histologic and molecular genetic properties were evaluated. The biological tumour volume (BTV) was segmented in static 20-40 min p.i. 18F-FET PET images using the established threshold of 1.6 × background activity. For each enclosed voxel, the time-to-peak (TTP), the late slope (Slope15-40), and the tumour-to-background ratios (TBR5-15, TBR20-40) obtained from 5 to 15 min p.i. and 20 to 40 min p.i. images were determined. The percentage portion of these values within the BTV was evaluated with percentage volume fractions (PVFs) and cumulated percentage volume histograms (PVHs). The ability to differentiate histologic and molecular genetic classes was assessed and compared to volume-of-interest (VOI)-based parameters. RESULTS Aggressive WHO grades III and IV and IDH-wildtype gliomas were dominated by a high proportion of voxels with an early peak, negative slope, and high TBR, whereby the PVHs with TTP < 20 min p.i., Slope15-40 < 0 SUV/h, and TBR5-15 and TBR20-40 > 2 yielded the most significant differences between glioma grades. We found significant differences of the parameters between WHO grades and IDH mutation status, where the effect size was predominantly higher for voxel-based PVHs compared to the corresponding VOI-based parameters. A low overlap of BTV sub-volumes defined by TTP < 20 min p.i. and negative Slope15-40 with TBR5-15 > 2- and TBR20-40 > 2-defined hotspots was observed. CONCLUSIONS The presented approach applying voxel-wise analysis of dynamic 18F-FET PET enables an enhanced characterisation of gliomas and might potentially provide a fast identification of aggressive sub-volumes within the BTV. Parametric 3D 18F-FET PET information as investigated in this study has the potential to guide individual therapy instrumentation and may be included in future biopsy studies.
Collapse
|
23
|
Lohmann P, Lerche C, Bauer EK, Steger J, Stoffels G, Blau T, Dunkl V, Kocher M, Viswanathan S, Filss CP, Stegmayr C, Ruge MI, Neumaier B, Shah NJ, Fink GR, Langen KJ, Galldiks N. Predicting IDH genotype in gliomas using FET PET radiomics. Sci Rep 2018; 8:13328. [PMID: 30190592 PMCID: PMC6127131 DOI: 10.1038/s41598-018-31806-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/28/2018] [Indexed: 01/22/2023] Open
Abstract
Mutations in the isocitrate dehydrogenase (IDH mut) gene have gained paramount importance for the prognosis of glioma patients. To date, reliable techniques for a preoperative evaluation of IDH genotype remain scarce. Therefore, we investigated the potential of O-(2-[18F]fluoroethyl)-L-tyrosine (FET) PET radiomics using textural features combined with static and dynamic parameters of FET uptake for noninvasive prediction of IDH genotype. Prior to surgery, 84 patients with newly diagnosed and untreated gliomas underwent FET PET using a standard scanner (15 of 56 patients with IDH mut) or a dedicated high-resolution hybrid PET/MR scanner (11 of 28 patients with IDH mut). Static, dynamic and textural parameters of FET uptake in the tumor area were evaluated. Diagnostic accuracy of the parameters was evaluated using the neuropathological result as reference. Additionally, FET PET and textural parameters were combined to further increase the diagnostic accuracy. The resulting models were validated using cross-validation. Independent of scanner type, the combination of standard PET parameters with textural features increased significantly diagnostic accuracy. The highest diagnostic accuracy of 93% for prediction of IDH genotype was achieved with the hybrid PET/MR scanner. Our findings suggest that the combination of conventional FET PET parameters with textural features provides important diagnostic information for the non-invasive prediction of the IDH genotype.
Collapse
Affiliation(s)
- Philipp Lohmann
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany.
- Dept. of Stereotaxy and Functional Neurosurgery, University of Cologne, Cologne, Germany.
| | - Christoph Lerche
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
| | - Elena K Bauer
- Dept. of Neurology, University of Cologne, Cologne, Germany
| | - Jan Steger
- Dept. of Neurology, University of Cologne, Cologne, Germany
| | - Gabriele Stoffels
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
| | - Tobias Blau
- Dept. of Neuropathology, University of Cologne, Cologne, Germany
| | - Veronika Dunkl
- Dept. of Neurology, University of Cologne, Cologne, Germany
| | - Martin Kocher
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
- Dept. of Stereotaxy and Functional Neurosurgery, University of Cologne, Cologne, Germany
| | - Shivakumar Viswanathan
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
| | - Christian P Filss
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
| | - Carina Stegmayr
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
| | - Maximillian I Ruge
- Dept. of Stereotaxy and Functional Neurosurgery, University of Cologne, Cologne, Germany
| | - Bernd Neumaier
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
| | - Nadim J Shah
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
- Dept. of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Gereon R Fink
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
- Dept. of Neurology, University of Cologne, Cologne, Germany
| | - Karl-Josef Langen
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
- Dept. of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Norbert Galldiks
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
- Dept. of Neurology, University of Cologne, Cologne, Germany
- Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany
| |
Collapse
|
24
|
Lohmann P, Kocher M, Ceccon G, Bauer EK, Stoffels G, Viswanathan S, Ruge MI, Neumaier B, Shah NJ, Fink GR, Langen KJ, Galldiks N. Combined FET PET/MRI radiomics differentiates radiation injury from recurrent brain metastasis. NEUROIMAGE-CLINICAL 2018; 20:537-542. [PMID: 30175040 PMCID: PMC6118093 DOI: 10.1016/j.nicl.2018.08.024] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/01/2018] [Accepted: 08/17/2018] [Indexed: 12/12/2022]
Abstract
Background The aim of this study was to investigate the potential of combined textural feature analysis of contrast-enhanced MRI (CE-MRI) and static O-(2-[18F]fluoroethyl)-L-tyrosine (FET) PET for the differentiation between local recurrent brain metastasis and radiation injury since CE-MRI often remains inconclusive. Methods Fifty-two patients with new or progressive contrast-enhancing brain lesions on MRI after radiotherapy (predominantly stereotactic radiosurgery) of brain metastases were additionally investigated using FET PET. Based on histology (n = 19) or clinicoradiological follow-up (n = 33), local recurrent brain metastases were diagnosed in 21 patients (40%) and radiation injury in 31 patients (60%). Forty-two textural features were calculated on both unfiltered and filtered CE-MRI and summed FET PET images (20–40 min p.i.), using the software LIFEx. After feature selection, logistic regression models using a maximum of five features to avoid overfitting were calculated for each imaging modality separately and for the combined FET PET/MRI features. The resulting models were validated using cross-validation. Diagnostic accuracies were calculated for each imaging modality separately as well as for the combined model. Results For the differentiation between radiation injury and recurrence of brain metastasis, textural features extracted from CE-MRI had a diagnostic accuracy of 81% (sensitivity, 67%; specificity, 90%). FET PET textural features revealed a slightly higher diagnostic accuracy of 83% (sensitivity, 88%; specificity, 75%). However, the highest diagnostic accuracy was obtained when combining CE-MRI and FET PET features (accuracy, 89%; sensitivity, 85%; specificity, 96%). Conclusions Our findings suggest that combined FET PET/CE-MRI radiomics using textural feature analysis offers a great potential to contribute significantly to the management of patients with brain metastases. Differentiation between brain metastasis recurrence and radiation injury is of high clinical importance. Differentiation based on contrast-enhanced conventional MRI is often inconclusive. Radiomics and hybrid amino acid PET/MR imaging are increasingly gaining attention in Neuro-Oncology. We investigated the potential of combined PET/MRI radiomics analysis using MRI and FET PET in patients with brain metastases. Combined PET/MRI radiomics allows the differentiation of brain metastasis recurrence from radiation injury with high accuracy.
Collapse
Affiliation(s)
- Philipp Lohmann
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Stereotaxy and Functional Neurosurgery, University of Cologne, Cologne, Germany.
| | - Martin Kocher
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Stereotaxy and Functional Neurosurgery, University of Cologne, Cologne, Germany
| | - Garry Ceccon
- Dept. of Neurology, University of Cologne, Cologne, Germany
| | - Elena K Bauer
- Dept. of Neurology, University of Cologne, Cologne, Germany
| | - Gabriele Stoffels
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
| | - Shivakumar Viswanathan
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
| | - Maximilian I Ruge
- Dept. of Stereotaxy and Functional Neurosurgery, University of Cologne, Cologne, Germany
| | - Bernd Neumaier
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany
| | - Nadim J Shah
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Gereon R Fink
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Neurology, University of Cologne, Cologne, Germany
| | - Karl-Josef Langen
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Norbert Galldiks
- Inst. of Neuroscience and Medicine (INM-3, -4, -5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Neurology, University of Cologne, Cologne, Germany; Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany
| |
Collapse
|
25
|
Lohmann P, Piroth MD, Sellhaus B, Weis J, Geisler S, Oros-Peusquens AM, Mohlberg H, Amunts K, Shah NJ, Galldiks N, Langen KJ. Correlation of Dynamic O-(2-[ 18F]Fluoroethyl)-L-Tyrosine Positron Emission Tomography, Conventional Magnetic Resonance Imaging, and Whole-Brain Histopathology in a Pretreated Glioblastoma: A Postmortem Study. World Neurosurg 2018; 119:e653-e660. [PMID: 30077752 DOI: 10.1016/j.wneu.2018.07.232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Amino acid positron emission tomography (PET) using O-(2-[18F]fluoroethyl)-L-tyrosine (FET) provides important additional information on the extent of viable tumor tissue of glioblastoma compared with magnetic resonance imaging (MRI). Especially after radiochemotherapy, progression of contrast enhancement in MRI is equivocal and may represent either tumor progression or treatment-related changes. Here, the first case comparing postmortem whole-brain histology of a patient with pretreated glioblastoma with dynamic in vivo FET PET and MRI is presented. METHODS A 61-year-old patient with glioblastoma initially underwent partial tumor resection and died 11 weeks after completion of chemoradiation with concurrent temozolomide. Three days before the patient died, a follow-up FET PET and MRI scan indicated tumor progression. Autopsy was performed 48 hours after death. After formalin fixation, a 7-cm bihemispherical segment of the brain containing the entire tumor mass was cut into 3500 consecutive 20μm coronal sections. Representative sections were stained with hematoxylin and eosin stain, cresyl violet, and glial fibrillary acidic protein immunohistochemistry. An experienced neuropathologist identified areas of dense and diffuse neoplastic infiltration, astrogliosis, and necrosis. In vivo FET PET, MRI datasets, and postmortem histology were co-registered and compared by 3 experienced physicians. RESULTS Increased uptake of FET in the area of equivocal contrast enhancement on MRI correlated very well with dense infiltration by vital tumor cells and showed tracer kinetics typical for malignant gliomas. An area of predominantly reactive astrogliosis showed only moderate uptake of FET and tracer kinetics usually observed in benign lesions. CONCLUSIONS This case report impressively documents the correct imaging of a progressive glioblastoma by FET PET.
Collapse
Affiliation(s)
- Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany.
| | - Marc D Piroth
- Department of Radiation Oncology, HELIOS Hospital Wuppertal, Wuppertal, Germany; Department of Radiation Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Bernd Sellhaus
- Institute of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Joachim Weis
- Institute of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Stefanie Geisler
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany
| | - Ana-Maria Oros-Peusquens
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany
| | - Hartmut Mohlberg
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany
| | - Katrin Amunts
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany; Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Center of Integrated Oncology, Universities of Cologne and Bonn, Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany; Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
26
|
Koopman T, Verburg N, Schuit RC, Pouwels PJW, Wesseling P, Windhorst AD, Hoekstra OS, de Witt Hamer PC, Lammertsma AA, Boellaard R, Yaqub M. Quantification of O-(2-[ 18F]fluoroethyl)-L-tyrosine kinetics in glioma. EJNMMI Res 2018; 8:72. [PMID: 30066053 PMCID: PMC6068050 DOI: 10.1186/s13550-018-0418-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND This study identified the optimal tracer kinetic model for quantification of dynamic O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) positron emission tomography (PET) studies in seven patients with diffuse glioma (four glioblastoma, three lower grade glioma). The performance of more simplified approaches was evaluated by comparison with the optimal compartment model. Additionally, the relationship with cerebral blood flow-determined by [15O]H2O PET-was investigated. RESULTS The optimal tracer kinetic model was the reversible two-tissue compartment model. Agreement analysis of binding potential estimates derived from reference tissue input models with the distribution volume ratio (DVR)-1 derived from the plasma input model showed no significant average difference and limits of agreement of - 0.39 and 0.37. Given the range of DVR-1 (- 0.25 to 1.5), these limits are wide. For the simplified methods, the 60-90 min tumour-to-blood ratio to parent plasma concentration yielded the highest correlation with volume of distribution VT as calculated by the plasma input model (r = 0.97). The 60-90 min standardized uptake value (SUV) showed better correlation with VT (r = 0.77) than SUV based on earlier intervals. The 60-90 min SUV ratio to contralateral healthy brain tissue showed moderate agreement with DVR with no significant average difference and limits of agreement of - 0.24 and 0.30. A significant but low correlation was found between VT and CBF in the tumour regions (r = 0.61, p = 0.007). CONCLUSION Uptake of [18F]FET was best modelled by a reversible two-tissue compartment model. Reference tissue input models yielded estimates of binding potential which did not correspond well with plasma input-derived DVR-1. In comparison, SUV ratio to contralateral healthy brain tissue showed slightly better performance, if measured at the 60-90 min interval. SUV showed only moderate correlation with VT. VT shows correlation with CBF in tumour.
Collapse
Affiliation(s)
- Thomas Koopman
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Niels Verburg
- Neurosurgical Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Robert C. Schuit
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Petra J. W. Pouwels
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Pathology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Albert D. Windhorst
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Otto S. Hoekstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Philip C. de Witt Hamer
- Neurosurgical Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Adriaan A. Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| |
Collapse
|
27
|
Parent EE, Benayoun M, Ibeanu I, Olson JJ, Hadjipanayis CG, Brat DJ, Adhikarla V, Nye J, Schuster DM, Goodman MM. [ 18F]Fluciclovine PET discrimination between high- and low-grade gliomas. EJNMMI Res 2018; 8:67. [PMID: 30046944 PMCID: PMC6060188 DOI: 10.1186/s13550-018-0415-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/27/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The ability to accurately and non-invasively distinguish high-grade glioma from low-grade glioma remains a challenge despite advances in molecular and magnetic resonance imaging. We investigated the ability of fluciclovine (18F) PET as a means to identify and distinguish these lesions in patients with known gliomas and to correlate uptake with Ki-67. RESULTS Sixteen patients with a total of 18 newly diagnosed low-grade gliomas (n = 6) and high grade gliomas (n = 12) underwent fluciclovine PET imaging after histopathologic assessment. Fluciclovine PET analysis comprised tumor SUVmax and SUVmean, as well as metabolic tumor thresholds (1.3*, 1.6*, 1.9*) to normal brain background (TBmax, and TBmean). Comparison was additionally made to the proliferative status of the tumor as indicated by Ki-67 values. Fluciclovine uptake greater than normal brain parenchyma was found in all lesions studied. Time activity curves demonstrated statistically apparent flattening of the curves for both high-grade gliomas and low-grade gliomas starting 30 min after injection, suggesting an influx/efflux equilibrium. The best semiquantitative metric in discriminating HGG from LGG was obtained utilizing a metabolic 1 tumor threshold of 1.3* contralateral normal brain parenchyma uptake to create a tumor: background (TBmean1.3) cutoff of 2.15 with an overall sensitivity of 97.5% and specificity of 95.5%. Additionally, using a SUVmax > 4.3 cutoff gave a sensitivity of 90.9% and specificity of 97.5%. Tumor SUVmean and tumor SUVmax as a ratio to mean normal contralateral brain were both found to be less relevant predictors of tumor grade. Both SUVmax (R = 0.71, p = 0.0227) and TBmean (TBmean1.3: R = 0.81, p = 0.00081) had a high correlation with the tumor proliferative index Ki-67. CONCLUSIONS Fluciclovine PET produces high-contrast images between both low-grade and high grade gliomas and normal brain by visual and semiquantitative analysis. Fluciclovine PET appears to discriminate between low-grade glioma and high-grade glioma, but must be validated with a larger sample size.
Collapse
Affiliation(s)
- Ephraim E. Parent
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1841 Clifton Rd. NE, 2nd floor, Atlanta, GA 30329 USA
| | - Marc Benayoun
- Department of Radiology, Massachusetts General Hospital, Boston, MA USA
| | - Ijeoma Ibeanu
- Department of Radiology, Texas Tech University Health Sciences Center Foster School of Medicine, El Paso, TX USA
| | - Jeffrey J. Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA USA
| | | | - Daniel J. Brat
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Vikram Adhikarla
- Department of Information Sciences, City of Hope National Medical Center, Duarte, CA USA
| | - Jonathon Nye
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1841 Clifton Rd. NE, 2nd floor, Atlanta, GA 30329 USA
| | - David M. Schuster
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1841 Clifton Rd. NE, 2nd floor, Atlanta, GA 30329 USA
| | - Mark M. Goodman
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1841 Clifton Rd. NE, 2nd floor, Atlanta, GA 30329 USA
| |
Collapse
|
28
|
Galldiks N, Dunkl V, Ceccon G, Tscherpel C, Stoffels G, Law I, Henriksen OM, Muhic A, Poulsen HS, Steger J, Bauer EK, Lohmann P, Schmidt M, Shah NJ, Fink GR, Langen KJ. Early treatment response evaluation using FET PET compared to MRI in glioblastoma patients at first progression treated with bevacizumab plus lomustine. Eur J Nucl Med Mol Imaging 2018; 45:2377-2386. [PMID: 29982845 DOI: 10.1007/s00259-018-4082-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/27/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND The goal of this prospective study was to compare the value of both conventional MRI and O-(2-18F-fluoroethyl)-L-tyrosine (FET) PET for response evaluation in glioblastoma patients treated with bevacizumab plus lomustine (BEV/LOM) at first progression. METHODS After chemoradiation with concomitant and adjuvant temozolomide, 21 IDH wild-type glioblastoma patients at first progression (age range, 33-75 years; MGMT promoter unmethylated, 81%) were treated with BEV/LOM. Contrast-enhanced MRI and FET-PET scans were performed at baseline and after 8-10 weeks. We obtained FET metabolic tumor volumes (MTV) and tumor/brain ratios. Threshold values of FET-PET parameters for treatment response were established by ROC analyses using the post-progression overall survival (OS) ≤/>9 months as the reference. MRI response assessment was based on RANO criteria. The predictive ability of FET-PET thresholds and MRI changes on early response assessment was evaluated subsequently concerning OS using uni- and multivariate survival estimates. RESULTS Early treatment response as assessed by RANO criteria was not predictive for an OS>9 months (P = 0.203), whereas relative reductions of all FET-PET parameters significantly predicted an OS>9 months (P < 0.05). The absolute MTV at follow-up enabled the most significant OS prediction (sensitivity, 85%; specificity, 88%; P = 0.001). Patients with an absolute MTV below 5 ml at follow-up survived significantly longer (12 vs. 6 months, P < 0.001), whereas early responders defined by RANO criteria lived only insignificantly longer (9 vs. 6 months; P = 0.072). The absolute MTV at follow-up remained significant in the multivariate survival analysis (P = 0.006). CONCLUSIONS FET-PET appears to be useful for identifying responders to BEV/LOM early after treatment initiation.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany. .,Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany. .,Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany.
| | - Veronika Dunkl
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany
| | - Garry Ceccon
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany
| | - Caroline Tscherpel
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Otto M Henriksen
- Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Aida Muhic
- Department of Oncology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hans S Poulsen
- Department of Oncology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jan Steger
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany
| | - Elena K Bauer
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Matthias Schmidt
- Dept. of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany.,Department of Neurology, University Hospital Aachen, Aachen, Germany
| | - Gereon R Fink
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany.,Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| |
Collapse
|
29
|
Use of FET PET in glioblastoma patients undergoing neurooncological treatment including tumour-treating fields: initial experience. Eur J Nucl Med Mol Imaging 2018; 45:1626-1635. [DOI: 10.1007/s00259-018-3992-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/06/2018] [Indexed: 10/17/2022]
|
30
|
Unterrainer M, Winkelmann I, Suchorska B, Giese A, Wenter V, Kreth FW, Herms J, Bartenstein P, Tonn JC, Albert NL. Biological tumour volumes of gliomas in early and standard 20-40 min 18F-FET PET images differ according to IDH mutation status. Eur J Nucl Med Mol Imaging 2018; 45:1242-1249. [PMID: 29487977 DOI: 10.1007/s00259-018-3969-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/02/2018] [Indexed: 01/18/2023]
Abstract
PURPOSE For the clinical evaluation of O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET) PET images, the use of standard summation images obtained 20-40 min after injection is recommended. However, early summation images obtained 5-15 min after injection have been reported to allow better differentiation between low-grade glioma (LGG) and high-grade glioma (HGG) by capturing the early 18F-FET uptake peak specific for HGG. We compared early and standard summation images with regard to delineation of the PET-derived biological tumour volume (BTV) in correlation with the molecular genetic profile according the updated 2016 WHO classification. METHODS The analysis included 245 patients with newly diagnosed, histologically verified glioma and a positive 18F-FET PET scan prior to any further treatment. BTVs were delineated during the early 5-15 min and standard 20-40 min time frames using a threshold of 1.6 × background activity and were compared intraindividually. Volume differences between early and late summation images of >20% were considered significant and were correlated with WHO grade and the molecular genetic profile (IDH mutation and 1p/19q codeletion status). RESULTS In 52.2% of the patients (128/245), a significant difference in BTV of >20% between early and standard summation images was found. While 44.3% of WHO grade II gliomas (31 of 70) showed a significantly smaller BTV in the early summation images, 35.0% of WHO grade III gliomas (28/80) and 37.9% of WHO grade IV gliomas (36/95) had a significantly larger BTVs. Among IDH-wildtype gliomas, an even higher portion (44.4%, 67/151) showed significantly larger BTVs in the early summation images, which was observed in 5.3% (5/94) of IDH-mutant gliomas only: most of the latter had significantly smaller BTVs in the early summation images, i.e. 51.2% of IDH-mutant gliomas without 1p/19q codeletion (21/41) and 39.6% with 1p/19q codeletion (21/53). CONCLUSION BTVs delineated in early and standard summation images differed significantly in more than half of gliomas. While the standard summation images seem appropriate for delineation of LGG as well as IDH-mutant gliomas, a remarkably high percentage of HGG and, particularly, IDH-wildtype gliomas were depicted with significantly larger volumes in early summation images. This finding might be of interest for optimization of treatment planning (e.g. radiotherapy) in accordance with the individual IDH mutation status.
Collapse
Affiliation(s)
- M Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - I Winkelmann
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - B Suchorska
- Department of Neurosurgery, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - A Giese
- Department of Neuropathology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - V Wenter
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - F W Kreth
- Department of Neurosurgery, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - J Herms
- Department of Neuropathology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - P Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - J C Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - N L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
31
|
Hybrid MR-PET of brain tumours using amino acid PET and chemical exchange saturation transfer MRI. Eur J Nucl Med Mol Imaging 2018; 45:1031-1040. [PMID: 29478081 DOI: 10.1007/s00259-018-3940-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/04/2018] [Indexed: 10/18/2022]
Abstract
PURPOSE PET using radiolabelled amino acids has become a promising tool in the diagnostics of gliomas and brain metastasis. Current research is focused on the evaluation of amide proton transfer (APT) chemical exchange saturation transfer (CEST) MR imaging for brain tumour imaging. In this hybrid MR-PET study, brain tumours were compared using 3D data derived from APT-CEST MRI and amino acid PET using O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET). METHODS Eight patients with gliomas were investigated simultaneously with 18F-FET PET and APT-CEST MRI using a 3-T MR-BrainPET scanner. CEST imaging was based on a steady-state approach using a B1 average power of 1μT. B0 field inhomogeneities were corrected a Prametric images of magnetisation transfer ratio asymmetry (MTRasym) and differences to the extrapolated semi-solid magnetisation transfer reference method, APT# and nuclear Overhauser effect (NOE#), were calculated. Statistical analysis of the tumour-to-brain ratio of the CEST data was performed against PET data using the non-parametric Wilcoxon test. RESULTS A tumour-to-brain ratio derived from APT# and 18F-FET presented no significant differences, and no correlation was found between APT# and 18F-FET PET data. The distance between local hot spot APT# and 18F-FET were different (average 20 ± 13 mm, range 4-45 mm). CONCLUSION For the first time, CEST images were compared with 18F-FET in a simultaneous MR-PET measurement. Imaging findings derived from18F-FET PET and APT CEST MRI seem to provide different biological information. The validation of these imaging findings by histological confirmation is necessary, ideally using stereotactic biopsy.
Collapse
|
32
|
Papp L, Pötsch N, Grahovac M, Schmidbauer V, Woehrer A, Preusser M, Mitterhauser M, Kiesel B, Wadsak W, Beyer T, Hacker M, Traub-Weidinger T. Glioma Survival Prediction with Combined Analysis of In Vivo 11C-MET PET Features, Ex Vivo Features, and Patient Features by Supervised Machine Learning. J Nucl Med 2017; 59:892-899. [DOI: 10.2967/jnumed.117.202267] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/31/2017] [Indexed: 01/03/2023] Open
|
33
|
Evaluation of factors influencing 18F-FET uptake in the brain. NEUROIMAGE-CLINICAL 2017; 17:491-497. [PMID: 29159062 PMCID: PMC5684535 DOI: 10.1016/j.nicl.2017.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/24/2017] [Accepted: 11/07/2017] [Indexed: 01/20/2023]
Abstract
PET using the amino-acid O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) is gaining increasing interest for brain tumour management. Semi-quantitative analysis of tracer uptake in brain tumours is based on the standardized uptake value (SUV) and the tumour-to-brain ratio (TBR). The aim of this study was to explore physiological factors that might influence the relationship of SUV of 18F-FET uptake in various brain areas, and thus affect quantification of 18F-FET uptake in brain tumours. Negative 18F-FET PET scans of 107 subjects, showing an inconspicuous brain distribution of 18F-FET, were evaluated retrospectively. Whole-brain quantitative analysis with Statistical Parametric Mapping (SPM) using parametric SUV PET images, and volumes of interest (VOIs) analysis with fronto-parietal, temporal, occipital, and cerebellar SUV background areas were performed to study the effect of age, gender, height, weight, injected activity, body mass index (BMI), and body surface area (BSA). After multivariate analysis, female gender and high BMI were found to be two independent factors associated with increased SUV of 18F-FET uptake in the brain. In women, SUVmean of 18F-FET uptake in the brain was 23% higher than in men (p < 0.01). SUVmean of 18F-FET uptake in the brain was positively correlated with BMI (r = 0.29; p < 0.01). The influence of these factors on SUV of 18F-FET was similar in all brain areas. In conclusion, SUV of 18F-FET in the normal brain is influenced by gender and weakly by BMI, but changes are similar in all brain areas. SUVmean of 18F-FET in the normal brain is influenced by gender. SUVmean of 18F-FET in the normal brain is weekly influenced by BMI. The influence of these factors on SUV of 18F-FET is similar in all brain areas.
Collapse
|
34
|
Imaging of amino acid transport in brain tumours: Positron emission tomography with O-(2-[ 18 F]fluoroethyl)- L -tyrosine (FET). Methods 2017; 130:124-134. [DOI: 10.1016/j.ymeth.2017.05.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/08/2017] [Accepted: 05/21/2017] [Indexed: 01/01/2023] Open
|
35
|
Oros-Peusquens A, Loução R, Zimmermann M, Langen KJ, Shah N. Methods for molecular imaging of brain tumours in a hybrid MR-PET context: Water content, T 2 ∗ , diffusion indices and FET-PET. Methods 2017; 130:135-151. [DOI: 10.1016/j.ymeth.2017.07.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/22/2017] [Accepted: 07/27/2017] [Indexed: 11/27/2022] Open
|
36
|
Verger A, Stoffels G, Bauer EK, Lohmann P, Blau T, Fink GR, Neumaier B, Shah NJ, Langen KJ, Galldiks N. Static and dynamic 18F–FET PET for the characterization of gliomas defined by IDH and 1p/19q status. Eur J Nucl Med Mol Imaging 2017; 45:443-451. [DOI: 10.1007/s00259-017-3846-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/02/2017] [Indexed: 01/01/2023]
|
37
|
Ceccon G, Lohmann P, Stoffels G, Judov N, Filss CP, Rapp M, Bauer E, Hamisch C, Ruge MI, Kocher M, Kuchelmeister K, Sellhaus B, Sabel M, Fink GR, Shah NJ, Langen KJ, Galldiks N. Dynamic O-(2-18F-fluoroethyl)-L-tyrosine positron emission tomography differentiates brain metastasis recurrence from radiation injury after radiotherapy. Neuro Oncol 2017; 19:281-288. [PMID: 27471107 DOI: 10.1093/neuonc/now149] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/02/2016] [Indexed: 11/14/2022] Open
Abstract
Background The aim of this study was to investigate the potential of dynamic O-(2-[18F]fluoroethyl)-L-tyrosine (18F-FET) PET for differentiating local recurrent brain metastasis from radiation injury after radiotherapy since contrast-enhanced MRI often remains inconclusive. Methods Sixty-two patients (mean age, 55 ± 11 y) with single or multiple contrast-enhancing brain lesions (n = 76) on MRI after radiotherapy of brain metastases (predominantly stereotactic radiosurgery) were investigated with dynamic 18F-FET PET. Maximum and mean tumor-to-brain ratios (TBRmax, TBRmean) of 18F-FET uptake were determined (20-40 min postinjection) as well as tracer uptake kinetics (ie, time-to-peak and slope of time-activity curves). Diagnoses were confirmed histologically (34%; 26 lesions in 25 patients) or by clinical follow-up (66%; 50 lesions in 37 patients). Diagnostic accuracies of PET parameters for the correct identification of recurrent brain metastasis were evaluated by receiver-operating-characteristic analyses or the chi-square test. Results TBRs were significantly higher in recurrent metastases (n = 36) than in radiation injuries (n = 40) (TBRmax 3.3 ± 1.0 vs 2.2 ± 0.4, P < .001; TBRmean 2.2 ± 0.4 vs 1.7 ± 0.3, P < .001). The highest accuracy (88%) for diagnosing local recurrent metastasis could be obtained with TBRs in combination with the slope of time-activity curves (P < .001). Conclusions The results of this study confirm previous preliminary observations that the combined evaluation of the TBRs of 18F-FET uptake and the slope of time-activity curves can differentiate local brain metastasis recurrence from radiation-induced changes with high accuracy. 18F-FET PET may thus contribute significantly to the management of patients with brain metastases.
Collapse
Affiliation(s)
- Garry Ceccon
- Department of Neurology, University of Cologne, Cologne, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany
| | - Natalie Judov
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany
| | - Christian P Filss
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany.,Department of Neurology, University of Aachen, Aachen, Germany
| | - Marion Rapp
- Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany
| | - Elena Bauer
- Department of Neurology, University of Cologne, Cologne, Germany
| | | | - Maximilian I Ruge
- Department of Stereotaxy and Functional Neurosurgery, University of Cologne, Cologne, Germany
| | - Martin Kocher
- Department of Radiation Oncology, University of Cologne, Cologne, Germany
| | | | - Bernd Sellhaus
- Department of Neuropathology, University of Aachen, Aachen, Germany
| | - Michael Sabel
- Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany
| | - Gereon R Fink
- Department of Neurology, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany.,Department of Neurology, University of Aachen, Aachen, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Jülich and Aachen, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany.,Department of Neuropathology, University of Aachen, Aachen, Germany.,Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Norbert Galldiks
- Department of Neurology, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany.,Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany
| |
Collapse
|
38
|
Ladefoged CN, Andersen FL, Kjær A, Højgaard L, Law I. RESOLUTE PET/MRI Attenuation Correction for O-(2- 18F-fluoroethyl)-L-tyrosine (FET) in Brain Tumor Patients with Metal Implants. Front Neurosci 2017; 11:453. [PMID: 28848379 PMCID: PMC5554515 DOI: 10.3389/fnins.2017.00453] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/25/2017] [Indexed: 01/06/2023] Open
Abstract
Aim: Positron emission tomography (PET) imaging is a useful tool for assisting in correct differentiation of tumor progression from reactive changes, and the radiolabeled amino acid analog tracer O-(2-18F-fluoroethyl)-L-tyrosine (FET)-PET is amongst the most frequently used. The FET-PET images need to be quantitatively correct in order to be used clinically, which require accurate attenuation correction (AC) in PET/MRI. The aim of this study was to evaluate the use of the subject-specific MR-derived AC method RESOLUTE in post-operative brain tumor patients. Methods: We analyzed 51 post-operative brain tumor patients (68 examinations, 200 MBq [18F]-FET) investigated in a PET/MRI scanner. MR-AC maps were acquired using: (1) the Dixon water fat separation sequence, (2) the ultra short echo time (UTE) sequences, (3) calculated using our new RESOLUTE methodology, and (4) a same day low-dose CT used as reference “gold standard.” For each subject and each AC method the tumor was delineated by isocontouring tracer uptake above a tumor(T)-to-brain background (B) activity ratio of 1.6. We measured B, tumor mean and maximal activity (TMEAN, TMAX), biological tumor volume (BTV), and calculated the clinical metrics TMEAN/B and TMAX/B. Results: When using RESOLUTE 5/68 studies did not meet our predefined acceptance criteria of TMAX/B difference to CT-AC < ±0.1 or 5%, TMEAN/B < ±0.05 or 5%, and BTV < ±2 mL or 10%. In total, 46/68 studies failed our acceptance criteria using Dixon, and 26/68 using UTE. The 95% limits of agreement for TMAX/B was for RESOLUTE (−3%; 4%), Dixon (−9%; 16%), and UTE (−7%; 10%). The absolute error when measuring BTV was 0.7 ± 1.9 mL (N.S) with RESOLUTE, 5.3 ± 10 mL using Dixon, and 1.7 ± 3.7 mL using UTE. RESOLUTE performed best in the identification of the location of peak activity and in brain tumor follow-up monitoring using clinical FET PET metrics. Conclusions: Overall, we found RESOLUTE to be the AC method that most robustly reproduced the CT-AC clinical metrics per se, during follow-up, and when interpreted into defined clinical use cut-off criteria and into the patient history. RESOLUTE is especially suitable for brain tumor patients, as these often present with distorted anatomy where other methods based on atlas/template information might fail.
Collapse
Affiliation(s)
- Claes N Ladefoged
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of CopenhagenCopenhagen, Denmark
| | - Flemming L Andersen
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of CopenhagenCopenhagen, Denmark
| | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of CopenhagenCopenhagen, Denmark
| | - Liselotte Højgaard
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of CopenhagenCopenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of CopenhagenCopenhagen, Denmark
| |
Collapse
|
39
|
Unterrainer M, Vettermann F, Brendel M, Holzgreve A, Lifschitz M, Zähringer M, Suchorska B, Wenter V, Illigens BM, Bartenstein P, Albert NL. Towards standardization of 18F-FET PET imaging: do we need a consistent method of background activity assessment? EJNMMI Res 2017; 7:48. [PMID: 28560582 PMCID: PMC5449315 DOI: 10.1186/s13550-017-0295-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/18/2017] [Indexed: 11/25/2022] Open
Abstract
Background PET with O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET) has reached increasing clinical significance for patients with brain neoplasms. For quantification of standard PET-derived parameters such as the tumor-to-background ratio, the background activity is assessed using a region of interest (ROI) or volume of interest (VOI) in unaffected brain tissue. However, there is no standardized approach regarding the assessment of the background reference. Therefore, we evaluated the intra- and inter-reader variability of commonly applied approaches for clinical 18F-FET PET reading. The background activity of 20 18F-FET PET scans was independently evaluated by 6 readers using a (i) simple 2D-ROI, (ii) spherical VOI with 3.0 cm diameter, and (iii) VOI consisting of crescent-shaped ROIs; each in the contralateral, non-affected hemisphere including white and gray matter in line with the European Association of Nuclear Medicine (EANM) and German guidelines. To assess intra-reader variability, each scan was evaluated 10 times by each reader. The coefficient of variation (CoV) was assessed for determination of intra- and inter-reader variability. In a second step, the best method was refined by instructions for a guided background activity assessment and validated by 10 further scans. Results Compared to the other approaches, the crescent-shaped VOIs revealed most stable results with the lowest intra-reader variabilities (median CoV 1.52%, spherical VOI 4.20%, 2D-ROI 3.69%; p < 0.001) and inter-reader variabilities (median CoV 2.14%, spherical VOI 4.02%, 2D-ROI 3.83%; p = 0.001). Using the guided background assessment, both intra-reader variabilities (median CoV 1.10%) and inter-reader variabilities (median CoV 1.19%) could be reduced even more. Conclusions The commonly applied methods for background activity assessment show different variability which might hamper 18F-FET PET quantification and comparability in multicenter settings. The proposed background activity assessment using a (guided) crescent-shaped VOI allows minimization of both intra- and inter-reader variability and might facilitate comprehensive methodological standardization of amino acid PET which is of interest in the light of the anticipated EANM technical guidelines.
Collapse
Affiliation(s)
- Marcus Unterrainer
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,Center for Clinical Research and Management Education, Division of Health Care Sciences, Dresden International University, Dresden, Germany
| | - Franziska Vettermann
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,Department of Neurosurgery, LMU Munich, Munich, Germany
| | - Michael Lifschitz
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Matthias Zähringer
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | | | - Vera Wenter
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Ben M Illigens
- Center for Clinical Research and Management Education, Division of Health Care Sciences, Dresden International University, Dresden, Germany.,Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| |
Collapse
|
40
|
Tscherpel C, Dunkl V, Ceccon G, Stoffels G, Judov N, Rapp M, Meyer PT, Kops ER, Ermert J, Fink GR, Shah NJ, Langen KJ, Galldiks N. The use of O-(2-18F-fluoroethyl)-L-tyrosine PET in the diagnosis of gliomas located in the brainstem and spinal cord. Neuro Oncol 2017; 19:710-718. [PMID: 28039366 DOI: 10.1093/neuonc/now243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background Despite an increasing number of O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET) PET studies in supratentorial gliomas, studies regarding the usefulness of 18F-FET PET in brainstem and spinal cord gliomas to date remain scarce. Methods Thirty-six 18F-FET PET scans were performed in 29 patients with brainstem (n = 29 scans) or spinal cord glioma (n = 7 scans). In 32 of 36 PET scans, a dynamic acquisition was performed. Fifteen scans in 15 patients were performed to assess newly diagnosed lesions, and 21 scans were obtained during follow-up: for diagnosing tumor progression (n = 15 scans in 14 patients) as well as for treatment monitoring (n = 6 scans in 3 patients). Four patients underwent additional serial scans (range, 1-2), and 3 of these 4 patients were examined for more than one indication. Maximum and mean tumor/brain ratios (TBRmax/mean) of 18F-FET uptake (20-40 min post injection) as well as kinetic 18F-FET uptake parameters were determined. Final diagnoses were confirmed histologically (54%) or by clinical follow-up (46%). Results In all newly diagnosed high-grade (n = 3 patients) and in 5 of 11 patients with low-grade gliomas, 18F-FET uptake was increased (TBRmax ≥2.5 and/or TBRmean ≥1.9). In 2 patients with newly diagnosed gliomas without MR contrast enhancement, 18F-FET PET nevertheless showed increased metabolism. At suspected progression, the combination of TBRs with kinetic 18F-FET parameters correctly identified presence or absence of progressive disease in 9 of 11 patients (82%). Conclusions This preliminary study suggests that 18F-FET PET adds valuable diagnostic information in brainstem and spinal cord glioma, particularly when the diagnostic information derived from MRI is equivocal.
Collapse
Affiliation(s)
- Caroline Tscherpel
- Department of Neurology, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Veronika Dunkl
- Department of Neurology, University of Cologne, Cologne, Germany
| | - Garry Ceccon
- Department of Neurology, University of Cologne, Cologne, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Natalie Judov
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Marion Rapp
- Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Elena Rota Kops
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Johannes Ermert
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Gereon R Fink
- Department of Neurology, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany.,Departments of Neurology, University of Aachen, Aachen, Germany.,Section JARA-Brain, Jülich-Aachen Research Alliance (JARA), Jülich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany.,Section JARA-Brain, Jülich-Aachen Research Alliance (JARA), Jülich, Germany.,Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Norbert Galldiks
- Department of Neurology, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany.,Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany
| |
Collapse
|
41
|
Marner L, Henriksen OM, Lundemann M, Larsen VA, Law I. Clinical PET/MRI in neurooncology: opportunities and challenges from a single-institution perspective. Clin Transl Imaging 2016; 5:135-149. [PMID: 28936429 PMCID: PMC5581366 DOI: 10.1007/s40336-016-0213-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 10/31/2016] [Indexed: 01/17/2023]
Abstract
Purpose Magnetic resonance imaging (MRI) plays a key role in neurooncology, i.e., for diagnosis, treatment evaluation and detection of recurrence. However, standard MRI cannot always separate malignant tissue from other pathologies or treatment-induced changes. Advanced MRI techniques such as diffusion-weighted imaging, perfusion imaging and spectroscopy show promising results in discriminating malignant from benign lesions. Further, supplemental imaging with amino acid positron emission tomography (PET) has been shown to increase accuracy significantly and is used routinely at an increasing number of sites. Several centers are now implementing hybrid PET/MRI systems allowing for multiparametric imaging, combining conventional MRI with advanced MRI and amino acid PET imaging. Neurooncology is an obvious focus area for PET/MR imaging. Methods Based on the literature and our experience from more than 300 PET/MRI examinations of brain tumors with 18F-fluoro-ethyl-tyrosine, the clinical use of PET/MRI in adult and pediatric neurooncology is critically reviewed. Results Although the results are increasingly promising, the added value and range of indications for multiparametric imaging with PET/MRI are yet to be established. Robust solutions to overcome the number of issues when using a PET/MRI scanner are being developed, which is promising for a more routine use in the future. Conclusions In a clinical setting, a PET/MRI scan may increase accuracy in discriminating recurrence from treatment changes, although sequential same-day imaging on separate systems will often constitute a reliable and cost-effective alternative. Pediatric patients who require general anesthesia will benefit the most from simultaneous PET and MR imaging.
Collapse
Affiliation(s)
- Lisbeth Marner
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark
| | - Otto M Henriksen
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark
| | - Michael Lundemann
- Department of Oncology, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark
| | - Vibeke Andrée Larsen
- Department of Radiology, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark
| |
Collapse
|
42
|
Radiation injury vs. recurrent brain metastasis: combining textural feature radiomics analysis and standard parameters may increase 18F-FET PET accuracy without dynamic scans. Eur Radiol 2016; 27:2916-2927. [PMID: 27853813 DOI: 10.1007/s00330-016-4638-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 09/29/2016] [Accepted: 10/07/2016] [Indexed: 10/20/2022]
Abstract
OBJECTIVES We investigated the potential of textural feature analysis of O-(2-[18F]fluoroethyl)-L-tyrosine (18F-FET) PET to differentiate radiation injury from brain metastasis recurrence. METHODS Forty-seven patients with contrast-enhancing brain lesions (n = 54) on MRI after radiotherapy of brain metastases underwent dynamic 18F-FET PET. Tumour-to-brain ratios (TBRs) of 18F-FET uptake and 62 textural parameters were determined on summed images 20-40 min post-injection. Tracer uptake kinetics, i.e., time-to-peak (TTP) and patterns of time-activity curves (TAC) were evaluated on dynamic PET data from 0-50 min post-injection. Diagnostic accuracy of investigated parameters and combinations thereof to discriminate between brain metastasis recurrence and radiation injury was compared. RESULTS Diagnostic accuracy increased from 81 % for TBRmean alone to 85 % when combined with the textural parameter Coarseness or Short-zone emphasis. The accuracy of TBRmax alone was 83 % and increased to 85 % after combination with the textural parameters Coarseness, Short-zone emphasis, or Correlation. Analysis of TACs resulted in an accuracy of 70 % for kinetic pattern alone and increased to 83 % when combined with TBRmax. CONCLUSIONS Textural feature analysis in combination with TBRs may have the potential to increase diagnostic accuracy for discrimination between brain metastasis recurrence and radiation injury, without the need for dynamic 18F-FET PET scans. KEY POINTS • Textural feature analysis provides quantitative information about tumour heterogeneity • Textural features help improve discrimination between brain metastasis recurrence and radiation injury • Textural features might be helpful to further understand tumour heterogeneity • Analysis does not require a more time consuming dynamic PET acquisition.
Collapse
|
43
|
Unterrainer M, Galldiks N, Suchorska B, Kowalew LC, Wenter V, Schmid-Tannwald C, Niyazi M, Bartenstein P, Langen KJ, Albert NL. 18F-FET PET Uptake Characteristics in Patients with Newly Diagnosed and Untreated Brain Metastasis. J Nucl Med 2016; 58:584-589. [PMID: 27754904 DOI: 10.2967/jnumed.116.180075] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/12/2016] [Indexed: 01/03/2023] Open
Abstract
In patients with brain metastasis, PET using labeled amino acids has gained clinical importance, mainly regarding the differentiation of viable tumor tissue from treatment-related effects. However, there is still limited knowledge concerning the uptake characteristics in patients with newly diagnosed and untreated brain metastases. Hence, we evaluated the uptake characteristics in these patients using dynamic O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) PET. Methods: Patients with newly diagnosed brain metastases without prior local therapy and 18F-FET PET scanning were retrospectively identified in 2 centers. Static and dynamic PET parameters (maximal/mean tumor-to-brain-ratio [TBRmax/TBRmean], biologic tumor volume [BTV], and time-activity curves with minimal time to peak [TTPmin]) were evaluated and correlated with MRI parameters (maximal lesion diameter, volume of contrast enhancement) and originating primary tumor. Results: Forty-five brain metastases in 30 patients were included. Forty of 45 metastases (89%) had a TBRmax ≥ 1.6 and were classified as 18F-FET-positive (median TBRmax, 2.53 [range, 1.64-9.47]; TBRmean, 1.86 [range, 1.63-5.48]; and BTV, 3.59 mL [range, 0.04-23.98 mL], respectively). In 39 of 45 brain metastases eligible for dynamic analysis, a wide range of TTPmin was observed (median, 22.5 min; range, 4.5-47.5 min). All 18F-FET-negative metastases had a diameter of ≤ 1.0 cm, whereas metastases with a > 1.0 cm diameter all showed pathologic 18F-FET uptake, which did not correlate with lesion size. The highest variability of uptake intensity was observed within the group of melanoma metastases. Conclusion: Untreated metastases predominantly show increased 18F-FET uptake, and only a third of metastases < 1.0 cm were 18F-FET-negative, most likely because of scanner resolution and partial-volume effects. In metastases > 1.0 cm, 18F-FET uptake intensity was highly variable and independent of tumor size (even intraindividually). 18F-FET PET might provide additional information beyond the tumor extent by reflecting molecular features of a metastasis and might be a useful tool for future clinical applications, for example, response assessment.
Collapse
Affiliation(s)
- Marcus Unterrainer
- Department of Nuclear Medicine, University of Munich (LMU), Munich, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany.,Department of Neurology, University Hospital Cologne, Cologne, Germany
| | | | | | - Vera Wenter
- Department of Nuclear Medicine, University of Munich (LMU), Munich, Germany
| | | | - Maximilian Niyazi
- Department of Radiation Oncology, LMU, Munich, Germany.,German Cancer Consortium (DKTK) & German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University of Munich (LMU), Munich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, University of Aachen, Aachen, Germany; and.,JARA-Brain Section, Juelich-Aachen-Research-Alliance (JARA), Juelich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University of Munich (LMU), Munich, Germany
| |
Collapse
|
44
|
Hutterer M, Ebner Y, Riemenschneider MJ, Willuweit A, McCoy M, Egger B, Schröder M, Wendl C, Hellwig D, Grosse J, Menhart K, Proescholdt M, Fritsch B, Urbach H, Stockhammer G, Roelcke U, Galldiks N, Meyer PT, Langen KJ, Hau P, Trinka E. Epileptic Activity Increases Cerebral Amino Acid Transport Assessed by 18F-Fluoroethyl-l-Tyrosine Amino Acid PET: A Potential Brain Tumor Mimic. J Nucl Med 2016; 58:129-137. [PMID: 27469356 DOI: 10.2967/jnumed.116.176610] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/29/2016] [Indexed: 11/16/2022] Open
Abstract
O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) PET is a well-established method increasingly used for diagnosis, treatment planning, and monitoring in gliomas. Epileptic activity, frequently occurring in glioma patients, can influence MRI findings. Whether seizures also affect 18F-FET PET imaging is currently unknown. The aim of this retrospective analysis was to investigate the brain amino acid metabolism during epileptic seizures by 18F-FET PET and to elucidate the pathophysiologic background. METHODS Ten patients with 11 episodes of serial seizures or status epilepticus, who underwent MRI and 18F-FET PET, were studied. The main diagnosis was glioma World Health Organization grade II-IV (n = 8); 2 patients suffered from nonneoplastic diseases. Immunohistochemical assessment of LAT1/LAT2/CD98 amino acid transporters was performed in seizure-affected cortex (n = 2) and compared with glioma tissues (n = 3). RESULTS All patients exhibited increased seizure-associated strict gyral 18F-FET uptake, which was reversible in follow-up studies or negative shortly before and without any histologic or clinical signs of tumor recurrence. 18F-FET uptake corresponded to structural MRI changes, compatible with cortical vasogenic and cytotoxic edema, partial contrast enhancement, and hyperperfusion. Patients with prolonged postictal symptoms lasting up to 8 wk displayed intensive and widespread (≥ 1 lobe) cortical 18F-FET uptake. LAT1/LAT2/CD98 was strongly expressed in neurons and endothelium of seizure-affected brains and less in reactive astrocytosis. CONCLUSION Seizure activity, in particular status epilepticus, increases cerebral amino acid transport with a strict gyral 18F-FET uptake pattern. Such periictal pseudoprogression represents a potential pitfall of 18F-FET PET and may mimic brain tumor. Our data also indicate a seizure-induced upregulation of neuronal, endothelial, and less astroglial LAT1/LAT2/CD98 amino acid transporter expression.
Collapse
Affiliation(s)
- Markus Hutterer
- Department of Neurology, University of Regensburg Medical School, Regensburg, Germany .,Wilhelm Sander-Neurooncology Unit, University of Regensburg Medical School, Regensburg, Germany.,Department of Neurology and Centre for Cognitive Neuroscience, Christian-Doppler Klinik, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Yvonne Ebner
- Department of Neurology and Centre for Cognitive Neuroscience, Christian-Doppler Klinik, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Markus J Riemenschneider
- Wilhelm Sander-Neurooncology Unit, University of Regensburg Medical School, Regensburg, Germany.,Department of Neuropathology, University of Regensburg Medical School, Regensburg, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Mark McCoy
- Department of Radiology and Division of Neuroradiology, Christian-Doppler Klinik, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Barbara Egger
- Department of Nuclear Medicine, Landeskrankenhaus Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Michael Schröder
- Department of Neurology, University of Regensburg Medical School, Regensburg, Germany
| | - Christina Wendl
- Department of Radiology and Division of Neuroradiology, University of Regensburg Medical School, Regensburg, Germany
| | - Dirk Hellwig
- Department of Nuclear Medicine, University of Regensburg Medical School, Regensburg, Germany
| | - Jirka Grosse
- Department of Nuclear Medicine, University of Regensburg Medical School, Regensburg, Germany
| | - Karin Menhart
- Department of Nuclear Medicine, University of Regensburg Medical School, Regensburg, Germany
| | - Martin Proescholdt
- Wilhelm Sander-Neurooncology Unit, University of Regensburg Medical School, Regensburg, Germany.,Department of Neurosurgery, University of Regensburg Medical School, Regensburg, Germany
| | - Brita Fritsch
- Department of Neurology, University Hospital Freiburg, Freiburg, Germany
| | - Horst Urbach
- Department of Neuroradiology, University Hospital Freiburg, Freiburg, Germany
| | | | - Ulrich Roelcke
- Department of Neurology and Brain Tumor Center, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, University Hospital Freiburg, Freiburg, Germany; and
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany.,Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Peter Hau
- Department of Neurology, University of Regensburg Medical School, Regensburg, Germany.,Wilhelm Sander-Neurooncology Unit, University of Regensburg Medical School, Regensburg, Germany
| | - Eugen Trinka
- Department of Neurology and Centre for Cognitive Neuroscience, Christian-Doppler Klinik, Paracelsus Medical University Salzburg, Salzburg, Austria
| |
Collapse
|
45
|
Early static 18F-FET-PET scans have a higher accuracy for glioma grading than the standard 20–40 min scans. Eur J Nucl Med Mol Imaging 2015; 43:1105-14. [DOI: 10.1007/s00259-015-3276-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/27/2015] [Indexed: 12/31/2022]
|
46
|
Galldiks N, Stoffels G, Filss C, Rapp M, Blau T, Tscherpel C, Ceccon G, Dunkl V, Weinzierl M, Stoffel M, Sabel M, Fink GR, Shah NJ, Langen KJ. The use of dynamic O-(2-18F-fluoroethyl)-l-tyrosine PET in the diagnosis of patients with progressive and recurrent glioma. Neuro Oncol 2015; 17:1293-300. [PMID: 26008606 DOI: 10.1093/neuonc/nov088] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 04/11/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND We evaluated the diagnostic value of static and dynamic O-(2-[(18)F]fluoroethyl)-L-tyrosine ((18)F-FET) PET parameters in patients with progressive or recurrent glioma. METHODS We retrospectively analyzed 132 dynamic (18)F-FET PET and conventional MRI scans of 124 glioma patients (primary World Health Organization grade II, n = 55; grade III, n = 19; grade IV, n = 50; mean age, 52 ± 14 y). Patients had been referred for PET assessment with clinical signs and/or MRI findings suggestive of tumor progression or recurrence based on Response Assessment in Neuro-Oncology criteria. Maximum and mean tumor/brain ratios of (18)F-FET uptake were determined (20-40 min post-injection) as well as tracer uptake kinetics (ie, time to peak and patterns of the time-activity curves). Diagnoses were confirmed histologically (95%) or by clinical follow-up (5%). Diagnostic accuracies of PET and MR parameters for the detection of tumor progression or recurrence were evaluated by receiver operating characteristic analyses/chi-square test. RESULTS Tumor progression or recurrence could be diagnosed in 121 of 132 cases (92%). MRI and (18)F-FET PET findings were concordant in 84% and discordant in 16%. Compared with the diagnostic accuracy of conventional MRI to diagnose tumor progression or recurrence (85%), a higher accuracy (93%) was achieved by (18)F-FET PET when a mean tumor/brain ratio ≥2.0 or time to peak <45 min was present (sensitivity, 93%; specificity, 100%; accuracy, 93%; positive predictive value, 100%; P < .001). CONCLUSION Static and dynamic (18)F-FET PET parameters differentiate progressive or recurrent glioma from treatment-related nonneoplastic changes with higher accuracy than conventional MRI.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Gabriele Stoffels
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Christian Filss
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Marion Rapp
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Tobias Blau
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Caroline Tscherpel
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Garry Ceccon
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Veronika Dunkl
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Martin Weinzierl
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Michael Stoffel
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Michael Sabel
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Gereon R Fink
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Nadim J Shah
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| | - Karl-Josef Langen
- Department of Neurology, University of Cologne, Cologne, Germany (N.G., C.T., G.C., V.D., G.R.F.); Department of Neuropathology, University of Cologne, Cologne, Germany (T.B.); Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany (N.G., G.S., C.F., V.D., G.R.F., N.J.S., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany (M.R., M.Sa.); Department of Neurosurgery, Helios Kliniken, Krefeld, Germany (M.W., M.St.); Department of Neurology, University of Aachen, Aachen, Germany (N.J.S.); Department of Nuclear Medicine, University of Aachen, Aachen, Germany (K.-J.L.); Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain (N.J.S., K.-J.L.)
| |
Collapse
|
47
|
Lohmann P, Herzog H, Rota Kops E, Stoffels G, Judov N, Filss C, Galldiks N, Tellmann L, Weiss C, Sabel M, Coenen HH, Shah NJ, Langen KJ. Dual-time-point O-(2-[(18)F]fluoroethyl)-L-tyrosine PET for grading of cerebral gliomas. Eur Radiol 2015; 25:3017-24. [PMID: 25813014 DOI: 10.1007/s00330-015-3691-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/19/2015] [Accepted: 02/23/2015] [Indexed: 11/27/2022]
Abstract
OBJECTIVE We aimed to evaluate the diagnostic potential of dual-time-point imaging with positron emission tomography (PET) using O-(2-[(18)F]fluoroethyl)-L-tyrosine ((18)F-FET) for non-invasive grading of cerebral gliomas compared with a dynamic approach. METHODS Thirty-six patients with histologically confirmed cerebral gliomas (21 primary, 15 recurrent; 24 high-grade, 12 low-grade) underwent dynamic PET from 0 to 50 min post-injection (p.i.) of (18)F-FET, and additionally from 70 to 90 min p.i. Mean tumour-to-brain ratios (TBRmean) of (18)F-FET uptake were determined in early (20-40 min p.i.) and late (70-90 min p.i.) examinations. Time-activity curves (TAC) of the tumours from 0 to 50 min after injection were assigned to different patterns. The diagnostic accuracy of changes of (18)F-FET uptake between early and late examinations for tumour grading was compared to that of curve pattern analysis from 0 to 50 min p.i. of (18)F-FET. RESULTS The diagnostic accuracy of changes of the TBRmean of (18)F-FET PET uptake between early and late examinations for the identification of HGG was 81% (sensitivity 83%; specificity 75%; cutoff - 8%; p < 0.001), and 83% for curve pattern analysis (sensitivity 88%; specificity 75%; p < 0.001). CONCLUSION Dual-time-point imaging of (18)F-FET uptake in gliomas achieves diagnostic accuracy for tumour grading that is similar to the more time-consuming dynamic data acquisition protocol. KEY POINTS • Dual-time-point imaging is equivalent to dynamic FET PET for grading of gliomas. • Dual-time-point imaging is less time consuming than dynamic FET PET. • Costs can be reduced due to higher patient throughput. • Reduced imaging time increases patient comfort and sedation might be avoided. • Quicker image interpretation is possible, as no curve evaluation is necessary.
Collapse
Affiliation(s)
- Philipp Lohmann
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Wilhelm-Johnen-Str., Jülich, 52428, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lapa C, Linsenmann T, Lückerath K, Samnick S, Herrmann K, Stoffer C, Ernestus RI, Buck AK, Löhr M, Monoranu CM. Tumor-associated macrophages in glioblastoma multiforme-a suitable target for somatostatin receptor-based imaging and therapy? PLoS One 2015; 10:e0122269. [PMID: 25807228 PMCID: PMC4373835 DOI: 10.1371/journal.pone.0122269] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 02/11/2015] [Indexed: 12/22/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. Tumor-associated macrophages (TAM) have been shown to promote malignant growth and to correlate with poor prognosis. [1,4,7,10-tetraazacyclododecane-NN′,N″,N′″-tetraacetic acid]-d-Phe1,Tyr3-octreotate (DOTATATE) labeled with Gallium-68 selectively binds to somatostatin receptor 2A (SSTR2A) which is specifically expressed and up-regulated in activated macrophages. On the other hand, the role of SSTR2A expression on the cell surface of glioma cells has not been fully elucidated yet. The aim of this study was to non-invasively assess SSTR2A expression of both glioma cells as well as macrophages in GBM. Methods 15 samples of patient-derived GBM were stained immunohistochemically for macrophage infiltration (CD68), proliferative activity (Ki67) as well as expression of SSTR2A. Anti-CD45 staining was performed to distinguish between resident microglia and tumor-infiltrating macrophages. In a subcohort, positron emission tomography (PET) imaging using 68Ga-DOTATATE was performed and the semiquantitatively evaluated tracer uptake was compared to the results of immunohistochemistry. Results The amount of microglia/macrophages ranged from <10% to >50% in the tumor samples with the vast majority being resident microglial cells. A strong SSTR2A immunostaining was observed in endothelial cells of proliferating vessels, in neurons and neuropile. Only faint immunostaining was identified on isolated microglial and tumor cells. Somatostatin receptor imaging revealed areas of increased tracer accumulation in every patient. However, retention of the tracer did not correlate with immunohistochemical staining patterns. Conclusion SSTR2A seems not to be overexpressed in GBM samples tested, neither on the cell surface of resident microglia or infiltrating macrophages, nor on the surface of tumor cells. These data suggest that somatostatin receptor directed imaging and treatment strategies are less promising in GBM.
Collapse
Affiliation(s)
- Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- * E-mail:
| | - Thomas Linsenmann
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Katharina Lückerath
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Carolin Stoffer
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Ralf-Ingo Ernestus
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Andreas K. Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Mario Löhr
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Camelia-Maria Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
49
|
Dunkl V, Cleff C, Stoffels G, Judov N, Sarikaya-Seiwert S, Law I, Bøgeskov L, Nysom K, Andersen SB, Steiger HJ, Fink GR, Reifenberger G, Shah NJ, Coenen HH, Langen KJ, Galldiks N. The usefulness of dynamic O-(2-18F-fluoroethyl)-L-tyrosine PET in the clinical evaluation of brain tumors in children and adolescents. J Nucl Med 2014; 56:88-92. [PMID: 25525183 DOI: 10.2967/jnumed.114.148734] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Experience regarding O-(2-(18)F-fluoroethyl)-L-tyrosine ((18)F-FET) PET in children and adolescents with brain tumors is limited. METHODS Sixty-nine (18)F-FET PET scans of 48 children and adolescents (median age, 13 y; range, 1-18 y) were analyzed retrospectively. Twenty-six scans to assess newly diagnosed cerebral lesions, 24 scans for diagnosing tumor progression or recurrence, 8 scans for monitoring of chemotherapy effects, and 11 scans for the detection of residual tumor after resection were obtained. Maximum and mean tumor-to-brain ratios (TBRs) were determined at 20-40 min after injection, and time-activity curves of (18)F-FET uptake were assigned to 3 different patterns: constant increase; peak at greater than 20-40 min after injection, followed by a plateau; and early peak (≤ 20 min), followed by a constant descent. The diagnostic accuracy of (18)F-FET PET was assessed by receiver-operating-characteristic curve analyses using histology or clinical course as a reference. RESULTS In patients with newly diagnosed cerebral lesions, the highest accuracy (77%) to detect neoplastic tissue (19/26 patients) was obtained when the maximum TBR was 1.7 or greater (area under the curve, 0.80 ± 0.09; sensitivity, 79%; specificity, 71%; positive predictive value, 88%; P = 0.02). For diagnosing tumor progression or recurrence, the highest accuracy (82%) was obtained when curve patterns 2 or 3 were present (area under the curve, 0.80 ± 0.11; sensitivity, 75%; specificity, 90%; positive predictive value, 90%; P = 0.02). During chemotherapy, a decrease of TBRs was associated with a stable clinical course, and in 2 patients PET detected residual tumor after presumably complete tumor resection. CONCLUSION Our findings suggest that (18)F-FET PET can add valuable information for clinical decision making in pediatric brain tumor patients.
Collapse
Affiliation(s)
- Veronika Dunkl
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany Department of Neurology, University of Cologne, Cologne, Germany
| | - Corvin Cleff
- Department of Neurology, University of Cologne, Cologne, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany
| | - Natalie Judov
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany
| | | | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| | - Lars Bøgeskov
- Department of Neurosurgery, Rigshospitalet, Copenhagen, Denmark
| | - Karsten Nysom
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark
| | - Sofie B Andersen
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| | - Hans-Jakob Steiger
- Department of Neurosurgery, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Gereon R Fink
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany Department of Neurology, University of Cologne, Cologne, Germany
| | - Guido Reifenberger
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany
| | - Heinz H Coenen
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany Department of Nuclear Medicine, University of Aachen, Aachen, Germany; and
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany Department of Neurology, University of Cologne, Cologne, Germany Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany
| |
Collapse
|
50
|
Galldiks N, Dunkl V, Stoffels G, Hutterer M, Rapp M, Sabel M, Reifenberger G, Kebir S, Dorn F, Blau T, Herrlinger U, Hau P, Ruge MI, Kocher M, Goldbrunner R, Fink GR, Drzezga A, Schmidt M, Langen KJ. Diagnosis of pseudoprogression in patients with glioblastoma using O-(2-[18F]fluoroethyl)-L-tyrosine PET. Eur J Nucl Med Mol Imaging 2014; 42:685-95. [PMID: 25411133 DOI: 10.1007/s00259-014-2959-4] [Citation(s) in RCA: 190] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/03/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE The follow-up of glioblastoma patients after radiochemotherapy with conventional MRI can be difficult since reactive alterations to the blood-brain barrier with contrast enhancement may mimic tumour progression (i.e. pseudoprogression, PsP). The aim of this study was to assess the clinical value of O-(2-(18)F-fluoroethyl)-L-tyrosine ((18)F-FET) PET in the differentiation of PsP and early tumour progression (EP) after radiochemotherapy of glioblastoma. METHODS A group of 22 glioblastoma patients with new contrast-enhancing lesions or lesions showing increased enhancement (>25 %) on standard MRI within the first 12 weeks after completion of radiochemotherapy with concomitant temozolomide (median 7 weeks) were additionally examined using amino acid PET with (18)F-FET. Maximum and mean tumour-to-brain ratios (TBRmax, TBRmean) were determined. (18)F-FET uptake kinetic parameters (i.e. patterns of time-activity curves, TAC) were also evaluated. Classification as PsP or EP was based on the clinical course (no treatment change at least for 6 months), follow-up MR imaging and/or histopathological findings. Imaging results were also related to overall survival (OS). RESULTS PsP was confirmed in 11 of the 22 patients. In patients with PsP, (18)F-FET uptake was significantly lower than in patients with EP (TBRmax 1.9 ± 0.4 vs. 2.8 ± 0.5, TBRmean 1.8 ± 0.2 vs. 2.3 ± 0.3; both P < 0.001) and presence of MGMT promoter methylation was significantly more frequent (P = 0.05). Furthermore, a TAC type II or III was more frequently present in patients with EP (P = 0.04). Receiver operating characteristic analysis showed that the optimal (18)F-FET TBRmax cut-off value for identifying PsP was 2.3 (sensitivity 100 %, specificity 91 %, accuracy 96 %, AUC 0.94 ± 0.06; P < 0.001). Univariate survival analysis showed that a TBRmax <2.3 predicted a significantly longer OS (median OS 23 vs. 12 months; P = 0.046). CONCLUSION (18)F-FET PET may facilitate the diagnosis of PsP following radiochemotherapy of glioblastoma.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, University of Cologne, Cologne, Germany,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|