1
|
Weller J, Unterrainer M, Sonderer M, Katzendobler S, Holzgreve A, Biczok A, Harter PN, Tonn JC, Albert NL, Suchorska B. Patterns of intersectional tumor volumes in T2-weighted MRI and [ 18F]FET PET in adult glioma: a prospective, observational study. Sci Rep 2024; 14:23071. [PMID: 39367019 PMCID: PMC11452397 DOI: 10.1038/s41598-024-73681-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 09/19/2024] [Indexed: 10/06/2024] Open
Abstract
Brain tumor volumes as assessed by magnetic resonance imaging (MRI) do not always spatially overlap with biological tumor volumes (BTV) measured by [18F]Fluoroethyltyrosine positron emission tomography ([18F]FET PET). We prospectively investigated volumetric patterns based on the extent of tumor volume overlap between the two modalities. Eighty-six patients with newly diagnosed glioma who had undergone MRI and [18F]FET PET between 2007 and 2009 were included in this prospective study and (re-)classified according to CNS WHO 2021 (Classification of Tumors of the Central Nervous System by the World Health Organization). Four different patterns of volume overlap were defined mathematically according to the extent of overlap between MRI-based T2 tumor volume (non-enhancing tumor volume, nCEV) and BTVs. Progression-free (PFS) and overall survival (OS) were determined. Seventy patients were diagnosed with isocitrate dehydrogenase wildtype (IDHwt) glioblastoma and 16 with IDH-mutant glioma, respectively. The most common pattern was characterized by a larger non-contrast-enhancing tumor volume (nCEV) that enclosed all or most of the BTV and was observed in 46 patients (54%) (pattern 1). This pattern was more frequent in IDH-mutant gliomas than in IDH-wildtype glioblastoma (81% versus 47%, p = 0.02). In multivariate analyses, pattern 1 was associated with prolonged PFS (HR 0.59; 95 CI 0.34-1.0; p = 0.05), but not OS (HR 0.66; 95 CI 0.4-1.08; p = 0.1). For OS, presence of an IDH mutation (p = 0.05) and lower age (p = 0.03) were associated with prolonged OS. The spatial relation between nCEV and BTV varies within and between glioma entities. Most frequently, a larger nCEV encases the BTV. Some patients show spatially dissociated nCEVs and BTVs. Not accounting for this phenomenon in surgery or radiotherapy planning might lead to undertreatment.
Collapse
Affiliation(s)
- Jonathan Weller
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Marcus Unterrainer
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Markéta Sonderer
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sophie Katzendobler
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Annamaria Biczok
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Patrick N Harter
- Center for Neuropathology and Prion Research, LMU University Hospital, LMU Munich, Munich, Germany
| | - Joerg-Christian Tonn
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner site Munich, Heidelberg, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Bogdana Suchorska
- German Consortium for Translational Cancer Research (DKTK), Partner site Munich, Heidelberg, Germany.
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
2
|
Robert JA, Leclerc A, Ducloie M, Emery E, Agostini D, Vigne J. Contribution of [ 18F]FET PET in the Management of Gliomas, from Diagnosis to Follow-Up: A Review. Pharmaceuticals (Basel) 2024; 17:1228. [PMID: 39338390 PMCID: PMC11435125 DOI: 10.3390/ph17091228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/14/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Gliomas, the most common type of primary malignant brain tumors in adults, pose significant challenges in diagnosis and management due to their heterogeneity and potential aggressiveness. This review evaluates the utility of O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) positron emission tomography (PET), a promising imaging modality, to enhance the clinical management of gliomas. We reviewed 82 studies involving 4657 patients, focusing on the application of [18F]FET in several key areas: diagnosis, grading, identification of IDH status and presence of oligodendroglial component, guided resection or biopsy, detection of residual tumor, guided radiotherapy, detection of malignant transformation in low-grade glioma, differentiation of recurrence versus treatment-related changes and prognostic factors, and treatment response evaluation. Our findings confirm that [18F]FET helps delineate tumor tissue, improves diagnostic accuracy, and aids in therapeutic decision-making by providing crucial insights into tumor metabolism. This review underscores the need for standardized parameters and further multicentric studies to solidify the role of [18F]FET PET in routine clinical practice. By offering a comprehensive overview of current research and practical implications, this paper highlights the added value of [18F]FET PET in improving management of glioma patients from diagnosis to follow-up.
Collapse
Affiliation(s)
- Jade Apolline Robert
- CHU de Caen Normandie, UNICAEN, Department of Nuclear Medicine, Normandie Université, 14000 Caen, France; (J.A.R.)
| | - Arthur Leclerc
- Department of Neurosurgery, Caen University Hospital, 14000 Caen, France
- Caen Normandie University, ISTCT UMR6030, GIP Cyceron, 14000 Caen, France
| | - Mathilde Ducloie
- Department of Neurology, Caen University Hospital, 14000 Caen, France
- Centre François Baclesse, Department of Neurology, 14000 Caen, France
| | - Evelyne Emery
- Department of Neurosurgery, Caen University Hospital, 14000 Caen, France
| | - Denis Agostini
- CHU de Caen Normandie, UNICAEN, Department of Nuclear Medicine, Normandie Université, 14000 Caen, France; (J.A.R.)
| | - Jonathan Vigne
- CHU de Caen Normandie, UNICAEN, Department of Nuclear Medicine, Normandie Université, 14000 Caen, France; (J.A.R.)
- CHU de Caen Normandie, UNICAEN Department of Pharmacy, Normandie Université, 14000 Caen, France
- Centre Cyceron, Institut Blood and Brain @ Caen-Normandie, Normandie Université, UNICAEN, INSERM U1237, PhIND, 14000 Caen, France
| |
Collapse
|
3
|
Chehri S, Henriksen OM, Marner L, Christensen M, Muhic A, Poulsen HS, Law I. A prospective clinical study of the influence of oral protein intake on [ 18F]FET-PET uptake and test-retest repeatability in glioma. EJNMMI Res 2024; 14:58. [PMID: 38922458 PMCID: PMC11208353 DOI: 10.1186/s13550-024-01119-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND O-(2-[18F]fluoroethyl)-L-tyrosine positron emission tomography ([18F]FET PET) scanning is used in routine clinical management and evaluation of gliomas with a recommended 4 h prior fasting. Knowledge of test-retest variation of [18F]FET PET imaging uptake metrics and the impact of accidental protein intake can be critical for interpretation. The aim of this study was to investigate the repeatability of [18F]FET-PET metrics and to assess the impact of protein-intake prior to [18F]FET PET scanning of gliomas. RESULTS Test-retest variability in the non-protein group was good with absolute (and relative) upper and lower limits of agreement of + 0.15 and - 0.13 (+ 9.7% and - 9.0%) for mean tumour-to-background ratio (TBRmean), + 0.43 and - 0.28 (+ 19.6% and - 11.8%) for maximal tumour-to-background ratio (TBRmax), and + 2.14 cm3 and - 1.53 ml (+ 219.8% and - 57.3%) for biological tumour volume (BTV). Variation was lower for uptake ratios than for BTV. Protein intake was associated with a 27% increase in the total sum of plasma concentration of the L-type amino acid transporter 1 (LAT1) relevant amino acids and with decreased standardized uptake value (SUV) in both healthy appearing background brain tissue (mean SUV - 25%) and in tumour (maximal SUV - 14%). Oral intake of 24 g of protein 1 h prior to injection of tracer tended to increase variability, but the effects on derived tumour metrics TBRmean and TBRmax were only borderline significant, and changes generally within the variability observed in the group with no protein intake. CONCLUSION The test-retest repeatability was found to be good, and better for TBRmax and TBRmean than BTV, with the methodological limitation that tumour growth may have influenced results. Oral intake of 24 g of protein one hour before a [18F]FET PET scan decreases uptake of [18F]FET in both tumour and in healthy appearing brain, with no clinically significant difference on the most commonly used tumour metrics.
Collapse
Affiliation(s)
- Sarah Chehri
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Radiation Biology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Otto Mølby Henriksen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Lisbeth Marner
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, 2400, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mette Christensen
- Department of Clinical Genetics, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Aida Muhic
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Hans Skovgaard Poulsen
- Department of Radiation Biology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Harat M, Miechowicz I, Rakowska J, Zarębska I, Małkowski B. A Biopsy-Controlled Prospective Study of Contrast-Enhancing Diffuse Glioma Infiltration Based on FET-PET and FLAIR. Cancers (Basel) 2024; 16:1265. [PMID: 38610944 PMCID: PMC11010945 DOI: 10.3390/cancers16071265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 04/14/2024] Open
Abstract
Accurately defining glioma infiltration is crucial for optimizing radiotherapy and surgery, but glioma infiltration is heterogeneous and MRI imperfectly defines the tumor extent. Currently, it is impossible to determine the tumor infiltration gradient within a FLAIR signal. O-(2-[18F]fluoroethyl)-L-tyrosine (FET)-PET often reveals high-grade glioma infiltration beyond contrast-enhancing areas on MRI. Here, we studied FET uptake dynamics in tumor and normal brain structures by dual-timepoint (10 min and 40-60 min post-injection) acquisition to optimize analysis protocols for defining glioma infiltration. Over 300 serial stereotactic biopsies from 23 patients (mean age 47, 12 female/11 male) of diffuse contrast-enhancing gliomas were taken from areas inside and outside contrast enhancement or outside the FET hotspot but inside FLAIR. The final diagnosis was G4 in 11, grade 3 in 10, and grade 2 in 2 patients. The target-to-background (TBRs) ratios and standardized uptake values (SUVs) were calculated in areas used for biopsy planning and in background structures. The optimal method and threshold values were determined to find a preferred strategy for defining glioma infiltration. Standard thresholding (1.6× uptake in the contralateral brain) in standard acquisition PET images differentiated a tumor of any grade from astrogliosis, although the uptake in astrogliosis and grade 2 glioma was similar. Analyzing an optimal strategy for infiltration volume definition astrogliosis could be accurately differentiated from tumor samples using a choroid plexus as a background. Early acquisition improved the AUC in many cases, especially within FLAIR, from 56% to 90% sensitivity and 41% to 61% specificity (standard TBR 1.6 vs. early TBR plexus). The current FET-PET evaluation protocols for contrast-enhancing gliomas are limited, especially at the tumor border where grade 2 tumor and astrogliosis have similar uptake, but using choroid plexus uptake in early acquisitions as a background, we can precisely define a tumor within FLAIR that was outside of the scope of current FET-PET protocols.
Collapse
Affiliation(s)
- Maciej Harat
- Department of Neurooncology and Radiosurgery, Franciszek Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
- Department of Clinical Medicine, Faculty of Medicine, University of Science and Technology, 85-796 Bydgoszcz, Poland
| | - Izabela Miechowicz
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | - Józefina Rakowska
- Department of Neurosurgery, 10th Military Research Hospital, 85-681 Bydgoszcz, Poland;
| | - Izabela Zarębska
- Department of Radiotherapy, Franciszek Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland;
| | - Bogdan Małkowski
- Department of Nuclear Medicine, Franciszek Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
- Department of Diagnostic Imaging, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland
| |
Collapse
|
5
|
Mardanshahi A, Vaseghi S, Hosseinimehr SJ, Abedi SM, Molavipordanjani S. 99mTc(CO) 3-labeled 1-(2-Pyridyl)piperazine derivatives as radioligands for 5-HT 7 receptors. Ann Nucl Med 2024; 38:139-153. [PMID: 38032496 DOI: 10.1007/s12149-023-01885-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND The 5-hydroxytryptamine receptor (5-HTR) family includes seven classes of receptors. The 5-HT7R is the newest member of this family and contributes to different physiological and pathological processes. As a pathology, glioblastoma multiform (GBM) overexpresses 5-HT7R; hence, this study aims to develop radiolabeled aryl piperazine derivatives as 5-HT7R imaging agents. METHODS: Compounds 6 and 7 as 1-(3-nitropyridin-2-yl)piperazine derivatives were radiolabeled with fac-[99mTc(CO)3(H2O)3]+ and 99mTc(CO)3-[6] and 99mTc(CO)3-[7] were obtained with high radiochemical purity (RCP > 94%). The stability of the radiotracers was evaluated in both saline and mouse serum. Specific binding on different cell lines including U-87 MG, MCF-7, SKBR3, and HT-29 was performed. The biodistribution of these radiotracers was evaluated in normal and U-87 MG Xenografted models. Finally, 99mTc(CO)3-[6] and 99mTc(CO)3-[7] were applied for in vivo imaging in U-87 MG Xenografted models. RESULTS Specific binding study indicates that 99mTc(CO)3-[6] and 99mTc(CO)3-[7] can recognize 5-HT7R of U87-MG cell line. The biodistribution study in normal mice indicates that the brain uptake of 99mTc(CO)3-[6] and 99mTc(CO)3-[7] is the highest at 30 min post-injection (0.8 ± 0.25 and 0.64 ± 0.18%ID/g, respectively). The data of the biodistribution study in the U87-MG xenograft model revealed that these radiotracers could accumulate in the tumor site, and the highest tumor uptake was observed at 60 min post-injection (3.38 ± 0.65 and 3.27 ± 0.5%ID/g, respectively). The injection of pimozide can block the tumor's radiotracer uptake, indicating the binding of these radiotracers to the 5-HT7R. The imaging study in the xenograft model also confirms the biodistribution data. The acquired images clearly show the tumor site, and the tumor-to-muscle ratio for 99mTc(CO)3-[6] and 99mTc(CO)3-[7] at 60 min was 3.33 and 3.88, respectively. CONCLUSIONS: 99mTc(CO)3-[6] and 99mTc(CO)3-[7] can visualize tumor in the U87-MG xenograft model due to their affinity toward 5-HT7R.
Collapse
Affiliation(s)
- Alireza Mardanshahi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Samaneh Vaseghi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mohammad Abedi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sajjad Molavipordanjani
- Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
6
|
Filss CP, Cramer J, Löher S, Lohmann P, Stoffels G, Stegmayr C, Kocher M, Heinzel A, Galldiks N, Wittsack HJ, Sabel M, Neumaier B, Scheins J, Shah NJ, Meyer PT, Mottaghy FM, Langen KJ. Assessment of Brain Tumour Perfusion Using Early-Phase 18F-FET PET: Comparison with Perfusion-Weighted MRI. Mol Imaging Biol 2024; 26:36-44. [PMID: 37848641 PMCID: PMC10827807 DOI: 10.1007/s11307-023-01861-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/10/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023]
Abstract
PURPOSE Morphological imaging using MRI is essential for brain tumour diagnostics. Dynamic susceptibility contrast (DSC) perfusion-weighted MRI (PWI), as well as amino acid PET, may provide additional information in ambiguous cases. Since PWI is often unavailable in patients referred for amino acid PET, we explored whether maps of relative cerebral blood volume (rCBV) in brain tumours can be extracted from the early phase of PET using O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET). PROCEDURE Using a hybrid brain PET/MRI scanner, PWI and dynamic 18F-FET PET were performed in 33 patients with cerebral glioma and four patients with highly vascularized meningioma. The time interval from 0 to 2 min p.i. was selected to best reflect the blood pool phase in 18F-FET PET. For each patient, maps of MR-rCBV, early 18F-FET PET (0-2 min p.i.) and late 18F-FET PET (20-40 min p.i.) were generated and coregistered. Volumes of interest were placed on the tumour (VOI-TU) and normal-appearing brain (VOI-REF). The correlation between tumour-to-brain ratios (TBR) of the different parameters was analysed. In addition, three independent observers evaluated MR-rCBV and early 18F-FET maps (18F-FET-rCBV) for concordance in signal intensity, tumour extent and intratumoural distribution. RESULTS TBRs calculated from MR-rCBV and 18F-FET-rCBV showed a significant correlation (r = 0.89, p < 0.001), while there was no correlation between late 18F-FET PET and MR-rCBV (r = 0.24, p = 0.16) and 18F-FET-rCBV (r = 0.27, p = 0.11). Visual rating yielded widely agreeing findings or only minor differences between MR-rCBV maps and 18F-FET-rCBV maps in 93 % of the tumours (range of three independent raters 91-94%, kappa among raters 0.78-1.0). CONCLUSION Early 18F-FET maps (0-2 min p.i.) in gliomas provide similar information to MR-rCBV maps and may be helpful when PWI is not possible or available. Further studies in gliomas are needed to evaluate whether 18F-FET-rCBV provides the same clinical information as MR-rCBV.
Collapse
Affiliation(s)
- Christian P Filss
- Department of Nuclear Medicine, RWTH University Hospital, Aachen, Germany.
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany.
- Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany.
| | - Julian Cramer
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
- Faculty of Medical Engineering and Technomathematics, FH Aachen University of Applied Sciences, Campus Juelich, Jülich, Germany
| | - Saskia Löher
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
- Faculty of Medical Engineering and Technomathematics, FH Aachen University of Applied Sciences, Campus Juelich, Jülich, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Martin Kocher
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
- Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
- Department of Stereotactic and Functional Neurosurgery, Center for Neurosurgery, University Hospital Cologne, Cologne, Germany
| | - Alexander Heinzel
- Department of Nuclear Medicine, RWTH University Hospital, Aachen, Germany
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
- Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
- Department of Nuclear Medicine, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
- Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Hans J Wittsack
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Michael Sabel
- Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
- Department of Neurosurgery, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
- Institute of Radiochemistry and Experimental Molecular Imaging, University Hospital Cologne, Cologne, Germany
| | - Jürgen Scheins
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - N Jon Shah
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
- JARA - BRAIN - Translational Medicine, RWTH Aachen University, Aachen, Germany
- Department of Neurology, RWTH Aachen University Hospital, Aachen, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH University Hospital, Aachen, Germany
- Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands
| | - Karl-Josef Langen
- Department of Nuclear Medicine, RWTH University Hospital, Aachen, Germany
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
- Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
- JARA - BRAIN - Translational Medicine, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
7
|
Barry N, Francis RJ, Ebert MA, Koh ES, Rowshanfarzad P, Hassan GM, Kendrick J, Gan HK, Lee ST, Lau E, Moffat BA, Fitt G, Moore A, Thomas P, Pattison DA, Akhurst T, Alipour R, Thomas EL, Hsiao E, Schembri GP, Lin P, Ly T, Yap J, Kirkwood I, Vallat W, Khan S, Krishna D, Ngai S, Yu C, Beuzeville S, Yeow TC, Bailey D, Cook O, Whitehead A, Dykyj R, Rossi A, Grose A, Scott AM. Delineation and agreement of FET PET biological volumes in glioblastoma: results of the nuclear medicine credentialing program from the prospective, multi-centre trial evaluating FET PET In Glioblastoma (FIG) study-TROG 18.06. Eur J Nucl Med Mol Imaging 2023; 50:3970-3981. [PMID: 37563351 PMCID: PMC10611835 DOI: 10.1007/s00259-023-06371-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023]
Abstract
PURPOSE The O-(2-[18F]-fluoroethyl)-L-tyrosine (FET) PET in Glioblastoma (FIG) trial is an Australian prospective, multi-centre study evaluating FET PET for glioblastoma patient management. FET PET imaging timepoints are pre-chemoradiotherapy (FET1), 1-month post-chemoradiotherapy (FET2), and at suspected progression (FET3). Before participant recruitment, site nuclear medicine physicians (NMPs) underwent credentialing of FET PET delineation and image interpretation. METHODS Sites were required to complete contouring and dynamic analysis by ≥ 2 NMPs on benchmarking cases (n = 6) assessing biological tumour volume (BTV) delineation (3 × FET1) and image interpretation (3 × FET3). Data was reviewed by experts and violations noted. BTV definition includes tumour-to-background ratio (TBR) threshold of 1.6 with crescent-shaped background contour in the contralateral normal brain. Recurrence/pseudoprogression interpretation (FET3) required assessment of maximum TBR (TBRmax), dynamic analysis (time activity curve [TAC] type, time to peak), and qualitative assessment. Intraclass correlation coefficient (ICC) assessed volume agreement, coefficient of variation (CoV) compared maximum/mean TBR (TBRmax/TBRmean) across cases, and pairwise analysis assessed spatial (Dice similarity coefficient [DSC]) and boundary agreement (Hausdorff distance [HD], mean absolute surface distance [MASD]). RESULTS Data was accrued from 21 NMPs (10 centres, n ≥ 2 each) and 20 underwent review. The initial pass rate was 93/119 (78.2%) and 27/30 requested resubmissions were completed. Violations were found in 25/72 (34.7%; 13/12 minor/major) of FET1 and 22/74 (29.7%; 14/8 minor/major) of FET3 reports. The primary reasons for resubmission were as follows: BTV over-contour (15/30, 50.0%), background placement (8/30, 26.7%), TAC classification (9/30, 30.0%), and image interpretation (7/30, 23.3%). CoV median and range for BTV, TBRmax, and TBRmean were 21.53% (12.00-30.10%), 5.89% (5.01-6.68%), and 5.01% (3.37-6.34%), respectively. BTV agreement was moderate to excellent (ICC = 0.82; 95% CI, 0.63-0.97) with good spatial (DSC = 0.84 ± 0.09) and boundary (HD = 15.78 ± 8.30 mm; MASD = 1.47 ± 1.36 mm) agreement. CONCLUSION The FIG study credentialing program has increased expertise across study sites. TBRmax and TBRmean were robust, with considerable variability in BTV delineation and image interpretation observed.
Collapse
Affiliation(s)
- Nathaniel Barry
- School of Physics, Mathematics and Computing, University of Western Australia, WA, Crawley, Australia.
- Centre for Advanced Technologies in Cancer Research (CATCR), WA, Perth, Australia.
| | - Roslyn J Francis
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- Australian Centre for Quantitative Imaging, Medical School, University of Western Australia, Crawley, WA, Australia
| | - Martin A Ebert
- School of Physics, Mathematics and Computing, University of Western Australia, WA, Crawley, Australia
- Centre for Advanced Technologies in Cancer Research (CATCR), WA, Perth, Australia
- Australian Centre for Quantitative Imaging, Medical School, University of Western Australia, Crawley, WA, Australia
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Eng-Siew Koh
- Department of Radiation Oncology, Liverpool and Macarthur Cancer Therapy Centres, Liverpool, NSW, Australia
- South Western Sydney Clinical School, UNSW Medicine, University of New South Wales, Liverpool, NSW, Australia
| | - Pejman Rowshanfarzad
- School of Physics, Mathematics and Computing, University of Western Australia, WA, Crawley, Australia
- Centre for Advanced Technologies in Cancer Research (CATCR), WA, Perth, Australia
| | - Ghulam Mubashar Hassan
- School of Physics, Mathematics and Computing, University of Western Australia, WA, Crawley, Australia
| | - Jake Kendrick
- School of Physics, Mathematics and Computing, University of Western Australia, WA, Crawley, Australia
- Centre for Advanced Technologies in Cancer Research (CATCR), WA, Perth, Australia
| | - Hui K Gan
- Department of Medical Oncology, Austin Hospital, Melbourne, VIC, Australia
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Sze T Lee
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia
| | - Eddie Lau
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia
- Department of Radiology, Austin Health, Melbourne, VIC, Australia
- Department of Radiology, University of Melbourne, Melbourne, VIC, Australia
| | - Bradford A Moffat
- Department of Radiology, University of Melbourne, Melbourne, VIC, Australia
| | - Greg Fitt
- Department of Radiology, Austin Health, Melbourne, VIC, Australia
| | - Alisha Moore
- Trans Tasman Radiation Oncology Group (TROG Cancer Research), University of Newcastle, Callaghan, NSW, Australia
| | - Paul Thomas
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, St Lucia, QLD, Australia
| | - David A Pattison
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, St Lucia, QLD, Australia
| | - Tim Akhurst
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, Melbourne, VIC, Australia
| | - Ramin Alipour
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, Melbourne, VIC, Australia
| | - Elizabeth L Thomas
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Edward Hsiao
- Department of Nuclear Medicine, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Geoffrey P Schembri
- Department of Nuclear Medicine, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Peter Lin
- South Western Sydney Clinical School, UNSW Medicine, University of New South Wales, Liverpool, NSW, Australia
- Department of Nuclear Medicine, Liverpool Hospital, Liverpool, NSW, Australia
| | - Tam Ly
- Department of Nuclear Medicine, Liverpool Hospital, Liverpool, NSW, Australia
| | - June Yap
- Department of Nuclear Medicine, Liverpool Hospital, Liverpool, NSW, Australia
| | - Ian Kirkwood
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Wilson Vallat
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Shahroz Khan
- Department of Nuclear Medicine, Canberra Hospital, Woden, ACT, Australia
| | - Dayanethee Krishna
- Department of Nuclear Medicine, Canberra Hospital, Woden, ACT, Australia
| | - Stanley Ngai
- Department of Nuclear Medicine, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Chris Yu
- Department of Nuclear Medicine, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Scott Beuzeville
- Department of Nuclear Medicine, St George Hospital, Kogarah, NSW, Australia
| | - Tow C Yeow
- Department of Nuclear Medicine, St George Hospital, Kogarah, NSW, Australia
| | - Dale Bailey
- Department of Nuclear Medicine, Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine 7 Health, University of Sydney, Sydney, NSW, Australia
| | - Olivia Cook
- Trans Tasman Radiation Oncology Group (TROG Cancer Research), University of Newcastle, Callaghan, NSW, Australia
| | - Angela Whitehead
- Trans Tasman Radiation Oncology Group (TROG Cancer Research), University of Newcastle, Callaghan, NSW, Australia
| | - Rachael Dykyj
- Trans Tasman Radiation Oncology Group (TROG Cancer Research), University of Newcastle, Callaghan, NSW, Australia
| | - Alana Rossi
- Trans Tasman Radiation Oncology Group (TROG Cancer Research), University of Newcastle, Callaghan, NSW, Australia
| | - Andrew Grose
- Trans Tasman Radiation Oncology Group (TROG Cancer Research), University of Newcastle, Callaghan, NSW, Australia
| | - Andrew M Scott
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia
| |
Collapse
|
8
|
van den Bent MJ, Geurts M, French PJ, Smits M, Capper D, Bromberg JEC, Chang SM. Primary brain tumours in adults. Lancet 2023; 402:1564-1579. [PMID: 37738997 DOI: 10.1016/s0140-6736(23)01054-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 05/06/2023] [Accepted: 05/16/2023] [Indexed: 09/24/2023]
Abstract
The most frequent adult-type primary CNS tumours are diffuse gliomas, but a large variety of rarer CNS tumour types exists. The classification of these tumours is increasingly based on molecular diagnostics, which is reflected in the extensive molecular foundation of the recent WHO 2021 classification of CNS tumours. Resection as extensive as is safely possible is the cornerstone of treatment in most gliomas, and is now also recommended early in the treatment of patients with radiological evidence of histologically low-grade tumours. For the adult-type diffuse glioma, standard of care is a combination of radiotherapy and chemotherapy. Although treatment with curative intent is not available, combined modality treatment has resulted in long-term survival (>10-20 years) for some patients with isocitrate dehydrogenase (IDH) mutant tumours. Other rarer tumours require tailored approaches, best delivered in specialised centres. Targeted treatments based on molecular alterations still only play a minor role in the treatment landscape of adult-type diffuse glioma, and today are mainly limited to patients with tumours with BRAFV600E (ie, Val600Glu) mutations. Immunotherapy for CNS tumours is still in its infancy, and so far, trials with checkpoint inhibitors and vaccination studies have not shown improvement in patient outcomes in glioblastoma. Current research is focused on improving our understanding of the immunosuppressive tumour environment, the molecular heterogeneity of tumours, and the role of tumour microtube network connections between cells in the tumour microenvironment. These factors all appear to play a role in treatment resistance, and indicate that novel approaches are needed to further improve outcomes of patients with CNS tumours.
Collapse
Affiliation(s)
- Martin J van den Bent
- Department of Neurology, Brain Tumor Center, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Netherlands.
| | - Marjolein Geurts
- Department of Neurology, Brain Tumor Center, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Netherlands
| | - Pim J French
- Department of Neurology, Brain Tumor Center, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Netherlands
| | - Marion Smits
- Department of Radiology & Nuclear Medicine, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Netherlands; Medical Delta, Delft, Netherlands
| | - David Capper
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium, Berlin, Germany; German Cancer Research Center, Heidelberg, Germany
| | - Jacoline E C Bromberg
- Department of Neurology, Brain Tumor Center, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Netherlands
| | - Susan M Chang
- Brain Tumor Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
9
|
Harat M, Rakowska J, Harat M, Szylberg T, Furtak J, Miechowicz I, Małkowski B. Combining amino acid PET and MRI imaging increases accuracy to define malignant areas in adult glioma. Nat Commun 2023; 14:4572. [PMID: 37516762 PMCID: PMC10387066 DOI: 10.1038/s41467-023-39731-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/24/2023] [Indexed: 07/31/2023] Open
Abstract
Accurate determination of the extent and grade of adult-type diffuse gliomas is critical to patient management. In clinical practice, contrast-enhancing areas of diffuse gliomas in magnetic resonance imaging (MRI) sequences are usually used to target biopsy, surgery, and radiation therapy, but there can be discrepancies between these areas and the actual tumor extent. Here we show that adding 18F-fluoro-ethyl-tyrosine positron emission tomography (FET-PET) to MRI sequences accurately locates the most malignant areas of contrast-enhancing gliomas, potentially impacting subsequent management and outcomes. We present a prospective analysis of over 300 serial biopsy specimens from 23 patients with contrast-enhancing adult-type diffuse gliomas using a hybrid PET-MRI scanner to compare T2-weighted and contrast-enhancing MRI images with FET-PET. In all cases, we observe and confirm high FET uptake in early PET acquisitions (5-15 min after 18F-FET administration) outside areas of contrast enhancement on MRI, indicative of high-grade glioma. In 30% cases, inclusion of FET-positive sites changes the biopsy result to a higher tumor grade.
Collapse
Affiliation(s)
- Maciej Harat
- Department of Neurooncology and Radiosurgery, Franciszek Lukaszczyk Oncology Center, Bydgoszcz, Poland.
- Department of Oncology and Brachytherapy, Faculty of Medicine, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland.
| | - Józefina Rakowska
- Department of Neurosurgery, 10th Military Research Hospital, Bydgoszcz, Poland
| | - Marek Harat
- Department of Neurosurgery, 10th Military Research Hospital, Bydgoszcz, Poland
- Centre of Medical Sciences, Bydgoszcz, University of Science and Technology, Bydgoszcz, Poland
| | - Tadeusz Szylberg
- Department of Pathomorphology, 10th Military Research Hospital, Bydgoszcz, Poland
| | - Jacek Furtak
- Department of Neurosurgery, 10th Military Research Hospital, Bydgoszcz, Poland
| | - Izabela Miechowicz
- Department of Computer Science and Statistics, University of Medical Sciences, Poznan, Poland
| | - Bogdan Małkowski
- Department of Nuclear Medicine, Franciszek Lukaszczyk Oncology Center, Bydgoszcz, Poland.
- Department of Positron Emission Tomography and Molecular Imaging, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland.
| |
Collapse
|
10
|
Laudicella R, Mantarro C, Catalfamo B, Alongi P, Gaeta M, Minutoli F, Baldari S, Bisdas S. PET Imaging in Gliomas. RADIOLOGY‐NUCLEAR MEDICINE DIAGNOSTIC IMAGING 2023:194-218. [DOI: 10.1002/9781119603627.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Karimi M, Mardanshahi A, Irannejad H, Mohammad Abedi S, Molavipordanjani S. Synthesis and evaluation of 99mTc-labeled 1-(2-Pyridyl)piperazine derivatives as radioligands for 5HT 7 receptors. Bioorg Chem 2023; 135:106486. [PMID: 36965286 DOI: 10.1016/j.bioorg.2023.106486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/07/2023] [Accepted: 03/17/2023] [Indexed: 03/27/2023]
Abstract
Glioblastoma multiform (GBM) is one of the most aggressive tumors of the central nervous system in humans. GBM overexpresses serotonin-7 receptors (5-HT7Rs); hence, this study aims to develop 5-HT7R targeted radiotracers. Aryl piperazine derivatives can act as ligands for 5-HT7R. Therefore, compounds 6 and 7 as 1-(3-nitropyridin-2-yl)piperazine derivatives were synthesized and radiolabeled with 99mTcN2+ core. Radiolabeled 6 and 7 (99mTcN-[6] and 99mTcN-[7]) were prepared with high radiochemical purity (RCP > 96%). They displayed high affinity toward U-87 MG cell line 5-HT7R. The calculated Ki for 99mTcN-[7] was lower than that of 99mTcN-[6] (14.85 ± 0.32 vs 22.57 ± 0.73 nM) which indicates the higher affinity of 99mTcN-[7] toward 5-HT7R. A molecular docking study also confirmed the binding of these radiotracers to 5-HT7R. The biodistribution study in normal mice revealed that 99mTcN-[7] has the highest brain accumulation at 30 min post-injection (0.54 ± 0.12 %ID/g) while the uptake of 99mTcN-[6] is much lower (0.14 ± 0.02 %ID/g). The biodistribution study in the xenograft model confirms that the radiotracers recognize the tumor site. 99mTcN-[6], and 99mTcN-[7] showed the highest tumor uptake at 1-hour post-injection (5.44 ± 0.58 vs 4.94 ± 1.65 %ID/g) and tumor-to-muscle ratios were (4.61 vs. 5.61). The injection of pimozide blocks the receptors and significantly reduces the tumor-to-muscle ratios at 1-hour post-injection to 0.81 and 0.31, respectively. In correlation with in vitro study, 99mTcN-[6] and 99mTcN-[7] visualize the tumor site in U-87 MG glioma xenografted nude mice and display the tumor-to-muscle ratios of 7.05 and 6.03.
Collapse
Affiliation(s)
- Maryam Karimi
- Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Mardanshahi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamid Irannejad
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mohammad Abedi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sajjad Molavipordanjani
- Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
12
|
Kertels O, Krauß J, Monoranu CM, Samnick S, Dierks A, Kircher M, Mihovilovic MI, Pham M, Buck AK, Eyrich M, Schlegel PG, Frühwald MC, Bison B, Lapa C. [ 18F]FET-PET in children and adolescents with central nervous system tumors: does it support difficult clinical decision-making? Eur J Nucl Med Mol Imaging 2023; 50:1699-1708. [PMID: 36670283 PMCID: PMC10119036 DOI: 10.1007/s00259-023-06114-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/11/2023] [Indexed: 01/22/2023]
Abstract
PURPOSE Positron emission tomography (PET) with O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) is a well-established tool for non-invasive assessment of adult central nervous system (CNS) tumors. However, data on its diagnostic utility and impact on clinical management in children and adolescents are limited. METHODS Twenty-one children and young adults (13 males; mean age, 8.6 ± 5.2 years; range, 1-19 at initial diagnosis) with either newly diagnosed (n = 5) or pretreated (n = 16) CNS tumors were retrospectively analyzed. All patients had previously undergone neuro-oncological work-up including cranial magnetic resonance imaging. In all cases, [18F]FET-PET was indicated in a multidisciplinary team conference. The impact of PET imaging on clinical decision-making was assessed. Histopathology (n = 12) and/or clinical and imaging follow-up (n = 9) served as the standard of reference. RESULTS The addition of [18F]FET-PET to the available information had an impact on further patient management in 14 out of 21 subjects, with avoidance of invasive surgery or biopsy in four patients, biopsy guidance in four patients, change of further treatment in another five patients, and confirmation of diagnosis in one patient. CONCLUSION [18F]FET-PET may provide important additional information for treatment guidance in pediatric and adolescent patients with CNS tumors.
Collapse
Affiliation(s)
- Olivia Kertels
- Institute of Diagnostic and Interventional Radiology, University Hospital Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany
| | - Jürgen Krauß
- Section Pediatric Neurosurgery, Department of Neurosurgery, University Hospital Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany
| | - Camelia Maria Monoranu
- Department of Neuropathology, Institute for Pathology, University of Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany
| | - Alexander Dierks
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Malte Kircher
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Milena I. Mihovilovic
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany
| | - Mirko Pham
- Institute of Diagnostic and Interventional Neuroradiology, University Hospital Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| | - Andreas K. Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany
| | - Matthias Eyrich
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Würzburg, Josef-Schneider- Str. 2, 97080 Würzburg, Germany
| | - Paul-Gerhardt Schlegel
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Würzburg, Josef-Schneider- Str. 2, 97080 Würzburg, Germany
| | - Michael C. Frühwald
- Paediatric and Adolescent Medicine, University Medical Center Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Brigitte Bison
- Diagnostic and Interventional Neuroradiology, Neuroradiological Reference Center for Pediatric Brain Tumor (HIT) Studies of the German Society of Pediatric Oncology and Hematology, Faculty of Medicine, University of Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| |
Collapse
|
13
|
TSPO PET signal using [ 18F]GE180 is associated with survival in recurrent gliomas. Eur J Nucl Med Mol Imaging 2023; 50:859-869. [PMID: 36329288 PMCID: PMC9852133 DOI: 10.1007/s00259-022-06006-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE Glioma patients, especially recurrent glioma, suffer from a poor prognosis. While advances to classify glioma on a molecular level improved prognostication at initial diagnosis, markers to prognosticate survival in the recurrent situation are still needed. As 18 kDa translocator protein (TSPO) was previously reported to be associated with aggressive histopathological glioma features, we correlated the TSPO positron emission tomography (PET) signal using [18F]GE180 in a large cohort of recurrent glioma patients with their clinical outcome. METHODS In patients with [18F]GE180 PET at glioma recurrence, [18F]GE180 PET parameters (e.g., SUVmax) as well as other imaging features (e.g., MRI volume, [18F]FET PET parameters when available) were evaluated together with patient characteristics (age, sex, Karnofsky-Performance score) and neuropathological features (e.g. WHO 2021 grade, IDH-mutation status). Uni- and multivariate Cox regression and Kaplan-Meier survival analyses were performed to identify prognostic factors for post-recurrence survival (PRS) and time to treatment failure (TTF). RESULTS Eighty-eight consecutive patients were evaluated. TSPO tracer uptake correlated with tumor grade at recurrence (p < 0.05), with no significant differences in IDH-wild-type versus IDH-mutant tumors. Within the subgroup of IDH-mutant glioma (n = 46), patients with low SUVmax (median split, ≤ 1.60) had a significantly longer PRS (median 41.6 vs. 25.3 months, p = 0.031) and TTF (32.2 vs 8.7 months, p = 0.001). Also among IDH-wild-type glioblastoma (n = 42), patients with low SUVmax (≤ 1.89) had a significantly longer PRS (median not reached vs 8.2 months, p = 0.002). SUVmax remained an independent prognostic factor for PRS in the multivariate analysis including CNS WHO 2021 grade, IDH status, and age. Tumor volume defined by [18F]FET PET or contrast-enhanced MRI correlated weakly with TSPO tracer uptake. Treatment regimen did not differ among the median split subgroups. CONCLUSION Our data suggest that TSPO PET using [18F]GE180 can help to prognosticate recurrent glioma patients even among homogeneous molecular subgroups and may therefore serve as valuable non-invasive biomarker for individualized patient management.
Collapse
|
14
|
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
|
15
|
Henssen D, Meijer F, Verburg FA, Smits M. Challenges and opportunities for advanced neuroimaging of glioblastoma. Br J Radiol 2023; 96:20211232. [PMID: 36062962 PMCID: PMC10997013 DOI: 10.1259/bjr.20211232] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 08/10/2022] [Accepted: 08/25/2022] [Indexed: 11/05/2022] Open
Abstract
Glioblastoma is the most aggressive of glial tumours in adults. On conventional magnetic resonance (MR) imaging, these tumours are observed as irregular enhancing lesions with areas of infiltrating tumour and cortical expansion. More advanced imaging techniques including diffusion-weighted MRI, perfusion-weighted MRI, MR spectroscopy and positron emission tomography (PET) imaging have found widespread application to diagnostic challenges in the setting of first diagnosis, treatment planning and follow-up. This review aims to educate readers with regard to the strengths and weaknesses of the clinical application of these imaging techniques. For example, this review shows that the (semi)quantitative analysis of the mentioned advanced imaging tools was found useful for assessing tumour aggressiveness and tumour extent, and aids in the differentiation of tumour progression from treatment-related effects. Although these techniques may aid in the diagnostic work-up and (post-)treatment phase of glioblastoma, so far no unequivocal imaging strategy is available. Furthermore, the use and further development of artificial intelligence (AI)-based tools could greatly enhance neuroradiological practice by automating labour-intensive tasks such as tumour measurements, and by providing additional diagnostic information such as prediction of tumour genotype. Nevertheless, due to the fact that advanced imaging and AI-diagnostics is not part of response assessment criteria, there is no harmonised guidance on their use, while at the same time the lack of standardisation severely hampers the definition of uniform guidelines.
Collapse
Affiliation(s)
- Dylan Henssen
- Department of Medical Imaging, Radboud university medical
center, Nijmegen, The Netherlands
| | - Frederick Meijer
- Department of Medical Imaging, Radboud university medical
center, Nijmegen, The Netherlands
| | - Frederik A. Verburg
- Department of Medical Imaging, Radboud university medical
center, Nijmegen, The Netherlands
| | - Marion Smits
- Department of Medical Imaging, Radboud university medical
center, Nijmegen, The Netherlands
| |
Collapse
|
16
|
Chen S, Jiang J, Shen A, Miao Y, Cao Y, Zhang Y, Cong P, Gao P. Rewired Metabolism of Amino Acids and Its Roles in Glioma Pathology. Metabolites 2022; 12:918. [PMID: 36295820 PMCID: PMC9611130 DOI: 10.3390/metabo12100918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/21/2022] Open
Abstract
Amino acids (AAs) are indispensable building blocks of diverse bio-macromolecules as well as functional regulators for various metabolic processes. The fact that cancer cells live with a voracious appetite for specific AAs has been widely recognized. Glioma is one of the most lethal malignancies occurring in the central nervous system. The reprogrammed metabolism of AAs benefits glioma proliferation, signal transduction, epigenetic modification, and stress tolerance. Metabolic alteration of specific AAs also contributes to glioma immune escape and chemoresistance. For clinical consideration, fluctuations in the concentrations of AAs observed in specific body fluids provides opportunities to develop new diagnosis and prognosis markers. This review aimed at providing an extra dimension to understanding glioma pathology with respect to the rewired AA metabolism. A deep insight into the relevant fields will help to pave a new way for new therapeutic target identification and valuable biomarker development.
Collapse
Affiliation(s)
- Sirui Chen
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Jingjing Jiang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ao Shen
- HE University, Shenyang 110163, China
| | - Ying Miao
- E&M College, Shenyang Aerospace University, Shenyang 110136, China
| | - Yunfeng Cao
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
| | - Ying Zhang
- Internal Medicine Department, Dalian Public Health Clinical Center, Dalian 116033, China
| | - Peiyu Cong
- Neurosurgery Department, Affiliated Dalian Municipal Central Hospital of Dalian Medical University, Dalian 116022, China
| | - Peng Gao
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, China
| |
Collapse
|
17
|
Castello A, Castellani M, Florimonte L, Ciccariello G, Mansi L, Lopci E. PET radiotracers in glioma: a review of clinical indications and evidence. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00523-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
18
|
Xu J, Meng Y, Qiu K, Topatana W, Li S, Wei C, Chen T, Chen M, Ding Z, Niu G. Applications of Artificial Intelligence Based on Medical Imaging in Glioma: Current State and Future Challenges. Front Oncol 2022; 12:892056. [PMID: 35965542 PMCID: PMC9363668 DOI: 10.3389/fonc.2022.892056] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/22/2022] [Indexed: 12/24/2022] Open
Abstract
Glioma is one of the most fatal primary brain tumors, and it is well-known for its difficulty in diagnosis and management. Medical imaging techniques such as magnetic resonance imaging (MRI), positron emission tomography (PET), and spectral imaging can efficiently aid physicians in diagnosing, treating, and evaluating patients with gliomas. With the increasing clinical records and digital images, the application of artificial intelligence (AI) based on medical imaging has reduced the burden on physicians treating gliomas even further. This review will classify AI technologies and procedures used in medical imaging analysis. Additionally, we will discuss the applications of AI in glioma, including tumor segmentation and classification, prediction of genetic markers, and prediction of treatment response and prognosis, using MRI, PET, and spectral imaging. Despite the benefits of AI in clinical applications, several issues such as data management, incomprehension, safety, clinical efficacy evaluation, and ethical or legal considerations, remain to be solved. In the future, doctors and researchers should collaborate to solve these issues, with a particular emphasis on interdisciplinary teamwork.
Collapse
Affiliation(s)
- Jiaona Xu
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuting Meng
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kefan Qiu
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Win Topatana
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shijie Li
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Wei
- Department of Neurology, Affiliated Ningbo First Hospital, Ningbo, China
| | - Tianwen Chen
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingyu Chen
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| | - Guozhong Niu
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| |
Collapse
|
19
|
Borja AJ, Saini J, Raynor WY, Ayubcha C, Werner TJ, Alavi A, Revheim ME, Nagaraj C. Role of Molecular Imaging with PET/MR Imaging in the Diagnosis and Management of Brain Tumors. PET Clin 2022; 17:431-451. [PMID: 35662494 DOI: 10.1016/j.cpet.2022.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gliomas are the most common primary brain tumors. Hybrid PET/MR imaging has revolutionized brain tumor imaging, allowing for noninvasive, simultaneous assessment of morphologic, functional, metabolic, and molecular parameters within the brain. Molecular information obtained from PET imaging may aid in the detection, classification, prognostication, and therapeutic decision making for gliomas. 18F-fluorodeoxyglucose (FDG) has been widely used in the setting of brain tumor imaging, and multiple techniques may be employed to optimize this methodology. More recently, a number of non-18F-FDG-PET radiotracers have been applied toward brain tumor imaging and are used in clinical practice.
Collapse
Affiliation(s)
- Austin J Borja
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Jitender Saini
- Department of Neuro Imaging and Interventional Radiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, Karnataka 560-029, India
| | - William Y Raynor
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Cyrus Ayubcha
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Mona-Elisabeth Revheim
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Sognsvannsveien 20, Oslo 0372, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Problemveien 7, Oslo 0315, Norway
| | - Chandana Nagaraj
- Department of Neuro Imaging and Interventional Radiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, Karnataka 560-029, India.
| |
Collapse
|
20
|
Bernstock JD, Gary SE, Klinger N, Valdes PA, Ibn Essayed W, Olsen HE, Chagoya G, Elsayed G, Yamashita D, Schuss P, Gessler FA, Peruzzi PP, Bag A, Friedman GK. Standard clinical approaches and emerging modalities for glioblastoma imaging. Neurooncol Adv 2022; 4:vdac080. [PMID: 35821676 PMCID: PMC9268747 DOI: 10.1093/noajnl/vdac080] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary adult intracranial malignancy and carries a dismal prognosis despite an aggressive multimodal treatment regimen that consists of surgical resection, radiation, and adjuvant chemotherapy. Radiographic evaluation, largely informed by magnetic resonance imaging (MRI), is a critical component of initial diagnosis, surgical planning, and post-treatment monitoring. However, conventional MRI does not provide information regarding tumor microvasculature, necrosis, or neoangiogenesis. In addition, traditional MRI imaging can be further confounded by treatment-related effects such as pseudoprogression, radiation necrosis, and/or pseudoresponse(s) that preclude clinicians from making fully informed decisions when structuring a therapeutic approach. A myriad of novel imaging modalities have been developed to address these deficits. Herein, we provide a clinically oriented review of standard techniques for imaging GBM and highlight emerging technologies utilized in disease characterization and therapeutic development.
Collapse
Affiliation(s)
- Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Sam E Gary
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Neil Klinger
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Pablo A Valdes
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Walid Ibn Essayed
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Hannah E Olsen
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Gustavo Chagoya
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Galal Elsayed
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Daisuke Yamashita
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Patrick Schuss
- Department of Neurosurgery, Unfallkrankenhaus Berlin , Berlin, Germany
| | | | - Pier Paolo Peruzzi
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Asim Bag
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital , Memphis, TN USA
| | - Gregory K Friedman
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham , Birmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham , AL, USA
| |
Collapse
|
21
|
Carrete LR, Young JS, Cha S. Advanced Imaging Techniques for Newly Diagnosed and Recurrent Gliomas. Front Neurosci 2022; 16:787755. [PMID: 35281485 PMCID: PMC8904563 DOI: 10.3389/fnins.2022.787755] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Management of gliomas following initial diagnosis requires thoughtful presurgical planning followed by regular imaging to monitor treatment response and survey for new tumor growth. Traditional MR imaging modalities such as T1 post-contrast and T2-weighted sequences have long been a staple of tumor diagnosis, surgical planning, and post-treatment surveillance. While these sequences remain integral in the management of gliomas, advances in imaging techniques have allowed for a more detailed characterization of tumor characteristics. Advanced MR sequences such as perfusion, diffusion, and susceptibility weighted imaging, as well as PET scans have emerged as valuable tools to inform clinical decision making and provide a non-invasive way to help distinguish between tumor recurrence and pseudoprogression. Furthermore, these advances in imaging have extended to the operating room and assist in making surgical resections safer. Nevertheless, surgery, chemotherapy, and radiation treatment continue to make the interpretation of MR changes difficult for glioma patients. As analytics and machine learning techniques improve, radiomics offers the potential to be more quantitative and personalized in the interpretation of imaging data for gliomas. In this review, we describe the role of these newer imaging modalities during the different stages of management for patients with gliomas, focusing on the pre-operative, post-operative, and surveillance periods. Finally, we discuss radiomics as a means of promoting personalized patient care in the future.
Collapse
Affiliation(s)
- Luis R. Carrete
- University of California San Francisco School of Medicine, San Francisco, CA, United States
| | - Jacob S. Young
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Jacob S. Young,
| | - Soonmee Cha
- Department of Radiology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
22
|
Vettermann FJ, Diekmann C, Weidner L, Unterrainer M, Suchorska B, Ruf V, Dorostkar M, Wenter V, Herms J, Tonn JC, Bartenstein P, Riemenschneider MJ, Albert NL. L-type amino acid transporter (LAT) 1 expression in 18F-FET-negative gliomas. EJNMMI Res 2021; 11:124. [PMID: 34905134 PMCID: PMC8671595 DOI: 10.1186/s13550-021-00865-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/26/2021] [Indexed: 12/24/2022] Open
Abstract
Background O-(2-[18F]-fluoroethyl)-L-tyrosine (18F-FET) is a highly sensitive PET tracer for glioma imaging, and its uptake is suggested to be driven by an overexpression of the L-type amino-acid transporter 1 (LAT1). However, 30% of low- and 5% of high-grade gliomas do not present enhanced 18F-FET uptake at primary diagnosis (“18F-FET-negative gliomas”) and the pathophysiologic basis for this phenomenon remains unclear. The aim of this study was to determine the expression of LAT1 in a homogeneous group of newly diagnosed 18F-FET-negative gliomas and to compare them to a matched group of 18F-FET-positive gliomas. Forty newly diagnosed IDH-mutant astrocytomas without 1p/19q codeletion were evaluated (n = 20 18F-FET-negative (tumour-to-background ratio (TBR) < 1.6), n = 20 18F-FET-positive gliomas (TBR > 1.6)). LAT1 immunohistochemistry (IHC) was performed using SLC7A5/LAT1 antibody. The percentage of LAT1-positive tumour cells (%) and the staining intensity (range 0–2) were multiplied to an overall score (H-score; range 0–200) and correlated to PET findings as well as progression-free survival (PFS). Results IHC staining of LAT1 expression was positive in both, 18F-FET-positive as well as 18F-FET-negative gliomas. No differences were found between the 18F-FET-negative and 18F-FET-positive group with regard to percentage of LAT1-positive tumour cells, staining intensity or H-score. Interestingly, the LAT1 expression showed a significant negative correlation with the PFS (p = 0.031), whereas no significant correlation was found for TBRmax, neither in the overall group nor in the 18F-FET-positive group only (p = 0.651 and p = 0.140). Conclusion Although LAT1 is reported to mediate the uptake of 18F-FET into tumour cells, the levels of LAT1 expression do not correlate with the levels of 18F-FET uptake in IDH-mutant astrocytomas. In particular, the lack of tracer uptake in 18F-FET-negative gliomas cannot be explained by a reduced LAT1 expression. A higher LAT1 expression in IDH-mutant astrocytomas seems to be associated with a short PFS. Further studies regarding mechanisms influencing the uptake of 18F-FET are necessary. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-021-00865-9.
Collapse
Affiliation(s)
- Franziska J Vettermann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
| | - Caroline Diekmann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lorraine Weidner
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Marcus Unterrainer
- Department of Radiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Bogdana Suchorska
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany.,Department of Neurosurgery, Sana Hospital, Duisburg, Germany
| | - Viktoria Ruf
- Center for Neuropathology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Mario Dorostkar
- Center for Neuropathology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Vera Wenter
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jörg-Christian Tonn
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
23
|
Tatkovic A, McBean R, Perkins E, Wong D. 18 F-FET PET maximum standard uptake value and WHO tumour classification grade in glioma. J Med Imaging Radiat Oncol 2021; 66:332-336. [PMID: 34510758 DOI: 10.1111/1754-9485.13322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/09/2021] [Accepted: 08/22/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND In the area of oncology, molecular imaging techniques are becoming increasingly utilised. In neuro-oncology imaging, 18 Fluoro-O-(2) fluoroethyl-L-tyrosine (18 F-FET) is one of the molecular tracers used in positron-emission tomography (PET). Here, we investigated the correlation between maximum standard uptake value (SUV) of 18 F-FET PET and histologically determined World Health Organization (WHO) grade in glioma. PATIENTS AND METHODS This was a retrospective review of all 18 F-FET PET studies conducted between August 2014 and August 2019. Review was conducted to identify imaging studies performed on patients who had a glioma with histopathology results from surgical resection or biopsy available. RESULTS A total of 31 18 F-FET PET studies of histologically confirmed glioma were included. WHO grades ranged from II-IV. A positive correlation between maximum SUV uptake on 18 F-FET PET and WHO grade was observed. CONCLUSIONS There was a correlation identified between WHO glioma grade and maximum SUV on 18 F-FET PET. Further studies are recommended to explore this relationship.
Collapse
Affiliation(s)
- Annaleis Tatkovic
- I-MED Radiology, The Wesley Hospital, Auchenflower, Queensland, Australia
| | - Rhiannon McBean
- I-MED Radiology, The Wesley Hospital, Auchenflower, Queensland, Australia
| | - Ebony Perkins
- I-MED Radiology, The Wesley Hospital, Auchenflower, Queensland, Australia
| | - David Wong
- I-MED Radiology, The Wesley Hospital, Auchenflower, Queensland, Australia
| |
Collapse
|
24
|
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
|
25
|
Repeatability of image features extracted from FET PET in application to post-surgical glioblastoma assessment. Phys Eng Sci Med 2021; 44:1131-1140. [PMID: 34436751 DOI: 10.1007/s13246-021-01049-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/18/2021] [Indexed: 11/27/2022]
Abstract
Positron emission tomography (PET) imaging using the amino acid tracer O-[2-(18F)fluoroethyl]-L-tyrosine (FET) has gained increased popularity within the past decade in the management of glioblastoma (GBM). Radiomics features extracted from FET PET images may be sensitive to variations when imaging at multiple time points. It is therefore necessary to assess feature robustness to test-retest imaging. Eight patients with histologically confirmed GBM that had undergone post-surgical test-retest FET PET imaging were recruited. In total, 1578 radiomic features were extracted from biological tumour volumes (BTVs) delineated using a semi-automatic contouring method. Feature repeatability was assessed using the intraclass correlation coefficient (ICC). The effect of both bin width and filter choice on feature repeatability was also investigated. 59/106 (55.7%) features from the original image and 843/1472 (57.3%) features from filtered images had an ICC ≥ 0.85. Shape and first order features were most stable. Choice of bin width showed minimal impact on features defined as stable. The Laplacian of Gaussian (LoG, σ = 5 mm) and Wavelet filters (HLL and LHL) significantly improved feature repeatability (p ≪ 0.0001, p = 0.003, p = 0.002, respectively). Correlation of textural features with tumour volume was reported for transparency. FET PET radiomic features extracted from post-surgical images of GBM patients that are robust to test-retest imaging were identified. An investigation with a larger dataset is warranted to validate the findings in this study.
Collapse
|
26
|
Stegmayr C, Surges R, Choi CH, Burda N, Stoffels G, Filß C, Willuweit A, Neumaier B, Heinzel A, Shah NJ, Mottaghy FM, Langen KJ. Investigation of Cerebral O-(2-[ 18F]Fluoroethyl)-L-Tyrosine Uptake in Rat Epilepsy Models. Mol Imaging Biol 2021; 22:1255-1265. [PMID: 32409931 PMCID: PMC7497431 DOI: 10.1007/s11307-020-01503-x] [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] [Indexed: 12/19/2022]
Abstract
PURPOSE A recent study reported on high, longer lasting and finally reversible cerebral uptake of O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) induced by epileptic activity. Therefore, we examined cerebral [18F]FET uptake in two chemically induced rat epilepsy models and in patients with focal epilepsy to further investigate whether this phenomenon represents a major pitfall in brain tumor diagnostics and whether [18F]FET may be a potential marker to localize epileptic foci. PROCEDURES Five rats underwent kainic acid titration to exhibit 3 to 3.5 h of class IV-V motor seizures (status epilepticus, SE). Rats underwent 4× [18F]FET PET and 4× MRI on the following 25 days. Six rats underwent kindling with pentylenetetrazol (PTZ) 3 to 8×/week over 10 weeks, and hence, seizures increased from class I to class IV. [18F]FET PET and MRI were performed regularly on days with and without seizures. Four rats served as healthy controls. Additionally, five patients with focal epilepsy underwent [18F]FET PET within 12 days after the last documented seizure. RESULTS No abnormalities in [18F]FET PET or MRI were detected in the kindling model. The SE model showed significantly decreased [18F]FET uptake 3 days after SE in all examined brain regions, and especially in the amygdala region, which normalized within 2 weeks. Corresponding signal alterations in T2-weighted MRI were noted in the amygdala and hippocampus, which recovered 24 days post-SE. No abnormality of cerebral [18F]FET uptake was noted in the epilepsy patients. CONCLUSIONS There was no evidence for increased cerebral [18F]FET uptake after epileptic seizures neither in the rat models nor in patients. The SE model even showed decreased [18F]FET uptake throughout the brain. We conclude that epileptic seizures per se do not cause a longer lasting increased [18F]FET accumulation and are unlikely to be a major cause of pitfall for brain tumor diagnostics.
Collapse
Affiliation(s)
- Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.
| | - Rainer Surges
- Department of Neurology, RWTH University Aachen, Aachen, Germany.,Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Chang-Hoon Choi
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Nicole Burda
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Christian Filß
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Alexander Heinzel
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany
| | - N Jon Shah
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Neurology, RWTH University Aachen, Aachen, Germany.,JARA - BRAIN - Translational Medicine, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany.,Centre of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany.,JARA - BRAIN - Translational Medicine, Aachen, Germany.,Centre of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
| |
Collapse
|
27
|
Nose-to-brain delivery: exploring newer domains for glioblastoma multiforme management. Drug Deliv Transl Res 2021; 10:1044-1056. [PMID: 32221847 DOI: 10.1007/s13346-020-00747-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive form of the primary brain tumors in humans. The intricate pathophysiology, the development of resistance by tumor cells, and the inability of the drugs to effectively cross the blood-brain and blood-tumor barriers result in poor prognosis for GBM patients, with a median survival time of only 1 to 2 years. Nose-to-brain delivery offers an attractive, noninvasive strategy to enhance drug penetration or transport novel drug/gene carriers into the brain. Although the exact mechanism of intranasal delivery remains elusive, the olfactory and trigeminal nerve pathways have been found to play a vital role in circumventing the traditional barriers of brain targeting. This review discusses the intranasal pathway as a novel domain for delivering drugs and nanocarriers encapsulating drugs/genes, as well as stem cell carriers specifically to the glioma cells. Considering the fact that most of these studies are still in preclinical stage, translating such intranasal delivery strategies from bench to bedside would be a critical step for better management and prognosis of GBM. Graphical abstract.
Collapse
|
28
|
18F-FET PET Uptake Characteristics of Long-Term IDH-Wildtype Diffuse Glioma Survivors. Cancers (Basel) 2021; 13:cancers13133163. [PMID: 34202726 PMCID: PMC8268019 DOI: 10.3390/cancers13133163] [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: 05/14/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 11/28/2022] Open
Abstract
Simple Summary IDH-wildtype (IDHwt) gliomas represent a tumor entity with poor overall survival. Only rare cases have an overall survival over several years. Dynamic and static 18F-FET PET is recommended as valuable complementary tool for glioma imaging in gliomas. This study shows that, besides molecular genetic prognosticators, long survival (≥36 months survival) in IDHwt gliomas is associated with a longer time-to-peak and smaller volume on 18F-FET PET at initial diagnosis compared to glioma patients with a short-term survival (≤15 months survival). 18F-FET uptake intensity and MRI-derived tumor size do not differ in patients with long-term survival compared to patient with a short-term survival. Abstract Background: IDHwt diffuse gliomas represent the tumor entity with one of the worst clinical outcomes. Only rare cases present with a long-term survival of several years. Here we aimed at comparing the uptake characteristics on dynamic 18F-FET PET, clinical and molecular genetic parameters of long-term survivors (LTS) versus short-term survivors (STS): Methods: Patients with de-novo IDHwt glioma (WHO grade III/IV) and 18F-FET PET prior to any therapy were stratified into LTS (≥36 months survival) and STS (≤15 months survival). Static and dynamic 18F-FET PET parameters (mean/maximal tumor-to-background ratio (TBRmean/max), biological tumor volume (BTV), minimal time-to-peak (TTPmin)), diameter and volume of contrast-enhancement on MRI, clinical parameters (age, sex, Karnofksy-performance-score), mode of surgery; initial treatment and molecular genetics were assessed and compared between LTS and STS. Results: Overall, 75 IDHwt glioma patients were included (26 LTS, 49 STS). LTS were significantly younger (p < 0.001), had a higher rate of WHO grade III glioma (p = 0.032), of O(6)-Methylguanine-DNA methyltransferase (MGMT) promoter methylation (p < 0.001) and missing Telomerase reverse transcriptase promoter (TERTp) mutations (p = 0.004) compared to STS. On imaging, LTS showed a smaller median BTV (p = 0.017) and a significantly longer TTPmin (p = 0.008) on 18F-FET PET than STS, while uptake intensity (TBRmean/max) did not differ. In contrast to the tumor-volume on PET, MRI-derived parameters such as tumor size as well as all other above-mentioned parameters did not differ between LTS and STS (p > 0.05 each). Conclusion: Besides molecular genetic prognosticators, a long survival time in IDHwt glioma patients is associated with a longer TTPmin as well as a smaller BTV on 18F-FET PET at initial diagnosis. 18F-FET uptake intensity as well as the MRI-derived tumor size (volume and maximal diameter) do not differ in patients with long-term survival.
Collapse
|
29
|
Saednia S, Emami S, Molavipordanjani S, Abedi SM, Amiri FT, Hosseinimehr SJ. Synthesis and Biological Evaluation of 99mTc-Labeled Phenylpiperazine Derivatives as Selective Serotonin-7 Receptor Ligands for Brain Tumor Imaging. Mol Pharm 2021; 18:2360-2374. [PMID: 34027660 DOI: 10.1021/acs.molpharmaceut.1c00172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
With a poor prognosis, glioblastoma multiforme is the most aggressive tumor of the central nervous system in humans. The aim of this study was to develop novel tracers for the tumor targeting and imaging of overexpressed serotonin-7 receptors (5-HT7Rs) in U-87 MG glioma xenografted nude mice. Two phenylpiperazine derivatives named as PHH and MPHH were designed, and the corresponding radiotracers 99mTc-PHH and 99mTc-MPHH were synthesized in high radiochemical purity (>95%). 99mTc-MPHH showed a higher affinity to 5-HT7Rs on U-87 MG cells compared to 99mTc-PHH. In biodistribution studies, the radiocomplexes showed good brain uptake at 15 min combined with good radioactivity retention in the brain for 240 min. Regional rabbit brain studies indicated a higher radioactivity concentration in the hippocampus and diencephalon than in the cerebellum. Compared to 99mTc-MPHH, the 99mTc-PHH exhibited a significantly increased tumor uptake at 15 and 60 min, but the rapid blood clearance of 99mTc-MPHH led to enhanced tumor-to-muscle ratios at 240 min. A significant reduction in tumor uptake 60 min after an injection of pimozide (5-HT7 receptor antagonist) confirms the tumor uptake was receptor-mediated specifically. The tumor-to-contralateral muscle tissue ratio of 99mTc-PHH and 99mTc-MPHH in nude mice with U-87 MG xenograft was measured (5.25 and 4.65) at 60 min as well as (6.25 and 6.76) at 240 min, respectively.
Collapse
Affiliation(s)
- Shahnaz Saednia
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sajjad Molavipordanjani
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mohammad Abedi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
30
|
Martens C, Debeir O, Decaestecker C, Metens T, Lebrun L, Leurquin-Sterk G, Trotta N, Goldman S, Van Simaeys G. Voxelwise Principal Component Analysis of Dynamic [S-Methyl- 11C]Methionine PET Data in Glioma Patients. Cancers (Basel) 2021; 13:cancers13102342. [PMID: 34066294 PMCID: PMC8152079 DOI: 10.3390/cancers13102342] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 05/09/2021] [Indexed: 01/08/2023] Open
Abstract
Recent works have demonstrated the added value of dynamic amino acid positron emission tomography (PET) for glioma grading and genotyping, biopsy targeting, and recurrence diagnosis. However, most of these studies are based on hand-crafted qualitative or semi-quantitative features extracted from the mean time activity curve within predefined volumes. Voxelwise dynamic PET data analysis could instead provide a better insight into intra-tumor heterogeneity of gliomas. In this work, we investigate the ability of principal component analysis (PCA) to extract relevant quantitative features from a large number of motion-corrected [S-methyl-11C]methionine ([11C]MET) PET frames. We first demonstrate the robustness of our methodology to noise by means of numerical simulations. We then build a PCA model from dynamic [11C]MET acquisitions of 20 glioma patients. In a distinct cohort of 13 glioma patients, we compare the parametric maps derived from our PCA model to these provided by the classical one-compartment pharmacokinetic model (1TCM). We show that our PCA model outperforms the 1TCM to distinguish characteristic dynamic uptake behaviors within the tumor while being less computationally expensive and not requiring arterial sampling. Such methodology could be valuable to assess the tumor aggressiveness locally with applications for treatment planning and response evaluation. This work further supports the added value of dynamic over static [11C]MET PET in gliomas.
Collapse
Affiliation(s)
- Corentin Martens
- Department of Nuclear Medicine, Hôpital Erasme, Université libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium; (G.L.-S.); (N.T.); (S.G.); (G.V.S.)
- Laboratory of Image Synthesis and Analysis (LISA), École Polytechnique de Bruxelles, Université libre de Bruxelles, Avenue Franklin Roosevelt 50, 1050 Brussels, Belgium; (O.D.); (C.D.); (T.M.)
- Correspondence:
| | - Olivier Debeir
- Laboratory of Image Synthesis and Analysis (LISA), École Polytechnique de Bruxelles, Université libre de Bruxelles, Avenue Franklin Roosevelt 50, 1050 Brussels, Belgium; (O.D.); (C.D.); (T.M.)
| | - Christine Decaestecker
- Laboratory of Image Synthesis and Analysis (LISA), École Polytechnique de Bruxelles, Université libre de Bruxelles, Avenue Franklin Roosevelt 50, 1050 Brussels, Belgium; (O.D.); (C.D.); (T.M.)
| | - Thierry Metens
- Laboratory of Image Synthesis and Analysis (LISA), École Polytechnique de Bruxelles, Université libre de Bruxelles, Avenue Franklin Roosevelt 50, 1050 Brussels, Belgium; (O.D.); (C.D.); (T.M.)
- Department of Radiology, Hôpital Erasme, Université libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium
| | - Laetitia Lebrun
- Department of Pathology, Hôpital Erasme, Université libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium;
| | - Gil Leurquin-Sterk
- Department of Nuclear Medicine, Hôpital Erasme, Université libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium; (G.L.-S.); (N.T.); (S.G.); (G.V.S.)
| | - Nicola Trotta
- Department of Nuclear Medicine, Hôpital Erasme, Université libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium; (G.L.-S.); (N.T.); (S.G.); (G.V.S.)
| | - Serge Goldman
- Department of Nuclear Medicine, Hôpital Erasme, Université libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium; (G.L.-S.); (N.T.); (S.G.); (G.V.S.)
| | - Gaetan Van Simaeys
- Department of Nuclear Medicine, Hôpital Erasme, Université libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium; (G.L.-S.); (N.T.); (S.G.); (G.V.S.)
| |
Collapse
|
31
|
Sadaghiani MS, Sheikhbahaei S, Rowe SP, Pomper MG, Solnes LB. Cellular and Molecular Imaging with SPECT and PET in Brain Tumors. Radiol Clin North Am 2021; 59:363-375. [PMID: 33926683 DOI: 10.1016/j.rcl.2021.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This review highlights the 2 major molecular imaging modalities that are used in clinics, namely single-photon emission computed tomography (SPECT) and positron emission tomography (PET), and their added value in management of patients with brain tumors. There are a variety of SPECT and PET radiotracers that can allow imaging of different molecular processes. Those radiotracers target specific molecular features of tumors, resulting in improved specificity of these agents. Potential applications include staging of brain tumors and evaluating post-therapeutic changes.
Collapse
Affiliation(s)
- Mohammad S Sadaghiani
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3150, Baltimore, MD 21287, USA
| | - Sara Sheikhbahaei
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3150, Baltimore, MD 21287, USA
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3150, Baltimore, MD 21287, USA
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3150, Baltimore, MD 21287, USA
| | - Lilja B Solnes
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3150, Baltimore, MD 21287, USA.
| |
Collapse
|
32
|
Unterrainer M, Ruf V, von Rohr K, Suchorska B, Mittlmeier LM, Beyer L, Brendel M, Wenter V, Kunz WG, Bartenstein P, Herms J, Niyazi M, Tonn JC, Albert NL. TERT-Promoter Mutational Status in Glioblastoma - Is There an Association With Amino Acid Uptake on Dynamic 18F-FET PET? Front Oncol 2021; 11:645316. [PMID: 33996563 PMCID: PMC8121001 DOI: 10.3389/fonc.2021.645316] [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: 12/22/2020] [Accepted: 03/26/2021] [Indexed: 12/19/2022] Open
Abstract
Objective The mutation of the ‘telomerase reverse transcriptase gene promoter’ (TERTp) has been identified as an important factor for individual prognostication and tumorigenesis and will be implemented in upcoming glioma classifications. Uptake characteristics on dynamic 18F-FET PET have been shown to serve as additional imaging biomarker for prognosis. However, data on the correlation of TERTp-mutational status and amino acid uptake on dynamic 18F-FET PET are missing. Therefore, we aimed to analyze whether static and dynamic 18F-FET PET parameters are associated with the TERTp-mutational status in de-novo IDH-wildtype glioblastoma and whether a TERTp-mutation can be predicted by dynamic 18F-FET PET. Methods Patients with de-novo IDH-wildtype glioblastoma, WHO grade IV, available TERTp-mutational status and dynamic 18F-FET PET scan prior to any therapy were included. Here, established clinical parameters maximal and mean tumor-to-background-ratios (TBRmax/TBRmean), the biological-tumor-volume (BTV) and minimal-time-to-peak (TTPmin) on dynamic PET were analyzed and correlated with the TERTp-mutational status. Results One hundred IDH-wildtype glioblastoma patients were evaluated; 85/100 of the analyzed tumors showed a TERTp-mutation (C228T or C250T), 15/100 were classified as TERTp-wildtype. None of the static PET parameters was associated with the TERTp-mutational status (median TBRmax 3.41 vs. 3.32 (p=0.362), TBRmean 2.09 vs. 2.02 (p=0.349) and BTV 26.1 vs. 22.4 ml (p=0.377)). Also, the dynamic PET parameter TTPmin did not differ in both groups (12.5 vs. 12.5 min, p=0.411). Within the TERTp-mutant subgroups (i.e., C228T (n=23) & C250T (n=62)), the median TBRmax (3.33 vs. 3.69, p=0.095), TBRmean (2.08 vs. 2.09, p=0.352), BTV (25.4 vs. 30.0 ml, p=0.130) and TTPmin (12.5 vs. 12.5 min, p=0.190) were comparable, too. Conclusion Uptake characteristics on dynamic 18F-FET PET are not associated with the TERTp-mutational status in glioblastoma However, as both, dynamic 18F-FET PET parameters as well as the TERTp-mutation status are well-known prognostic biomarkers, future studies should investigate the complementary and independent prognostic value of both factors in order to further stratify patients into risk groups.
Collapse
Affiliation(s)
- Marcus Unterrainer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.,Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Viktoria Ruf
- Department of Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Katharina von Rohr
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Bogdana Suchorska
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | | | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Vera Wenter
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jochen Herms
- Department of Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Maximilian Niyazi
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Jörg C Tonn
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Nathalie Lisa Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
33
|
Stegmayr C, Stoffels G, Filß C, Heinzel A, Lohmann P, Willuweit A, Ermert J, Coenen HH, Mottaghy FM, Galldiks N, Langen KJ. Current trends in the use of O-(2-[ 18F]fluoroethyl)-L-tyrosine ([ 18F]FET) in neurooncology. Nucl Med Biol 2021; 92:78-84. [PMID: 32113820 DOI: 10.1016/j.nucmedbio.2020.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/16/2020] [Indexed: 12/14/2022]
Abstract
The diagnostic potential of PET using the amino acid analogue O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) in brain tumor diagnostics has been proven in many studies during the last two decades and is still the subject of multiple studies every year. In addition to standard magnetic resonance imaging (MRI), positron emission tomography (PET) using [18F]FET provides important diagnostic data concerning brain tumor delineation, therapy planning, treatment monitoring, and improved differentiation between treatment-related changes and tumor recurrence. The pharmacokinetics, uptake mechanisms and metabolism have been well described in various preclinical studies. The accumulation of [18F]FET in most benign lesions and healthy brain tissue has been shown to be low, thus providing a high contrast between tumor tissue and benign tissue alterations. Based on logistic advantages of F-18 labelling and convincing clinical results, [18F]FET has widely replaced short lived amino acid tracers such as L-[11C]methyl-methionine ([11C]MET) in many centers across Western Europe. This review summarizes the basic knowledge on [18F]FET and its contribution to the care of patients with brain tumors. In particular, recent studies about specificity, possible pitfalls, and the utility of [18F]FET PET in tumor grading and prognostication regarding the revised WHO classification of brain tumors are addressed.
Collapse
Affiliation(s)
- Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Christian Filß
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany
| | - Alexander Heinzel
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany; Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Johannes Ermert
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Heinz H Coenen
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Felix M Mottaghy
- Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany; Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Germany; Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Duesseldorf, Germany; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Duesseldorf, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany; Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Germany; Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Duesseldorf, Germany.
| |
Collapse
|
34
|
Solnes LB, Jacobs AH, Coughlin JM, Du Y, Goel R, Hammoud DA, Pomper MG. Central Nervous System Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00088-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
35
|
Galldiks N, Langen KJ, Albert NL, Chamberlain M, Soffietti R, Kim MM, Law I, Le Rhun E, Chang S, Schwarting J, Combs SE, Preusser M, Forsyth P, Pope W, Weller M, Tonn JC. PET imaging in patients with brain metastasis-report of the RANO/PET group. Neuro Oncol 2020; 21:585-595. [PMID: 30615138 DOI: 10.1093/neuonc/noz003] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/11/2018] [Accepted: 01/03/2019] [Indexed: 12/23/2022] Open
Abstract
Brain metastases (BM) from extracranial cancer are associated with significant morbidity and mortality. Effective local treatment options are stereotactic radiotherapy, including radiosurgery or fractionated external beam radiotherapy, and surgical resection. The use of systemic treatment for intracranial disease control also is improving. BM diagnosis, treatment planning, and follow-up is most often based on contrast-enhanced magnetic resonance imaging (MRI). However, anatomic imaging modalities including standard MRI have limitations in accurately characterizing posttherapeutic reactive changes and treatment response. Molecular imaging techniques such as positron emission tomography (PET) characterize specific metabolic and cellular features of metastases, potentially providing clinically relevant information supplementing anatomic MRI. Here, the Response Assessment in Neuro-Oncology working group provides recommendations for the use of PET imaging in the clinical management of patients with BM based on evidence from studies validated by histology and/or clinical outcome.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, University Hospital Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine 3, 4, Research Center Juelich, Juelich, Germany.,Center of Integrated Oncology, Universities of Cologne and Bonn, Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine 3, 4, Research Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig Maximilians-University of Munich, Munich, Germany
| | - Marc Chamberlain
- Departments of Neurology and Neurological Surgery, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington, USA
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Denmark
| | - Emilie Le Rhun
- Department of Neurosurgery, University Hospital Lille, Lille, France
| | - Susan Chang
- Department of Neurosurgery, University of California, San Francisco, California, USA
| | - Julian Schwarting
- Department of Neurosurgery, Ludwig Maximilians-University of Munich, Munich, Germany.,German Cancer Consortium, Partner Site Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Technical University Munich, Munich, Germany
| | - Matthias Preusser
- Department of Medicine I and Comprehensive Cancer Centre CNS Tumours Unit, Medical University of Vienna, Vienna, Austria
| | - Peter Forsyth
- Moffitt Cancer Center, University of South Florida, Tampa, Florida, USA
| | - Whitney Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California , USA
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Jörg C Tonn
- Department of Neurosurgery, Ludwig Maximilians-University of Munich, Munich, Germany.,German Cancer Consortium, Partner Site Munich, Germany
| |
Collapse
|
36
|
Ponisio MR, McConathy JE, Dahiya SM, Miller-Thomas MM, Rich KM, Salter A, Wang Q, LaMontagne PJ, Guzmán Pérez-Carrillo GJ, Benzinger TLS. Dynamic 18F-FDOPA-PET/MRI for the preoperative evaluation of gliomas: correlation with stereotactic histopathology. Neurooncol Pract 2020; 7:656-667. [PMID: 33312679 DOI: 10.1093/nop/npaa044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background MRI alone has limited accuracy for delineating tumor margins and poorly predicts the aggressiveness of gliomas, especially when tumors do not enhance. This study evaluated simultaneous 3,4-dihydroxy-6-[18F]fluoro-L-phenylalanine (FDOPA)-PET/MRI to define tumor volumes compared to MRI alone more accurately, assessed its role in patient management, and correlated PET findings with histopathology. Methods Ten patients with known or suspected gliomas underwent standard of care surgical resection and/or stereotactic biopsy. FDOPA-PET/MRI was performed prior to surgery, allowing for precise co-registration of PET, MR, and biopsies. The biopsy sites were modeled as 5-mm spheres, and the local FDOPA uptake at each site was determined. Correlations were performed between measures of tumor histopathology, and static and dynamic PET values: standardized uptake values (SUVs), tumor to brain ratios, metabolic tumor volumes, and tracer kinetics at volumes of interest (VOIs) and biopsy sites. Results Tumor FDOPA-PET uptake was visualized in 8 patients. In 2 patients, tracer uptake was similar to normal brain reference with no histological findings of malignancy. Eight biopsy sites confirmed for glioma had FDOPA uptake without T1 contrast enhancement. The PET parameters were highly correlated only with the cell proliferation marker, Ki-67 (SUVmax: r = 0.985, P = .002). In this study, no statistically significant difference between high-grade and low-grade tumors was demonstrated. The dynamic PET analysis of VOIs and biopsy sites showed decreasing time-activity curves patterns. FDOPA-PET imaging directly influenced patient management. Conclusions Simultaneous FDOPA-PET/MRI allowed for more accurate visualization and delineation of gliomas, enabling more appropriate patient management and simplified validation of PET findings with histopathology.
Collapse
Affiliation(s)
- Maria R Ponisio
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Jonathan E McConathy
- Department of Radiology, Division of Molecular Imaging and Therapeutics, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Sonika M Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Michelle M Miller-Thomas
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Keith M Rich
- Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri
| | - Amber Salter
- Department of Biostatistics, Washington University School of Medicine, St Louis, Missouri
| | - Qing Wang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Pamela J LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| | | | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
37
|
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
|
38
|
Evangelista L, Cuppari L, Bellu L, Bertin D, Caccese M, Reccia P, Zagonel V, Lombardi G. Comparison Between 18F-Dopa and 18F-Fet PET/CT in Patients with Suspicious Recurrent High Grade Glioma: A Literature Review and Our Experience. Curr Radiopharm 2020; 12:220-228. [PMID: 30644351 DOI: 10.2174/1874471012666190115124536] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/13/2018] [Accepted: 12/19/2018] [Indexed: 12/16/2022]
Abstract
PURPOSES The aims of the present study were to: 1- critically assess the utility of L-3,4- dihydroxy-6-18Ffluoro-phenyl-alanine (18F-DOPA) and O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET) Positron Emission Tomography (PET)/Computed Tomography (CT) in patients with high grade glioma (HGG) and 2- describe the results of 18F-DOPA and 18F-FET PET/CT in a case series of patients with recurrent HGG. METHODS We searched for studies using the following databases: PubMed, Web of Science and Scopus. The search terms were: glioma OR brain neoplasm and DOPA OR DOPA PET OR DOPA PET/CT and FET OR FET PET OR FET PET/CT. From a mono-institutional database, we retrospectively analyzed the 18F-DOPA and 18F-FET PET/CT of 29 patients (age: 56 ± 12 years) with suspicious for recurrent HGG. All patients underwent 18F-DOPA or 18F-FET PET/CT for a multidisciplinary decision. The final definition of recurrence was made by magnetic resonance imaging (MRI) and/or multidisciplinary decision, mainly based on the clinical data. RESULTS Fifty-one articles were found, of which 49 were discarded, therefore 2 studies were finally selected. In both the studies, 18F-DOPA and 18F-FET as exchangeable in clinical practice particularly for HGG patients. From our institutional experience, in 29 patients, we found that sensitivity, specificity and accuracy of 18F-DOPA PET/CT in HGG were 100% (95% confidence interval- 95%CI - 81-100%), 63% (95%CI: 39-82%) and 62% (95%CI: 39-81%), respectively. 18F-FET PET/CT was true positive in 4 and true negative in 4 patients. Sensitivity, specificity and accuracy for 18F-FET PET/CT in HGG were 100%. CONCLUSION 18F-DOPA and 18F-FET PET/CT have a similar diagnostic accuracy in patients with recurrent HGG. However, 18F-DOPA PET/CT could be affected by inflammation conditions (false positive) that can alter the final results. Large comparative trials are warranted in order to better understand the utility of 18F-DOPA or 18F-FET PET/CT in patients with HGG.
Collapse
Affiliation(s)
- Laura Evangelista
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Lea Cuppari
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Luisa Bellu
- Radiation Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Daniele Bertin
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Mario Caccese
- Oncology 1 Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Pasquale Reccia
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Vittorina Zagonel
- Oncology 1 Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Giuseppe Lombardi
- Oncology 1 Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| |
Collapse
|
39
|
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
|
40
|
Dissaux G, Basse V, Schick U, EL Kabbaj O, Auberger B, Magro E, Kassoul A, Abgral R, Salaun PY, Bourhis D, Querellou S. Prognostic value of 18F-FET PET/CT in newly diagnosed WHO 2016 high-grade glioma. Medicine (Baltimore) 2020; 99:e19017. [PMID: 32000446 PMCID: PMC7004648 DOI: 10.1097/md.0000000000019017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
O-(2-[F]fluoroethyl)-L-tyrosine positron-emission tomography/computed tomography (F-FET PET/CT) is well known in brain tumor management. Our study aimed to identify the prognostic value of F-FET PET/CT in high-grade gliomas (HGG) according the current 2016 World Health Organization (WHO) classification.Patients with histologically proven WHO 2016 HGG were prospectively included. A dynamic F-FET PET/CT was performed allowing to obtain 2 static PET frames (static frame 1: 20-40 minutes and static frame 2: 2-22 minutes). We analyzed static parameters (standard uptake value [SUV]max, SUVmean, SUVpeak, TBRmax, TBRmean, tumoral lesion glycolysis, and metabolic tumoral volume) for various isocontours (from 10% to 90%). PET parameters, clinical features, and molecular biomarkers were compared with progression-free survival (PFS) and overall survival (OS) in univariate and multivariate analysis.Twenty-nine patients were included (grade III n = 3, grade IV n = 26). Mean PFS and OS were, respectively, 8.8 and 13.9 months. According to univariate analysis, SUVmean, SUVpeak, TBRmax, and TBRmean were significantly correlated with OS. In static 1 analysis, TBRmax seemed to be the best OS prognostic parameter (P = .004). In static 2 analysis, TBRmean was the best parameter (P = .01). In static 1 analysis, only SUVpeak was significant (P = .05) for PFS. Good performance status (PS < 2; P < .0001) and extent of resection (P = .019) identified the subgroup of patients with the best OS. Only TBRmax (P = .026) and extent of resection (P = .025) remained significant parameters in multivariate analysis.Our data suggested that high TBRmax seemed to be the most significant OS independent prognostic factor in patients with newly diagnosed HGG.
Collapse
Affiliation(s)
| | - Victor Basse
- Oncology Department, University Hospital Morvan, Brest Cedex
| | | | | | | | - Elsa Magro
- Neurosurgery Department University Hospital Cavale Blanche
| | - Aboubakr Kassoul
- Nuclear Medicine Department, University Hospitam Morvan, Brest cedex
| | - Ronan Abgral
- Nuclear Medicine Department, University Hospitam Morvan, Brest cedex
- EA 3878 GETBO IFR 148
- University of Bretagne Occidentale, Brest, France
| | - Pierre-Yves Salaun
- Nuclear Medicine Department, University Hospitam Morvan, Brest cedex
- EA 3878 GETBO IFR 148
- University of Bretagne Occidentale, Brest, France
| | - David Bourhis
- Nuclear Medicine Department, University Hospitam Morvan, Brest cedex
- EA 3878 GETBO IFR 148
- University of Bretagne Occidentale, Brest, France
| | - Solène Querellou
- Nuclear Medicine Department, University Hospitam Morvan, Brest cedex
- EA 3878 GETBO IFR 148
- University of Bretagne Occidentale, Brest, France
| |
Collapse
|
41
|
Ginet M, Zaragori T, Marie PY, Roch V, Gauchotte G, Rech F, Blonski M, Lamiral Z, Taillandier L, Imbert L, Verger A. Integration of dynamic parameters in the analysis of 18F-FDopa PET imaging improves the prediction of molecular features of gliomas. Eur J Nucl Med Mol Imaging 2019; 47:1381-1390. [PMID: 31529264 DOI: 10.1007/s00259-019-04509-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/23/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE 18F-FDopa PET imaging of gliomas is routinely interpreted with standardized uptake value (SUV)-derived indices. This study aimed to determine the added value of dynamic 18F-FDopa PET parameters for predicting the molecular features of newly diagnosed gliomas. METHODS We retrospectively included 58 patients having undergone an 18F-FDopa PET for establishing the initial diagnosis of gliomas, whose molecular features were additionally characterized according to the WHO 2016 classification. Dynamic parameters, involving time-to-peak (TTP) values and curve slopes, were tested for the prediction of glioma types in addition to current static parameters, i.e., tumor-to-normal brain or tumor-to-striatum SUV ratios and metabolic tumor volume (MTV). RESULTS There were 21 IDH mutant without 1p/19q co-deletion (IDH+/1p19q-) gliomas, 16 IDH mutants with 1p/19q co-deletion (IDH+/1p19q+) gliomas, and 21 IDH wildtype (IDH-) gliomas. Dynamic parameters enabled differentiating the gliomas according to these molecular features, whereas static parameters did not. In particular, a longer TTP was the single best independent predictor for identifying (1) IDH mutation status (area under the curve (AUC) of 0.789, global accuracy of 74% for the criterion of a TTP ≥ 5.4 min) and (2) 1p/19q co-deletion status (AUC of 0.679, global accuracy of 69% for the criterion of a TTP ≥ 6.9 min). Moreover, the TTP from IDH- gliomas was significantly shorter than those from both IDH+/1p19q- and IDH+/1p19q+ (p ≤ 0.007). CONCLUSION Prediction of the molecular features of newly diagnosed gliomas with 18F-FDopa PET and especially of the presence or not of an IDH mutation, may be obtained with dynamic but not with current static uptake parameters.
Collapse
Affiliation(s)
- Merwan Ginet
- CHRU-Nancy, Department of Nuclear Medicine & Nancyclotep Imaging platform, Université de Lorraine, F-54000, Nancy, France
| | - Timothée Zaragori
- CHRU-Nancy, Department of Nuclear Medicine & Nancyclotep Imaging platform, Université de Lorraine, F-54000, Nancy, France
- IADI, INSERM, UMR 1254, Université de Lorraine, F-54000, Nancy, France
| | - Pierre-Yves Marie
- CHRU-Nancy, Department of Nuclear Medicine & Nancyclotep Imaging platform, Université de Lorraine, F-54000, Nancy, France
- Université de Lorraine, INSERM U1116, F-54000, Nancy, France
| | - Véronique Roch
- CHRU-Nancy, Department of Nuclear Medicine & Nancyclotep Imaging platform, Université de Lorraine, F-54000, Nancy, France
| | - Guillaume Gauchotte
- CHRU-Nancy, Department of Pathology, Université de Lorraine, F-54000, Nancy, France
- INSERM U1256, Université de Lorraine, F-54000, Nancy, France
| | - Fabien Rech
- Department of Neurosurgery, CHU-Nancy, F-54000, Nancy, France
- Centre de Recherche en Automatique de Nancy CRAN, CNRS UMR 7039, Université de Lorraine, F-54000, Nancy, France
| | - Marie Blonski
- Department of Neurosurgery, CHU-Nancy, F-54000, Nancy, France
- Centre de Recherche en Automatique de Nancy CRAN, CNRS UMR 7039, Université de Lorraine, F-54000, Nancy, France
| | - Zohra Lamiral
- Université de Lorraine, INSERM U1116, F-54000, Nancy, France
| | - Luc Taillandier
- Centre de Recherche en Automatique de Nancy CRAN, CNRS UMR 7039, Université de Lorraine, F-54000, Nancy, France
- CHRU-Nancy, Department of Neuro-oncology, Université de Lorraine, F-54000, Nancy, France
| | - Laëtitia Imbert
- CHRU-Nancy, Department of Nuclear Medicine & Nancyclotep Imaging platform, Université de Lorraine, F-54000, Nancy, France
- IADI, INSERM, UMR 1254, Université de Lorraine, F-54000, Nancy, France
| | - Antoine Verger
- CHRU-Nancy, Department of Nuclear Medicine & Nancyclotep Imaging platform, Université de Lorraine, F-54000, Nancy, France.
- IADI, INSERM, UMR 1254, Université de Lorraine, F-54000, Nancy, France.
| |
Collapse
|
42
|
Abstract
PURPOSE This pilot study aimed to evaluate the amino acid tracer F-FACBC with simultaneous PET/MRI in diagnostic assessment and neurosurgery of gliomas. MATERIALS AND METHODS Eleven patients with suspected primary or recurrent low- or high-grade glioma received an F-FACBC PET/MRI examination before surgery. PET and MRI were used for diagnostic assessment, and for guiding tumor resection and histopathological tissue sampling. PET uptake, tumor-to-background ratios (TBRs), time-activity curves, as well as PET and MRI tumor volumes were evaluated. The sensitivities of lesion detection and to detect glioma tissue were calculated for PET, MRI, and combined PET/MRI with histopathology (biopsies for final diagnosis and additional image-localized biopsies) as reference. RESULTS Overall sensitivity for lesion detection was 54.5% (95% confidence interval [CI], 23.4-83.3) for PET, 45.5% (95% CI, 16.7-76.6) for contrast-enhanced MRI (MRICE), and 100% (95% CI, 71.5-100.0) for combined PET/MRI, with a significant difference between MRICE and combined PET/MRI (P = 0.031). TBRs increased with tumor grade (P = 0.004) and were stable from 10 minutes post injection. PET tumor volumes enclosed most of the MRICE volumes (>98%) and were generally larger (1.5-2.8 times) than the MRICE volumes. Based on image-localized biopsies, combined PET/MRI demonstrated higher concurrence with malignant findings at histopathology (89.5%) than MRICE (26.3%). CONCLUSIONS Low- versus high-grade glioma differentiation may be possible with F-FACBC using TBR. F-FACBC PET/MRI outperformed MRICE in lesion detection and in detection of glioma tissue. More research is required to evaluate F-FACBC properties, especially in grade II and III tumors, and for different subtypes of gliomas.
Collapse
|
43
|
Non-invasive prediction of IDH-wildtype genotype in gliomas using dynamic 18F-FET PET. Eur J Nucl Med Mol Imaging 2019; 46:2581-2589. [PMID: 31410540 DOI: 10.1007/s00259-019-04477-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/02/2019] [Indexed: 01/01/2023]
Abstract
PURPOSE According to the updated WHO classification of gliomas with its emphasis on molecular parameters, tumours with an IDH-wildtype status have a dismal prognosis. To ensure timely adjustment of treatment, demand for non-invasive prediction methods is high. 18F-FET PET has been shown to be an important diagnostic tool for glioma management. The aim of this study was to assess the value of dynamic 18F-FET PET for the non-invasive prediction of the IDH-mutation status. METHODS Newly diagnosed WHO grade II-IV glioma patients with MRI and dynamic 18F-FET PET were included. The 18F-FET PET parameters mean and maximal tumour-to-background ratio (TBRmean, TBRmax) and minimal time-to-peak (TTPmin) were evaluated. The diagnostic power for the prediction of the IDH genotype (positive/negative predictive value) was tested in the overall study group and in the subgroup of non-contrast enhancing gliomas. RESULTS Three hundred forty-one patients were evaluated. Molecular analyses revealed 178 IDH-mutant and 163 IDH-wildtype tumours. Overall, 270/341 gliomas were classified as 18F-FET-positive (TBRmax > 1.6), 90.2% of the IDH-wildtype and 69.1% of IDH-mutant gliomas. Median TBRmax was significantly higher in IDH-wildtype compared with IDH-mutant gliomas (2.9 vs. 2.3, p < 0.001); however, ROC-analyses revealed no reliable cutoff due to a high overlap (range 1.0-7.1 vs. 1.1-7.9). Dynamic analysis revealed a significantly shorter TTPmin in IDH-wildtype gliomas; using TTPmin ≤ 12.5 min as indicator for IDH-wildtype gliomas, a positive predictive value of 87% was reached (negative predictive value 72%, AUC = 0.796, p ≤ 0.001). A total of 161/341 gliomas did not show contrast enhancement on MRI; even within this subgroup, TTPmin ≤ 12.5 min remained a good predictor of IDH-wildtype glioma (positive predictive value 83%, negative predictive value 90%; AUC = 0.868, p < 0.001). CONCLUSION A short TTPmin in dynamic 18F-FET PET serves as good predictor of highly aggressive IDH-wildtype status in gliomas. In particular, a high diagnostic power was observed in the subgroup of non-contrast enhancing gliomas, which helps to identify patients with worse prognosis.
Collapse
|
44
|
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: 14] [Impact Index Per Article: 2.3] [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
|
45
|
|
46
|
Clinical Utility of Different Approaches for Detection of Late Pseudoprogression in Glioblastoma With O-(2-[18F]Fluoroethyl)-l-Tyrosine PET. Clin Nucl Med 2019; 44:695-701. [DOI: 10.1097/rlu.0000000000002652] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
Yang Y, He MZ, Li T, Yang X. MRI combined with PET-CT of different tracers to improve the accuracy of glioma diagnosis: a systematic review and meta-analysis. Neurosurg Rev 2019; 42:185-195. [PMID: 28918564 PMCID: PMC6503074 DOI: 10.1007/s10143-017-0906-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 09/01/2017] [Accepted: 09/06/2017] [Indexed: 12/18/2022]
Abstract
Based on studies focusing on positron emission tomography (PET)-computed tomography (CT) combined with magnetic resonance imaging (MRI) in the diagnosis of glioma, we conducted a systematic review and meta-analysis evaluating the pros and cons and the accuracy of different examinations. PubMed and Cochrane Library were searched. The search was conducted until April 2017. Two reviewers independently conducted the literature search according to the criteria set initially. Based on the exclusion criteria, 15 articles are included in this study. Of all studies that used MRI examination, there are five involving 18F-fluorodeoxyglucose-PET, five involving 11C-methionine-PET, five involving 18F-fluoro-ethyl-tyrosine-PET, and three involving 18F-fluorothymidine-PET. Due to the limitations such as lack of data, small sample size, and unrepresentative studies, we use a non-quantitative methodology. MRI examination can provide the anatomy information of glioma more clearly. PET-CT examinations based on tumor metabolism using different tracers have more advantages in determining the degree of glioma malignancy and boundaries. However, information provided by PET-CT of different tracers is not the same. With respect to the novel hybrid MRI/PET examination equipment proposed in recent years, the combination of MRI and PET-CT can definitively improve the diagnostic accuracy of glioma.
Collapse
Affiliation(s)
- Yihan Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Mike Z He
- Columbia University Mailman School of Public Health, New York, NY, USA
| | - Tao Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
48
|
|
49
|
Zhang Q, Gao X, Wei G, Qiu C, Qu H, Zhou X. Prognostic Value of MTV, SUVmax and the T/N Ratio of PET/CT in Patients with Glioma: A Systematic Review and Meta-Analysis. J Cancer 2019; 10:1707-1716. [PMID: 31205526 PMCID: PMC6548003 DOI: 10.7150/jca.28605] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/13/2019] [Indexed: 12/11/2022] Open
Abstract
Background: In the past decade, positron emission tomography/computed tomography (PET/CT) has become an important imaging tool for clinical assessment of tumor patients. Our meta-analysis aimed to compare the predictive value of PET/CT parameters regard to overall survival (OS) and progression-free survival (PFS) outcomes in glioma. Methods: Relevant articles were systematically searched in PMC, PubMed, EMBASE and WEB of science. Studies involving the prognostic roles of PET/CT parameters with OS and PFS in glioma patients were evaluated. The impact of metabolic tumor volume (MTV), maximal standard uptake value (SUVmax), and the ratio of uptake in tumor to normal (T/N ratio) on survival was measured by calculating combined hazard ratios (HRs) and 95% confidence intervals (CIs). Results: A total of 32 articles with 1715 patients were included. The combined HRs of higher MTV, higher SUVmax and higher T/N ratio for OS were 1.14 (95% CI: 0.98-1.32, P heterogeneity<0.001), 1.69 (95% CI: 1.18-2.41, P heterogeneity<0.001) and 1.68 (95% CI: 1.40-2.01, P heterogeneity< 0.001), respectively. Regarding PFS, the combined HRs were 1.04 (95% CI: 0.97-1.11, P heterogeneity=0.002) with higher MTV, 1.45 (95% CI: 1.11-1.90, P heterogeneity<0.001) with higher SUVmax and 2.07 (95% CI: 1.45-2.95, P heterogeneity<0.001) with higher T/N ratio. Results remained similar in the sub-group analyses. Conclusion: PET/CT parameters T/N ratio may be a significant prognostic factor in patients with glioma. Evidence of SUVmax and MTV needed more large-scale studies performed to validate. PET/CT scan could be a promising technique to provide prognostic information for these patients.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Neurosurgery, Xinghua People's Hospital, Xinghua 225700, Jiangsu, P.R. China
| | - Xian Gao
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Guohua Wei
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Cheng Qiu
- Department of Neurosurgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, P.R. China
| | - Hongyi Qu
- Department of Neurosurgery, Xinghua People's Hospital, Xinghua 225700, Jiangsu, P.R. China
| | - Xin Zhou
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
50
|
Harat M, Małkowski B, Roszkowski K. Prognostic value of subventricular zone involvement in relation to tumor volumes defined by fused MRI and O-(2-[ 18F]fluoroethyl)-L-tyrosine (FET) PET imaging in glioblastoma multiforme. Radiat Oncol 2019; 14:37. [PMID: 30832691 PMCID: PMC6398237 DOI: 10.1186/s13014-019-1241-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/21/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Subventricular zone (SVZ) involvement is associated with a dismal prognosis in patients with glioblastoma multiforme (GBM). Dual-time point (dtp) O-(2-[18F]fluoroethyl)-L-tyrosine (FET) PET/CT (PET) may be a time- and cost-effective alternative to dynamic FET PET, but its prognostic value, particularly with respect to SVZ involvement, is unknown. METHODS Thirty-five patients had two scans 5-15 and 50-60 min after i.v. FET injection to define tumor volumes and SVZ involvement before starting radiotherapy. Associations between clinical progression markers, MRI- and dtp FET PET-based tumor volumes, or SVZ involvement and progression-free (PFS) and overall survival (OS) were assessed in univariable and multivariable analyses. RESULTS The extent of resection was not related to outcomes. Albeit non-significant, dtp FET PET detected more SVZ infiltration than MRI (60% vs. 51%, p = 0.25) and was significantly associated with poor survival (p < 0.03), but PET-T1-Gad volumes were larger in this group (p < 0.002). Survival was shorter in patients with larger MRI tumor volumes, larger PET tumor volumes, and worse Karnofsky performance status (KPS), with fused PET-T1-Gad and KPS significant in multivariable analysis (p < 0.03). Uptake kinetics was not associated with treatment outcomes. CONCLUSIONS FET PET-based tumor volumes may be useful for predicting worse prognosis glioblastoma. Although the presence of SVZ infiltration is linked to higher PET/MRI-based tumor volumes, the independent value of dtp FET PET parameters and SVZ infiltration as prognostic markers pre-irradiation has not been confirmed.
Collapse
Affiliation(s)
- Maciej Harat
- Department of Oncology and Brachytherapy, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Romanowskiej 2 St, ,85-796, Bydgoszcz, Poland. .,Department of Radiotherapy, Unit of Radiosurgery and Radiotherapy of CNS, Franciszek Lukaszczyk Oncology Center, Bydgoszcz, Poland.
| | - Bogdan Małkowski
- Department of Positron Emission Tomography and Molecular Imaging, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Krzysztof Roszkowski
- Department of Oncology, Radiotherapy and Gynecologic Oncology, Faculty of Health Sciences, Nicolaus Copernicus University Toruń, Bydgoszcz, Poland
| |
Collapse
|