1
|
Li Y, Wang R, Chen J, Zhu Z, Wang Y, Ma W. 68Ga-NOTA-RM26 PET/CT in the evaluation of glioma: a pilot prospective study. EJNMMI Res 2025; 15:6. [PMID: 39821814 PMCID: PMC11748694 DOI: 10.1186/s13550-025-01198-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Gliomas are the most common malignant primary tumors of the central nervous system. There is an urgent need for new convenient, targeted and specific imaging agents for gliomas. This study aimed to firstly evaluate the feasibility of 68Ga-NOTA-RM26 PET/CT imaging in glioma and analyze the relationship between the imaging characteristics and glioma grade, classification and molecular alterations. RESULTS Twenty-two patients were confirmed as glioma by surgery or biopsy. All patients exhibited 68Ga-NOTA-RM26 uptake. SUVmax was chosen as the imaging marker for analysis. For all glioma patients, there were significant differences between grades (P = 0.047). For primary gliomas, SUVmax had good discrimination for both tumor classifications (P = 0.045) and grades (P = 0.03). There was a positive correlation (P < 0.01) between GRPR expression level and SUVmax. P53 mutations caused significant differences in SUVmax (P = 0.03). CONCLUSIONS This study is the first application of 68Ga-NOTA-RM26 in glioma patients and confirmed the safety and efficacy in glioma patients. 68Ga-NOTA-RM26 PET/CT has potential value in tumor grade, classification, and molecular alterations. TRIAL REGISTRATION ClinicalTrials.gov: NCT06412952. Registered 26 April 2024, https://clinicaltrials.gov/study/NCT06412952.
Collapse
Affiliation(s)
- Yilin Li
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Rongxi Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jingci Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhaohui Zhu
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yu Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Wenbin Ma
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
2
|
Dev ID, Puranik AD, Rangarajan V, Patra S, Purandare N, Sahu A, Choudhary A, Bhattacharya K, Gupta T, Chatterjee A, Dasgupta A, Moiyadi A, Shetty P, Singh V, Sridhar E, Sahay A, Shah A, Ghosh S, Choudhury S, Shah S, Agrawal A. Preliminary evaluation of FAPI-04-PET/CT for differentiating recurrence and post-treatment changes in high-grade gliomas. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1289-1296. [PMID: 39759219 PMCID: PMC11700622 DOI: 10.37349/etat.2024.00276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/30/2024] [Indexed: 01/07/2025] Open
Abstract
Fibroblast-activated protein (FAP) expression in glial cells is attributed to FAP-positive foci on tumor vessels and neoplastic cells. Preclinical and pilot studies have shown FAP expression in high-grade gliomas. We aimed at comparing PET imaging with FAP-inhibitor (FAPI-PET) with current standard, i.e., fluoro-ethyl tyrosine (FET) PET in post-treatment setting to differentiate recurrence and post-treatment changes. 6 patients with WHO Grade III and IV glioma who received standard treatment underwent Ga-68-FAPI-04 PET/CT (FAPI-PET/CT). Tracer uptake greater than background was considered positive. FET PET was performed and interpreted as per institutional standards, which formed the basis of treatment decision. There was concordance between FAPI expression and FET uptake in 5 patients suggestive of disease recurrence. There was no FAPI expression seen in 1 patient, in whom FET PET was suggestive of post-treatment changes. FAPI PET uptake correlated with amino acid expression to differentiate post treatment changes from recurrence in high-grade glial tumors; further validation with prospective study and histopathological confirmation is needed.
Collapse
Affiliation(s)
- Indraja D. Dev
- Depts of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Ameya D. Puranik
- Depts of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Venkatesh Rangarajan
- Depts of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Sukriti Patra
- Depts of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Nilendu Purandare
- Depts of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Arpita Sahu
- Radiodiagnosis, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Amitkumar Choudhary
- Radiodiagnosis, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Kajari Bhattacharya
- Radiodiagnosis, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Tejpal Gupta
- Radiation Oncology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Abhishek Chatterjee
- Radiation Oncology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Archya Dasgupta
- Radiation Oncology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Aliasgar Moiyadi
- Neurosurgery, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Prakash Shetty
- Neurosurgery, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Vikas Singh
- Neurosurgery, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Epari Sridhar
- Pathology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Ayushi Sahay
- Pathology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Aekta Shah
- Pathology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Suchismita Ghosh
- Depts of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Sayak Choudhury
- Depts of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Sneha Shah
- Depts of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| | - Archi Agrawal
- Depts of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Dr E Borges Marg, Parel, Mumbai 400012, India
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Zhou M, Yin X, Chen B, Hu S, Zhou W. A PET probe targeting polyamine transport system for precise tumor diagnosis and therapy. Asian J Pharm Sci 2024; 19:100924. [PMID: 38903130 PMCID: PMC11186966 DOI: 10.1016/j.ajps.2024.100924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/02/2024] [Accepted: 03/04/2024] [Indexed: 06/22/2024] Open
Abstract
Polyamine metabolism dysregulation is a hallmark of many cancers, offering a promising avenue for early tumor theranostics. This study presents the development of a nuclear probe derived from spermidine (SPM) for dual-purpose tumor PET imaging and internal radiation therapy. The probe, radiolabeled with either [68Ga]Ga for diagnostic applications or [177Lu]Lu for therapeutic use, was synthesized with exceptional purity, stability, and specific activity. Extensive testing involving 12 different tumor cell lines revealed remarkable specificity towards B16 melanoma cells, showcasing outstanding tumor localization and target-to-non-target ratio. Mechanistic investigations employing polyamines, non-labeled precursor, and polyamine transport system (PTS) inhibitor, consistently affirmed the probe's targetability through recognition of the PTS. Notably, while previous reports indicated PTS upregulation in various tumor types for targeted therapy, this study observed no positive signals, highlighting a concentration-dependent discrepancy between targeting for therapy and diagnosis. Furthermore, when labeled with [177Lu], the probe demonstrated its therapeutic potential by effectively controlling tumor growth and extending mouse survival. Investigations into biodistribution, excretion, and biosafety in healthy humans laid a robust foundation for clinical translation. This study introduces a versatile SPM-based nuclear probe with applications in precise tumor theranostics, offering promising prospects for clinical implementation.
Collapse
Affiliation(s)
- Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Xiaoqin Yin
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Bei Chen
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha 410008, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha 410008, China
| |
Collapse
|
6
|
Lai TH, Wenzel B, Dukić-Stefanović S, Teodoro R, Arnaud L, Maisonial-Besset A, Weber V, Moldovan RP, Meister S, Pietzsch J, Kopka K, Juratli TA, Deuther-Conrad W, Toussaint M. Radiosynthesis and biological evaluation of [ 18F]AG-120 for PET imaging of the mutant isocitrate dehydrogenase 1 in glioma. Eur J Nucl Med Mol Imaging 2024; 51:1085-1096. [PMID: 37982850 PMCID: PMC10881675 DOI: 10.1007/s00259-023-06515-7] [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: 07/28/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023]
Abstract
Glioma are clinically challenging tumors due to their location and invasiveness nature, which often hinder complete surgical resection. The evaluation of the isocitrate dehydrogenase mutation status has become crucial for effective patient stratification. Through a transdisciplinary approach, we have developed an 18F-labeled ligand for non-invasive assessment of the IDH1R132H variant by using positron emission tomography (PET) imaging. In this study, we have successfully prepared diastereomerically pure [18F]AG-120 by copper-mediated radiofluorination of the stannyl precursor 6 on a TRACERlab FX2 N radiosynthesis module. In vitro internalization studies demonstrated significantly higher uptake of [18F]AG-120 in U251 human high-grade glioma cells with stable overexpression of mutant IDH1 (IDH1R132H) compared to their wild-type IDH1 counterpart (0.4 vs. 0.013% applied dose/µg protein at 120 min). In vivo studies conducted in mice, exhibited the excellent metabolic stability of [18F]AG-120, with parent fractions of 85% and 91% in plasma and brain at 30 min p.i., respectively. Dynamic PET studies with [18F]AG-120 in naïve mice and orthotopic glioma rat model reveal limited blood-brain barrier permeation along with a low uptake in the brain tumor. Interestingly, there was no significant difference in uptake between mutant IDH1R132H and wild-type IDH1 tumors (tumor-to-blood ratio[40-60 min]: ~1.7 vs. ~1.3). In conclusion, our preclinical evaluation demonstrated a target-specific internalization of [18F]AG-120 in vitro, a high metabolic stability in vivo in mice, and a slightly higher accumulation of activity in IDH1R132H-glioma compared to IDH1-glioma. Overall, our findings contribute to advancing the field of molecular imaging and encourage the evaluation of [18F]AG-120 to improve diagnosis and management of glioma and other IDH1R132H-related tumors.
Collapse
Affiliation(s)
- Thu Hang Lai
- Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Helmholtz-Zentrum Dresden-Rossendorf, Research site Leipzig, Leipzig, Germany
- Department of Research and Development, ROTOP Pharmaka GmbH, Dresden, Germany
| | - Barbara Wenzel
- Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Helmholtz-Zentrum Dresden-Rossendorf, Research site Leipzig, Leipzig, Germany
| | - Sladjana Dukić-Stefanović
- Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Helmholtz-Zentrum Dresden-Rossendorf, Research site Leipzig, Leipzig, Germany
| | - Rodrigo Teodoro
- Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Helmholtz-Zentrum Dresden-Rossendorf, Research site Leipzig, Leipzig, Germany
| | - Lucie Arnaud
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, Inserm, Clermont- Ferrand, France
| | - Aurélie Maisonial-Besset
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, Inserm, Clermont- Ferrand, France
| | - Valérie Weber
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, Inserm, Clermont- Ferrand, France
| | - Rareş-Petru Moldovan
- Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Helmholtz-Zentrum Dresden-Rossendorf, Research site Leipzig, Leipzig, Germany
| | - Sebastian Meister
- Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Helmholtz-Zentrum Dresden-Rossendorf, Research site Leipzig, Leipzig, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Tareq A Juratli
- National Center for Tumor Diseases (NCT) Dresden, University Hospital Carl Gustav Carus, Dresden, Germany
- Department of Neurosurgery, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Winnie Deuther-Conrad
- Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Helmholtz-Zentrum Dresden-Rossendorf, Research site Leipzig, Leipzig, Germany
| | - Magali Toussaint
- Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Helmholtz-Zentrum Dresden-Rossendorf, Research site Leipzig, Leipzig, Germany.
| |
Collapse
|
7
|
Husby T, Johannessen K, Berntsen EM, Johansen H, Giskeødegård GF, Karlberg A, Fagerli UM, Eikenes L. 18F-FACBC and 18F-FDG PET/MRI in the evaluation of 3 patients with primary central nervous system lymphoma: a pilot study. EJNMMI REPORTS 2024; 8:2. [PMID: 38748286 PMCID: PMC10962628 DOI: 10.1186/s41824-024-00189-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/06/2023] [Indexed: 05/19/2024]
Abstract
BACKGROUND This PET/MRI study compared contrast-enhanced MRI, 18F-FACBC-, and 18F-FDG-PET in the detection of primary central nervous system lymphomas (PCNSL) in patients before and after high-dose methotrexate chemotherapy. Three immunocompetent PCNSL patients with diffuse large B-cell lymphoma received dynamic 18F-FACBC- and 18F-FDG-PET/MRI at baseline and response assessment. Lesion detection was defined by clinical evaluation of contrast enhanced T1 MRI (ce-MRI) and visual PET tracer uptake. SUVs and tumor-to-background ratios (TBRs) (for 18F-FACBC and 18F-FDG) and time-activity curves (for 18F-FACBC) were assessed. RESULTS At baseline, seven ce-MRI detected lesions were also detected with 18F-FACBC with high SUVs and TBRs (SUVmax:mean, 4.73, TBRmax: mean, 9.32, SUVpeak: mean, 3.21, TBRpeak:mean: 6.30). High TBR values of 18F-FACBC detected lesions were attributed to low SUVbackground. Baseline 18F-FDG detected six lesions with high SUVs (SUVmax: mean, 13.88). In response scans, two lesions were detected with ce-MRI, while only one was detected with 18F-FACBC. The lesion not detected with 18F-FACBC was a small atypical MRI detected lesion, which may indicate no residual disease, as this patient was still in complete remission 12 months after initial diagnosis. No lesions were detected with 18F-FDG in the response scans. CONCLUSIONS 18F-FACBC provided high tumor contrast, outperforming 18F-FDG in lesion detection at both baseline and in response assessment. 18F-FACBC may be a useful supplement to ce-MRI in PCNSL detection and response assessment, but further studies are required to validate these findings. Trial registration ClinicalTrials.gov. Registered 15th of June 2017 (Identifier: NCT03188354, https://clinicaltrials.gov/study/NCT03188354 ).
Collapse
Affiliation(s)
- Trine Husby
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postboks 8905, Trondheim, Norway
- Department of Oncology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Knut Johannessen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postboks 8905, Trondheim, Norway
| | - Erik Magnus Berntsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postboks 8905, Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Håkon Johansen
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Guro Fanneløb Giskeødegård
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna Karlberg
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postboks 8905, Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Unn-Merete Fagerli
- Department of Oncology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Live Eikenes
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postboks 8905, Trondheim, Norway.
| |
Collapse
|
8
|
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
|
9
|
von Rohr K, Unterrainer M, Holzgreve A, Kirchner MA, Li Z, Unterrainer LM, Suchorska B, Brendel M, Tonn JC, Bartenstein P, Ziegler S, Albert NL, Kaiser L. Can Radiomics Provide Additional Information in [18F]FET-Negative Gliomas? Cancers (Basel) 2022; 14:cancers14194860. [PMID: 36230783 PMCID: PMC9612387 DOI: 10.3390/cancers14194860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Amino acid positron emission tomography (PET) complements standard magnetic resonance imaging (MRI) since it directly visualizes the increased amino acid transport into tumor cells. Amino acid PET using O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) has proven to be relevant, for example, for glioma classification, identification of tumor progression or recurrence, or for the delineation of tumor extent. Nevertheless, a relevant proportion of low-grade gliomas (30%) and few high-grade gliomas (5%) were found to show no or even decreased amino acid uptake by conventional visual analysis of PET images. Advanced image analysis with the extraction of radiomic features is known to provide more detailed information on tumor characteristics than conventional analyses. Hence, this study aimed to investigate whether radiomic features derived from dynamic [18F]FET PET data differ between [18F]FET-negative glioma and healthy background and thus provide information that cannot be extracted by visual read. Abstract The purpose of this study was to evaluate the possibility of extracting relevant information from radiomic features even in apparently [18F]FET-negative gliomas. A total of 46 patients with a newly diagnosed, histologically verified glioma that was visually classified as [18F]FET-negative were included. Tumor volumes were defined using routine T2/FLAIR MRI data and applied to extract information from dynamic [18F]FET PET data, i.e., early and late tumor-to-background (TBR5–15, TBR20–40) and time-to-peak (TTP) images. Radiomic features of healthy background were calculated from the tumor volume of interest mirrored in the contralateral hemisphere. The ability to distinguish tumors from healthy tissue was assessed using the Wilcoxon test and logistic regression. A total of 5, 15, and 69% of features derived from TBR20–40, TBR5–15, and TTP images, respectively, were significantly different. A high number of significantly different TTP features was even found in isometabolic gliomas (after exclusion of photopenic gliomas) with visually normal [18F]FET uptake in static images. However, the differences did not reach satisfactory predictability for machine-learning-based identification of tumor tissue. In conclusion, radiomic features derived from dynamic [18F]FET PET data may extract additional information even in [18F]FET-negative gliomas, which should be investigated in larger cohorts and correlated with histological and outcome features in future studies.
Collapse
Affiliation(s)
- Katharina von Rohr
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Marcus Unterrainer
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | | | - Zhicong Li
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Lena M. Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | | | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Joerg-Christian Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
- Correspondence: (N.L.A.); (L.K.)
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
- Correspondence: (N.L.A.); (L.K.)
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Cicone F, Galldiks N, Papa A, Langen KJ, Cascini GL, Minniti G. Repeated amino acid PET imaging for longitudinal monitoring of brain tumors. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00504-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
13
|
Cheng Y, Song S, Wei Y, Xu G, An Y, Ma J, Yang H, Qi Z, Xiao X, Bai J, Xu L, Hu Z, Sun T, Wang L, Lu J, Lin Q. Glioma Imaging by O-(2-18F-Fluoroethyl)-L-Tyrosine PET and Diffusion-Weighted MRI and Correlation With Molecular Phenotypes, Validated by PET/MR-Guided Biopsies. Front Oncol 2021; 11:743655. [PMID: 34912706 PMCID: PMC8666958 DOI: 10.3389/fonc.2021.743655] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/11/2021] [Indexed: 12/05/2022] Open
Abstract
Gliomas exhibit high intra-tumoral histological and molecular heterogeneity. Introducing stereotactic biopsy, we achieved a superior molecular analysis of glioma using O-(2-18F-fluoroethyl)-L-tyrosine (FET)-positron emission tomography (PET) and diffusion-weighted magnetic resonance imaging (DWI). Patients underwent simultaneous DWI and FET-PET scans. Correlations between biopsy-derived tumor tissue values, such as the tumor-to-background ratio (TBR) and apparent diffusion coefficient (ADC)/exponential ADC (eADC) and histopathological diagnoses and those between relevant genes and TBR and ADC values were determined. Tumor regions with human telomerase reverse transcriptase (hTERT) mutation had higher TBR and lower ADC values. Tumor protein P53 mutation correlated with lower TBR and higher ADC values. α-thalassemia/mental-retardation-syndrome-X-linked gene (ATRX) correlated with higher ADC values. 1p/19q codeletion and epidermal growth factor receptor (EGFR) mutations correlated with lower ADC values. Isocitrate dehydrogenase 1 (IDH1) mutations correlated with higher TBRmean values. No correlation existed between TBRmax/TBRmean/ADC/eADC values and phosphatase and tensin homolog mutations (PTEN) or O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation. Furthermore, TBR/ADC combination had a higher diagnostic accuracy than each single imaging method for high-grade and IDH1-, hTERT-, and EGFR-mutated gliomas. This is the first study establishing the accurate diagnostic criteria for glioma based on FET-PET and DWI.
Collapse
Affiliation(s)
- Ye Cheng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China.,Department of Neurosurgery, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shuangshuang Song
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China.,Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yukui Wei
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Geng Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Yang An
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Jie Ma
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Hongwei Yang
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Zhigang Qi
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xinru Xiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Jie Bai
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Lixin Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Zeliang Hu
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tingting Sun
- Department of Medicine, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Leiming Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Qingtang Lin
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
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: 5] [Impact Index Per Article: 1.3] [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
|
17
|
Johannessen K, Berntsen EM, Johansen H, Solheim TS, Karlberg A, Eikenes L. 18F-FACBC PET/MRI in the evaluation of human brain metastases: a case report. Eur J Hybrid Imaging 2021; 5:7. [PMID: 34181107 PMCID: PMC8218039 DOI: 10.1186/s41824-021-00101-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/28/2021] [Indexed: 12/31/2022] Open
Abstract
Background Patients with metastatic cancer to the brain have a poor prognosis. In clinical practice, MRI is used to delineate, diagnose and plan treatment of brain metastases. However, MRI alone is limited in detecting micro-metastases, delineating lesions and discriminating progression from pseudo-progression. Combined PET/MRI utilises superior soft tissue images from MRI and metabolic data from PET to evaluate tumour structure and function. The amino acid PET tracer 18F-FACBC has shown promising results in discriminating high- and low-grade gliomas, but there are currently no reports on its use on brain metastases. This is the first study to evaluate the use of 18F-FACBC on brain metastases. Case presentation A middle-aged female patient with brain metastases was evaluated using hybrid PET/MRI with 18F-FACBC before and after stereotactic radiotherapy, and at suspicion of recurrence. Static/dynamic PET and contrast-enhanced T1 MRI data were acquired and analysed. This case report includes the analysis of four 18F-FACBC PET/MRI examinations, investigating their utility in evaluating functional and structural metastasis properties. Conclusion Analysis showed high tumour-to-background ratios in brain metastases compared to other amino acid PET tracers, including high uptake in a very small cerebellar metastasis, suggesting that 18F-FACBC PET can provide early detection of otherwise overlooked metastases. Further studies to determine a threshold for 18F-FACBC brain tumour boundaries and explore its utility in clinical practice should be performed.
Collapse
Affiliation(s)
- Knut Johannessen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Postboks 8905, 7491, Trondheim, Norway
| | - Erik Magnus Berntsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Postboks 8905, 7491, Trondheim, Norway.,Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Håkon Johansen
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tora S Solheim
- Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna Karlberg
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Postboks 8905, 7491, Trondheim, Norway.,Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Live Eikenes
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Postboks 8905, 7491, Trondheim, Norway.
| |
Collapse
|
18
|
Tatekawa H, Uetani H, Hagiwara A, Yao J, Oughourlian TC, Ueda I, Raymond C, Lai A, Cloughesy TF, Nghiemphu PL, Liau LM, Bahri S, Pope WB, Salamon N, Ellingson BM. Preferential tumor localization in relation to 18F-FDOPA uptake for lower-grade gliomas. J Neurooncol 2021; 152:573-582. [PMID: 33704629 DOI: 10.1007/s11060-021-03730-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/01/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Although tumor localization and 3,4-dihydroxy-6-18F-fluoro-L-phenylalanine (FDOPA) uptake may have an association, preferential tumor localization in relation to FDOPA uptake is yet to be investigated in lower-grade gliomas (LGGs). This study aimed to identify differences in the frequency of tumor localization between FDOPA hypometabolic and hypermetabolic LGGs using a probabilistic radiographic atlas. METHODS Fifty-one patients with newly diagnosed LGG (WHO grade II, 29; III, 22; isocitrate dehydrogenase wild-type, 21; mutant 1p19q non-codeleted,16; mutant codeleted, 14) who underwent FDOPA positron emission tomography (PET) were retrospectively selected. Semiautomated tumor segmentation on FLAIR was performed. Patients with LGGs were separated into two groups (FDOPA hypometabolic and hypermetabolic LGGs) according to the normalized maximum standardized uptake value of FDOPA PET (a threshold of the uptake in the striatum) within the segmented regions. Spatial normalization procedures to build a 3D MRI-based atlas using each segmented region were validated by an analysis of differential involvement statistical mapping. RESULTS Superimposition of regions of interest showed a high number of hypometabolic LGGs localized in the frontal lobe, while a high number of hypermetabolic LGGs was localized in the insula, putamen, and temporal lobe. The statistical mapping revealed that hypometabolic LGGs occurred more frequently in the superior frontal gyrus (close to the supplementary motor area), while hypermetabolic LGGs occurred more frequently in the insula. CONCLUSION Radiographic atlases revealed preferential frontal lobe localization for FDOPA hypometabolic LGGs, which may be associated with relatively early detection.
Collapse
Affiliation(s)
- Hiroyuki Tatekawa
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.,Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Diagnostic and Interventional Radiology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hiroyuki Uetani
- Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Akifumi Hagiwara
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.,Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.,Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Bioengineering, Henry Samueli School of Engineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Talia C Oughourlian
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.,Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Neuroscience Interdepartmental Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Issei Ueda
- Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.,Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Albert Lai
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Phioanh L Nghiemphu
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Linda M Liau
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Shadfar Bahri
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Whitney B Pope
- Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Noriko Salamon
- Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA. .,Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. .,Department of Bioengineering, Henry Samueli School of Engineering, University of California Los Angeles, Los Angeles, CA, USA. .,Neuroscience Interdepartmental Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. .,UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Li Z, Kong Z, Chen J, Li J, Li N, Yang Z, Wang Y, Liu Z. 18F-Boramino acid PET/CT in healthy volunteers and glioma patients. Eur J Nucl Med Mol Imaging 2021; 48:3113-3121. [PMID: 33590273 DOI: 10.1007/s00259-021-05212-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/18/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE In this work, the safety, biodistribution, and radiation dosimetry of large neutral amino acid transporter type-1 (LAT-1) targeting PET tracer 18F-trifluorobborate-derived tyrosine (denoted as 18F-FBY) has been investigated. It is designed as a first-in-human study in healthy volunteers and to assay LAT-1 expression level in glioma patients. METHODS Six healthy volunteers (3 M, 3 F) underwent whole-body PET acquisitions at multiple time points after bolus injection of 18F-FBY. Regions of interest (ROIs) were mapped manually on major organs, and then the time-activity curves (TACs) were obtained. Dosimetry was calculated with the OLINDA/EXM software. Thirteen patients who were suspected of glioma were scanned with PET/CT at 30 min after 18F-FBY injection. Within 7 days after PET/CT, the tumor was removed surgically, and LAT-1 immunohistochemical staining for LAT-1 was performed on tumor samples and correlated with 18F-FBY PET imaging. RESULTS 18F-FBY was well tolerated by all healthy volunteers, and no adverse symptoms were observed or reported. 18F-FBY is rapidly cleared from the blood circulation and excreted mainly through the kidneys and urinary tract. The effective dose (ED) was 0.0039 ± 0.0006 mSv/MBq. In 14 surgical confirmed gliomas (one of the patiens had two gliomas), 18F-FBY uptake increased consistently with tumor grade, with maximum standard uptake values (SUVmax) of 0.28 ± 0.14 and 2.84 ± 0.46 and tumor-to-normal contralateral activity (T/N) ratio of 2.30 ± 1.26 and 24.56 ± 6.32 in low- and high-grade tumors, respectively. In addition to the significant difference in the uptakes between low- and high-grade gliomas (P < 0.001), the immunohistochemical staining confirmed the positive correlations between the SUVmax, LAT-1 expression (r2 = 0.80, P < 0.001), and Ki-67 labeling index (r2 = 0.79, P < 0.001). CONCLUSION 18F-FBY is a PET tracer with favorable dosimetry profile and pharmacokinetics. It has the potential to assay LAT-1 expression in glioma patients and may provide imaging guidance for further boron neutron capture therapy of gliomas. TRIAL REGISTRATION clinicaltrials.gov (NCT03980431).
Collapse
Affiliation(s)
- Zhu Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of nuclear medicine, Peking University Cancer Hospital & Institute, Beijing, 100871, China
| | - Ziren Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junyi Chen
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Jiyuan Li
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Nan Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of nuclear medicine, Peking University Cancer Hospital & Institute, Beijing, 100871, China
| | - Zhi Yang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of nuclear medicine, Peking University Cancer Hospital & Institute, Beijing, 100871, China.
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Zhibo Liu
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China. .,Peking University-Tsinghua University Center for Life Sciences, Beijing, 100871, China.
| |
Collapse
|
20
|
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
|
21
|
Maximum Uptake and Hypermetabolic Volume of 18F-FDOPA PET Estimate Molecular Status and Overall Survival in Low-Grade Gliomas: A PET and MRI Study. Clin Nucl Med 2020; 45:e505-e511. [PMID: 33031233 DOI: 10.1097/rlu.0000000000003318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE We evaluated F-FDOPA PET and MRI characteristics in association with the molecular status and overall survival (OS) in a large number of low-grade gliomas (LGGs). METHODS Eighty-six patients who underwent F-FDOPA PET and MRI and were diagnosed with new or recurrent LGGs were retrospectively evaluated with respect to their isocitrate dehydrogenase (IDH) and 1p19q status (10 IDH wild type, 57 mutant, 19 unknown; 1p19q status in IDH mutant: 20 noncodeleted, 37 codeleted). After segmentation of the hyperintense area on fluid-attenuated inversion recovery image (FLAIRROI), the following were calculated: normalized SUVmax (nSUVmax) of F-FDOPA relative to the striatum, F-FDOPA hypermetabolic volume (tumor-to-striatum ratios >1), FLAIRROI volume, relative cerebral blood volume, and apparent diffusion coefficient within FLAIRROI. Receiver operating characteristic curve and Cox regression analyses were performed. RESULTS PET and MRI metrics combined with age predicted the IDH mutation and 1p19q codeletion statuses with sensitivities of 73% and 76% and specificities of 100% and 94%, respectively. Significant correlations were found between OS and the IDH mutation status (hazard ratio [HR] = 4.939), nSUVmax (HR = 2.827), F-FDOPA hypermetabolic volume (HR = 1.048), and FLAIRROI volume (HR = 1.006). The nSUVmax (HR = 151.6) for newly diagnosed LGGs and the F-FDOPA hypermetabolic volume (HR = 1.038) for recurrent LGGs demonstrated significant association with OS. CONCLUSIONS Combining F-FDOPA PET and MRI with age proved useful for predicting the molecular status in patients with LGGs, whereas the nSUVmax and F-FDOPA hypermetabolic volume may be useful for prognostication.
Collapse
|
22
|
Galldiks N, Unterrainer M, Judov N, Stoffels G, Rapp M, Lohmann P, Vettermann F, Dunkl V, Suchorska B, Tonn JC, Kreth FW, Fink GR, Bartenstein P, Langen KJ, Albert NL. Photopenic defects on O-(2-[18F]-fluoroethyl)-L-tyrosine PET: clinical relevance in glioma patients. Neuro Oncol 2020; 21:1331-1338. [PMID: 31077276 DOI: 10.1093/neuonc/noz083] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND O-(2-[18F]-fluoroethyl)-L-tyrosine (FET) PET has a sensitivity of more than 90% to detect gliomas. In the remaining small fraction of gliomas without increased tracer uptake, some tumors even show photopenic defects whose clinical significance is unclear. METHODS Glioma patients with a negative FET PET scan prior to neuropathological confirmation were identified retrospectively. Gliomas were rated visually as (i) having indifferent FET uptake or (ii) photopenic, if FET uptake was below background activity. FET uptake in the area of signal hyperintensity on the T2/fluid attenuated inversion recovery-weighted MRI was evaluated by mean standardized uptake value (SUV) and mean tumor-to-brain ratio (TBR). The progression-free survival (PFS) of photopenic gliomas was compared with that of gliomas with indifferent FET uptake. RESULTS Of 100 FET-negative gliomas, 40 cases with photopenic defects were identified. Fifteen of these 40 cases (38%) had World Health Organization (WHO) grades III and IV gliomas. FET uptake in photopenic gliomas was significantly decreased compared with both the healthy-appearing brain tissue (SUV, 0.89 ± 0.26 vs 1.08 ± 0.23; P < 0.001) and gliomas with indifferent FET uptake (TBR, 0.82 ± 0.09 vs 0.96 ± 0.13; P < 0.001). Irrespective of the applied treatment, isocitrate dehydrogenase (IDH)-mutated WHO grade II diffuse astrocytoma patients with indifferent FET uptake (n = 25) had a significantly longer PFS than patients with IDH-mutated diffuse astrocytomas (WHO grade II) with photopenic defects (n = 11) (51 vs 24 mo; P = 0.027). The multivariate survival analysis indicated that photopenic defects predict an unfavorable PFS (P = 0.009). CONCLUSION Photopenic gliomas in negative FET PET scans should be managed more actively, as they seem to have a higher risk of harboring a higher-grade glioma and an unfavorable outcome.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany.,Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Düsseldorf, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich (LMU), Munich, Germany
| | - Natalie Judov
- Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany
| | - Marion Rapp
- Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany
| | - Franziska Vettermann
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich (LMU), Munich, Germany
| | - Veronika Dunkl
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | | | - Jörg C Tonn
- Department of Neurosurgery, LMU, Munich, Germany
| | | | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich (LMU), Munich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3/-4), Reseach Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich (LMU), Munich, Germany
| |
Collapse
|
23
|
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
|
24
|
Kamson DO. Hypometabolic gliomas on FET-PET-is there an inverted U-curve for survival? Neuro Oncol 2020; 21:1221-1222. [PMID: 31278863 DOI: 10.1093/neuonc/noz122] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- David Olayinka Kamson
- Department of Neurology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
25
|
Giammarile F, Castellucci P, Dierckx R, Estrada Lobato E, Farsad M, Hustinx R, Jalilian A, Pellet O, Rossi S, Paez D. Non-FDG PET/CT in Diagnostic Oncology: a pictorial review. Eur J Hybrid Imaging 2019; 3:20. [PMID: 34191163 PMCID: PMC8218094 DOI: 10.1186/s41824-019-0066-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/22/2019] [Indexed: 11/25/2022] Open
Abstract
Positron emission tomography/computed tomography (PET/CT) is currently one of the main imaging modalities for cancer patients worldwide. Fluorodeoxyglucose (FDG) PET/CT has earned its global recognition in the modern management of cancer patients and is rapidly becoming an important imaging modality for patients with cardiac, neurological, and infectious/inflammatory conditions. Despite its proven benefits, FDG has limitations in the assessment of several relevant tumours such as prostate cancer. Therefore, there has been a pressing need for the development and clinical application of different PET radiopharmaceuticals that could image these tumours more precisely. Accordingly, several non-FDG PET radiopharmaceuticals have been introduced into the clinical arena for management of cancer. This trend will undoubtedly continue to spread internationally. The use of PET/CT with different PET radiopharmaceuticals specific to tumour type and biological process being assessed is part of the personalised precision medicine approach. The objective of this publication is to provide a case-based method of understanding normal biodistribution, variants, and pitfalls, including several examples of different imaging appearances for the main oncological indications for each of the new non-FDG PET radiopharmaceuticals. This should facilitate the interpretation and recognition of common variants and pitfalls to ensure that, in clinical practice, the official report is accurate and helpful. Some of these radiopharmaceuticals are already commercially available in many countries (e.g. 68Ga-DOTATATE and DOTATOC), others are in the process of becoming available (e.g. 68Ga-PSMA), and some are still being researched. However, this list is subject to change as some radiopharmaceuticals are increasingly utilised, while others gradually decrease in use.
Collapse
Affiliation(s)
- Francesco Giammarile
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria.
| | - Paolo Castellucci
- Department of Nuclear Medicine, Sant'Orsola-Malpighi Hospital, 40138, Bologna, Italy
| | - Rudi Dierckx
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| | - Enrique Estrada Lobato
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Mohsen Farsad
- Department of Nuclear Medicine, Bolzano Hospital, Bolzano, Italy
| | - Roland Hustinx
- Department of Nuclear Medicine, CHU Liège, University of Liège, Liège, Belgium
| | - Amirreza Jalilian
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Olivier Pellet
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Susana Rossi
- Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Diana Paez
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| |
Collapse
|
26
|
Abstract
PURPOSE To retrospectively investigate the uptake of F-fluciclovine on PET/CT in patients with suspected recurrent high-grade glioma (HGG). METHODS Twenty-one patients were included. The standard of truth was histopathologic interpretation if available. When histopathology was not available or rebiopsy did not show signs of malignancy, clinical follow-up including MRI and clinical outcome was considered the standard of truth. RESULTS All 21 patients met the reference standard of either histopathologic proof of HGG recurrence (n = 10) or disease progression clinically and with tumor growth corresponding to the primary tumor sites on follow-up MRI (n = 11). Median time from PET/CT to death was 5 months (range, 1-20 months). Median time from primary diagnosis to death was 14.5 months (range, 6 to >400). Average SUVmax of the lesions was 8.3 ± 5.3 (SD) and 0.34 ± 0.13 for normal brain tissue. Median lesion-to-background ratio was 21.6 (range, 3.1-84.4). In 4 patients, F-fluciclovine PET/CT detected small satellite tumors that had not been reported on MR. CONCLUSIONS The uptake of F-fluciclovine in clinically and/or histopathologically confirmed recurrent HGG is high compared with the uptake reported for other amino acid PET tracers. Because of the high tumor uptake and thus high tracer contrast, small satellite tumors with a diameter below usual reported PET spatial resolution and not reported on MRI were detected in 4 patients. As no patients with confirmed treatment-related changes were included, we cannot as of yet ascertain the ability of F-fluciclovine PET to discriminate between recurrent HGG and treatment-related changes, for example, pseudoprogression and radionecrosis.
Collapse
|
27
|
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
|
28
|
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
|
29
|
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: 34] [Impact Index Per Article: 5.7] [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
|
30
|
Röhrich M, Loktev A, Wefers AK, Altmann A, Paech D, Adeberg S, Windisch P, Hielscher T, Flechsig P, Floca R, Leitz D, Schuster JP, Huber PE, Debus J, von Deimling A, Lindner T, Haberkorn U. IDH-wildtype glioblastomas and grade III/IV IDH-mutant gliomas show elevated tracer uptake in fibroblast activation protein-specific PET/CT. Eur J Nucl Med Mol Imaging 2019; 46:2569-2580. [PMID: 31388723 DOI: 10.1007/s00259-019-04444-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Targeting fibroblast activation protein (FAP) is a new diagnostic approach allowing the visualization of tumor stroma. Here, we applied FAP-specific PET imaging to gliomas. We analyzed the target affinity and specificity of two FAP ligands (FAPI-02 and FAPI-04) in vitro, and the pharmacokinetics and biodistribution in mice in vivo. Clinically, we used 68Ga-labeled FAPI-02/04 for PET imaging in 18 glioma patients (five IDH-mutant gliomas, 13 IDH-wildtype glioblastomas). METHODS For binding studies with 177Lu-radiolabeled FAPI-02/04, we used the glioblastoma cell line U87MG, FAP-transfected fibrosarcoma cells, and CD26-transfected human embryonic kidney cells. For pharmacokinetic and biodistribution studies, U87MG-xenografted mice were injected with 68Ga-labeled compounds followed by small-animal PET imaging and 177Lu-labeled FAPI-02/04, respectively. Clinical PET/CT scans were performed 30 min post intravenous administration of 68Ga-FAPI-02/04. PET and MRI scans were co-registrated. Immunohistochemistry was done on 14 gliomas using a FAP-specific antibody. RESULTS FAPI-02 and FAPI-04 showed high binding specificity to FAP. FAPI-04 demonstrated higher tumor accumulation and delayed elimination compared with FAPI-02 in preclinical studies. IDH-wildtype glioblastomas and grade III/IV, but not grade II, IDH-mutant gliomas showed elevated tracer uptake. In glioblastomas, we observed spots with increased uptake in projection on contrast-enhancing areas. Immunohistochemistry showed FAP-positive cells with mainly elongated cell bodies and perivascular FAP-positive cells in glioblastomas and an anaplastic IDH-mutant astrocytoma. CONCLUSIONS Using FAP-specific PET imaging, increased tracer uptake in IDH-wildtype glioblastomas and high-grade IDH-mutant astrocytomas, but not in diffuse astrocytomas, may allow non-invasive distinction between low-grade IDH-mutant and high-grade gliomas. Therefore, FAP-specific imaging in gliomas may be useful for follow-up studies although further clinical evaluation is required.
Collapse
Affiliation(s)
- Manuel Röhrich
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.
| | - Anastasia Loktev
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Annika K Wefers
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Annette Altmann
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Paech
- Division of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Adeberg
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Paul Windisch
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Hielscher
- Department of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paul Flechsig
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Ralf Floca
- Division of Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominik Leitz
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Julius P Schuster
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Radiooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Radiooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Lindner
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Uwe Haberkorn
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
31
|
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]
|
32
|
Abstract
PURPOSE OF REVIEW The aim of this study was to give an update on the emerging role of PET using radiolabelled amino acids in the diagnostic workup and management of patients with cerebral gliomas and brain metastases. RECENT FINDINGS Numerous studies have demonstrated the potential of PET using radiolabelled amino acids for differential diagnosis of brain tumours, delineation of tumour extent for treatment planning and biopsy guidance, differentiation between tumour progression and recurrence versus treatment-related changes, and for monitoring of therapy. The Response Assessment in Neuro-Oncology (RANO) working group - an international effort to develop new standardized response criteria for clinical trials in brain tumours - has recently recommended the use of amino acid PET imaging for brain tumour management in addition to MRI at every stage of disease. With the introduction of F-18 labelled amino acids, a broader clinical application has become possible, but is still hampered by the lack of regulatory approval and of reimbursement in many countries. SUMMARY PET using radiolabelled amino acids is a rapidly evolving method that can significantly enhance the diagnostic value of MRI in brain tumours. Current developments suggest that this imaging technique will become an indispensable tool in neuro-oncological centres in the near future.
Collapse
|
33
|
Suchorska B, Giese A, Biczok A, Unterrainer M, Weller M, Drexler M, Bartenstein P, Schüller U, Tonn JC, Albert NL. Identification of time-to-peak on dynamic 18F-FET-PET as a prognostic marker specifically in IDH1/2 mutant diffuse astrocytoma. Neuro Oncol 2019; 20:279-288. [PMID: 29016996 DOI: 10.1093/neuonc/nox153] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Stratification of glioma according to isocitrate dehydrogenase 1/2 (IDH1/2) mutation and 1p/19q codeletion status has gained major importance in the new World Health Organization (WHO) classification. Parameters derived from uptake dynamics of 18F-fluoro-ethyl-tyrosine PET (18F-FET-PET) such as minimal time-to-peak (TTPmin) allow discrimination between different prognostic glioma subgroups, too. The present study is aimed at exploring whether TTPmin analysis provides prognostic information beyond the WHO classification. Methods Three hundred patients with newly diagnosed WHO 2007 grades II-IV gliomas with 18F-FET-PET imaging at diagnosis were grouped into 4 subgroups (IDH1/2 mut-1p/19q codel; IDH1/2 mut-1p/19q non-codel; IDH1/2 wildtype WHO grade II and III tumors; and glioblastoma). Clinical and imaging factors such as age, Karnofsky performance score, treatment, TTPmin, and maximal tumor-to-brain ratio (TBRmax) were analyzed with regard to progression-free and overall survival (PFS and OS) via univariate and multivariate regression analysis. Results PFS and OS were longest in the IDH1/2 mut-1p/19q codel subgroup, followed by IDH1/2 mut-1p/19q non-codel, IDH1/2 wildtype, and GBM (P < 0.001). Further, outcome stratified by TTPmin with a cutoff of 17.5 minutes revealed significantly longer PFS and OS in patients with TTPmin >17.5 minutes (P < 0.001 for PFS and OS). Lower TBRmax values or the absence of 18F-FET uptake was also associated with favorable outcome in the entire group. In the subgroup analyses, longer median TTPmin was associated with improved outcome specifically in the IDH1/2 mut-1p/19q non-codel group. Conclusion 18F-FET-PET-derived dynamic analysis defines prognostically distinct subgroups of IDH1/2 mutant-1p/19q non-codel gliomas which cannot be distinguished as yet by molecular marker analysis.
Collapse
Affiliation(s)
- Bogdana Suchorska
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany (B.S., A.B., J.C.T.); German Cancer Consortium, partner site Munich, German Cancer Research Center, Heidelberg, Germany (B.S., A.G., A.B., M.U., M.D., P.B., U.S., J.C.T., N.L.A.); Department of Neuropathology (A.G., U.S.) and Department of Nuclear Medicine (M.U., M.D., P.B., N.L.A.), Ludwig-Maximilians-University, Munich, Germany; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.)
| | - Armin Giese
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany (B.S., A.B., J.C.T.); German Cancer Consortium, partner site Munich, German Cancer Research Center, Heidelberg, Germany (B.S., A.G., A.B., M.U., M.D., P.B., U.S., J.C.T., N.L.A.); Department of Neuropathology (A.G., U.S.) and Department of Nuclear Medicine (M.U., M.D., P.B., N.L.A.), Ludwig-Maximilians-University, Munich, Germany; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.)
| | - Annamaria Biczok
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany (B.S., A.B., J.C.T.); German Cancer Consortium, partner site Munich, German Cancer Research Center, Heidelberg, Germany (B.S., A.G., A.B., M.U., M.D., P.B., U.S., J.C.T., N.L.A.); Department of Neuropathology (A.G., U.S.) and Department of Nuclear Medicine (M.U., M.D., P.B., N.L.A.), Ludwig-Maximilians-University, Munich, Germany; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.)
| | - Marcus Unterrainer
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany (B.S., A.B., J.C.T.); German Cancer Consortium, partner site Munich, German Cancer Research Center, Heidelberg, Germany (B.S., A.G., A.B., M.U., M.D., P.B., U.S., J.C.T., N.L.A.); Department of Neuropathology (A.G., U.S.) and Department of Nuclear Medicine (M.U., M.D., P.B., N.L.A.), Ludwig-Maximilians-University, Munich, Germany; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.)
| | - Michael Weller
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany (B.S., A.B., J.C.T.); German Cancer Consortium, partner site Munich, German Cancer Research Center, Heidelberg, Germany (B.S., A.G., A.B., M.U., M.D., P.B., U.S., J.C.T., N.L.A.); Department of Neuropathology (A.G., U.S.) and Department of Nuclear Medicine (M.U., M.D., P.B., N.L.A.), Ludwig-Maximilians-University, Munich, Germany; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.)
| | - Mark Drexler
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany (B.S., A.B., J.C.T.); German Cancer Consortium, partner site Munich, German Cancer Research Center, Heidelberg, Germany (B.S., A.G., A.B., M.U., M.D., P.B., U.S., J.C.T., N.L.A.); Department of Neuropathology (A.G., U.S.) and Department of Nuclear Medicine (M.U., M.D., P.B., N.L.A.), Ludwig-Maximilians-University, Munich, Germany; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.)
| | - Peter Bartenstein
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany (B.S., A.B., J.C.T.); German Cancer Consortium, partner site Munich, German Cancer Research Center, Heidelberg, Germany (B.S., A.G., A.B., M.U., M.D., P.B., U.S., J.C.T., N.L.A.); Department of Neuropathology (A.G., U.S.) and Department of Nuclear Medicine (M.U., M.D., P.B., N.L.A.), Ludwig-Maximilians-University, Munich, Germany; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.)
| | - Ulrich Schüller
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany (B.S., A.B., J.C.T.); German Cancer Consortium, partner site Munich, German Cancer Research Center, Heidelberg, Germany (B.S., A.G., A.B., M.U., M.D., P.B., U.S., J.C.T., N.L.A.); Department of Neuropathology (A.G., U.S.) and Department of Nuclear Medicine (M.U., M.D., P.B., N.L.A.), Ludwig-Maximilians-University, Munich, Germany; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.)
| | - Jörg-Christian Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany (B.S., A.B., J.C.T.); German Cancer Consortium, partner site Munich, German Cancer Research Center, Heidelberg, Germany (B.S., A.G., A.B., M.U., M.D., P.B., U.S., J.C.T., N.L.A.); Department of Neuropathology (A.G., U.S.) and Department of Nuclear Medicine (M.U., M.D., P.B., N.L.A.), Ludwig-Maximilians-University, Munich, Germany; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.)
| | - Nathalie L Albert
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany (B.S., A.B., J.C.T.); German Cancer Consortium, partner site Munich, German Cancer Research Center, Heidelberg, Germany (B.S., A.G., A.B., M.U., M.D., P.B., U.S., J.C.T., N.L.A.); Department of Neuropathology (A.G., U.S.) and Department of Nuclear Medicine (M.U., M.D., P.B., N.L.A.), Ludwig-Maximilians-University, Munich, Germany; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.)
| |
Collapse
|
34
|
The Emerging Role of Amino Acid PET in Neuro-Oncology. Bioengineering (Basel) 2018; 5:bioengineering5040104. [PMID: 30487391 PMCID: PMC6315339 DOI: 10.3390/bioengineering5040104] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/12/2022] Open
Abstract
Imaging plays a critical role in the management of the highly complex and widely diverse central nervous system (CNS) malignancies in providing an accurate diagnosis, treatment planning, response assessment, prognosis, and surveillance. Contrast-enhanced magnetic resonance imaging (MRI) is the primary modality for CNS disease management due to its high contrast resolution, reasonable spatial resolution, and relatively low cost and risk. However, defining tumor response to radiation treatment and chemotherapy by contrast-enhanced MRI is often difficult due to various factors that can influence contrast agent distribution and perfusion, such as edema, necrosis, vascular alterations, and inflammation, leading to pseudoprogression and pseudoresponse assessments. Amino acid positron emission tomography (PET) is emerging as the method of resolving such equivocal lesion interpretations. Amino acid radiotracers can more specifically differentiate true tumor boundaries from equivocal lesions based on their specific and active uptake by the highly metabolic cellular component of CNS tumors. These therapy-induced metabolic changes detected by amino acid PET facilitate early treatment response assessments. Integrating amino acid PET in the management of CNS malignancies to complement MRI will significantly improve early therapy response assessment, treatment planning, and clinical trial design.
Collapse
|
35
|
Neurosarcoidosis Mimics High-Grade Glioma in Dynamic 18F-FET PET Due to LAT Expression. Clin Nucl Med 2018; 43:840-841. [DOI: 10.1097/rlu.0000000000002266] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Comparison of 18F-GE-180 and dynamic 18F-FET PET in high grade glioma: a double-tracer pilot study. Eur J Nucl Med Mol Imaging 2018; 46:580-590. [DOI: 10.1007/s00259-018-4166-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/11/2018] [Indexed: 12/20/2022]
|
37
|
Freyschlag CF, Krieg SM, Kerschbaumer J, Pinggera D, Forster MT, Cordier D, Rossi M, Miceli G, Roux A, Reyes A, Sarubbo S, Smits A, Sierpowska J, Robe PA, Rutten GJ, Santarius T, Matys T, Zanello M, Almairac F, Mondot L, Jakola AS, Zetterling M, Rofes A, von Campe G, Guillevin R, Bagatto D, Lubrano V, Rapp M, Goodden J, De Witt Hamer PC, Pallud J, Bello L, Thomé C, Duffau H, Mandonnet E. Imaging practice in low-grade gliomas among European specialized centers and proposal for a minimum core of imaging. J Neurooncol 2018; 139:699-711. [PMID: 29992433 PMCID: PMC6132968 DOI: 10.1007/s11060-018-2916-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/29/2018] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Imaging studies in diffuse low-grade gliomas (DLGG) vary across centers. In order to establish a minimal core of imaging necessary for further investigations and clinical trials in the field of DLGG, we aimed to establish the status quo within specialized European centers. METHODS An online survey composed of 46 items was sent out to members of the European Low-Grade Glioma Network, the European Association of Neurosurgical Societies, the German Society of Neurosurgery and the Austrian Society of Neurosurgery. RESULTS A total of 128 fully completed surveys were received and analyzed. Most centers (n = 96, 75%) were academic and half of the centers (n = 64, 50%) adhered to a dedicated treatment program for DLGG. There were national differences regarding the sequences enclosed in MRI imaging and use of PET, however most included T1 (without and with contrast, 100%), T2 (100%) and TIRM or FLAIR (20, 98%). DWI is performed by 80% of centers and 61% of centers regularly performed PWI. CONCLUSION A minimal core of imaging composed of T1 (w/wo contrast), T2, TIRM/FLAIR, PWI and DWI could be identified. All morphologic images should be obtained in a slice thickness of ≤ 3 mm. No common standard could be obtained regarding advanced MRI protocols and PET. IMPORTANCE OF THE STUDY We believe that our study makes a significant contribution to the literature because we were able to determine similarities in numerous aspects of LGG imaging. Using the proposed "minimal core of imaging" in clinical routine will facilitate future cooperative studies.
Collapse
Affiliation(s)
- Christian F Freyschlag
- Department of Neurosurgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Sandro M Krieg
- Department of Neurosurgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Johannes Kerschbaumer
- Department of Neurosurgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Daniel Pinggera
- Department of Neurosurgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | | | - Dominik Cordier
- Department of Neurosurgery, Universitätsspital Basel, Basel, Switzerland
| | - Marco Rossi
- Neurosurgical Oncology Unit, Humanitas Research Hospital, IRCCS, Milan, Italy
| | - Gabriele Miceli
- Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - Alexandre Roux
- Department of Neurosurgery, Sainte-Anne Hospital, Paris Descartes University, Sorbonne Paris Cité, Paris, France
- Inserm U894, IMA-Brain, Centre de Psychiatrie et Neurosciences, Paris, France
| | - Andrés Reyes
- European Master's in Clinical Linguistics (EMCL), University of Groningen, Groningen, The Netherlands
- EMCL University of Potsdam, Potsdam, Germany
- Neuroscience Institute, and Laboratory of Experimental Psychology, Faculty of Psychology, El Bosque University, Bogotá, Colombia
| | - Silvio Sarubbo
- Division of Neurosurgery, Structural and Functional Connectivity Lab Project, "S. Chiara" Hospital, APSS, Trento, Italy
| | - Anja Smits
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden
| | - Joanna Sierpowska
- Cognition and Brain Plasticity Unit, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, Barcelona, Spain
- Department of Cognition, Development and Education Psychology, Barcelona, Spain
| | - Pierre A Robe
- Department of Neurology and Neurosurgery, Rudolf Magnus Brain Institute, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Geert-Jan Rutten
- Department of Neurosurgery, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands
| | - Thomas Santarius
- Department of Neurosurgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Tomasz Matys
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Marc Zanello
- Department of Neurosurgery, Sainte-Anne Hospital, Paris Descartes University, Sorbonne Paris Cité, Paris, France
- Inserm U894, IMA-Brain, Centre de Psychiatrie et Neurosciences, Paris, France
| | - Fabien Almairac
- Neurosurgery Department, Hôpital Pasteur 2, University Hospital of Nice, Nice, France
| | - Lydiane Mondot
- Radiology Department, Hôpital Pasteur 2, University Hospital of Nice, Nice, France
| | - Asgeir S Jakola
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg, Sweden
| | - Maria Zetterling
- Department of Neurosurgery, Institution of Neuroscience, Uppsala University Hospital, Uppsala, Sweden
| | - Adrià Rofes
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
- Department of Cognitive Science, Johns Hopkins University, Baltimore, USA
| | - Gord von Campe
- Department of Neurosurgery, Medical University Graz, Graz, Austria
| | - Remy Guillevin
- DACTIM, UMR CNRS 7348, Université de Poitiers et CHU de Poitiers, Poitiers, France
| | - Daniele Bagatto
- Neuroradiology Department, University Hospital Santa Maria della Misericordia, Udine, Italy
| | - Vincent Lubrano
- Department of Neurosurgery, CHU Toulouse, Toulouse, France
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Marion Rapp
- Department of Neurosurgery, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - John Goodden
- Department of Neurosurgery, The General Infirmary at Leeds, Leeds, West Yorkshire, UK
| | | | - Johan Pallud
- Department of Neurosurgery, Sainte-Anne Hospital, Paris Descartes University, Sorbonne Paris Cité, Paris, France
- Inserm U894, IMA-Brain, Centre de Psychiatrie et Neurosciences, Paris, France
| | - Lorenzo Bello
- Neurosurgical Oncology Unit, Humanitas Research Hospital, IRCCS, Milan, Italy
| | - Claudius Thomé
- Department of Neurosurgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Hugues Duffau
- Department of Neurosurgery, Hôpital Gui de Chauliac, Montpellier Medical University Center, Montpellier, France
- Institute of Neuroscience of Montpellier, INSERM U1051, University of Montpellier, Montpellier, France
| | - Emmanuel Mandonnet
- Department of Neurosurgery, Lariboisière Hospital, APHP, Paris, France
- University Paris 7, Paris, France
- IMNC, UMR 8165, Orsay, France
| |
Collapse
|
38
|
Näslund O, Smits A, Förander P, Laesser M, Bartek J, Gempt J, Liljegren A, Daxberg EL, Jakola AS. Amino acid tracers in PET imaging of diffuse low-grade gliomas: a systematic review of preoperative applications. Acta Neurochir (Wien) 2018; 160:1451-1460. [PMID: 29797098 PMCID: PMC5995993 DOI: 10.1007/s00701-018-3563-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/09/2018] [Indexed: 11/28/2022]
Abstract
Positron emission tomography (PET) imaging using amino acid tracers has in recent years become widely used in the diagnosis and prediction of disease course in diffuse low-grade gliomas (LGG). However, implications of preoperative PET for treatment and prognosis in this patient group have not been systematically studied. The aim of this systematic review was to evaluate the preoperative diagnostic and prognostic value of amino acid PET in suspected diffuse LGG. Medline, Cochrane Library, and Embase databases were systematically searched using keywords "PET," "low-grade glioma," and "amino acids tracers" with their respective synonyms. Out of 2137 eligible studies, 28 met the inclusion criteria. Increased amino acid uptake (lesion/brain) was consistently reported among included studies; in 25-92% of subsequently histopathology-verified LGG, in 83-100% of histopathology-verified HGG, and also in some non-neoplastic lesions. No consistent results were found in studies reporting hot spot areas on PET in MRI-suspected LGG. Thus, the diagnostic value of amino acid PET imaging in suspected LGG has proven difficult to interpret, showing clear overlap and inconsistencies among reported results. Similarly, the results regarding the prognostic value of PET in suspected LGG and the correlation between uptake ratios and the molecular tumor status of LGG were conflicting. This systematic review illustrates the difficulties with prognostic studies presenting data on group-level without adjustment for established clinical prognostic factors, leading to a loss of additional prognostic information. We conclude that the prognostic value of PET is limited to analysis of histological subgroups of LGG and is probably strongest when using kinetic analysis of dynamic FET uptake parameters.
Collapse
Affiliation(s)
- Olivia Näslund
- Sahlgrenska Academy, Medicinaregatan 3, 41390, Gothenburg, Sweden.
| | - Anja Smits
- Institute of Physiology and Neuroscience, Sahlgrenska Academy, Gothenburg, Sweden
- Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden
| | - Petter Förander
- Department of Clinical Neuroscience and Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| | - Mats Laesser
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
- Department of Radiology, Institute of Clinical Sciences, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jiri Bartek
- Department of Clinical Neuroscience and Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Neurosurgery, St. Olavs Hospital, Trondheim, Norway
- Department of Neurosurgery, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Ann Liljegren
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Eva-Lotte Daxberg
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Asgeir Store Jakola
- Institute of Physiology and Neuroscience, Sahlgrenska Academy, Gothenburg, Sweden
- Medical Library, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
39
|
Ly KI, Gerstner ER. The Role of Advanced Brain Tumor Imaging in the Care of Patients with Central Nervous System Malignancies. Curr Treat Options Oncol 2018; 19:40. [PMID: 29931476 DOI: 10.1007/s11864-018-0558-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OPINION STATEMENT T1-weighted post-contrast and T2-weighted fluid-attenuated inversion recovery (FLAIR) magnetic resonance imaging (MRI) constitute the gold standard for diagnosis and response assessment in neuro-oncologic patients but are limited in their ability to accurately reflect tumor biology and metabolism, particularly over the course of a patient's treatment. Advanced MR imaging methods are sensitized to different biophysical processes in tissue, including blood perfusion, tumor metabolism, and chemical composition of tissue, and provide more specific information on tissue physiology than standard MRI. This review provides an overview of the most common and emerging advanced imaging modalities in the field of brain tumor imaging and their applications in the care of neuro-oncologic patients.
Collapse
Affiliation(s)
- K Ina Ly
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9E, Boston, MA, 02114, USA
| | - Elizabeth R Gerstner
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9E, Boston, MA, 02114, USA.
| |
Collapse
|
40
|
Verger A, Arbizu J, Law I. Role of amino-acid PET in high-grade gliomas: limitations and perspectives. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2018; 62:254-266. [PMID: 29696948 DOI: 10.23736/s1824-4785.18.03092-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Positron emission tomography (PET) using radiolabeled amino-acids was recently recommended by the Response Assessment in Neuro-Oncology (RANO) working group as an additional tool in the diagnostic assessment of brain tumors. The aim of this review is to summarize available literature data on the role of amino-acid PET imaging in high-grade gliomas (HGGs), with regard to diagnosis, treatment planning and follow-up of these tumors. Indeed, amino-acid PET applications are multiple throughout the evolution of HGGs. However, certain limitations such as lack of specificity, uncertain value for grading and prognostication or the limited data for treatment monitoring should to be taken into account, the latter of which are further developed in this review. Notwithstanding these limitations, amino-acid PET is becoming increasingly accessible in many nuclear medicine centers. Larger prospective cohort prospective studies are thus needed in order to increase the clinical value of this modality and enable its extended use to the largest number of patients.
Collapse
Affiliation(s)
- Antoine Verger
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, CHRU Nancy, Lorraine University, Nancy, France - .,IADI, INSERM, Lorraine University, Nancy, France -
| | - Javier Arbizu
- Department of Nuclear Medicine, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
41
|
Bashir A, Brennum J, Broholm H, Law I. The diagnostic accuracy of detecting malignant transformation of low-grade glioma using O-(2-[18F]fluoroethyl)-l-tyrosine positron emission tomography: a retrospective study. J Neurosurg 2018; 130:451-464. [PMID: 29624154 DOI: 10.3171/2017.8.jns171577] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/02/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The diagnostic accuracy of O-(2-[18F]fluoroethyl)-l-tyrosine (FET) PET scanning in detecting the malignant transformation of low-grade gliomas (LGGs) is controversial. In this study, the authors retrospectively assessed the diagnostic potential of FET PET in patients with MRI-suspected malignant progression of LGGs that had previously been treated and the relationship between FET uptake and MRI and molecular biomarkers. METHODS Forty-two patients who had previously undergone surgical or multimodal treatment for a histologically verified LGG were referred for FET PET assessment because of clinical signs and/or MRI findings suggestive of tumor progression. Maximal and mean tumor-to-brain ratios (TBRmax and TBRmean, respectively) on FET PET as well as kinetic FET PET parameters (time to peak [TTP] and time-activity curve [TAC]) were determined. Final diagnoses were confirmed histologically. The diagnostic accuracy of FET parameters, separately and combined, for the detection of malignant progression was evaluated using receiver operating characteristic (ROC) curve analysis. Possible predictors that might influence the diagnostic accuracy of FET PET were assessed using multiple linear regression analysis. Spearman’s rank correlation r method was applied to determine the correlation between TBRmax and TAC, and molecular biomarkers from tumor tissues. RESULTS A total of 47 FET PET scans were obtained and showed no significant association between FET parameters and contrast enhancement on MRI. ROC curve analyses overall were unable to demonstrate any significant differentiation between nontransformed LGGs and LGGs that had transformed to high-grade gliomas when evaluating FET parameters separately or combined. After excluding the oligodendroglial subgroup, a significant difference was observed between nontransformed and transformed LGGs when combining FET parameters (i.e., TBRmax > 1.6, TAC describing a plateau or decreasing pattern, and TTP < 25 minutes), with the best result yielded by a combined analysis of TBRmax > 1.6 and TAC with a plateau or decreasing pattern (sensitivity 75% and specificity 83%, p = 0.003). The difference was even greater when patients who had previously undergone oncological treatment were also excluded (sensitivity 93% and specificity 100%, p = 0.001). Multiple linear regression analysis revealed that the presence of an oligodendroglial component (p = 0.029), previous oncological treatment (p = 0.039), and the combined FET parameters (p = 0.027) were significant confounding factors in the detection of malignant progression. TBRmax was positively correlated with increasing cell density (p = 0.040) and inversely correlated with IDH1 mutation (p = 0.006). CONCLUSIONS A single FET PET scan obtained at the time of radiological and/or clinical progression seems to be of limited value in distinguishing transformed from nontransformed LGGs, especially if knowledge of the primary tumor histopathology is not known. Therefore, FET PET imaging alone is not adequate to replace histological confirmation, but it may provide valuable information on the location and delineation of active tumor tissue, as well as an assessment of tumor biology in a subgroup of LGGs.
Collapse
Affiliation(s)
- Asma Bashir
- Departments of1Clinical Physiology, Nuclear Medicine & PET
| | | | - Helle Broholm
- 3Pathology, National University Hospital, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Ian Law
- Departments of1Clinical Physiology, Nuclear Medicine & PET
| |
Collapse
|
42
|
Unterrainer M, Winkelmann I, Suchorska B, Giese A, Wenter V, Kreth FW, Herms J, Bartenstein P, Tonn JC, Albert NL. Biological tumour volumes of gliomas in early and standard 20-40 min 18F-FET PET images differ according to IDH mutation status. Eur J Nucl Med Mol Imaging 2018; 45:1242-1249. [PMID: 29487977 DOI: 10.1007/s00259-018-3969-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/02/2018] [Indexed: 01/18/2023]
Abstract
PURPOSE For the clinical evaluation of O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET) PET images, the use of standard summation images obtained 20-40 min after injection is recommended. However, early summation images obtained 5-15 min after injection have been reported to allow better differentiation between low-grade glioma (LGG) and high-grade glioma (HGG) by capturing the early 18F-FET uptake peak specific for HGG. We compared early and standard summation images with regard to delineation of the PET-derived biological tumour volume (BTV) in correlation with the molecular genetic profile according the updated 2016 WHO classification. METHODS The analysis included 245 patients with newly diagnosed, histologically verified glioma and a positive 18F-FET PET scan prior to any further treatment. BTVs were delineated during the early 5-15 min and standard 20-40 min time frames using a threshold of 1.6 × background activity and were compared intraindividually. Volume differences between early and late summation images of >20% were considered significant and were correlated with WHO grade and the molecular genetic profile (IDH mutation and 1p/19q codeletion status). RESULTS In 52.2% of the patients (128/245), a significant difference in BTV of >20% between early and standard summation images was found. While 44.3% of WHO grade II gliomas (31 of 70) showed a significantly smaller BTV in the early summation images, 35.0% of WHO grade III gliomas (28/80) and 37.9% of WHO grade IV gliomas (36/95) had a significantly larger BTVs. Among IDH-wildtype gliomas, an even higher portion (44.4%, 67/151) showed significantly larger BTVs in the early summation images, which was observed in 5.3% (5/94) of IDH-mutant gliomas only: most of the latter had significantly smaller BTVs in the early summation images, i.e. 51.2% of IDH-mutant gliomas without 1p/19q codeletion (21/41) and 39.6% with 1p/19q codeletion (21/53). CONCLUSION BTVs delineated in early and standard summation images differed significantly in more than half of gliomas. While the standard summation images seem appropriate for delineation of LGG as well as IDH-mutant gliomas, a remarkably high percentage of HGG and, particularly, IDH-wildtype gliomas were depicted with significantly larger volumes in early summation images. This finding might be of interest for optimization of treatment planning (e.g. radiotherapy) in accordance with the individual IDH mutation status.
Collapse
Affiliation(s)
- M Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - I Winkelmann
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - B Suchorska
- Department of Neurosurgery, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - A Giese
- Department of Neuropathology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - V Wenter
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - F W Kreth
- Department of Neurosurgery, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - J Herms
- Department of Neuropathology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - P Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - J C Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - N L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
43
|
Karlberg A, Berntsen EM, Johansen H, Myrthue M, Skjulsvik AJ, Reinertsen I, Esmaeili M, Dai HY, Xiao Y, Rivaz H, Borghammer P, Solheim O, Eikenes L. Multimodal 18 F-Fluciclovine PET/MRI and Ultrasound-Guided Neurosurgery of an Anaplastic Oligodendroglioma. World Neurosurg 2017; 108:989.e1-989.e8. [DOI: 10.1016/j.wneu.2017.08.085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 08/10/2017] [Accepted: 08/12/2017] [Indexed: 11/28/2022]
|
44
|
Imaging of amino acid transport in brain tumours: Positron emission tomography with O-(2-[ 18 F]fluoroethyl)- L -tyrosine (FET). Methods 2017; 130:124-134. [DOI: 10.1016/j.ymeth.2017.05.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/08/2017] [Accepted: 05/21/2017] [Indexed: 01/01/2023] Open
|
45
|
Unterrainer M, Vettermann F, Brendel M, Holzgreve A, Lifschitz M, Zähringer M, Suchorska B, Wenter V, Illigens BM, Bartenstein P, Albert NL. Towards standardization of 18F-FET PET imaging: do we need a consistent method of background activity assessment? EJNMMI Res 2017; 7:48. [PMID: 28560582 PMCID: PMC5449315 DOI: 10.1186/s13550-017-0295-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/18/2017] [Indexed: 11/25/2022] Open
Abstract
Background PET with O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET) has reached increasing clinical significance for patients with brain neoplasms. For quantification of standard PET-derived parameters such as the tumor-to-background ratio, the background activity is assessed using a region of interest (ROI) or volume of interest (VOI) in unaffected brain tissue. However, there is no standardized approach regarding the assessment of the background reference. Therefore, we evaluated the intra- and inter-reader variability of commonly applied approaches for clinical 18F-FET PET reading. The background activity of 20 18F-FET PET scans was independently evaluated by 6 readers using a (i) simple 2D-ROI, (ii) spherical VOI with 3.0 cm diameter, and (iii) VOI consisting of crescent-shaped ROIs; each in the contralateral, non-affected hemisphere including white and gray matter in line with the European Association of Nuclear Medicine (EANM) and German guidelines. To assess intra-reader variability, each scan was evaluated 10 times by each reader. The coefficient of variation (CoV) was assessed for determination of intra- and inter-reader variability. In a second step, the best method was refined by instructions for a guided background activity assessment and validated by 10 further scans. Results Compared to the other approaches, the crescent-shaped VOIs revealed most stable results with the lowest intra-reader variabilities (median CoV 1.52%, spherical VOI 4.20%, 2D-ROI 3.69%; p < 0.001) and inter-reader variabilities (median CoV 2.14%, spherical VOI 4.02%, 2D-ROI 3.83%; p = 0.001). Using the guided background assessment, both intra-reader variabilities (median CoV 1.10%) and inter-reader variabilities (median CoV 1.19%) could be reduced even more. Conclusions The commonly applied methods for background activity assessment show different variability which might hamper 18F-FET PET quantification and comparability in multicenter settings. The proposed background activity assessment using a (guided) crescent-shaped VOI allows minimization of both intra- and inter-reader variability and might facilitate comprehensive methodological standardization of amino acid PET which is of interest in the light of the anticipated EANM technical guidelines.
Collapse
Affiliation(s)
- Marcus Unterrainer
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,Center for Clinical Research and Management Education, Division of Health Care Sciences, Dresden International University, Dresden, Germany
| | - Franziska Vettermann
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,Department of Neurosurgery, LMU Munich, Munich, Germany
| | - Michael Lifschitz
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Matthias Zähringer
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | | | - Vera Wenter
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Ben M Illigens
- Center for Clinical Research and Management Education, Division of Health Care Sciences, Dresden International University, Dresden, Germany.,Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| |
Collapse
|
46
|
Abstract
Despite the fact that MRI has evolved to become the standard method for diagnosis and monitoring of patients with brain tumours, conventional MRI sequences have two key limitations: the inability to show the full extent of the tumour and the inability to differentiate neoplastic tissue from nonspecific, treatment-related changes after surgery, radiotherapy, chemotherapy or immunotherapy. In the past decade, PET involving the use of radiolabelled amino acids has developed into an important diagnostic tool to overcome some of the shortcomings of conventional MRI. The Response Assessment in Neuro-Oncology working group - an international effort to develop new standardized response criteria for clinical trials in brain tumours - has recommended the additional use of amino acid PET imaging for brain tumour management. Concurrently, a number of advanced MRI techniques such as magnetic resonance spectroscopic imaging and perfusion weighted imaging are under clinical evaluation to target the same diagnostic problems. This Review summarizes the clinical role of amino acid PET in relation to advanced MRI techniques for differential diagnosis of brain tumours; delineation of tumour extent for treatment planning and biopsy guidance; post-treatment differentiation between tumour progression or recurrence versus treatment-related changes; and monitoring response to therapy. An outlook for future developments in PET and MRI techniques is also presented.
Collapse
Affiliation(s)
- Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4) Forschungszentrum Jülich, Wilhelm-Johnen-Strasse, D-52425 Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4) Forschungszentrum Jülich, Wilhelm-Johnen-Strasse, D-52425 Jülich, Germany.,Department of Neurology, University of Cologne, Kerpener Strasse 62, D-50937 Cologne, Germany.,Center for Integrated Oncology, Josef-Stelzmann-Strasse 9, D-50937 Cologne, Germany
| | - Elke Hattingen
- Department of Neuroradiology and Center for Integrated Oncology, University of Bonn, Sigmund-Freud-Strasse 25, D-53127 Bonn, Germany
| | - Nadim Jon Shah
- Institute of Neuroscience and Medicine (INM-3, INM-4) Forschungszentrum Jülich, Wilhelm-Johnen-Strasse, D-52425 Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Pauwelsstrasse 30, D-52074 Aachen, Germany.,Monash Institute of Medical Engineering, Department of Electrical and Computer Systems Engineering, and Monash Biomedical Imaging, School of Psychological Sciences, Monash University, 18 Innovation Walk, Clayton Campus, Wellington Road, Melbourne, Victoria 3800, Australia
| |
Collapse
|
47
|
Filss CP, Cicone F, Shah NJ, Galldiks N, Langen KJ. Amino acid PET and MR perfusion imaging in brain tumours. Clin Transl Imaging 2017; 5:209-223. [PMID: 28680873 PMCID: PMC5487907 DOI: 10.1007/s40336-017-0225-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/28/2017] [Indexed: 12/17/2022]
Abstract
Purpose Despite the excellent capacity of the conventional MRI to image brain tumours, problems remain in answering a number of critical diagnostic questions. To overcome these diagnostic shortcomings, PET using radiolabeled amino acids and perfusion-weighted imaging (PWI) are currently under clinical evaluation. The role of amino acid PET and PWI in different diagnostic challenges in brain tumours is controversial. Methods Based on the literature and experience of our centres in correlative imaging with PWI and PET using O-(2-[18F]fluoroethyl)-l-tyrosine or 3,4-dihydroxy-6-[18F]-fluoro-l-phenylalanine, the current role and shortcomings of amino acid PET and PWI in different diagnostic challenges in brain tumours are reviewed. Literature searches were performed on PubMed, and additional literature was retrieved from the reference lists of identified articles. In particular, all studies in which amino acid PET was directly compared with PWI were included. Results PWI is more readily available, but requires substantial expertise and is more sensitive to artifacts than amino acid PET. At initial diagnosis, PWI and amino acid PET can help to define a site for biopsy but amino acid PET appears to be more powerful to define the tumor extent. Both methods are helpful to differentiate progression or recurrence from unspecific posttherapeutic changes. Assessment of therapeutic efficacy can be achieved especially with amino acid PET, while the data with PWI are sparse. Conclusion Both PWI and amino acid PET add valuable diagnostic information to the conventional MRI in the assessment of patients with brain tumours, but further studies are necessary to explore the complementary nature of these two methods.
Collapse
Affiliation(s)
- Christian P Filss
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Aachen, Germany
| | - Francesco Cicone
- Unit of Nuclear Medicine, Department of Surgical and Medical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy.,Nuclear Medicine and Molecular Medicine Department, University Hospital of Lausanne, Lausanne, Switzerland
| | - Nadim Jon Shah
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Aachen, Germany.,JARA-Jülich Aachen Research Alliance, Jülich, Germany.,Monash Institute of Medical Engineering, Department of Electrical and Computer Systems Engineering, and Monash Biomedical Imaging, School of Psychological Sciences, Monash University, Melbourne, VIC Australia
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany.,Center of Integrated Oncology (CIO), University of Cologne and Bonn, Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Aachen, Germany.,JARA-Jülich Aachen Research Alliance, Jülich, Germany
| |
Collapse
|
48
|
Ferda J, Ferdová E, Hes O, Mraček J, Kreuzberg B, Baxa J. PET/MRI: Multiparametric imaging of brain tumors. Eur J Radiol 2017; 94:A14-A25. [PMID: 28283219 DOI: 10.1016/j.ejrad.2017.02.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/19/2017] [Accepted: 02/20/2017] [Indexed: 12/01/2022]
Abstract
A combination of morphological imaging of the brain with microstructural and functional imaging provides a comprehensive overview of the properties of individual tissues. While diffusion weighted imaging provides information about tissue cellularity, spectroscopic imaging allows us to evaluate the integrity of neurons and possible anaerobic glycolysis during tumor hypoxia, in addition to the presence of accelerated synthesis or degradation of cellular membranes; on the other hand, PET metabolic imaging is used to evaluate major metabolic pathways, determining the overall extent of the tumor (18F-FET, 18F-FDOPA, 18F-FCH) or the degree of differentiation (18F-FDG, 18F-FLT, 18F-FDOPA and 18F-FET). Multi-parameter analysis of tissue characteristics and determination of the phenotype of the tumor tissue is a natural advantage of PET/MRI scanning. The disadvantages are higher cost and limited availability in all centers with neuro-oncology surgery. PET/MRI scanning of brain tumors is one of the most promising indications since the earliest experiments with integrated PET/MRI imaging systems, and along with hybrid imaging of neurodegenerative diseases, represent a new direction in the development of neuroradiology on the path towards comprehensive imaging at the molecular level.
Collapse
Affiliation(s)
- Jiří Ferda
- Clinic of the Imaging Methods, University Hospital Plzen, Alej Svobody 80, 304 60 Plzeň, Czech Republic.
| | - Eva Ferdová
- Clinic of the Imaging Methods, University Hospital Plzen, Alej Svobody 80, 304 60 Plzeň, Czech Republic.
| | - Ondřej Hes
- Sikl's Institute of Pathological Anatomy, University Hospital Plzen, Alej Svobody 80;304 60 Plzeň, Czech Republic.
| | - Jan Mraček
- Clinic of the Neurosurgery, University Hospital Plzen, Alej Svobody 80, 304 60 Plzeň, Czech Republic.
| | - Boris Kreuzberg
- Clinic of the Imaging Methods, University Hospital Plzen, Alej Svobody 80, 304 60 Plzeň, Czech Republic.
| | - Jan Baxa
- Clinic of the Imaging Methods, University Hospital Plzen, Alej Svobody 80, 304 60 Plzeň, Czech Republic.
| |
Collapse
|
49
|
Unterrainer M, Galldiks N, Suchorska B, Kowalew LC, Wenter V, Schmid-Tannwald C, Niyazi M, Bartenstein P, Langen KJ, Albert NL. 18F-FET PET Uptake Characteristics in Patients with Newly Diagnosed and Untreated Brain Metastasis. J Nucl Med 2016; 58:584-589. [PMID: 27754904 DOI: 10.2967/jnumed.116.180075] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/12/2016] [Indexed: 01/03/2023] Open
Abstract
In patients with brain metastasis, PET using labeled amino acids has gained clinical importance, mainly regarding the differentiation of viable tumor tissue from treatment-related effects. However, there is still limited knowledge concerning the uptake characteristics in patients with newly diagnosed and untreated brain metastases. Hence, we evaluated the uptake characteristics in these patients using dynamic O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) PET. Methods: Patients with newly diagnosed brain metastases without prior local therapy and 18F-FET PET scanning were retrospectively identified in 2 centers. Static and dynamic PET parameters (maximal/mean tumor-to-brain-ratio [TBRmax/TBRmean], biologic tumor volume [BTV], and time-activity curves with minimal time to peak [TTPmin]) were evaluated and correlated with MRI parameters (maximal lesion diameter, volume of contrast enhancement) and originating primary tumor. Results: Forty-five brain metastases in 30 patients were included. Forty of 45 metastases (89%) had a TBRmax ≥ 1.6 and were classified as 18F-FET-positive (median TBRmax, 2.53 [range, 1.64-9.47]; TBRmean, 1.86 [range, 1.63-5.48]; and BTV, 3.59 mL [range, 0.04-23.98 mL], respectively). In 39 of 45 brain metastases eligible for dynamic analysis, a wide range of TTPmin was observed (median, 22.5 min; range, 4.5-47.5 min). All 18F-FET-negative metastases had a diameter of ≤ 1.0 cm, whereas metastases with a > 1.0 cm diameter all showed pathologic 18F-FET uptake, which did not correlate with lesion size. The highest variability of uptake intensity was observed within the group of melanoma metastases. Conclusion: Untreated metastases predominantly show increased 18F-FET uptake, and only a third of metastases < 1.0 cm were 18F-FET-negative, most likely because of scanner resolution and partial-volume effects. In metastases > 1.0 cm, 18F-FET uptake intensity was highly variable and independent of tumor size (even intraindividually). 18F-FET PET might provide additional information beyond the tumor extent by reflecting molecular features of a metastasis and might be a useful tool for future clinical applications, for example, response assessment.
Collapse
Affiliation(s)
- Marcus Unterrainer
- Department of Nuclear Medicine, University of Munich (LMU), Munich, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany.,Department of Neurology, University Hospital Cologne, Cologne, Germany
| | | | | | - Vera Wenter
- Department of Nuclear Medicine, University of Munich (LMU), Munich, Germany
| | | | - Maximilian Niyazi
- Department of Radiation Oncology, LMU, Munich, Germany.,German Cancer Consortium (DKTK) & German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University of Munich (LMU), Munich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, University of Aachen, Aachen, Germany; and.,JARA-Brain Section, Juelich-Aachen-Research-Alliance (JARA), Juelich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University of Munich (LMU), Munich, Germany
| |
Collapse
|
50
|
Abstract
A previous review published in 2012 demonstrated the role of clinical PET for diagnosis and management of brain tumors using mainly FDG, amino acid tracers, and 18F-fluorothymidine. This review provides an update on clinical PET studies, most of which are motivated by prediction of prognosis and planning and monitoring of therapy in gliomas. For FDG, there has been additional evidence supporting late scanning, and combination with 13N ammonia has yielded some promising results. Large neutral amino acid tracers have found widespread applications mostly based on 18F-labeled compounds fluoroethyltyrosine and fluorodopa for targeting biopsies, therapy planning and monitoring, and as outcome markers in clinical trials. 11C-alpha-methyltryptophan (AMT) has been proposed as an alternative to 11C-methionine, and there may also be a role for cyclic amino acid tracers. 18F-fluorothymidine has shown strengths for tumor grading and as an outcome marker. Studies using 18F-fluorocholine (FCH) and 68Ga-labeled compounds are promising but have not yet clearly defined their role. Studies on radiotherapy planning have explored the use of large neutral amino acid tracers to improve the delineation of tumor volume for irradiation and the use of hypoxia markers, in particular 18F-fluoromisonidazole. Many studies employed the combination of PET with advanced multimodal MR imaging methods, mostly demonstrating complementarity and some potential benefits of hybrid PET/MR.
Collapse
Affiliation(s)
- Karl Herholz
- The University of Manchester, Division of Neuroscience and Experimental Psychology Wolfson Molecular Imaging Centre, Manchester, England, United Kingdom.
| |
Collapse
|