1
|
Gröner B, Hoffmann C, Endepols H, Urusova EA, Brugger M, Neumaier F, Timmer M, Neumaier B, Zlatopolskiy BD. Radiosynthesis and Preclinical Evaluation of m-[ 18F]FET and [ 18F]FET-OMe as Novel [ 18F]FET Analogs for Brain Tumor Imaging. Mol Pharm 2024; 21:2795-2812. [PMID: 38747353 DOI: 10.1021/acs.molpharmaceut.3c01215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
O-([18F]Fluoroethyl)-l-tyrosine ([18F]FET) is actively transported into the brain and cancer cells by LAT1 and possibly other amino acid transporters, which enables brain tumor imaging by positron emission tomography (PET). However, tumor delivery of this probe in the presence of competing amino acids may be limited by a relatively low affinity for LAT1. The aim of the present work was to evaluate the meta-substituted [18F]FET analog m-[18F]FET and the methyl ester [18F]FET-OMe, which were designed to improve tumor delivery by altering the physicochemical, pharmacokinetic, and/or transport properties. Both tracers could be prepared with good radiochemical yields of 41-56% within 66-90 min. Preclinical evaluation with [18F]FET as a reference tracer demonstrated reduced in vitro uptake of [18F]FET-OMe by U87 glioblastoma cells and no advantage for in vivo tumor imaging. In contrast, m-[18F]FET showed significantly improved in vitro uptake and accelerated in vivo tumor accumulation in an orthotopic glioblastoma model. As such, our work identifies m-[18F]FET as a promising alternative to [18F]FET for brain tumor imaging that deserves further evaluation with regard to its transport properties and in vivo biodistribution.
Collapse
Affiliation(s)
- Benedikt Gröner
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Chris Hoffmann
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Heike Endepols
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
- Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Elizaveta A Urusova
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Melanie Brugger
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
| | - Felix Neumaier
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Marco Timmer
- Faculty of Medicine and University Hospital Cologne, Center for Neurosurgery, Department of General Neurosurgery, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Bernd Neumaier
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Boris D Zlatopolskiy
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| |
Collapse
|
2
|
Ekmekcioglu O, Albert NL, Heinrich K, Tolboom N, Van Weehaeghe D, Traub-Weidinger T, Atay LO, Garibotto V, Morbelli S. Neurological Disorders and Women's Health: Contribution of Molecular Neuroimaging Techniques. Semin Nucl Med 2024; 54:237-246. [PMID: 38365546 DOI: 10.1053/j.semnuclmed.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/18/2024]
Abstract
Sex differences in brain physiology and the mechanisms of drug action have been extensively reported. These biological variances, from structure to hormonal and genetic aspects, can profoundly influence healthy functioning and disease mechanisms and might have implications for treatment and drug development. Molecular neuroimaging techniques may help to disclose sex's impact on brain functioning, as well as the neuropathological changes underpinning several diseases. This narrative review summarizes recent lines of evidence based on PET and SPECT imaging, highlighting sex differences in normal conditions and various neurological disorders.
Collapse
Affiliation(s)
- Ozgul Ekmekcioglu
- Department of Nuclear Medicine, University of Health Sciences, Sisli Hamidiye Etfal Education and Research Hospital, Istanbul, Turkey.
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Kathrin Heinrich
- Department of Medicine III, LMU University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Tatiana Traub-Weidinger
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, University Hospitals of Geneva, Faculty of Medicine, University of Geneva, CIBM Center for Biomedical Imaging, Geneva, Switzerland
| | - Silvia Morbelli
- Nuclear Medicine Unit, AOU Città Della Salute e Della Scienza di Torino, University of Turin, Turin, Italy
| |
Collapse
|
3
|
Galldiks N, Lohmann P, Fink GR, Langen KJ. Amino Acid PET in Neurooncology. J Nucl Med 2023; 64:693-700. [PMID: 37055222 DOI: 10.2967/jnumed.122.264859] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/10/2023] [Indexed: 04/15/2023] Open
Abstract
For decades, several amino acid PET tracers have been used to optimize diagnostics in patients with brain tumors. In clinical routine, the most important clinical indications for amino acid PET in brain tumor patients are differentiation of neoplasm from nonneoplastic etiologies, delineation of tumor extent for further diagnostic and treatment planning (i.e., diagnostic biopsy, resection, or radiotherapy), differentiation of treatment-related changes such as pseudoprogression or radiation necrosis after radiation or chemoradiation from tumor progression at follow-up, and assessment of response to anticancer therapy, including prediction of patient outcome. This continuing education article addresses the diagnostic value of amino acid PET for patients with either glioblastoma or metastatic brain cancer.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany;
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany; and
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany; and
- Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
4
|
Laudicella R, Mantarro C, Catalfamo B, Alongi P, Gaeta M, Minutoli F, Baldari S, Bisdas S. PET Imaging in Gliomas. RADIOLOGY‐NUCLEAR MEDICINE DIAGNOSTIC IMAGING 2023:194-218. [DOI: 10.1002/9781119603627.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
5
|
Novruzov E, Mori Y, Antke C, Dabir M, Schmitt D, Kratochwil C, Koerber SA, Haberkorn U, Giesel FL. A Role of Non-FDG Tracers in Lung Cancer? Semin Nucl Med 2022; 52:720-733. [PMID: 35803770 DOI: 10.1053/j.semnuclmed.2022.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/11/2022]
Abstract
Since the introduction of PET/CT hybrid imaging about two decades ago the landscape of oncological imaging has fundamentally changed, opening a new era of molecular imaging with emphasis on functional characterization of biological processes such as metabolism, cellular proliferation, hypoxia, apoptosis, angiogenesis and immune response. The most commonly assessed functional hallmark of cancer is the increased metabolism in tumor cells due to well-known Warburg effect, because of which FDG has been the most employed radiotracer, the so-called pan-cancer agent, in oncological imaging. However, several limitations such as low specificity and low sensitivity for several histopathological forms of lung cancer as well as high background uptake in the normal tissue of FDG imaging lead to numerous serious pitfalls. This restricts its utilization and diagnostic value in lung cancer imaging, even though this is currently considered to be the method of choice in pulmonary cancer imaging. Accurate initial tumor staging and therapy response monitoring with respect to the TNM criteria plays a crucial role in therapy planning and management in patients with lung cancer. To this end, many efforts have been made for decades to develop novel PET radiopharmaceuticals with innovative approaches that go beyond the assessment of increased glycolytic activity alone. Radiopharmaceuticals targeting DNA synthesis, amino acid metabolism, angiogenesis, or hypoxia have been extensively studied, leading to the emergence of indications for specific clinical questions or as a complementary imaging tool alongside existing conventional or FDG imaging. Nevertheless, despite some initial encouraging results, these tracers couldn't gain a widespread use and acceptance in clinical routine. However, given its mechanism of action and some initial pilot studies regarding lung cancer imaging, FAPI has emerged as a very promising alternative tool that could provide superior or comparable diagnostic performance to FDG imaging in lung cancer entities. Thus, in this review article, we summarized the current PET radiopharmaceuticals, different imaging approaches and discussed the potential benefits and clinical applications of these agents in lung cancer imaging.
Collapse
Affiliation(s)
- Emil Novruzov
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Yuriko Mori
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Christina Antke
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Mardjan Dabir
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Dominik Schmitt
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan A Koerber
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Frederik L Giesel
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany.
| |
Collapse
|
6
|
Tong J, Wang C, Liu H. Temporal information guided dynamic dual-tracer PET signal separation network. Med Phys 2022; 49:4585-4598. [PMID: 35396705 DOI: 10.1002/mp.15566] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 02/21/2021] [Accepted: 01/24/2022] [Indexed: 11/09/2022] Open
Abstract
PURPOSE The difficulty of dynamic dual-tracer positron emission tomography (PET) technology is to separate the complete single-tracer information from mixed dual-tracer. Traditional methods cannot separate single injection single-scan dynamic dual-tracer PET images. In this paper, we propose a deep learning framework based on gated recurrent unit (GRU) network and evaluate its performance with simulation experiments and realistic monkey data. METHODS The proposed single-scan dynamic dual-tracer PET image separation network consists of three parts, including encoder, separation and decoder module. Encoder part is to map the mixed time activity curves (TACs) from the low-dimensional space to the high-dimensional space to get mixed weight vector matrix. Separation part is to capture the temporal information of mixed weight vector matrix using bi-directional GRU (bi-GRU) layer to obtain the single-tracer masks, and the decoding part remaps the high-dimensional single-tracer weight vector matrix to the low-dimensional space to obtain two separated single tracers. RESULTS In the simulation experiments under different tracers, phantoms, noise levels, arterial input function (AIF) and k-parameter with Gaussian random, compared to the stacked auto encoder (SAE) network and traditional background subtraction method, GRU-based network has better performance with low bias and mean squared error (MSE). In the realistic study, the image results of GRU network have higher mean structural similarity (MSSIM), and peak signal to noise ratio (PSNR). CONCLUSIONS This study demonstrates the feasibility of temporal information guided neural network in single-injection single-scan dynamic dual-tracer PET images separation. The GRU-based network uses TAC temporal information without AIFs to make the separation results more robust and accurate, which significantly outperforms state-of-the-art method qualitatively and quantitatively. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Junyi Tong
- State Key Laboratory of Modern Optical Instrumentation, Zhejiang University, Hangzhou, 310027, China
| | - Chunxia Wang
- State Key Laboratory of Modern Optical Instrumentation, Zhejiang University, Hangzhou, 310027, China
| | - Huafeng Liu
- State Key Laboratory of Modern Optical Instrumentation, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
7
|
Minamimoto R. Amino Acid and Proliferation PET/CT for the Diagnosis of Multiple Myeloma. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2022; 1:796357. [PMID: 39355641 PMCID: PMC11440849 DOI: 10.3389/fnume.2021.796357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 11/22/2021] [Indexed: 10/03/2024]
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by infiltration of monoclonal plasma cells in the bone marrow (BM). The standard examination performed for the assessment of bone lesions has progressed from radiographic skeletal survey to the more advanced imaging modalities of computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography/computed tomography (PET/CT). The Durie-Salmon PLUS staging system (upgraded from the Durie-Salmon staging system) applies 2-[18F]-fluoro-2-deoxy-glucose (18F-FDG) PET/CT, and MRI findings to the staging of MM, and 18F-FDG PET/CT has been incorporated into the International Myeloma Working Group (IMWG) guidelines for the diagnosis and staging of MM. However, 18F-FDG PET/CT has significant limitations in the assessment of diffuse BM infiltration and in the differentiation of MM lesions from inflammatory or infectious lesions. The potential of several new PET tracers that exploit the underlying disease mechanism of MM has been evaluated in terms of improving the diagnosis. L-type amino acid transporter 1 (LAT1), a membrane protein that transports neutral amino acids, is associated with cell proliferation and has strong ability to represent the status of MM. This review evaluates the potential of amino acid and proliferation PET tracers for diagnosis and compares the characteristics and accuracy of non-FDG tracers in the management of patients with MM.
Collapse
Affiliation(s)
- Ryogo Minamimoto
- Division of Nuclear Medicine, Department of Radiology, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Chiaravalloti A, Cimini A, Ricci M, Quartuccio N, Arnone G, Filippi L, Calabria F, Leporace M, Bagnato A, Schillaci O. Positron emission tomography imaging in primary brain tumors. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00042-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
9
|
Krämer F, Gröner B, Hoffmann C, Craig A, Brugger M, Drzezga A, Timmer M, Neumaier F, Zlatopolskiy BD, Endepols H, Neumaier B. Evaluation of 3-l- and 3-d-[ 18F]Fluorophenylalanines as PET Tracers for Tumor Imaging. Cancers (Basel) 2021; 13:cancers13236030. [PMID: 34885141 PMCID: PMC8656747 DOI: 10.3390/cancers13236030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The early detection and treatment of malignant brain tumors can significantly improve the survival time and life quality of affected patients. Whereas positron emission tomography (PET) with O-(2-[18F]fluoroethyl)tyrosine ([18F]FET) offers improved diagnostic accuracy compared to other imaging methods, there is still a need for PET tracers with better tumor-specificity. A higher protein incorporation rate, as well as a higher affinity for the amino acid transporter LAT1, could provide probes with superior image quality compared to [18F]FET. The aim of the present study was a preclinical evaluation of the two enantiomeric phenylalanine (Phe) analogues, 3-l- and 3-d-[18F]fluorophenylalanine ([18F]FPhes), as possible alternatives to [18F]FET. Based on promising in vitro evaluation results, the radiolabeled amino acids were studied in vivo in two subcutaneous and one orthotopic rodent tumor xenograft models using µPET. The results show that 3-l- and 3-d-[18F]FPhe enable high-quality visualization of tumors with certain advantages over [18F]FET, making them promising candidates for further preclinical and clinical evaluations. Abstract Purpose: The preclinical evaluation of 3-l- and 3-d-[18F]FPhe in comparison to [18F]FET, an established tracer for tumor imaging. Methods: In vitro studies were conducted with MCF-7, PC-3, and U87 MG human tumor cell lines. In vivo µPET studies were conducted in healthy rats with/without the inhibition of peripheral aromatic l-amino acid decarboxylase by benserazide pretreatment (n = 3 each), in mice bearing subcutaneous MCF-7 or PC-3 tumor xenografts (n = 10), and in rats bearing orthotopic U87 MG tumor xenografts (n = 14). Tracer accumulation was quantified by SUVmax, SUVmean and tumor-to-brain ratios (TBrR). Results: The uptake of 3-l-[18F]FPhe in MCF-7 and PC-3 cells was significantly higher relative to [18F]FET. The uptake of all three tracers was significantly reduced by the suppression of amino acid transport systems L or ASC. 3-l-[18F]FPhe but not 3-d-[18F]FPhe exhibited protein incorporation. In benserazide-treated healthy rats, brain uptake after 42–120 min was significantly higher for 3-d-[18F]FPhe vs. 3-l-[18F]FPhe. [18F]FET showed significantly higher uptake into subcutaneous MCF-7 tumors (52–60 min p.i.), while early uptake into orthotopic U87 MG tumors was significantly higher for 3-l-[18F]FPhe (SUVmax: 3-l-[18F]FPhe, 107.6 ± 11.3; 3-d-[18F]FPhe, 86.0 ± 4.3; [18F]FET, 90.2 ± 7.7). Increased tumoral expression of LAT1 and ASCT2 was confirmed immunohistologically. Conclusion: Both novel tracers enable accurate tumor delineation with an imaging quality comparable to [18F]FET.
Collapse
Affiliation(s)
- Felicia Krämer
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
| | - Benedikt Gröner
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
| | - Chris Hoffmann
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
| | - Austin Craig
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
| | - Melanie Brugger
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
| | - Alexander Drzezga
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn-Cologne, Germany
- Molecular Organization of the Brain (INM-2), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany
| | - Marco Timmer
- Center for Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
| | - Felix Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
| | - Boris D. Zlatopolskiy
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
- Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| | - Heike Endepols
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
- Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
- Correspondence:
| |
Collapse
|
10
|
Miner MWG, Liljenbäck H, Virta J, Helin S, Eskola O, Elo P, Teuho J, Seppälä K, Oikonen V, Yang G, Kindler-Röhrborn A, Minn H, Li XG, Roivainen A. Comparison of: (2 S,4 R)-4-[ 18F]Fluoroglutamine, [ 11C]Methionine, and 2-Deoxy-2-[ 18F]Fluoro- D-Glucose and Two Small-Animal PET/CT Systems Imaging Rat Gliomas. Front Oncol 2021; 11:730358. [PMID: 34692505 PMCID: PMC8530378 DOI: 10.3389/fonc.2021.730358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/14/2021] [Indexed: 12/04/2022] Open
Abstract
Purpose The three positron emission tomography (PET) imaging compounds: (2S,4R)-4-[18F]Fluoroglutamine ([18F]FGln), L-[methyl-11C]Methionine ([11C]Met), and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) were investigated to contrast their ability to image orthotopic BT4C gliomas in BDIX rats. Two separate small animal imaging systems were compared for their tumor detection potential. Dynamic acquisition of [18F]FGln was evaluated with multiple pharmacokinetic models for future quantitative comparison. Procedures Up to four imaging studies were performed on each orthotopically grafted BT4C glioma-bearing BDIX rat subject (n = 16) on four consecutive days. First, a DOTAREM® contrast enhanced MRI followed by attenuation correction CT and dynamic PET imaging with each radiopharmaceutical (20 min [11C]Met, 60 min [18F]FDG, and 60 min [18F]FGln with either the Molecubes PET/CT (n = 5) or Inveon PET/CT cameras (n = 11). Ex vivo brain autoradiography was completed for each radiopharmaceutical and [18F]FGln pharmacokinetics were studied by injecting 40 MBq into healthy BDIX rats (n = 10) and collecting blood samples between 5 and 60 min. Erythrocyte uptake, plasma protein binding and plasma parent-fraction were combined to estimate the total blood bioavailability of [18F]FGln over time. The corrected PET-image blood data was then applied to multiple pharmacokinetic models. Results Average BT4C tumor-to-healthy brain tissue uptake ratios (TBR) for PET images reached maxima of: [18F]FGln TBR: 1.99 ± 0.19 (n = 13), [18F]FDG TBR: 1.41 ± 0.11 (n = 6), and [11C]Met TBR: 1.08 ± 0.08, (n = 12) for the dynamic PET images. Pharmacokinetic modeling in dynamic [18F]FGln studies suggested both reversible and irreversible uptake play a similar role. Imaging with Inveon and Molecubes yielded similar end-result ratios with insignificant differences (p > 0.25). Conclusions In orthotopic BT4C gliomas, [18F]FGln may offer improved imaging versus [11C]Met and [18F]FDG. No significant difference in normalized end-result data was found between the Inveon and Molecubes camera systems. Kinetic modelling of [18F]FGln uptake suggests that both reversible and irreversible uptake play an important role in BDIX rat pharmacokinetics.
Collapse
Affiliation(s)
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Jenni Virta
- Turku PET Centre, University of Turku, Turku, Finland
| | - Semi Helin
- Turku PET Centre, University of Turku, Turku, Finland
| | - Olli Eskola
- Turku PET Centre, University of Turku, Turku, Finland
| | - Petri Elo
- Turku PET Centre, University of Turku, Turku, Finland
| | - Jarmo Teuho
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Kerttu Seppälä
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Vesa Oikonen
- Turku PET Centre, University of Turku, Turku, Finland
| | - Guangli Yang
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Andrea Kindler-Röhrborn
- Institute of Pathology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Heikki Minn
- Turku PET Centre, University of Turku, Turku, Finland.,Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
| | - Xiang-Guo Li
- Turku PET Centre, University of Turku, Turku, Finland.,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland.,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| |
Collapse
|
11
|
Celli M, Caroli P, Amadori E, Arpa D, Gurrieri L, Ghigi G, Cenni P, Paganelli G, Matteucci F. Diagnostic and Prognostic Potential of 18F-FET PET in the Differential Diagnosis of Glioma Recurrence and Treatment-Induced Changes After Chemoradiation Therapy. Front Oncol 2021; 11:721821. [PMID: 34671551 PMCID: PMC8521061 DOI: 10.3389/fonc.2021.721821] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/09/2021] [Indexed: 11/24/2022] Open
Abstract
Background MRI-based differential diagnosis of glioma recurrence (GR) and treatment-induced changes (TICs) remain elusive in up to 30% of treated glioma patients. We aimed to determine 18F-FET PET diagnostic performance in this clinical scenario, its outcome dependency on established prognostic factors, optimal 18F-FET semi-quantitative thresholds, and whether 18F-FET parameters may instantly predict progression-free survival (PFS) and overall survival (OS). Methods We retrospectively analyzed 45 glioma patients treated with chemoradiation therapy (32 males; mean age: 51 years, glioma grade: n=26 WHO4; n=15 WHO3; n=4 WHO2) who underwent 18F-FET PET to resolve differential diagnosis of GR and TICs raised by MRI performed in the preceding 2 weeks and depicting any of the following changes in their radiation field: volumetric increase of contrast-enhancing lesions; new contrast-enhancing lesion; significant increase in T2/FLAIR non-enhancing lesion without reducing corticosteroids. 18F-FET PET outcome relied on evaluation of maximum tumor-to-brain ratio (TBRmax), time-to-peak (TTP), and time-activity curve pattern (TAC). Metabolic tumor volume (MTV) and total tumor metabolism (TTM) were calculated for prognostic purposes. Standard of reference was repeat MRI performed 4–6 weeks after the previous MRI. Non-parametric statistics tested 18F-FET-based parameters for dependency on established prognostic markers. ROC curve analysis determined optimal cutoff values for 18F-FET semi-quantitative parameters. 18F-FET parameters and prognostic factors were evaluated for PFS and OS by Kaplan-Meier, univariate, and multivariate analyses. Results 18F-FET PET sensitivity, specificity, positive predictive value, negative predictive value were 86.2, 81.3, 89.3, 76.5%, respectively; higher diagnostic accuracy was yielded in IDH-wild-type glioma patients compared to IDH-mutant glioma patients (sensitivity: 81.8 versus 88.9%; specificity: 80.8 versus 81.8%). KPS was the only prognostic factor differing according to 18F-FET PET outcome (negative versus positive). Optimal 18F-FET cutoff values for GR were TBRmax ≥ 2.1, SUVmax ≥ 3.5, and TTP ≤ 29 min. PFS differed based on 18F-FET outcome and related metrics and according to KPS; a different OS was observed according to KPS only. On multivariate analysis, 18F-FET PET outcome was the only significant PFS factor; KPS and age the only significant OS factors. Conclusion 18F-FET PET demonstrated good diagnostic performance. 18F-FET PET outcome and metrics were significantly predictive only for PFS.
Collapse
Affiliation(s)
- Monica Celli
- Diagnostic Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Paola Caroli
- Diagnostic Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Elena Amadori
- Radiology MRI Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Donatella Arpa
- Radiation Therapy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Lorena Gurrieri
- Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giulia Ghigi
- Radiation Therapy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Patrizia Cenni
- Radiology MRI Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Paganelli
- Diagnostic Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Federica Matteucci
- Diagnostic Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
12
|
Stegmayr C, Surges R, Choi CH, Burda N, Stoffels G, Filß C, Willuweit A, Neumaier B, Heinzel A, Shah NJ, Mottaghy FM, Langen KJ. Investigation of Cerebral O-(2-[ 18F]Fluoroethyl)-L-Tyrosine Uptake in Rat Epilepsy Models. Mol Imaging Biol 2021; 22:1255-1265. [PMID: 32409931 PMCID: PMC7497431 DOI: 10.1007/s11307-020-01503-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE A recent study reported on high, longer lasting and finally reversible cerebral uptake of O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) induced by epileptic activity. Therefore, we examined cerebral [18F]FET uptake in two chemically induced rat epilepsy models and in patients with focal epilepsy to further investigate whether this phenomenon represents a major pitfall in brain tumor diagnostics and whether [18F]FET may be a potential marker to localize epileptic foci. PROCEDURES Five rats underwent kainic acid titration to exhibit 3 to 3.5 h of class IV-V motor seizures (status epilepticus, SE). Rats underwent 4× [18F]FET PET and 4× MRI on the following 25 days. Six rats underwent kindling with pentylenetetrazol (PTZ) 3 to 8×/week over 10 weeks, and hence, seizures increased from class I to class IV. [18F]FET PET and MRI were performed regularly on days with and without seizures. Four rats served as healthy controls. Additionally, five patients with focal epilepsy underwent [18F]FET PET within 12 days after the last documented seizure. RESULTS No abnormalities in [18F]FET PET or MRI were detected in the kindling model. The SE model showed significantly decreased [18F]FET uptake 3 days after SE in all examined brain regions, and especially in the amygdala region, which normalized within 2 weeks. Corresponding signal alterations in T2-weighted MRI were noted in the amygdala and hippocampus, which recovered 24 days post-SE. No abnormality of cerebral [18F]FET uptake was noted in the epilepsy patients. CONCLUSIONS There was no evidence for increased cerebral [18F]FET uptake after epileptic seizures neither in the rat models nor in patients. The SE model even showed decreased [18F]FET uptake throughout the brain. We conclude that epileptic seizures per se do not cause a longer lasting increased [18F]FET accumulation and are unlikely to be a major cause of pitfall for brain tumor diagnostics.
Collapse
Affiliation(s)
- Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.
| | - Rainer Surges
- Department of Neurology, RWTH University Aachen, Aachen, Germany.,Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Chang-Hoon Choi
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Nicole Burda
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Christian Filß
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Alexander Heinzel
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany
| | - N Jon Shah
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Neurology, RWTH University Aachen, Aachen, Germany.,JARA - BRAIN - Translational Medicine, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany.,Centre of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany.,JARA - BRAIN - Translational Medicine, Aachen, Germany.,Centre of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
| |
Collapse
|
13
|
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
|
14
|
Galldiks N, Langen KJ, Albert NL, Chamberlain M, Soffietti R, Kim MM, Law I, Le Rhun E, Chang S, Schwarting J, Combs SE, Preusser M, Forsyth P, Pope W, Weller M, Tonn JC. PET imaging in patients with brain metastasis-report of the RANO/PET group. Neuro Oncol 2020; 21:585-595. [PMID: 30615138 DOI: 10.1093/neuonc/noz003] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/11/2018] [Accepted: 01/03/2019] [Indexed: 12/23/2022] Open
Abstract
Brain metastases (BM) from extracranial cancer are associated with significant morbidity and mortality. Effective local treatment options are stereotactic radiotherapy, including radiosurgery or fractionated external beam radiotherapy, and surgical resection. The use of systemic treatment for intracranial disease control also is improving. BM diagnosis, treatment planning, and follow-up is most often based on contrast-enhanced magnetic resonance imaging (MRI). However, anatomic imaging modalities including standard MRI have limitations in accurately characterizing posttherapeutic reactive changes and treatment response. Molecular imaging techniques such as positron emission tomography (PET) characterize specific metabolic and cellular features of metastases, potentially providing clinically relevant information supplementing anatomic MRI. Here, the Response Assessment in Neuro-Oncology working group provides recommendations for the use of PET imaging in the clinical management of patients with BM based on evidence from studies validated by histology and/or clinical outcome.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, University Hospital Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine 3, 4, Research Center Juelich, Juelich, Germany.,Center of Integrated Oncology, Universities of Cologne and Bonn, Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine 3, 4, Research Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig Maximilians-University of Munich, Munich, Germany
| | - Marc Chamberlain
- Departments of Neurology and Neurological Surgery, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington, USA
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Denmark
| | - Emilie Le Rhun
- Department of Neurosurgery, University Hospital Lille, Lille, France
| | - Susan Chang
- Department of Neurosurgery, University of California, San Francisco, California, USA
| | - Julian Schwarting
- Department of Neurosurgery, Ludwig Maximilians-University of Munich, Munich, Germany.,German Cancer Consortium, Partner Site Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Technical University Munich, Munich, Germany
| | - Matthias Preusser
- Department of Medicine I and Comprehensive Cancer Centre CNS Tumours Unit, Medical University of Vienna, Vienna, Austria
| | - Peter Forsyth
- Moffitt Cancer Center, University of South Florida, Tampa, Florida, USA
| | - Whitney Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California , USA
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Jörg C Tonn
- Department of Neurosurgery, Ludwig Maximilians-University of Munich, Munich, Germany.,German Cancer Consortium, Partner Site Munich, Germany
| |
Collapse
|
15
|
Oliveira D, Stegmayr C, Heinzel A, Ermert J, Neumaier B, Shah NJ, Mottaghy FM, Langen KJ, Willuweit A. High uptake of 68Ga-PSMA and 18F-DCFPyL in the peritumoral area of rat gliomas due to activated astrocytes. EJNMMI Res 2020; 10:55. [PMID: 32451793 PMCID: PMC7378136 DOI: 10.1186/s13550-020-00642-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Recent studies reported on high uptake of the PSMA ligands [68Ga]HBED-CC (68Ga-PSMA) and 18F-DCFPyL in cerebral gliomas. This study explores the regional uptake and cellular targets of 68Ga-PSMA and 18F-DCFPyL in three different rat glioma models. METHODS F98, 9 L, or U87 rat gliomas were implanted into the brains of 38 rats. After 13 days of tumor growth, 68Ga-PSMA (n = 21) or 18F-DCFPyL (n = 17) was injected intravenously, and animals were sacrificed 40 min later. Five animals for each tracer and tumor model were additionally investigated by micro-PET at 20-40 min post injection. Cryosections of the tumor bearing brains were analyzed by ex vivo autoradiography and immunofluorescence staining for blood vessels, microglia, astrocytes, and presence of PSMA. Blood-brain barrier (BBB) permeability was tested by coinjection of Evans blue dye (EBD). 68Ga-PSMA uptake after restoration of BBB integrity by treatment with dexamethasone (Dex) was evaluated in four animals with U87 gliomas. Competition experiments using the PSMA-receptor inhibitor 2-(phosphonomethyl)pentane-1,5-dioic acid (PMPA) were performed for both tracers in two animals each. RESULTS Autoradiography demonstrated a strong 68Ga-PSMA and 18F-DCFPyL binding in the peritumoral area and moderate binding in the center of the tumors. PMPA administration led to complete inhibition of 68Ga-PSMA and 18F-DCFPyL binding in the peritumoral region. Restoration of BBB by Dex treatment reduced EBD extravasation but 68Ga-PSMA binding remained unchanged. Expression of activated microglia (CD11b) was low in the intra- and peritumoral area but GFAP staining revealed strong activation of astrocytes in congruency to the tracer binding in the peritumoral area. All tumors were visualized in micro PET, showing a lower tumor/brain contrast with 68Ga-PSMA than with 18F-DCFPyL. CONCLUSIONS High uptake of 68Ga-PSMA and 18F-DCFPyL in the peritumoral area of all glioma models is presumably caused by activated astrocytes. This may represent a limitation for the clinical application of PSMA ligands in gliomas.
Collapse
Affiliation(s)
- Dennis Oliveira
- Institute of Neuroscience and Medicine (INM), Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Carina Stegmayr
- Institute of Neuroscience and Medicine (INM), Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Alexander Heinzel
- Department of Nuclear Medicine, RWTH Aachen University, Aachen, Germany
- Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain, Aachen, Germany
| | - Johannes Ermert
- Institute of Neuroscience and Medicine (INM), Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine (INM), Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - N Jon Shah
- Institute of Neuroscience and Medicine (INM), Forschungszentrum Jülich, D-52425, Jülich, Germany
- Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain, Aachen, Germany
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH Aachen University, Aachen, Germany
- Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM), Forschungszentrum Jülich, D-52425, Jülich, Germany.
- Department of Nuclear Medicine, RWTH Aachen University, Aachen, Germany.
- Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain, Aachen, Germany.
| | - Antje Willuweit
- Institute of Neuroscience and Medicine (INM), Forschungszentrum Jülich, D-52425, Jülich, Germany
| |
Collapse
|
16
|
Stegmayr C, Willuweit A, Lohmann P, Langen KJ. O-(2-[18F]-Fluoroethyl)-L-Tyrosine (FET) in Neurooncology: A Review of Experimental Results. Curr Radiopharm 2020; 12:201-210. [PMID: 30636621 DOI: 10.2174/1874471012666190111111046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 11/22/2022]
Abstract
In recent years, PET using radiolabelled amino acids has gained considerable interest as an additional tool besides MRI to improve the diagnosis of cerebral gliomas and brain metastases. A very successful tracer in this field is O-(2-[18F]fluoroethyl)-L-tyrosine (FET) which in recent years has replaced short-lived tracers such as [11C]-methyl-L-methionine in many neuro-oncological centers in Western Europe. FET can be produced with high efficiency and distributed in a satellite concept like 2- [18F]fluoro-2-deoxy-D-glucose. Many clinical studies have demonstrated that FET PET provides important diagnostic information regarding the delineation of cerebral gliomas for therapy planning, an improved differentiation of tumor recurrence from treatment-related changes and sensitive treatment monitoring. In parallel, a considerable number of experimental studies have investigated the uptake mechanisms of FET on the cellular level and the behavior of the tracer in various benign lesions in order to clarify the specificity of FET uptake for tumor tissue. Further studies have explored the effects of treatment related tissue alterations on tracer uptake such as surgery, radiation and drug therapy. Finally, the role of blood-brain barrier integrity for FET uptake which presents an important aspect for PET tracers targeting neoplastic lesions in the brain has been investigated in several studies. Based on a literature research regarding experimental FET studies and corresponding clinical applications this article summarizes the knowledge on the uptake behavior of FET, which has been collected in more than 30 experimental studies during the last two decades and discusses the role of these results in the clinical context.
Collapse
Affiliation(s)
- Carina Stegmayr
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany.,Department of Nuclear Medicine, University of Aachen, Aachen, Germany.,Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Juelich, Germany
| |
Collapse
|
17
|
Ali JS, Ain NU, Naz S, Zia M. Biomarker selection and imaging design in cancer: A link with biochemical pathways for imminent engineering. Heliyon 2020; 6:e03340. [PMID: 32055737 PMCID: PMC7005466 DOI: 10.1016/j.heliyon.2020.e03340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 05/27/2019] [Accepted: 01/29/2020] [Indexed: 01/15/2023] Open
Abstract
Malignant cells reprogram metabolic pathways to meet the demands of growth and proliferation. These altered manners of metabolism are now identified as hallmarks of cancer. Studies have revealed tumor cells alter specific pathways such as glycolysis, fatty acid synthesis and amino acid synthesis to support their proliferation. In this review, we provide a theoretical framework to understand metabolic reprogramming and the mechanisms accompanying distorted metabolism to tumor progression. How these alterations will be assisting in cancer diagnostics and advances in standard techniques in marker identification and imagining are also discussed.
Collapse
Affiliation(s)
| | | | | | - Muhammad Zia
- Department of Biotechnology, Quaid-i-Azam University Islamabad 45320 Pakistan
| |
Collapse
|
18
|
Lisova K, Chen BY, Wang J, Fong KMM, Clark PM, van Dam RM. Rapid, efficient, and economical synthesis of PET tracers in a droplet microreactor: application to O-(2-[ 18F]fluoroethyl)-L-tyrosine ([ 18F]FET). EJNMMI Radiopharm Chem 2019; 5:1. [PMID: 31893318 PMCID: PMC6938530 DOI: 10.1186/s41181-019-0082-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/21/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Conventional scale production of small batches of PET tracers (e.g. for preclinical imaging) is an inefficient use of resources. Using O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET), we demonstrate that simple microvolume radiosynthesis techniques can improve the efficiency of production by consuming tiny amounts of precursor, and maintaining high molar activity of the tracers even with low starting activity. PROCEDURES The synthesis was carried out in microvolume droplets manipulated on a disposable patterned silicon "chip" affixed to a heater. A droplet of [18F]fluoride containing TBAHCO3 was first deposited onto a chip and dried at 100 °C. Subsequently, a droplet containing 60 nmol of precursor was added to the chip and the fluorination reaction was performed at 90 °C for 5 min. Removal of protecting groups was accomplished with a droplet of HCl heated at 90 °C for 3 min. Finally, the crude product was collected in a methanol-water mixture, purified via analytical-scale radio-HPLC and formulated in saline. As a demonstration, using [18F]FET produced on the chip, we prepared aliquots with different molar activities to explore the impact on preclinical PET imaging of tumor-bearing mice. RESULTS The microdroplet synthesis exhibited an overall decay-corrected radiochemical yield of 55 ± 7% (n = 4) after purification and formulation. When automated, the synthesis could be completed in 35 min. Starting with < 370 MBq of activity, ~ 150 MBq of [18F]FET could be produced, sufficient for multiple in vivo experiments, with high molar activities (48-119 GBq/μmol). The demonstration imaging study revealed the uptake of [18F]FET in subcutaneous tumors, but no significant differences in tumor uptake as a result of molar activity differences (ranging 0.37-48 GBq/μmol) were observed. CONCLUSIONS A microdroplet synthesis of [18F]FET was developed demonstrating low reagent consumption, high yield, and high molar activity. The approach can be expanded to tracers other than [18F]FET, and adapted to produce higher quantities of the tracer sufficient for clinical PET imaging.
Collapse
Affiliation(s)
- Ksenia Lisova
- Physics in Biology and Medicine Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bao Ying Chen
- Physics in Biology and Medicine Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jia Wang
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kelly Mun-Ming Fong
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peter M Clark
- Physics in Biology and Medicine Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - R Michael van Dam
- Physics in Biology and Medicine Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA, USA.
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Chiaravalloti A, Filippi L, Ricci M, Cimini A, Schillaci O. Molecular Imaging in Pediatric Brain Tumors. Cancers (Basel) 2019; 11:cancers11121853. [PMID: 31771237 PMCID: PMC6966547 DOI: 10.3390/cancers11121853] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 02/07/2023] Open
Abstract
In the last decade, several radiopharmaceuticals have been developed and investigated for imaging in vivo of pediatric brain tumors with the aim of exploring peculiar metabolic processes as glucose consumption, amino-acid metabolism, and protein synthesis with nuclear medicine techniques. Although the clinical shreds of evidence are limited, preliminary results are encouraging. In this review, we performed web-based and desktop research summarizing the most relevant findings of the literature published to date on this topic. Particular attention was given to the wide spectrum of nuclear medicine advances and trends in pediatric neurooncology and neurosurgery. Furthermore, the role of somatostatin receptor imaging through single-photon emission computed tomography (SPECT) and positron emission tomography (PET) probes, with reference to their potential therapeutic implications, was examined in the peculiar context. Preliminary results show that functional imaging in pediatric brain tumors might lead to significant improvements in terms of diagnostic accuracy and it could be of help in the management of the disease.
Collapse
Affiliation(s)
- Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy (O.S.)
- Nuclear Medicine Section, IRCCS Neuromed, 86077 Pozzilli, Italy
- Correspondence: or ; Tel.: +39-062-090-2457
| | - Luca Filippi
- Nuclear Medicine Section, “Santa Maria Goretti” Hospital, 04100 Latina, Italy;
| | - Maria Ricci
- Department of Radiological, Oncological and Pathological Sciences, Faculty of Medicine and Surgery, La Sapienza University, 00161 Rome, Italy;
| | - Andrea Cimini
- Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy (O.S.)
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy (O.S.)
- Nuclear Medicine Section, IRCCS Neuromed, 86077 Pozzilli, Italy
| |
Collapse
|
20
|
Bogni A, Laera L, Cucchi C, Iwata R, Seregni E, Pascali C. An improved automated one-pot synthesis of O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) based on a purification by cartridges. Nucl Med Biol 2019; 72-73:11-19. [DOI: 10.1016/j.nucmedbio.2019.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/09/2019] [Accepted: 05/14/2019] [Indexed: 10/26/2022]
|
21
|
Verhoeven J, Hulpia F, Kersemans K, Bolcaen J, De Lombaerde S, Goeman J, Descamps B, Hallaert G, Van den Broecke C, Deblaere K, Vanhove C, Van der Eycken J, Van Calenbergh S, Goethals I, De Vos F. New fluoroethyl phenylalanine analogues as potential LAT1-targeting PET tracers for glioblastoma. Sci Rep 2019; 9:2878. [PMID: 30814660 PMCID: PMC6393465 DOI: 10.1038/s41598-019-40013-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/07/2019] [Indexed: 02/07/2023] Open
Abstract
The use of O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) as a positron emission tomography (PET) tracer for brain tumor imaging might have some limitations because of the relatively low affinity for the L-type amino acid transporter 1 (LAT1). To assess the stereospecificity and evaluate the influence of aromatic ring modification of phenylalanine LAT1 targeting tracers, six different fluoroalkylated phenylalanine analogues were synthesized. After in vitro Ki determination, the most promising compound, 2-[18F]-2-fluoroethyl-L-phenylalanine (2-[18F]FELP), was selected for further evaluation and in vitro comparison with [18F]FET. Subsequently, 2-[18F]FELP was assessed in vivo and compared with [18F]FET and [18F]FDG in a F98 glioblastoma rat model. 2-[18F]FELP showed improved in vitro characteristics over [18F]FET, especially when the affinity and specificity for system L is concerned. Based on our results, 2-[18F]FELP is a promising new PET tracer for brain tumor imaging.
Collapse
Affiliation(s)
| | - Fabian Hulpia
- Laboratory for Medicinal Chemistry, Ghent University, Ghent, Belgium
| | - Ken Kersemans
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | - Julie Bolcaen
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | | | - Jan Goeman
- Laboratory for Organic and Bio-organic synthesis, Ghent University, Ghent, Belgium
| | - Benedicte Descamps
- IBiTech-MEDISIP Ghent University, Department of Electronics and Information Systems, Ghent, Belgium
| | - Giorgio Hallaert
- Ghent University Hospital, Department of Neurosurgery, Ghent, Belgium
| | | | - Karel Deblaere
- Ghent University Hospital, Department of Radiology and Medical Imaging, Ghent, Belgium
| | - Christian Vanhove
- IBiTech-MEDISIP Ghent University, Department of Electronics and Information Systems, Ghent, Belgium
| | - Johan Van der Eycken
- Laboratory for Organic and Bio-organic synthesis, Ghent University, Ghent, Belgium
| | | | - Ingeborg Goethals
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | - Filip De Vos
- Laboratory of Radiopharmacy, Ghent University, Ghent, Belgium
| |
Collapse
|
22
|
Voxel-wise analysis of dynamic 18F-FET PET: a novel approach for non-invasive glioma characterisation. EJNMMI Res 2018; 8:91. [PMID: 30203138 PMCID: PMC6131687 DOI: 10.1186/s13550-018-0444-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/26/2018] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Glioma grading with dynamic 18F-FET PET (0-40 min p.i.) is typically performed by analysing the mean time-activity curve of the entire tumour or a suspicious area within a heterogeneous tumour. This work aimed to ensure a reader-independent glioma characterisation and identification of aggressive sub-volumes by performing a voxel-based analysis with diagnostically relevant kinetic and static 18F-FET PET parameters. One hundred sixty-two patients with a newly diagnosed glioma classified according to histologic and molecular genetic properties were evaluated. The biological tumour volume (BTV) was segmented in static 20-40 min p.i. 18F-FET PET images using the established threshold of 1.6 × background activity. For each enclosed voxel, the time-to-peak (TTP), the late slope (Slope15-40), and the tumour-to-background ratios (TBR5-15, TBR20-40) obtained from 5 to 15 min p.i. and 20 to 40 min p.i. images were determined. The percentage portion of these values within the BTV was evaluated with percentage volume fractions (PVFs) and cumulated percentage volume histograms (PVHs). The ability to differentiate histologic and molecular genetic classes was assessed and compared to volume-of-interest (VOI)-based parameters. RESULTS Aggressive WHO grades III and IV and IDH-wildtype gliomas were dominated by a high proportion of voxels with an early peak, negative slope, and high TBR, whereby the PVHs with TTP < 20 min p.i., Slope15-40 < 0 SUV/h, and TBR5-15 and TBR20-40 > 2 yielded the most significant differences between glioma grades. We found significant differences of the parameters between WHO grades and IDH mutation status, where the effect size was predominantly higher for voxel-based PVHs compared to the corresponding VOI-based parameters. A low overlap of BTV sub-volumes defined by TTP < 20 min p.i. and negative Slope15-40 with TBR5-15 > 2- and TBR20-40 > 2-defined hotspots was observed. CONCLUSIONS The presented approach applying voxel-wise analysis of dynamic 18F-FET PET enables an enhanced characterisation of gliomas and might potentially provide a fast identification of aggressive sub-volumes within the BTV. Parametric 3D 18F-FET PET information as investigated in this study has the potential to guide individual therapy instrumentation and may be included in future biopsy studies.
Collapse
|
23
|
Wu Z, Ma J, Brownell AL, Wang H, Li C, Meng X, Yuan L, Liu H, Li S, Xie J. Synthesis and evaluation of an N-[ 18F]fluorodeoxyglycosyl amino acid for PET imaging of tumor metabolism. Nucl Med Biol 2018; 66:40-48. [PMID: 30248568 DOI: 10.1016/j.nucmedbio.2018.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/19/2018] [Accepted: 08/07/2018] [Indexed: 11/18/2022]
Abstract
INTRODUCTION The limitations of [18F]fluorodeoxyglucose ([18F]FDG), including producing false-positive or -negative results, low image contrast in brain tumor diagnosis and poor differentiation of tumor and inflammatory, necessitate the development of new radiopharmaceuticals. In the present study, a novel [18F]fluoroglycoconjugate tracer, [18F]FDGly-NH-Phe, for tumor metabolism imaging was prepared and evaluated. METHODS [18F]FDGly-NH-Phe was prepared by condensing [18F]FDG with L-4-aminophenylalanine in an acidic condition, and purified with semi-preparative-high performance liquid chromatography (HPLC). The in vitro stability study was conducted in phosphate-buffered saline (PBS, pH 4.0-9.18) at room temperature (RT) and in fetal bovine serum (FBS) at 37 °C. The preliminary cellular uptake studies were performed using Hep-2 cell. The bio-distribution studies, PET/CT imaging and metabolism studies were performed and compared with [18F]FDG on ICR or BALB/c nude model mice. RESULTS [18F]FDGly-NH-Phe was derived from a direct condensation of [18F]FDG with L-4-aminophenylalanine with high stability in FBS and PBS (pH of 6.5-9.18). In vitro cell experiments showed that [18F]FDGly-NH-Phe uptake in Hep-2 cells was primarily transported through amino acid transporters including Na+-dependent A system, ASC system, and system B0,+ system. The bio-distribution of [18F]FDGly-NH-Phe in normal ICR mice showed faster blood radioactivity clearance, and lower uptake in brain and heart than [18F]FDG. The performance of PET/CT imaging for [18F]FDGly-NH-Phe in the mice model manifested excellent tumor visualization, high tumor-to-background ratios, and low accumulation in inflammatory lesions. Metabolism studies for [18F]FDGly-NH-Phe indicated high in vivo stability in plasma and urine and decomposition into [18F]FDG in the tumor microenvironment. CONCLUSION The results demonstrated that [18F]FDGly-NH-Phe as a novel amino acid PET tracer showed the capability to differentiate tumor from inflammation, and the potentials for future clinical applications.
Collapse
Affiliation(s)
- Zhifang Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Department of Radiology, Massachusetts General Hospital, Boston, USA; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Jingxin Ma
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | | | - Hongliang Wang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Chaomin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xiaxia Meng
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Ling Yuan
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Haiyan Liu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Jun Xie
- Shanxi Medical University, Taiyuan, People's Republic of China.
| |
Collapse
|
24
|
Prakash S, Hazari PP, Meena VK, Mishra AK. Radiolabeling and Preclinical Evaluation of a New S-Alkylated Cysteine Derivative Conjugated to C-Substituted Macrocycle for Positron Emission Tomography. ACS OMEGA 2018; 3:6497-6505. [PMID: 30023950 PMCID: PMC6045381 DOI: 10.1021/acsomega.8b00059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 06/05/2018] [Indexed: 06/08/2023]
Abstract
A new S-alkylated cysteine-derivatized tumor targeting agent, 2,2'-(12-(2-((2-acetamido-2-carboxyethyl)thio)acetamido)-11,13-dioxo-1,4,7,10-tetraazacyclotridecane-4,7-diyl)diacetic acid was developed for positron emission tomography (PET) imaging. N-Acetyl cysteine (NAC) was conjugated to ATRIDAT as a specific targeting agent toward L-type and ASC amino acid transporter systems in the oncogenic cells. NAC was attached via S-alkylation to prevent its incorporation at undesired recognition sites affecting the signal-to-noise ratio. NAC-ATRIDAT was subjected to gallium-68 complexation with >75% radiolabeling yield. The radiocomplex was purified through the tc18 cartridge to obtain 99.89% radiochemical yield. IC-50 of the NAC-ATRIDAT conjugate was 0.8 mM in A549 cells as evaluated through 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazonium bromide assay. Binding affinity experiments on A549 cells showed noteworthy binding with KD in the nanomolar range. A time course study showed a Km value of 0.19 μM and Vmax value of 0.49 pmol/μg protein/min showing reasonable tumor kinetics. Efflux studies showed that the synthesized radioligand is transported majorly by LAT followed by the ASC system. Clearance was found to be renal with 7.67 ± 1.48% ID/g uptake at 30 min which substantially declined to 0.52 ± 0.% ID/g at 4 h. A significant uptake of 10.06 ± 1.056% ID/g was observed at the tumor site in mice at 1 h. μPET images revealed a high contrast with a tumor-to-kidney ratio of 4.8 and a tumor-to-liver ratio of 35.85 at 1 h after injection. These preclinical in vitro and in vivo evaluation supports its potential on the way of becoming a successful 68Ga-radiolabeled amino acid-based PET imaging agent.
Collapse
|
25
|
Radiological evaluation of response to immunotherapy in brain tumors: Where are we now and where are we going? Crit Rev Oncol Hematol 2018; 126:135-144. [PMID: 29759556 DOI: 10.1016/j.critrevonc.2018.03.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/14/2018] [Accepted: 03/29/2018] [Indexed: 11/21/2022] Open
|
26
|
Hayes AR, Jayamanne D, Hsiao E, Schembri GP, Bailey DL, Roach PJ, Khasraw M, Newey A, Wheeler HR, Back M. Utilizing 18F-fluoroethyltyrosine (FET) positron emission tomography (PET) to define suspected nonenhancing tumor for radiation therapy planning of glioblastoma. Pract Radiat Oncol 2018; 8:230-238. [PMID: 29730279 DOI: 10.1016/j.prro.2018.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/25/2018] [Indexed: 11/15/2022]
Abstract
AIM The authors sought to evaluate the impact of 18F-fluoroethyltyrosine (FET) positron emission tomography (PET) on radiation therapy planning for patients diagnosed with glioblastoma (GBM) and the presence of suspected nonenhancing tumors compared with standard magnetic resonance imaging (MRI). METHODS AND MATERIALS Patients with GBM and contrast-enhanced MRI scans showing regions suspicious of nonenhancing tumor underwent postoperative FET-PET before commencing radiation therapy. Two clinical target volumes (CTVs) were created using pre- and postoperative MRI: MRI fluid-attenuated inversion recovery (FLAIR) sequences (CTVFLAIR) and MRI contrast sequences with an expansion on the surgical cavity (CTVSx). FET-PET was used to create biological tumor volumes (BTVs) by encompassing FET-avid regions, forming BTVFLAIR and BTVSx. Volumetric analyses were conducted between CTVs and respective BTVs using Wilcoxon signed-rank tests. The volume increase with addition of FET was analyzed with respect to BTVFLAIR and BTVSx. Presence of focal gadolinium contrast enhancement within previously nonenhancing tumor or within the FET-avid region was noted on MRI scans at 1 and 3 months after radiation therapy. RESULTS Twenty-six patients were identified retrospectively from our database, of whom 24 had demonstrable FET uptake. The median CTVFLAIR, CTVSx, BTVFLAIR, and BTVSx were 57.1 mL (range, 1.1-217.4), 83.6 mL (range, 27.2-275.8), 62.8 mL (range, 1.1-307.3), and 94.7 mL (range, 27.2-285.5), respectively. When FET-PET was used, there was a mean increase in volume of 26.8% from CTVFLAIR to BTVFLAIR and 20.6% from CTVSx to BTVSx. A statistically significant difference was noted on Wilcoxon signed-rank test when assessing volumetric change between CTVFLAIR and BTVFLAIR (P < .0001) and CTVSx and BTVSx (P < .0001). Six of 24 patients (25%) with FET avidity before radiation therapy showed focal gadolinium enhancement within the radiation therapy portal. CONCLUSIONS FET-PET may help improve delineation of GBM in cases with a suspected nonenhancing component. This may result in improved radiation therapy target delineation and reduce the risk of potential geographical miss. SUMMARY We investigated the impact of 18F-fluoroethyltyrosine (FET) positron emission tomography (PET) on radiation therapy planning for patients diagnosed with glioblastoma (GBM) and a suspected nonenhancing tumor compared with standard magnetic resonance imaging. We performed volumetric analyses between clinical target volumes and respective biological target volumes using Wilcoxon signed-rank tests. FET-PET may help improve delineation of GBM in cases with a suspected nonenhancing component and reduce the risk of potential geographical miss.
Collapse
Affiliation(s)
- Aimee R Hayes
- Department of Nuclear Medicine, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Vital, Northern Translational Cancer Research Centre, St Leonards, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia.
| | - Dasantha Jayamanne
- Department of Radiation Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Edward Hsiao
- Department of Nuclear Medicine, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Geoffrey P Schembri
- Department of Nuclear Medicine, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Dale L Bailey
- Department of Nuclear Medicine, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Vital, Northern Translational Cancer Research Centre, St Leonards, NSW, Australia; Faculty of Health Sciences, Cumberland Campus, The University of Sydney, Lidcombe, NSW, Australia
| | - Paul J Roach
- Department of Nuclear Medicine, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Mustafa Khasraw
- Sydney Vital, Northern Translational Cancer Research Centre, St Leonards, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Sydney Neuro-Oncology Group, North Shore Private Hospital, St Leonards, NSW, Australia
| | - Allison Newey
- Department of Radiology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Helen R Wheeler
- Sydney Vital, Northern Translational Cancer Research Centre, St Leonards, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Sydney Neuro-Oncology Group, North Shore Private Hospital, St Leonards, NSW, Australia
| | - Michael Back
- Sydney Vital, Northern Translational Cancer Research Centre, St Leonards, NSW, Australia; Department of Radiation Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Sydney Neuro-Oncology Group, North Shore Private Hospital, St Leonards, NSW, Australia
| |
Collapse
|
27
|
Pronin AI, Dolgushin MB, Lyuosev AS, Odzharova AA, Nevzorov DI, Nechipay EA, Gasparyan TG. [Capabilities of 18F-FET PET/CT in a patient with brain glioma (a case report and literature review)]. ZHURNAL VOPROSY NEIROKHIRURGII IMENI N. N. BURDENKO 2018; 82:95-99. [PMID: 29795092 DOI: 10.17116/oftalma201882295-99] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Positron emission tomography combined with computed tomography (PET/CT) enables assessment of not only anatomical and structural but also metabolic changes in tumor mass. 18F-fluoroethyl tyrosine (18F-FET) PET/CT is based on evaluation of transport of 18F-labeled tyrosine in tissues. We present a clinical case of a patient with a newly diagnosed brain tumor, demonstrating the capabilities of 18F-FET PET/CT in assessing the reliable volume and degree of tumor anaplasia, which is important when choosing the treatment approach for a patient.
Collapse
Affiliation(s)
- A I Pronin
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - M B Dolgushin
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - A S Lyuosev
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - A A Odzharova
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - D I Nevzorov
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - E A Nechipay
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - T G Gasparyan
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| |
Collapse
|
28
|
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
|
29
|
Tang C, Nie D, Tang G, Gao S, Liu S, Wen F, Tang X. Radiosynthesis and biological evaluation of N-(2-[ 18F]fluoropropionyl)-3,4-dihydroxy-l-phenylalanine as a PET tracer for oncologic imaging. Nucl Med Biol 2017; 50:39-46. [PMID: 28456080 DOI: 10.1016/j.nucmedbio.2017.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/22/2017] [Accepted: 04/05/2017] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Several 11C and 18F labeled 3,4-dihydroxy-l-phenylalanine (l-DOPA) analogues have been used for neurologic and oncologic diseases, especially for brain tumors and neuroendocrine tumors PET imaging. However, 18F-labeled N-substituted l-DOPA analogues have not been reported so far. In the current study, radiosynthesis and biological evaluation of a new 18F-labeled l-DOPA analogue, N-(2-[18F]fluoropropionyl)-3,4-dihydroxy-l-phenylalanine ([18F]FPDOPA) for tumor PET imaging are performed. METHODS The synthesis of [18F]FPDOPA was via a two-step reaction sequence from 4-nitrophenyl-2-[18F]fluoropropionate ([18F]NFP). The biodistribution of [18F]FPDOPA was determined in normal Kunming mice. In vitro competitive inhibition and protein incorporation experiments were performed with SPC-A-1 lung adenocarcinoma cell lines. PET/CT studies of [18F]FPDOPA were conducted in C6 rat glioma and SPC-A-1 human lung adenocarcinoma and H460 human large cell lung cancer-bearing nude mice. RESULTS [18F]FPDOPA was prepared with a decay-corrected radiochemical yield of 28±5% and a specific activity of 50±15GBq/μmol (n=10) within 125min. In vitro cell experiments showed that [18F]FPDOPA uptake in SPC-A-1 cells was primarily transported through Na+-independent system L, with Na+-dependent system B0,+ and system ASC partly involved in it. Biodistribution data in mice showed that renal-bladder route was the main excretory system of [18F]FPDOPA. PET imaging demonstrated intense accumulation of [18F]FPDOPA in several tumor xenografts, with (8.50±0.40)%ID/g in C6 glioma, (6.30±0.12)%ID/g in SPC-A-1 lung adenocarcinoma, and (6.50±0.10)%ID/g in H460 large cell lung cancer, respectively. CONCLUSION A novel N-substituted 18F-labeled L-DOPA analogue [18F]FPDOPA is synthesized and evaluated in vitro and in vivo. The results support that [18F]FPDOPA seems to be a potential PET tracer for tumor imaging, especially be a better potential PET tracer than [18F]fluoro-2-deoxy-d-glucose ([18F]FDG) for brain tumor imaging.
Collapse
Affiliation(s)
- Caihua Tang
- Guangdong Engineering Research Center for Medical Radiopharmaceuticals Translational Application PET-CT Center and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Dahong Nie
- Guangdong Engineering Research Center for Medical Radiopharmaceuticals Translational Application PET-CT Center and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ganghua Tang
- Guangdong Engineering Research Center for Medical Radiopharmaceuticals Translational Application PET-CT Center and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Siyuan Gao
- Guangdong Engineering Research Center for Medical Radiopharmaceuticals Translational Application PET-CT Center and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Shaoyu Liu
- Guangdong Engineering Research Center for Medical Radiopharmaceuticals Translational Application PET-CT Center and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Fuhua Wen
- Guangdong Engineering Research Center for Medical Radiopharmaceuticals Translational Application PET-CT Center and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiaolan Tang
- College of Materials and Energy, Southern China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
30
|
Tscherpel C, Dunkl V, Ceccon G, Stoffels G, Judov N, Rapp M, Meyer PT, Kops ER, Ermert J, Fink GR, Shah NJ, Langen KJ, Galldiks N. The use of O-(2-18F-fluoroethyl)-L-tyrosine PET in the diagnosis of gliomas located in the brainstem and spinal cord. Neuro Oncol 2017; 19:710-718. [PMID: 28039366 DOI: 10.1093/neuonc/now243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background Despite an increasing number of O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET) PET studies in supratentorial gliomas, studies regarding the usefulness of 18F-FET PET in brainstem and spinal cord gliomas to date remain scarce. Methods Thirty-six 18F-FET PET scans were performed in 29 patients with brainstem (n = 29 scans) or spinal cord glioma (n = 7 scans). In 32 of 36 PET scans, a dynamic acquisition was performed. Fifteen scans in 15 patients were performed to assess newly diagnosed lesions, and 21 scans were obtained during follow-up: for diagnosing tumor progression (n = 15 scans in 14 patients) as well as for treatment monitoring (n = 6 scans in 3 patients). Four patients underwent additional serial scans (range, 1-2), and 3 of these 4 patients were examined for more than one indication. Maximum and mean tumor/brain ratios (TBRmax/mean) of 18F-FET uptake (20-40 min post injection) as well as kinetic 18F-FET uptake parameters were determined. Final diagnoses were confirmed histologically (54%) or by clinical follow-up (46%). Results In all newly diagnosed high-grade (n = 3 patients) and in 5 of 11 patients with low-grade gliomas, 18F-FET uptake was increased (TBRmax ≥2.5 and/or TBRmean ≥1.9). In 2 patients with newly diagnosed gliomas without MR contrast enhancement, 18F-FET PET nevertheless showed increased metabolism. At suspected progression, the combination of TBRs with kinetic 18F-FET parameters correctly identified presence or absence of progressive disease in 9 of 11 patients (82%). Conclusions This preliminary study suggests that 18F-FET PET adds valuable diagnostic information in brainstem and spinal cord glioma, particularly when the diagnostic information derived from MRI is equivocal.
Collapse
Affiliation(s)
- Caroline Tscherpel
- Department of Neurology, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Veronika Dunkl
- Department of Neurology, University of Cologne, Cologne, Germany
| | - Garry Ceccon
- Department of Neurology, University of Cologne, Cologne, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Natalie Judov
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Marion Rapp
- Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Elena Rota Kops
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Johannes Ermert
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Gereon R Fink
- Department of Neurology, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany.,Departments of Neurology, University of Aachen, Aachen, Germany.,Section JARA-Brain, Jülich-Aachen Research Alliance (JARA), Jülich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany.,Section JARA-Brain, Jülich-Aachen Research Alliance (JARA), Jülich, Germany.,Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Norbert Galldiks
- Department of Neurology, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany.,Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany
| |
Collapse
|
31
|
Yano H, Shinoda J, Iwama T. Clinical Utility of Positron Emission Tomography in Patients with Malignant Glioma. Neurol Med Chir (Tokyo) 2017; 57:312-320. [PMID: 28458384 PMCID: PMC5566704 DOI: 10.2176/nmc.ra.2016-0312] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Positron emission tomography (PET) is being increasingly utilized for the management of brain tumors. Herein, we primarily review our previous studies on the use of PET in glioma that utilize three types of tracers: 11C-methionine (MET), 11C-choline, and 18F-fluorodeoxyglucose. These studies included aspects such as tumor behavior, diagnosis, grade of malignancy, spread and invasion, viability, and genetic deletions; moreover, they also evaluated PET as a tool for planning radiation therapy (RT) and determining its outcome. MET-PET in particular is considered to be the most informative for diagnosis and therapeutic decision-making for glioma patients; it is therefore considered crucial for brain tumor therapy. MET-PET is expected to be widely used for brain tumor patients going forward.
Collapse
Affiliation(s)
- Hirohito Yano
- Department of Neurosurgery, Gifu University Graduate School of Medicine
| | - Jun Shinoda
- Chubu Medical Center for Prolonged Traumatic Brain Dysfunction, Department of Neurosurgery, Kizawa Memorial Hospital
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine
| |
Collapse
|
32
|
Stegmayr C, Oliveira D, Niemietz N, Willuweit A, Lohmann P, Galldiks N, Shah NJ, Ermert J, Langen KJ. Influence of Bevacizumab on Blood-Brain Barrier Permeability and O-(2- 18F-Fluoroethyl)-l-Tyrosine Uptake in Rat Gliomas. J Nucl Med 2017; 58:700-705. [PMID: 28153956 DOI: 10.2967/jnumed.116.187047] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/26/2016] [Indexed: 01/20/2023] Open
Abstract
Restoration of the blood-brain barrier (BBB) after antiangiogenic therapy of gliomas with bevacizumab may result in a decrease in contrast enhancement on MRI despite tumor progression. This so-called pseudoresponse is difficult to differentiate from a true tumor response with conventional MRI. Initial patient studies have indicated that PET using O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) may be helpful for solving this diagnostic problem. This study was performed to investigate the effects of bevacizumab on BBB permeability and 18F-FET uptake in a human xenograft model. Methods: Human U87 glioblastoma cells were implanted into the striatum of immunodeficient RNU rats. 18F-FET PET scans and ex vivo autoradiography were performed in animals receiving a single high dose of bevacizumab (45 mg/kg 2 d before PET; n = 9) or in animals receiving 2 lower doses (10 mg/kg 9 and 2 d before PET; n = 10) to evaluate short-term and long-term effects on the BBB, respectively, and in control animals without bevacizumab treatment (n = 8). Time-activity curves, slope, and tumor-to-brain ratios of 18F-FET uptake (18-61 min after injection) were evaluated using a volume-of-interest analysis. After PET scanning, Evans blue dye (EBD) was injected into animals, and cryosections of the brains were evaluated by autoradiography, by histology, and for EBD fluorescence to assess BBB permeability. Results: Compared with the control, short-term bevacizumab therapy resulted in a trend toward BBB restoration (P = 0.055) and long-term therapy resulted in a significant decrease (P = 0.004) in BBB permeability, as assessed by EBD fluorescence. In contrast, no significant differences in tumor-to-brain ratios or slope of 18F-FET uptake were observed in PET and autoradiography (P > 0.05). Conclusion:8F-FET uptake in glioblastomas seems to be largely independent of BBB permeability and reflects the viability of tumor tissue during antiangiogenic therapy more reliably than contrast-enhanced MRI.
Collapse
Affiliation(s)
- Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Dennis Oliveira
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Nicole Niemietz
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany
| | - N Jon Shah
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, RWTH University, Aachen, Germany.,Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain, RWTH Aachen University, Aachen, Germany; and
| | - Johannes Ermert
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany.,Department of Nuclear Medicine, RWTH University Hospital, Aachen, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany .,Department of Nuclear Medicine, RWTH University Hospital, Aachen, Germany
| |
Collapse
|
33
|
Kagawa S, Nishii R, Higashi T, Yamauchi H, Ogawa E, Okudaira H, Kobayashi M, Yoshimoto M, Shikano N, Kawai K. Relationship between [ 14C]MeAIB uptake and amino acid transporter family gene expression levels or proliferative activity in a pilot study in human carcinoma cells: Comparison with [ 3H]methionine uptake. Nucl Med Biol 2017; 49:8-15. [PMID: 28284101 DOI: 10.1016/j.nucmedbio.2017.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/18/2017] [Accepted: 01/18/2017] [Indexed: 01/09/2023]
Abstract
INTRODUCTION To clarify the difference between system A and L amino acid transport imaging in PET clinical imaging, we focused on the use of α-[N-methyl-11C]-methylaminoisobutyric acid ([11C]MeAIB), and compared it with [S-methyl-11C]-L-methionine ([11C]MET). The aim of this study was to assess the correlation of accumulation of these two radioactive amino acid analogs with expression of amino acid transporters and cell proliferative activity in carcinoma cells. METHODS Amino acid uptake inhibitor studies were performed in four human carcinoma cells (epidermal carcinoma A431, colorectal carcinoma LS180, and lung carcinomas PC14/GL and H441/GL) using the radioisotope analogs [3H]MET and [14C]MeAIB. MeAIB was used to inhibit the A system and 2-amino-2-norbornane-carboxylic acid (BCH) was used to inhibit the L system. The carcinoma gene expression levels of a number of amino acid transporters were measured by microarray and quantitative polymerase chain reaction. Carcinoma proliferative activity was assessed using accumulation of [methyl-3H]-3'-deoxy-3'-fluorothymidine ([3H]FLT). RESULTS AND CONCLUSION [14C]MeAIB uptake occurred principally via a Na+-dependent A type mechanism whereas [3H]MET uptake occurred predominantly via a Na+-independent L type mechanism although other transporters were also utilized depending on cell type. There was no correlation between [3H]MET uptake and total system L amino acid transporter (LAT) expression. In contrast, [14C]MeAIB uptake strongly correlated with total system A amino acid transporter (SNAT) expression and proliferative activity in this preliminary study using four human carcinoma cell lines. Carcinoma proliferative activity also correlated with total SNAT expression. Advances in Knowledge and Implications for Patient Care: Because there is a significant correlation between the accumulation of [14C]MeAIB and the gene expression level of total SNAT as well as the accumulation of [3H]FLT, it is suggested that use of the analog [11C]MeAIB in PET may provide an indication of tumor cell proliferative activity. [11C]MeAIB is therefore expected to be very useful in PET imaging.
Collapse
Affiliation(s)
- Shinya Kagawa
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan; Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Ryuichi Nishii
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan; Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, Chiba, Japan
| | - Tatsuya Higashi
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan; Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, Chiba, Japan
| | - Hiroshi Yamauchi
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan
| | - Emi Ogawa
- Department of Radiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | | | - Masato Kobayashi
- Wellness Promotion Science Center, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Mitsuyoshi Yoshimoto
- Division of Functional Imaging, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Naoto Shikano
- Department of Radiological Sciences, Ibaraki Prefectural University of Health Sciences, Ibaraki, Japan
| | - Keiichi Kawai
- Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan.
| |
Collapse
|
34
|
Galldiks N, Law I, Pope WB, Arbizu J, Langen KJ. The use of amino acid PET and conventional MRI for monitoring of brain tumor therapy. Neuroimage Clin 2016; 13:386-394. [PMID: 28116231 PMCID: PMC5226808 DOI: 10.1016/j.nicl.2016.12.020] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/09/2016] [Accepted: 12/16/2016] [Indexed: 12/03/2022]
Abstract
Routine diagnostics and treatment monitoring of brain tumors is usually based on contrast-enhanced MRI. However, the capacity of conventional MRI to differentiate tumor tissue from posttherapeutic effects following neurosurgical resection, chemoradiation, alkylating chemotherapy, radiosurgery, and/or immunotherapy may be limited. Metabolic imaging using PET can provide relevant additional information on tumor metabolism, which allows for more accurate diagnostics especially in clinically equivocal situations. This review article focuses predominantly on the amino acid PET tracers 11C-methyl-l-methionine (MET), O-(2-[18F]fluoroethyl)-l-tyrosine (FET) and 3,4-dihydroxy-6-[18F]-fluoro-l-phenylalanine (FDOPA) and summarizes investigations regarding monitoring of brain tumor therapy.
Collapse
Affiliation(s)
- Norbert Galldiks
- Dept. of Neurology, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
- Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany
| | - Ian Law
- Dept.of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Whitney B. Pope
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Javier Arbizu
- Dept. of Nuclear Medicine, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
- Dept. of Nuclear Medicine, University of Aachen, Aachen, Germany
| |
Collapse
|
35
|
Marner L, Henriksen OM, Lundemann M, Larsen VA, Law I. Clinical PET/MRI in neurooncology: opportunities and challenges from a single-institution perspective. Clin Transl Imaging 2016; 5:135-149. [PMID: 28936429 PMCID: PMC5581366 DOI: 10.1007/s40336-016-0213-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 10/31/2016] [Indexed: 01/17/2023]
Abstract
Purpose Magnetic resonance imaging (MRI) plays a key role in neurooncology, i.e., for diagnosis, treatment evaluation and detection of recurrence. However, standard MRI cannot always separate malignant tissue from other pathologies or treatment-induced changes. Advanced MRI techniques such as diffusion-weighted imaging, perfusion imaging and spectroscopy show promising results in discriminating malignant from benign lesions. Further, supplemental imaging with amino acid positron emission tomography (PET) has been shown to increase accuracy significantly and is used routinely at an increasing number of sites. Several centers are now implementing hybrid PET/MRI systems allowing for multiparametric imaging, combining conventional MRI with advanced MRI and amino acid PET imaging. Neurooncology is an obvious focus area for PET/MR imaging. Methods Based on the literature and our experience from more than 300 PET/MRI examinations of brain tumors with 18F-fluoro-ethyl-tyrosine, the clinical use of PET/MRI in adult and pediatric neurooncology is critically reviewed. Results Although the results are increasingly promising, the added value and range of indications for multiparametric imaging with PET/MRI are yet to be established. Robust solutions to overcome the number of issues when using a PET/MRI scanner are being developed, which is promising for a more routine use in the future. Conclusions In a clinical setting, a PET/MRI scan may increase accuracy in discriminating recurrence from treatment changes, although sequential same-day imaging on separate systems will often constitute a reliable and cost-effective alternative. Pediatric patients who require general anesthesia will benefit the most from simultaneous PET and MR imaging.
Collapse
Affiliation(s)
- Lisbeth Marner
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark
| | - Otto M Henriksen
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark
| | - Michael Lundemann
- Department of Oncology, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark
| | - Vibeke Andrée Larsen
- Department of Radiology, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100 Copenhagen, Denmark
| |
Collapse
|
36
|
Stegmayr C, Bandelow U, Oliveira D, Lohmann P, Willuweit A, Filss C, Galldiks N, Lübke JHR, Shah NJ, Ermert J, Langen KJ. Influence of blood-brain barrier permeability on O-(2- 18F-fluoroethyl)-L-tyrosine uptake in rat gliomas. Eur J Nucl Med Mol Imaging 2016; 44:408-416. [PMID: 27613541 DOI: 10.1007/s00259-016-3508-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/29/2016] [Indexed: 02/07/2023]
Abstract
PURPOSE O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET) is an established tracer for the diagnosis of brain tumors with PET. This study investigates the influence of blood-brain barrier (BBB) permeability on 18F-FET uptake in two rat glioma models and one human xenograft model. METHODS F98 glioma, 9L gliosarcoma or human U87 glioblastoma cells were implanted into the striatum of 56 Fischer or RNU rats. Thereafter, animals were divided into a control group and a group receiving injections of the glucocorticoid dexamethasone (Dex). After 12-13 days of tumor growth animals received injection of Evans blue dye (EBD) to visualize BBB disturbance and underwent 18F-FET PET followed by autoradiography. Time activity curves, standardized uptake values (SUV) and Tumor-to-brain ratios (TBR) of 18F-FET uptake [18-61 min post injection (p.i.)] were evaluated using a volume-of-Interest (VOI) analysis. BBB disturbance was quantitatively evaluated by EBD fluorescence. The membrane gaps of blood vessel endothelial tight junctions were measured using electron microscopy to visualize ultrastructural BBB alterations in one untreated and one Dex treated F98 glioma. Data were analyzed by two-way ANOVAs. RESULTS In Dex treated animals EBD extravasation was significantly reduced in 9L (P < 0.001) and U87 (P = 0.008) models and showed a trend in F98 models (P = 0.053). In contrast, no significant differences of 18F-FET uptake were observed between Dex treated animals and control group except a decrease of the TBR in the 9L tumor model in PET (P < 0.01). Ultrastructural evaluation of tumor blood vessel endothelia revealed significant reduction of the cleft diameter between endothelial cells after Dex treatment in F98 model (P = 0.010). CONCLUSION Despite a considerable reduction of BBB permeability in rat gliomas after Dex treatment, no relevant changes of 18F-FET uptake were noted in this experimental study. Thus, 18F-FET uptake in gliomas appears to be widely independent of the permeability of the BBB.
Collapse
Affiliation(s)
- Carina Stegmayr
- Forschungszentrum Jülich, Institute of Neuroscience and Medicine, D-52425, Jülich, Germany.
| | - Ulrike Bandelow
- Forschungszentrum Jülich, Institute of Neuroscience and Medicine, D-52425, Jülich, Germany
| | - Dennis Oliveira
- Forschungszentrum Jülich, Institute of Neuroscience and Medicine, D-52425, Jülich, Germany
| | - Philipp Lohmann
- Forschungszentrum Jülich, Institute of Neuroscience and Medicine, D-52425, Jülich, Germany
| | - Antje Willuweit
- Forschungszentrum Jülich, Institute of Neuroscience and Medicine, D-52425, Jülich, Germany
| | - Christian Filss
- Forschungszentrum Jülich, Institute of Neuroscience and Medicine, D-52425, Jülich, Germany.,Department of Nuclear Medicine and Neurology, RWTH/University Hospital Aachen, Aachen, Germany
| | - Norbert Galldiks
- Forschungszentrum Jülich, Institute of Neuroscience and Medicine, D-52425, Jülich, Germany
| | - Joachim H R Lübke
- Forschungszentrum Jülich, Institute of Neuroscience and Medicine, D-52425, Jülich, Germany
| | - N Jon Shah
- Forschungszentrum Jülich, Institute of Neuroscience and Medicine, D-52425, Jülich, Germany.,Department of Nuclear Medicine and Neurology, RWTH/University Hospital Aachen, Aachen, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Aachen, Germany
| | - Johannes Ermert
- Forschungszentrum Jülich, Institute of Neuroscience and Medicine, D-52425, Jülich, Germany.,Department of Nuclear Medicine and Neurology, RWTH/University Hospital Aachen, Aachen, Germany
| | - Karl-Josef Langen
- Forschungszentrum Jülich, Institute of Neuroscience and Medicine, D-52425, Jülich, Germany. .,Department of Nuclear Medicine and Neurology, RWTH/University Hospital Aachen, Aachen, Germany.
| |
Collapse
|
37
|
Harat M, Małkowski B, Makarewicz R. Pre-irradiation tumour volumes defined by MRI and dual time-point FET-PET for the prediction of glioblastoma multiforme recurrence: A prospective study. Radiother Oncol 2016; 120:241-7. [PMID: 27378734 DOI: 10.1016/j.radonc.2016.06.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 06/07/2016] [Accepted: 06/12/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND PURPOSE The diagnostic accuracy of magnetic resonance imaging (MRI) for glioblastoma multiforme (GBM) is suboptimal. We analysed pre-treatment MRI- and dual time-point 18F-fluoroethylthyrosine-PET (FET-PET)-based target volumes and GBM recurrence patterns following radiotherapy with temozolomide. MATERIALS AND METHODS Thirty-four patients with primary GBM were treated according to MRI-based treatment volumes (GTVRM). Patients underwent dual time-point FET-PET scans prior to treatment, and biological tumour volumes (GTVPET) were contoured but not used for target definition. Progressions were classified based on location of primary GTVs. Volume and uniformity of MRI- vs. FET-PET/CT-derived GTVs and progression patterns assessed by MRI were analysed. RESULTS FET-based GTVs measured 10min after radionuclide injection (a.r.i.; median 37.3cm(3)) were larger than GTVs measured 60min a.r.i. (median 27.7cm(3)). GTVPET volumes were significantly larger than corresponding MRI-based GTVs. MRI and PET concordance for the identification of glioblastoma GTVs was poor (mean uniformity index 0.4). 74% of failures were inside primary GTVPET volumes, with no solitary progressions inside the MRI-defined margin +20mm but outside the GTVPET detected. CONCLUSIONS The size and geometry of GTVs differed in the majority of patients. The GTVPET volume depends on time after radionuclide injection. FET-PET better defined failure site than MRI alone.
Collapse
Affiliation(s)
- Maciej Harat
- Department of Radiotherapy, The Franciszek Lukaszczyk Oncology Center, Bydgoszcz, Poland; Department of Oncology and Brachytherapy, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland.
| | - Bogdan Małkowski
- Department of Positron Emission Tomography and Molecular Imaging, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland; Department of Nuclear Medicine, The Franciszek Lukaszczyk Oncology Center, Bydgoszcz, Poland
| | - Roman Makarewicz
- Department of Oncology and Brachytherapy, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland; Department of Oncology and Brachytherapy, The Franciszek Lukaszczyk Oncology Center, Bydgoszcz, Poland
| |
Collapse
|
38
|
Stegmayr C, Schöneck M, Oliveira D, Willuweit A, Filss C, Galldiks N, Shah NJ, Coenen HH, Langen KJ. Reproducibility of O-(2-18F-fluoroethyl)-L-tyrosine uptake kinetics in brain tumors and influence of corticoid therapy: an experimental study in rat gliomas. Eur J Nucl Med Mol Imaging 2015; 43:1115-23. [DOI: 10.1007/s00259-015-3274-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/27/2015] [Indexed: 10/22/2022]
|
39
|
Malkowski B, Harat M, Zyromska A, Wisniewski T, Harat A, Lopatto R, Furtak J. The Sum of Tumour-to-Brain Ratios Improves the Accuracy of Diagnosing Gliomas Using 18F-FET PET. PLoS One 2015; 10:e0140917. [PMID: 26468649 PMCID: PMC4607373 DOI: 10.1371/journal.pone.0140917] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 10/01/2015] [Indexed: 11/19/2022] Open
Abstract
Gliomas are common brain tumours, but obtaining tissue for definitive diagnosis can be difficult. There is, therefore, interest in the use of non-invasive methods to diagnose and grade the disease. Although positron emission tomography (PET) with 18F-fluorethyltyrosine (18F-FET) can be used to differentiate between low-grade (LGG) and high-grade (HGG) gliomas, the optimal parameters to measure and their cut-points have yet to be established. We therefore assessed the value of single and dual time-point acquisition of 18F-FET PET parameters to differentiate between primary LGGs (n = 22) and HGGs (n = 24). PET examination was considered positive for glioma if the metabolic activity was 1.6-times higher than that of background (contralateral) brain, and maximum tissue-brain ratios (TBRmax) were calculated 10 and 60 min after isotope administration with their sums and differences calculated from individual time-point values. Using a threshold-based method, the overall sensitivity of PET was 97%. Several analysed parameters were significantly different between LGGs and HGGs. However, in a receiver operating characteristics analysis, TBR sum had the best diagnostic accuracy of 87% and sensitivity, specificity, and positive and negative predictive values of 100%, 72.7%, 80%, and 100%, respectively. 18F-FET PET is valuable for the non-invasive determination of glioma grade, especially when dual time-point metrics are used. TBR sum shows the greatest accuracy, sensitivity, and negative predictive value for tumour grade differentiation and is a simple method to implement. However, the cut-off may differ between institutions and calibration strategies would be useful.
Collapse
Affiliation(s)
- Bogdan Malkowski
- Department of Positron Emission Tomography and Molecular Imaging, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
- Department of Nuclear Medicine, Franciszek Lukaszczyk Oncology Centre, Bydgoszcz, Poland
| | - Maciej Harat
- Department of Radiotherapy, Franciszek Lukaszczyk Oncology Centre, Bydgoszcz, Poland
- Department of Oncology and Brachytherapy, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
- * E-mail:
| | - Agnieszka Zyromska
- Department of Radiotherapy, Franciszek Lukaszczyk Oncology Centre, Bydgoszcz, Poland
- Department of Oncology and Brachytherapy, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Tomasz Wisniewski
- Department of Radiotherapy, Franciszek Lukaszczyk Oncology Centre, Bydgoszcz, Poland
- Department of Oncology and Brachytherapy, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Aleksandra Harat
- Department of Public Health, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Rita Lopatto
- Department of Nuclear Medicine, Franciszek Lukaszczyk Oncology Centre, Bydgoszcz, Poland
| | - Jacek Furtak
- Department of Neurosurgery, 10th Military Research Hospital and Polyclinic, Bydgoszcz, Poland
| |
Collapse
|
40
|
Evaluation of trans-1-amino-3-18F-fluorocyclobutanecarboxylic acid accumulation in low-grade glioma in chemically induced rat models: PET and autoradiography compared with morphological images and histopathological findings. Nucl Med Biol 2015; 42:664-72. [DOI: 10.1016/j.nucmedbio.2015.04.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/14/2015] [Accepted: 04/15/2015] [Indexed: 11/17/2022]
|
41
|
Galldiks N, Langen KJ, Pope WB. From the clinician's point of view - What is the status quo of positron emission tomography in patients with brain tumors? Neuro Oncol 2015; 17:1434-44. [PMID: 26130743 DOI: 10.1093/neuonc/nov118] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/31/2015] [Indexed: 12/13/2022] Open
Abstract
The most common type of primary brain tumor is malignant glioma. Despite intensive therapeutic efforts, the majority of these neoplasms remain incurable. Imaging techniques are important for initial tumor detection and comprise indispensable tools for monitoring treatment. Structural imaging using contrast-enhanced MRI is the method of choice for brain tumor surveillance, but its capacity to differentiate tumor from nonspecific tissue changes can be limited, particularly with posttreatment gliomas. Metabolic imaging using positron-emission-tomography (PET) can provide relevant additional information, which may allow for better assessment of tumor burden in ambiguous cases. Specific PET tracers have addressed numerous molecular targets in the last decades, but only a few have achieved relevance in routine clinical practice. At present, PET studies using radiolabeled amino acids appear to improve clinical decision-making as these tracers can offer better delineation of tumor extent as well as improved targeting of biopsies, surgical interventions, and radiation therapy. Amino acid PET imaging also appears useful for distinguishing glioma recurrence or progression from postradiation treatment effects, particularly radiation necrosis and pseudoprogression, and provides information on histological grading and patient prognosis. In the last decade, the tracers O-(2-[(18)F]fluoroethyl)-L-tyrosine (FET) and 3,4-dihydroxy-6-[(18)F]-fluoro-L-phenylalanine (FDOPA) have been increasingly used for these indications. This review article focuses on these tracers and summarizes their recent applications for patients with brain tumors. Current uses of tracers other than FET and FDOPA are also discussed, and the most frequent practical questions regarding PET brain tumor imaging are reviewed.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, University of Cologne, Cologne, Germany (N.G.); Research Center Jülich, Institute of Neuroscience and Medicine, Jülich, Germany (N.G., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Nuclear Medicine, University of Aachen, Germany (K.-J.L.); Department of Radiological Sciences, David Geffen School of Medicine at UCLA., Los Angeles (W.B.P.)
| | - Karl-Josef Langen
- Department of Neurology, University of Cologne, Cologne, Germany (N.G.); Research Center Jülich, Institute of Neuroscience and Medicine, Jülich, Germany (N.G., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Nuclear Medicine, University of Aachen, Germany (K.-J.L.); Department of Radiological Sciences, David Geffen School of Medicine at UCLA., Los Angeles (W.B.P.)
| | - Whitney B Pope
- Department of Neurology, University of Cologne, Cologne, Germany (N.G.); Research Center Jülich, Institute of Neuroscience and Medicine, Jülich, Germany (N.G., K.-J.L.); Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany (N.G.); Department of Nuclear Medicine, University of Aachen, Germany (K.-J.L.); Department of Radiological Sciences, David Geffen School of Medicine at UCLA., Los Angeles (W.B.P.)
| |
Collapse
|
42
|
Yook CM, Lee SJ, Oh SJ, Ha HJ, Lee JJ. Simple preparation of new [(18) F]F-labeled synthetic amino acid derivatives with two click reactions in one-pot and SPE purification. J Labelled Comp Radiopharm 2015; 58:317-26. [PMID: 26031401 DOI: 10.1002/jlcr.3297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/01/2015] [Accepted: 04/12/2015] [Indexed: 01/23/2023]
Abstract
New [(18) F]fluorinated 1,2,3-triazolyl amino acid derivatives were efficiently prepared from Huisgen 1,3-dipolar cycloaddition reactions, well known as click reaction. We developed two simultaneous click reactions in one-pot with a simple solid-phase extraction (SPE) purification method. [(18) F]fluoro-1-propyne was obtained at a 45% non-decay corrected radiochemical yield based on the [(18) F]fluoride ion. The one-pot and simultaneous two click reactions were performed with unprotected azido-alkyl amino acid, [(18) F]fluoro-1-propyne, and lipophilic additive alkyne to produce three synthetic amino acid derivatives, AMC-101 ([(18) F]-6a), AMC-102 ([(18) F]-6b), and AMC-103 ([(18) F]-6c) with 29%, 28%, and 24% of non-decay corrected radiochemical yields, respectively. All radiotracers indicated that radiochemical purities were >95% without any residual organic solvent. Our new method involving two click reactions in one-pot showed high radiochemical and chemical purity by easy removal of the residual precursor from the simultaneous two click reactions.
Collapse
Affiliation(s)
- Cheol-Min Yook
- Department of Chemistry, Hankuk University of Foreign Studies, Yongin, Kyunggi-do, 449-719, South Korea
| | - Sang Ju Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 388-1 Pungnap-dong, Songpa-gu, Seoul, 138-736, South Korea
| | - Seung Jun Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 388-1 Pungnap-dong, Songpa-gu, Seoul, 138-736, South Korea
| | - Hyun-Joon Ha
- Department of Chemistry, Hankuk University of Foreign Studies, Yongin, Kyunggi-do, 449-719, South Korea
| | - Jong Jin Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 388-1 Pungnap-dong, Songpa-gu, Seoul, 138-736, South Korea
| |
Collapse
|
43
|
Sharma S, Singh B, Mishra AK, Rathod D, Hazari PP, Chuttani K, Chopra S, Singh PM, Abrar ML, Mittal BR, Singh G. LAT-1 based primary breast cancer detection by [99m]Tc-labeled DTPA-bis-methionine scintimammography: first results using indigenously developed single vial kit preparation. Cancer Biother Radiopharm 2015; 29:283-8. [PMID: 25203145 DOI: 10.1089/cbr.2014.1655] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE To evaluate the diagnostic utility of a single vial ready to label with [99m]Tc kit preparation of DTPA-bis-methionine (DTPA-bis-MET) for the detection of primary breast cancer. METHODS The conjugate (DTPA-bis-MET) was synthesized by covalently conjugating two molecules of methionine to DTPA and formulated as a single vial ready to label with [99m]Tc lyophilized kit preparations. Thirty female patients (mean age=47.5±11.8 years; range=21-69 years) with radiological/clinical evidence of having primary breast carcinoma were subjected to [99m]Tc-methionine scintigraphy. The whole body (anterior and posterior) imaging was performed on all the patients at 5 minutes, 10 minutes, 1 hour, 2 hours, and 4 hours following an intravenous administration of 555-740 MBq radioactivity of [99m]Tc-methionine. In addition, scintimammography (static images; 256×256 matrix) at 1, 2, and 4 hours was also performed on all the patients. RESULTS The resultant radiolabel, that is, [99m]Tc-DTPA-bis-MET, yielded high radiolabeling efficiency (>97.0%), radiochemical purity (166-296 MBq/μmol), and shelf life (>3 months). The radiotracer primarily gets excreted through the kidneys and localizes in the breast cancer lesions with high target-to-nontarget ratios. The mean±SD ratios on the scan-positive lesions acquired at 1, 2, and 4 hours postinjection were 3.6±0.48, 3.10±0.24, and 2.5±0.4, respectively. [99m]Tc-methionine scintimammography demonstrated an excellent sensitivity and positive predictive value of 96.0% each for the detection of primary breast cancer. CONCLUSION Ready to label single vial kit formulations of DTPA-bis-MET can be easily synthesized as in-house production and conveniently used for the scintigraphic detection of breast cancer and other methionine-dependent tumors expressing the L-type amino acid transporter-1 receptor. The imaging technique thus could be a potential substitute for the conventional single-photon emission computed tomography (SPECT)-based tumor imaging agents, especially for tracers with nonspecific mitochondrial uptake. However, the diagnostic efficacy of [99m]Tc-methionine needs to be evaluated in a large cohort of patients through further multicentric trials.
Collapse
Affiliation(s)
- Sarika Sharma
- 1 Department of Nuclear Medicine & PET, PGIMER , Chandigarh, India
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Huang T, Tang G, Wang H, Nie D, Tang X, Liang X, Hu K, Yi C, Yao B, Tang C. Synthesis and preliminary biological evaluation of S-11C-methyl-d-cysteine as a new amino acid PET tracer for cancer imaging. Amino Acids 2015; 47:719-727. [PMID: 25534431 DOI: 10.1007/s00726-014-1899-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022]
|
45
|
Jansen NL, Suchorska B, Wenter V, Schmid-Tannwald C, Todica A, Eigenbrod S, Niyazi M, Tonn JC, Bartenstein P, Kreth FW, la Fougère C. Prognostic significance of dynamic 18F-FET PET in newly diagnosed astrocytic high-grade glioma. J Nucl Med 2015; 56:9-15. [PMID: 25537990 DOI: 10.2967/jnumed.114.144675] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Despite advances in diagnosis and the use of different therapeutic regimens in astrocytic high-grade glioma (HGG), the prognosis for patients remains grim. Additional pretherapeutic information is needed to tailor management. To gain additional prognostic information at primary diagnosis, we investigated the value of dynamic O-(2-(18)F-fluoroethyl)-L-tyrosine ((18)F-FET) PET. METHODS We retrospectively evaluated 121 patients who had a primary diagnosis of astrocytic HGG (51 World Health Organization [WHO] grade III; 70 WHO IV) and underwent dynamic (18)F-FET PET before histopathologic assessment. We assessed static parameters (maximal and mean tumoral standardized uptake value corrected for mean background activity in the contralateral hemisphere [SUV(max)/BG and SUV(mean)/BG, respectively], biologic tumor volume) and dynamic time-activity curves, including minimal time to peak (TTP(min)). The prognostic influence of PET parameters and other clinical parameters on progression-free and overall survival was evaluated using uni- and multivariate Cox regression and Kaplan-Meier survival estimates. RESULTS In the group overall, median progression-free survival and overall survival were 12.2 and 21.9 mo. SUV(max)/BG, SUV(mean)/BG, and biologic tumor volume were significantly higher in WHO IV than in WHO III gliomas; median TTP(min) was 12.5 min in both groups. On univariate analysis, the factors age, WHO grade, O6-methylguanine-DNA methyltransferase promoter methylation status, contrast enhancement, initial treatment, and TTP(min) showed prognostic significance, with WHO grade, O6-methylguanine-DNA methyltransferase status, age, and TTP(min) remaining significant in the multivariate analysis. WHO grade and TTP(min) reached a similar fit for the prognostic evaluation. The prognosis of WHO III astrocytoma with an early TTP(min) of 12.5 min or less did not differ significantly from that of glioblastoma. CONCLUSION Early TTP(min) is associated with worse outcome in patients with newly diagnosed astrocytic HGG. In the preoperative setting, TTP(min) can be a valuable noninvasive prognostic marker with comparable significance to WHO grade. Additionally, TTP(min) can help identify highly aggressive WHO III astrocytoma tumors and may help in adjusting standard treatment toward an individualized, risk-adapted therapy regime.
Collapse
Affiliation(s)
- Nathalie L Jansen
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Germany
| | - Bogdana Suchorska
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany
| | - Vera Wenter
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Germany
| | | | - Andrei Todica
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Germany
| | - Sabina Eigenbrod
- Department of Neuropathology, Ludwig-Maximilians-University, Munich, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, Ludwig-Maximilians-University, Munich, Germany; and
| | | | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Germany
| | | | - Christian la Fougère
- Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
46
|
Rapp M, Floeth FW, Felsberg J, Steiger HJ, Sabel M, Langen KJ, Galldiks N. Clinical value of O-(2-[(18)F]-fluoroethyl)-L-tyrosine positron emission tomography in patients with low-grade glioma. Neurosurg Focus 2015; 34:E3. [PMID: 23373448 DOI: 10.3171/2012.12.focus12336] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Progress in morphological imaging has facilitated the diagnosis of low-grade glioma (LGG) and plays a decisive role in therapeutic decisions. To date, the method of choice is contrast-enhanced MRI including T1-/T2-weighted and FLAIR sequences. However, tumor delineation and the differentiation between neoplastic and normal brain tissue can be difficult when using morphological MRI and may complicate the identification of anaplastic foci for biopsy and further treatment planning. Furthermore, therapy monitoring and the differentiation of tumor recurrence from unspecific post-therapeutic changes in the tissue are challenging. Additional information about tumor metabolism may be very helpful for the diagnostic assessment of LGG and can be provided by PET. In recent years, the PET amino acid tracer O-(2-[(18)F]-fluoroethyl)-L-tyrosine ((18)F-FET) has been clinically validated for brain tumor diagnosis. This tracer has logistical advantages over the widely used PET tracer (11)C-methyl-L-methionine due to the longer half-life of the (18)F-label (109 vs 20 minutes, respectively). Additionally, it has been demonstrated that both tracers provide comparable diagnostic information. The authors provide an overview of the recent literature regarding the value of various clinical applications of (18)F-FET PET in patients with LGG.
Collapse
Affiliation(s)
- Marion Rapp
- Departments of Neurosurgery, University of Duesseldorf, Duesseldorf, Germany.
| | | | | | | | | | | | | |
Collapse
|
47
|
Munck Af Rosenschold P, Costa J, Engelholm SA, Lundemann MJ, Law I, Ohlhues L, Engelholm S. Impact of [18F]-fluoro-ethyl-tyrosine PET imaging on target definition for radiation therapy of high-grade glioma. Neuro Oncol 2014; 17:757-63. [PMID: 25537018 DOI: 10.1093/neuonc/nou316] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 10/20/2014] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND We sought to assess the impact of amino-acid (18)F-fluoro-ethyl-tyrosine (FET) positron emission tomography (PET) on the volumetric target definition for radiation therapy of high-grade glioma versus the current standard using MRI alone. Specifically, we investigated the influence of tumor grade, MR-defined tumor volume, and the extent of surgical resection on PET positivity. METHODS Fifty-four consecutive high-grade glioma patients (World Health Organization grades III-IV) with confirmed histology were scanned using FET-PET/CT and T1 and T2/fluid attenuated inversion recovery MRI. Gross tumor volume and clinical target volumes (CTVs) were defined in a blinded fashion based on MRI and subsequently PET, and volumetric analysis was performed. The extent of the surgical resection was reviewed using postoperative MRI. RESULTS Overall, for ∼ 90% of the patients, the PET-positive volumes were encompassed by T1 MRI with contrast-defined tumor plus a 20-mm margin. The tumor volume defined by PET was larger for glioma grade IV (P < .001) and smaller for patients with more extensive surgical resection (P = .004). The margin required to be added to the MRI-defined tumor in order to fully encompass the FET-PET positive volume tended to be larger for grade IV tumors (P = .018). CONCLUSION With an unchanged CTV margin and by including FET-PET for gross tumor volume definition, the CTV will increase moderately for most patients, and quite substantially for a minority of patients. Patients with grade IV glioma were found to be the primary candidates for PET-guided radiation therapy planning.
Collapse
Affiliation(s)
- Per Munck Af Rosenschold
- Department of Oncology, Section of Radiotherapy, Rigshospitalet, Copenhagen, Denmark (P.M.a.R., J.C., S.A.E., M.J.L., L.O.); Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark (P.M.a.R., M.J.L.); Department of Clinical Physiology, Nuclear Medicine, and PET, Rigshospitalet, Copenhagen, Denmark (I.L., J.C.); Section of Radiation Oncology, Skåne University Hospital, Lund, Sweden (S.E.)
| | - Junia Costa
- Department of Oncology, Section of Radiotherapy, Rigshospitalet, Copenhagen, Denmark (P.M.a.R., J.C., S.A.E., M.J.L., L.O.); Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark (P.M.a.R., M.J.L.); Department of Clinical Physiology, Nuclear Medicine, and PET, Rigshospitalet, Copenhagen, Denmark (I.L., J.C.); Section of Radiation Oncology, Skåne University Hospital, Lund, Sweden (S.E.)
| | - Svend Aage Engelholm
- Department of Oncology, Section of Radiotherapy, Rigshospitalet, Copenhagen, Denmark (P.M.a.R., J.C., S.A.E., M.J.L., L.O.); Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark (P.M.a.R., M.J.L.); Department of Clinical Physiology, Nuclear Medicine, and PET, Rigshospitalet, Copenhagen, Denmark (I.L., J.C.); Section of Radiation Oncology, Skåne University Hospital, Lund, Sweden (S.E.)
| | - Michael J Lundemann
- Department of Oncology, Section of Radiotherapy, Rigshospitalet, Copenhagen, Denmark (P.M.a.R., J.C., S.A.E., M.J.L., L.O.); Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark (P.M.a.R., M.J.L.); Department of Clinical Physiology, Nuclear Medicine, and PET, Rigshospitalet, Copenhagen, Denmark (I.L., J.C.); Section of Radiation Oncology, Skåne University Hospital, Lund, Sweden (S.E.)
| | - Ian Law
- Department of Oncology, Section of Radiotherapy, Rigshospitalet, Copenhagen, Denmark (P.M.a.R., J.C., S.A.E., M.J.L., L.O.); Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark (P.M.a.R., M.J.L.); Department of Clinical Physiology, Nuclear Medicine, and PET, Rigshospitalet, Copenhagen, Denmark (I.L., J.C.); Section of Radiation Oncology, Skåne University Hospital, Lund, Sweden (S.E.)
| | - Lars Ohlhues
- Department of Oncology, Section of Radiotherapy, Rigshospitalet, Copenhagen, Denmark (P.M.a.R., J.C., S.A.E., M.J.L., L.O.); Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark (P.M.a.R., M.J.L.); Department of Clinical Physiology, Nuclear Medicine, and PET, Rigshospitalet, Copenhagen, Denmark (I.L., J.C.); Section of Radiation Oncology, Skåne University Hospital, Lund, Sweden (S.E.)
| | - Silke Engelholm
- Department of Oncology, Section of Radiotherapy, Rigshospitalet, Copenhagen, Denmark (P.M.a.R., J.C., S.A.E., M.J.L., L.O.); Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark (P.M.a.R., M.J.L.); Department of Clinical Physiology, Nuclear Medicine, and PET, Rigshospitalet, Copenhagen, Denmark (I.L., J.C.); Section of Radiation Oncology, Skåne University Hospital, Lund, Sweden (S.E.)
| |
Collapse
|
48
|
System L amino acid transporter LAT1 accumulates O-(2-fluoroethyl)-L-tyrosine (FET). Amino Acids 2014; 47:335-44. [PMID: 25385314 DOI: 10.1007/s00726-014-1863-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/24/2014] [Indexed: 01/04/2023]
Abstract
O-(2-fluoroethyl)-L-tyrosine (FET) labeled with fluorine-18 is an important and specific tracer for diagnostics of glioblastoma via positron emission tomography (PET). However, the mechanism of its quite specific accumulation in tumor tissue has not been understood so far. In this work we demonstrate that [(3)H]L-tyrosine is primarily transported by the system L transporter LAT1 in human LN229 glioblastoma cells. FET reduced tyrosine transport, suggesting that it shares the same uptake pathway. More importantly, accumulation of FET was significantly reduced after siRNA-mediated downregulation of LAT1. Xenopus laevis oocytes expressing human LAT1 together with the glycoprotein 4F2hc (necessary to pull LAT-1 to the plasma membrane) exhibited a similar accumulation of FET as observed in glioblastoma cells. In contrast, no accumulation was observed in control oocytes, not overexpressing an exogenous transporter. Because LAT1 works exclusively as an exchanger of amino acids, substrates at one side of the membrane stimulate exchange against substrates at the other side. Extracellular FET stimulated the efflux of intracellular [(3)H]L-leucine, demonstrating that FET is indeed an influx substrate for LAT1. However, FET injected into oocytes was not able to stimulate uptake of extracellular [(3)H]L-leucine, indicating that FET is not a good efflux substrate. Our data, therefore, suggest that FET is trapped within cells due to the asymmetry of its intra- and extracellular recognition by LAT1. If also found for other transporters in tumor cells, asymmetric substrate recognition may be further exploited for tumor-specific accumulation of PET-tracers and/or other tumor-related drugs.
Collapse
|
49
|
Nedergaard MK, Kristoffersen K, Michaelsen SR, Madsen J, Poulsen HS, Stockhausen MT, Lassen U, Kjaer A. The use of longitudinal 18F-FET MicroPET imaging to evaluate response to irinotecan in orthotopic human glioblastoma multiforme xenografts. PLoS One 2014; 9:e100009. [PMID: 24918622 PMCID: PMC4053391 DOI: 10.1371/journal.pone.0100009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 05/21/2014] [Indexed: 11/26/2022] Open
Abstract
Objectives Brain tumor imaging is challenging. Although 18F-FET PET is widely used in the clinic, the value of 18F-FET MicroPET to evaluate brain tumors in xenograft has not been assessed to date. The aim of this study therefore was to evaluate the performance of in vivo18F-FET MicroPET in detecting a treatment response in xenografts. In addition, the correlations between the 18F-FET tumor accumulation and the gene expression of Ki67 and the amino acid transporters LAT1 and LAT2 were investigated. Furthermore, Ki67, LAT1 and LAT2 gene expression in xenograft and archival patient tumors was compared. Methods Human GBM cells were injected orthotopically in nude mice and 18F-FET uptake was followed by weekly MicroPET/CT. When tumor take was observed, mice were treated with CPT-11 or saline weekly. After two weeks of treatment the brain tumors were isolated and quantitative polymerase chain reaction were performed on the xenograft tumors and in parallel on archival patient tumor specimens. Results The relative tumor-to-brain (T/B) ratio of SUVmax was significantly lower after one week (123±6%, n = 7 vs. 147±6%, n = 7; p = 0.018) and after two weeks (142±8%, n = 5 vs. 204±27%, n = 4; p = 0.047) in the CPT-11 group compared with the control group. Strong negative correlations between SUVmax T/B ratio and LAT1 (r = −0.62, p = 0.04) and LAT2 (r = −0.67, p = 0.02) were observed. In addition, a strong positive correlation between LAT1 and Ki67 was detected in xenografts. Furthermore, a 1.6 fold higher expression of LAT1 and a 23 fold higher expression of LAT2 were observed in patient specimens compared to xenografts. Conclusions 18F-FET MicroPET can be used to detect a treatment response to CPT-11 in GBM xenografts. The strong negative correlation between SUVmax T/B ratio and LAT1/LAT2 indicates an export transport function. We suggest that 18F-FET PET may be used for detection of early treatment response in patients.
Collapse
Affiliation(s)
- Mette K. Nedergaard
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Karina Kristoffersen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Copenhagen, Denmark
| | - Signe R. Michaelsen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Copenhagen, Denmark
| | - Jacob Madsen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Hans S. Poulsen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Copenhagen, Denmark
| | | | - Ulrik Lassen
- Phase 1 Unit, Department of Oncology, The Finsen Center, Rigshospitalet, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
50
|
Synthesis and biological evaluation of N-(2-[18F]Fluoropropionyl)-L-methionine for tumor imaging. Nucl Med Biol 2013; 40:926-32. [PMID: 23886847 DOI: 10.1016/j.nucmedbio.2013.06.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 06/05/2013] [Accepted: 06/14/2013] [Indexed: 11/20/2022]
|