51
|
Donche S, Verhoeven J, Descamps B, Bolcaen J, Deblaere K, Boterberg T, Van den Broecke C, Vanhove C, Goethals I. The Path Toward PET-Guided Radiation Therapy for Glioblastoma in Laboratory Animals: A Mini Review. Front Med (Lausanne) 2019; 6:5. [PMID: 30761302 PMCID: PMC6361864 DOI: 10.3389/fmed.2019.00005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/10/2019] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is the most aggressive and malignant primary brain tumor in adults. Despite the current state-of-the-art treatment, which consists of maximal surgical resection followed by radiation therapy, concomitant, and adjuvant chemotherapy, progression remains rapid due to aggressive tumor characteristics. Several new therapeutic targets have been investigated using chemotherapeutics and targeted molecular drugs, however, the intrinsic resistance to induced cell death of brain cells impede the effectiveness of systemic therapies. Also, the unique immune environment of the central nervous system imposes challenges for immune-based therapeutics. Therefore, it is important to consider other approaches to treat these tumors. There is a well-known dose-response relationship for glioblastoma with increased survival with increasing doses, but this effect seems to cap around 60 Gy, due to increased toxicity to the normal brain. Currently, radiation treatment planning of glioblastoma patients relies on CT and MRI that does not visualize the heterogeneous nature of the tumor, and consequently, a homogenous dose is delivered to the entire tumor. Metabolic imaging, such as positron-emission tomography, allows to visualize the heterogeneous tumor environment. Using these metabolic imaging techniques, an approach called dose painting can be used to deliver a higher dose to the tumor regions with high malignancy and/or radiation resistance. Preclinical studies are required for evaluating the benefits of novel radiation treatment strategies, such as PET-based dose painting. The aim of this review is to give a brief overview of promising PET tracers that can be evaluated in laboratory animals to bridge the gap between PET-based dose painting in glioblastoma patients.
Collapse
Affiliation(s)
- Sam Donche
- Department of Radiology and Nuclear Medicine, Ghent University, Ghent, Belgium
| | - Jeroen Verhoeven
- Department of Pharmaceutical Analysis, Ghent University, Ghent, Belgium
| | - Benedicte Descamps
- Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Julie Bolcaen
- Department of Radiology and Nuclear Medicine, Ghent University, Ghent, Belgium
| | - Karel Deblaere
- Department of Radiology and Nuclear Medicine, Ghent University, Ghent, Belgium
| | - Tom Boterberg
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | | | - Christian Vanhove
- Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Ingeborg Goethals
- Department of Radiology and Nuclear Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
52
|
Emerging Functional Imaging Biomarkers of Tumour Responses to Radiotherapy. Cancers (Basel) 2019; 11:cancers11020131. [PMID: 30678055 PMCID: PMC6407112 DOI: 10.3390/cancers11020131] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/11/2019] [Accepted: 01/13/2019] [Indexed: 12/11/2022] Open
Abstract
Tumour responses to radiotherapy are currently primarily assessed by changes in size. Imaging permits non-invasive, whole-body assessment of tumour burden and guides treatment options for most tumours. However, in most tumours, changes in size are slow to manifest and can sometimes be difficult to interpret or misleading, potentially leading to prolonged durations of ineffective treatment and delays in changing therapy. Functional imaging techniques that monitor biological processes have the potential to detect tumour responses to treatment earlier and refine treatment options based on tumour biology rather than solely on size and staging. By considering the biological effects of radiotherapy, this review focusses on emerging functional imaging techniques with the potential to augment morphological imaging and serve as biomarkers of early response to radiotherapy.
Collapse
|
53
|
Debus C, Floca R, Ingrisch M, Kompan I, Maier-Hein K, Abdollahi A, Nolden M. MITK-ModelFit: A generic open-source framework for model fits and their exploration in medical imaging - design, implementation and application on the example of DCE-MRI. BMC Bioinformatics 2019; 20:31. [PMID: 30651067 PMCID: PMC6335810 DOI: 10.1186/s12859-018-2588-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/19/2018] [Indexed: 01/21/2023] Open
Abstract
Background Many medical imaging techniques utilize fitting approaches for quantitative parameter estimation and analysis. Common examples are pharmacokinetic modeling in dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI)/computed tomography (CT), apparent diffusion coefficient calculations and intravoxel incoherent motion modeling in diffusion-weighted MRI and Z-spectra analysis in chemical exchange saturation transfer MRI. Most available software tools are limited to a special purpose and do not allow for own developments and extensions. Furthermore, they are mostly designed as stand-alone solutions using external frameworks and thus cannot be easily incorporated natively in the analysis workflow. Results We present a framework for medical image fitting tasks that is included in the Medical Imaging Interaction Toolkit MITK, following a rigorous open-source, well-integrated and operating system independent policy. Software engineering-wise, the local models, the fitting infrastructure and the results representation are abstracted and thus can be easily adapted to any model fitting task on image data, independent of image modality or model. Several ready-to-use libraries for model fitting and use-cases, including fit evaluation and visualization, were implemented. Their embedding into MITK allows for easy data loading, pre- and post-processing and thus a natural inclusion of model fitting into an overarching workflow. As an example, we present a comprehensive set of plug-ins for the analysis of DCE MRI data, which we validated on existing and novel digital phantoms, yielding competitive deviations between fit and ground truth. Conclusions Providing a very flexible environment, our software mainly addresses developers of medical imaging software that includes model fitting algorithms and tools. Additionally, the framework is of high interest to users in the domain of perfusion MRI, as it offers feature-rich, freely available, validated tools to perform pharmacokinetic analysis on DCE MRI data, with both interactive and automatized batch processing workflows.
Collapse
Affiliation(s)
- Charlotte Debus
- German Cancer Consortium (DKTK), Heidelberg, Germany. .,Department of Translational Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Department of Radiation Oncology, Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany. .,National Center for Tumor Diseases (NCT), Heidelberg, Germany. .,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.
| | - Ralf Floca
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany. .,Division of Medical Image Computing, German Cancer Research Center DKFZ, Heidelberg, Germany.
| | - Michael Ingrisch
- Department of Radiology, University Hospital Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Ina Kompan
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,Division of Medical Image Computing, German Cancer Research Center DKFZ, Heidelberg, Germany
| | - Klaus Maier-Hein
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,Division of Medical Image Computing, German Cancer Research Center DKFZ, Heidelberg, Germany.,Section Pattern Recognition, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Amir Abdollahi
- German Cancer Consortium (DKTK), Heidelberg, Germany.,Department of Translational Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Marco Nolden
- Division of Medical Image Computing, German Cancer Research Center DKFZ, Heidelberg, Germany
| |
Collapse
|
54
|
Ladefoged CN, Marner L, Hindsholm A, Law I, Højgaard L, Andersen FL. Deep Learning Based Attenuation Correction of PET/MRI in Pediatric Brain Tumor Patients: Evaluation in a Clinical Setting. Front Neurosci 2019; 12:1005. [PMID: 30666184 PMCID: PMC6330282 DOI: 10.3389/fnins.2018.01005] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/13/2018] [Indexed: 11/13/2022] Open
Abstract
Aim: Positron emission tomography (PET) imaging is a useful tool for assisting in correct differentiation of tumor progression from reactive changes. O-(2-18F-fluoroethyl)-L-tyrosine (FET)-PET in combination with MRI can add valuable information for clinical decision making. Acquiring FET-PET/MRI simultaneously allows for a one-stop-shop that limits the need for a second sedation or anesthesia as with PET and MRI in sequence. PET/MRI is challenged by lack of a direct measure of photon attenuation. Accepted solutions for attenuation correction (AC) might not be applicable to pediatrics. The aim of this study was to evaluate the use of the subject-specific MR-derived AC method RESOLUTE, modified to a pediatric cohort, against the performance of an MR-AC technique based on deep learning in a pediatric brain tumor cohort. Methods: The modifications to RESOLUTE and the implementation of a deep learning method were performed using 79 pediatric patient examinations. We analyzed the 36 of these with active brain tumor area above 1 mL. We measured background (B), tumor mean and maximal activity (TMEAN, TMAX), biological tumor volume (BTV), and calculated the clinical metrics TMEAN/B and TMAX/B. Results: Overall, we found both RESOLUTE and our DeepUTE methodologies to accurately reproduce the CT-AC clinical metrics. Regardless of age, both methods were able to obtain AC maps similar to the CT-AC, albeit with DeepUTE producing the most similar based on both quantitative metrics and visual inspection. In the patient-by-patient analysis DeepUTE was the only technique with all patients inside the predefined acceptable clinical limits. It also had a higher precision with relative %-difference to the reference CT-AC (TMAX/B mean: -0.1%, CI: [-0.8%, 0.5%], p = 0.54) compared to RESOLUTE (TMAX/B mean: 0.3%, CI: [-0.6%, 1.2%], p = 0.67) and DIXON-AC (TMAX/B mean: 5.9%, CI: [4.5%, 7.4%], p < 0.0001). Conclusion: Overall, we found DeepUTE to be the AC method that most robustly reproduced the CT-AC clinical metrics per se, closely followed by RESOLUTE modified to pediatric cohorts. The added accuracy due to better noise handling of DeepUTE, ease of use, as well as the improved runtime makes DeepUTE the method of choice for PET/MRI attenuation correction.
Collapse
Affiliation(s)
- Claes Nøhr Ladefoged
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| | - Lisbeth Marner
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| | - Amalie Hindsholm
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| | - Liselotte Højgaard
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
55
|
Debus C, Afshar-Oromieh A, Floca R, Ingrisch M, Knoll M, Debus J, Haberkorn U, Abdollahi A. Feasibility and robustness of dynamic 18F-FET PET based tracer kinetic models applied to patients with recurrent high-grade glioma prior to carbon ion irradiation. Sci Rep 2018; 8:14760. [PMID: 30283013 PMCID: PMC6170489 DOI: 10.1038/s41598-018-33034-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 09/07/2018] [Indexed: 12/23/2022] Open
Abstract
The aim of this study was to analyze the robustness and diagnostic value of different compartment models for dynamic 18F-FET PET in recurrent high-grade glioma (HGG). Dynamic 18F-FET PET data of patients with recurrent WHO grade III (n:7) and WHO grade IV (n: 9) tumors undergoing re-irradiation with carbon ions were analyzed by voxelwise fitting of the time-activity curves with a simplified and an extended one-tissue compartment model (1TCM) and a two-tissue compartment model (2TCM), respectively. A simulation study was conducted to assess robustness and precision of the 2TCM. Parameter maps showed enhanced detail on tumor substructure. Neglecting the blood volume VB in the 1TCM yields insufficient results. Parameter K1 from both 1TCM and 2TCM showed correlation with overall patient survival after carbon ion irradiation (p = 0.043 and 0.036, respectively). The 2TCM yields realistic estimates for tumor blood volume, which was found to be significantly higher in WHO IV compared to WHO III (p = 0.031). Simulations on the 2TCM showed that K1 yields good accuracy and robustness while k2 showed lowest stability of all parameters. The 1TCM provides the best compromise between parameter stability and model accuracy; however application of the 2TCM is still feasible and provides a more accurate representation of tracer-kinetics at the cost of reduced robustness. Detailed tracer kinetic analysis of 18F-FET PET with compartment models holds valuable information on tumor substructures and provides additional diagnostic and prognostic value.
Collapse
Affiliation(s)
- Charlotte Debus
- German Cancer Consortium (DKTK), Heidelberg, Germany.
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Division of Molecular and Translational Radiation Oncology, Heidelberg University Medical School, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Heidelberg, Germany.
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ralf Floca
- Division of Molecular and Translational Radiation Oncology, Heidelberg University Medical School, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Heidelberg, Germany
- Division of Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Ingrisch
- Department of Radiology, University Hospital Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Maximilian Knoll
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Heidelberg University Medical School, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jürgen Debus
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Heidelberg University Medical School, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Amir Abdollahi
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Heidelberg University Medical School, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
56
|
Lohmann P, Piroth MD, Sellhaus B, Weis J, Geisler S, Oros-Peusquens AM, Mohlberg H, Amunts K, Shah NJ, Galldiks N, Langen KJ. Correlation of Dynamic O-(2-[ 18F]Fluoroethyl)-L-Tyrosine Positron Emission Tomography, Conventional Magnetic Resonance Imaging, and Whole-Brain Histopathology in a Pretreated Glioblastoma: A Postmortem Study. World Neurosurg 2018; 119:e653-e660. [PMID: 30077752 DOI: 10.1016/j.wneu.2018.07.232] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Amino acid positron emission tomography (PET) using O-(2-[18F]fluoroethyl)-L-tyrosine (FET) provides important additional information on the extent of viable tumor tissue of glioblastoma compared with magnetic resonance imaging (MRI). Especially after radiochemotherapy, progression of contrast enhancement in MRI is equivocal and may represent either tumor progression or treatment-related changes. Here, the first case comparing postmortem whole-brain histology of a patient with pretreated glioblastoma with dynamic in vivo FET PET and MRI is presented. METHODS A 61-year-old patient with glioblastoma initially underwent partial tumor resection and died 11 weeks after completion of chemoradiation with concurrent temozolomide. Three days before the patient died, a follow-up FET PET and MRI scan indicated tumor progression. Autopsy was performed 48 hours after death. After formalin fixation, a 7-cm bihemispherical segment of the brain containing the entire tumor mass was cut into 3500 consecutive 20μm coronal sections. Representative sections were stained with hematoxylin and eosin stain, cresyl violet, and glial fibrillary acidic protein immunohistochemistry. An experienced neuropathologist identified areas of dense and diffuse neoplastic infiltration, astrogliosis, and necrosis. In vivo FET PET, MRI datasets, and postmortem histology were co-registered and compared by 3 experienced physicians. RESULTS Increased uptake of FET in the area of equivocal contrast enhancement on MRI correlated very well with dense infiltration by vital tumor cells and showed tracer kinetics typical for malignant gliomas. An area of predominantly reactive astrogliosis showed only moderate uptake of FET and tracer kinetics usually observed in benign lesions. CONCLUSIONS This case report impressively documents the correct imaging of a progressive glioblastoma by FET PET.
Collapse
Affiliation(s)
- Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany.
| | - Marc D Piroth
- Department of Radiation Oncology, HELIOS Hospital Wuppertal, Wuppertal, Germany; Department of Radiation Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Bernd Sellhaus
- Institute of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Joachim Weis
- Institute of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Stefanie Geisler
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany
| | - Ana-Maria Oros-Peusquens
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany
| | - Hartmut Mohlberg
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany
| | - Katrin Amunts
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany; Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany; Department of Neurology, University of Cologne, Cologne, Germany; Center of Integrated Oncology, Universities of Cologne and Bonn, Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-1, -3, -4, -11), Forschungszentrum Juelich, Juelich, Germany; Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
57
|
Straightforward synthesis of fluorinated amino acids by Michael addition of ethyl bromodifluoroacetate to α,β-unsaturated α-amino acid derivatives. J Fluor Chem 2018. [DOI: 10.1016/j.jfluchem.2018.03.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
58
|
Blanc-Durand P, Van Der Gucht A, Verger A, Langen KJ, Dunet V, Bloch J, Brouland JP, Nicod-Lalonde M, Schaefer N, Prior JO. Voxel-based 18F-FET PET segmentation and automatic clustering of tumor voxels: A significant association with IDH1 mutation status and survival in patients with gliomas. PLoS One 2018; 13:e0199379. [PMID: 29953478 PMCID: PMC6023198 DOI: 10.1371/journal.pone.0199379] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 06/06/2018] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Aim was to develop a full automatic clustering approach of the time-activity curves (TAC) from dynamic 18F-FET PET and evaluate its association with IDH1 mutation status and survival in patients with gliomas. METHODS Thirty-seven patients (mean age: 45±13 y) with newly diagnosed gliomas and dynamic 18F-FET PET before any histopathologic investigation or treatment were retrospectively included. Each dynamic 18F-FET PET was realigned to the first image and spatially normalized in the Montreal Neurological Institute template. A tumor mask was semi-automatically generated from Z-score maps. Each brain tumor voxel was clustered in one of the 3 following centroids using dynamic time warping and k-means clustering (centroid #1: slowly increasing slope; centroid #2: rapidly increasing followed by slowly decreasing slope; and centroid #3: rapidly increasing followed by rapidly decreasing slope). The percentage of each dynamic 18F-FET TAC within tumors and other conventional 18F-FET PET parameters (maximum and mean tumor-to-brain ratios [TBRmax and TBRmean], time-to-peak [TTP] and slope) was compared between wild-type and IDH1 mutant tumors. Their prognostic value was assessed in terms of progression free-survival (PFS) and overall survival (OS) by Kaplan-Meier estimates. RESULTS Twenty patients were IDH1 wild-type and 17 IDH1 mutant. Higher percentage of centroid #1 and centroid #3 within tumors were positively (P = 0.016) and negatively (P = 0.01) correlated with IDH1 mutated status. Also, TBRmax, TBRmean, TTP, and slope discriminated significantly between tumors with and without IDH1 mutation (P range 0.01 to 0.04). Progression occurred in 22 patients (59%) at a median of 13.1 months (7.6-37.6 months) and 13 patients (35%) died from tumor progression. Patients with a percentage of centroid #1 > 90% had a longer survival compared with those with a percentage of centroid #1 < 90% (P = 0.003 for PFS and P = 0.028 for OS). This remained significant after stratification on IDH1 mutation status (P = 0.029 for PFS and P = 0.034 for OS). Compared to other conventional 18F-FET PET parameters, TTP and slope were associated with PFS and OS (P range 0.009 to 0.04). CONCLUSIONS Based on dynamic 18F-FET PET acquisition, we developed a full automatic clustering approach of TAC which appears to be a valuable noninvasive diagnostic and prognostic marker in patients with gliomas.
Collapse
Affiliation(s)
- Paul Blanc-Durand
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Axel Van Der Gucht
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Antoine Verger
- Department of Nuclear Medicine and Molecular Imaging, Nancy University Hospital, Nancy, France
| | - Karl-Josef Langen
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Vincent Dunet
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Jocelyne Bloch
- Department of Neurosurgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Jean-Philippe Brouland
- Department of Pathology and Laboratory Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Marie Nicod-Lalonde
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - John O. Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
59
|
Lohmann P, Kocher M, Steger J, Galldiks N. Radiomics derived from amino-acid PET and conventional MRI in patients with high-grade gliomas. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2018; 62:272-280. [PMID: 29869488 DOI: 10.23736/s1824-4785.18.03095-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Radiomics is a technique that uses high-throughput computing to extract quantitative features from tomographic medical images such as MRI and PET that usually are beyond visual perception. Importantly, the radiomics approach can be performed using neuroimages that have already been acquired during the routine follow-up of the patients allowing an additional data evaluation at low cost. In Neuro-Oncology, these features can potentially be used for differential diagnosis of newly diagnosed cerebral lesions suggestive for brain tumors or for the prediction of response to a neurooncological treatment option. Furthermore, especially in the light of the recent update of the World Health Organization classification of brain tumors, radiomics also has the potential to non-invasively assess important prognostic and predictive molecular markers such as a mutation in the isocitrate dehydrogenase gene or a 1p/19q codeletion which are not accessible by conventional visual interpretation of MRI or PET findings. This review summarizes the current status of the rapidly evolving field of radiomics with a special focus on patients with high-grade gliomas.
Collapse
Affiliation(s)
- Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Juelich, Germany -
| | - Martin Kocher
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Juelich, Germany.,Department of Stereotaxy and Functional Neurosurgery, University of Cologne, Cologne, Germany
| | - Jan Steger
- Department of Neurology, University of Cologne, Cologne, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Juelich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany.,Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany
| |
Collapse
|
60
|
|
61
|
Impact of 18F-FET PET on Target Volume Definition and Tumor Progression of Recurrent High Grade Glioma Treated with Carbon-Ion Radiotherapy. Sci Rep 2018; 8:7201. [PMID: 29740097 PMCID: PMC5940831 DOI: 10.1038/s41598-018-25350-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 04/17/2018] [Indexed: 11/26/2022] Open
Abstract
High-precision radiotherapy (HPR) of recurrent high grade glioma (HGG) requires accurate spatial allocation of these infiltrative tumors. We investigated the impact of 18F-FET PET on tumor delineation and progression of recurrent HGG after HPR with carbon ions. T1 contrast enhanced MRI and 18F-FET-PET scans of 26 HGG patients were fused with radiotherapy planning volumes. PET-positive (PET+) tumor volumes using different isocontours (I%) were systematically investigated and compared with MRI-derived gross tumor volumes (GTV). Standardized uptake ratios (SUR) were further correlated with GTV and tumor progression patterns. In grade IV glioma, SUR > 2.92 significantly correlated with poor median overall survival (6.5 vs 13.1 months, p = 0.00016). We found no reliable SUR cut-off criteria for definition of PET+ volumes. Overall conformity between PET and MRI-based contours was low, with maximum conformities between 0.42–0.51 at I40%. The maximum sensitivity and specificity for PET+ volumes outside of GTV predicting tumor progression were 0.16 (I40%) and 0.52 (I50%), respectively. In 75% of cases, FLAIR hyperintense area covered over 80% of PET+ volumes. 18F-FET-PET derived SUR has a prognostic impact in grade IV glioma. The value of substantial mismatches between MRI-based GTV and PET+ volumes to improve tumor delineation in radiotherapy awaits further validation in randomized prospective trials.
Collapse
|
62
|
Verger A, Arbizu J, Law I. Role of amino-acid PET in high-grade gliomas: limitations and perspectives. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2018; 62:254-266. [PMID: 29696948 DOI: 10.23736/s1824-4785.18.03092-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Positron emission tomography (PET) using radiolabeled amino-acids was recently recommended by the Response Assessment in Neuro-Oncology (RANO) working group as an additional tool in the diagnostic assessment of brain tumors. The aim of this review is to summarize available literature data on the role of amino-acid PET imaging in high-grade gliomas (HGGs), with regard to diagnosis, treatment planning and follow-up of these tumors. Indeed, amino-acid PET applications are multiple throughout the evolution of HGGs. However, certain limitations such as lack of specificity, uncertain value for grading and prognostication or the limited data for treatment monitoring should to be taken into account, the latter of which are further developed in this review. Notwithstanding these limitations, amino-acid PET is becoming increasingly accessible in many nuclear medicine centers. Larger prospective cohort prospective studies are thus needed in order to increase the clinical value of this modality and enable its extended use to the largest number of patients.
Collapse
Affiliation(s)
- Antoine Verger
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, CHRU Nancy, Lorraine University, Nancy, France - .,IADI, INSERM, Lorraine University, Nancy, France -
| | - Javier Arbizu
- Department of Nuclear Medicine, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
63
|
Blanc-Durand P, Van Der Gucht A, Schaefer N, Itti E, Prior JO. Automatic lesion detection and segmentation of 18F-FET PET in gliomas: A full 3D U-Net convolutional neural network study. PLoS One 2018; 13:e0195798. [PMID: 29652908 PMCID: PMC5898737 DOI: 10.1371/journal.pone.0195798] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/29/2018] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION Amino-acids positron emission tomography (PET) is increasingly used in the diagnostic workup of patients with gliomas, including differential diagnosis, evaluation of tumor extension, treatment planning and follow-up. Recently, progresses of computer vision and machine learning have been translated for medical imaging. Aim was to demonstrate the feasibility of an automated 18F-fluoro-ethyl-tyrosine (18F-FET) PET lesion detection and segmentation relying on a full 3D U-Net Convolutional Neural Network (CNN). METHODS All dynamic 18F-FET PET brain image volumes were temporally realigned to the first dynamic acquisition, coregistered and spatially normalized onto the Montreal Neurological Institute template. Ground truth segmentations were obtained using manual delineation and thresholding (1.3 x background). The volumetric CNN was implemented based on a modified Keras implementation of a U-Net library with 3 layers for the encoding and decoding paths. Dice similarity coefficient (DSC) was used as an accuracy measure of segmentation. RESULTS Thirty-seven patients were included (26 [70%] in the training set and 11 [30%] in the validation set). All 11 lesions were accurately detected with no false positive, resulting in a sensitivity and a specificity for the detection at the tumor level of 100%. After 150 epochs, DSC reached 0.7924 in the training set and 0.7911 in the validation set. After morphological dilatation and fixed thresholding of the predicted U-Net mask a substantial improvement of the DSC to 0.8231 (+ 4.1%) was noted. At the voxel level, this segmentation led to a 0.88 sensitivity [95% CI, 87.1 to, 88.2%] a 0.99 specificity [99.9 to 99.9%], a 0.78 positive predictive value: [76.9 to 78.3%], and a 0.99 negative predictive value [99.9 to 99.9%]. CONCLUSIONS With relatively high performance, it was proposed the first full 3D automated procedure for segmentation of 18F-FET PET brain images of patients with different gliomas using a U-Net CNN architecture.
Collapse
Affiliation(s)
- Paul Blanc-Durand
- Department of Nuclear Medicine, Henri Mondor University Hospital, Créteil, France
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
- * E-mail: (PBD); (AVDG)
| | - Axel Van Der Gucht
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
- * E-mail: (PBD); (AVDG)
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Emmanuel Itti
- Department of Nuclear Medicine, Henri Mondor University Hospital, Créteil, France
| | - John O. Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
64
|
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
|
65
|
Bashir A, Brennum J, Broholm H, Law I. The diagnostic accuracy of detecting malignant transformation of low-grade glioma using O-(2-[18F]fluoroethyl)-l-tyrosine positron emission tomography: a retrospective study. J Neurosurg 2018; 130:451-464. [PMID: 29624154 DOI: 10.3171/2017.8.jns171577] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/02/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The diagnostic accuracy of O-(2-[18F]fluoroethyl)-l-tyrosine (FET) PET scanning in detecting the malignant transformation of low-grade gliomas (LGGs) is controversial. In this study, the authors retrospectively assessed the diagnostic potential of FET PET in patients with MRI-suspected malignant progression of LGGs that had previously been treated and the relationship between FET uptake and MRI and molecular biomarkers. METHODS Forty-two patients who had previously undergone surgical or multimodal treatment for a histologically verified LGG were referred for FET PET assessment because of clinical signs and/or MRI findings suggestive of tumor progression. Maximal and mean tumor-to-brain ratios (TBRmax and TBRmean, respectively) on FET PET as well as kinetic FET PET parameters (time to peak [TTP] and time-activity curve [TAC]) were determined. Final diagnoses were confirmed histologically. The diagnostic accuracy of FET parameters, separately and combined, for the detection of malignant progression was evaluated using receiver operating characteristic (ROC) curve analysis. Possible predictors that might influence the diagnostic accuracy of FET PET were assessed using multiple linear regression analysis. Spearman’s rank correlation r method was applied to determine the correlation between TBRmax and TAC, and molecular biomarkers from tumor tissues. RESULTS A total of 47 FET PET scans were obtained and showed no significant association between FET parameters and contrast enhancement on MRI. ROC curve analyses overall were unable to demonstrate any significant differentiation between nontransformed LGGs and LGGs that had transformed to high-grade gliomas when evaluating FET parameters separately or combined. After excluding the oligodendroglial subgroup, a significant difference was observed between nontransformed and transformed LGGs when combining FET parameters (i.e., TBRmax > 1.6, TAC describing a plateau or decreasing pattern, and TTP < 25 minutes), with the best result yielded by a combined analysis of TBRmax > 1.6 and TAC with a plateau or decreasing pattern (sensitivity 75% and specificity 83%, p = 0.003). The difference was even greater when patients who had previously undergone oncological treatment were also excluded (sensitivity 93% and specificity 100%, p = 0.001). Multiple linear regression analysis revealed that the presence of an oligodendroglial component (p = 0.029), previous oncological treatment (p = 0.039), and the combined FET parameters (p = 0.027) were significant confounding factors in the detection of malignant progression. TBRmax was positively correlated with increasing cell density (p = 0.040) and inversely correlated with IDH1 mutation (p = 0.006). CONCLUSIONS A single FET PET scan obtained at the time of radiological and/or clinical progression seems to be of limited value in distinguishing transformed from nontransformed LGGs, especially if knowledge of the primary tumor histopathology is not known. Therefore, FET PET imaging alone is not adequate to replace histological confirmation, but it may provide valuable information on the location and delineation of active tumor tissue, as well as an assessment of tumor biology in a subgroup of LGGs.
Collapse
Affiliation(s)
- Asma Bashir
- Departments of1Clinical Physiology, Nuclear Medicine & PET
| | | | - Helle Broholm
- 3Pathology, National University Hospital, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Ian Law
- Departments of1Clinical Physiology, Nuclear Medicine & PET
| |
Collapse
|
66
|
Population Pharmacokinetic Approach Applied to Positron Emission Tomography: Computed Tomography for Tumor Tissue Identification in Patients with Glioma. Clin Pharmacokinet 2018; 56:953-961. [PMID: 27995528 DOI: 10.1007/s40262-016-0490-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND AIMS 18F-fluoro-ethyl-tyrosine (FET) is a radiopharmaceutical used in positron emission tomography (PET)-computed tomography in patients with glioma. We propose an original approach combining a radiotracer-pharmacokinetic exploration performed at the voxel level (three-dimensional pixel) and voxel classification to identify tumor tissue. Our methodology was validated using the standard FET-PET approach and magnetic resonance imaging (MRI) data acquired according to the current clinical practices. METHODS FET-PET and MRI data were retrospectively analyzed in ten patients presenting with progressive high-grade glioma. For FET-PET exploration, radioactivity acquisition started 15 min after radiotracer injection, and was measured each 5 min during 40 min. The tissue segmentation relies on population pharmacokinetic modeling with dependent individuals (voxels). This model can be approximated by a linear mixed-effects model. The tumor volumes estimated by our approach were compared with those determined with the current clinical techniques, FET-PET standard approach (i.e., a cumulated value of FET signal is computed during a time interval) and MRI sequences (T1 and T2/fluid-attenuated inversion recovery [FLAIR]), used as references. The T1 sequence is useful to identify highly vascular tumor and necrotic tissues, while the T2/FLAIR sequence is useful to isolate infiltration and edema tissue located around the tumor. RESULTS With our kinetic approach, the volumes of tumor tissue were larger than the tissues identified by the standard FET-PET and MRI T1, while they were smaller than those determined with MRI T2/FLAIR. CONCLUSION Our results revealed the presence of suspected tumor voxels not identified by the standard PET approach.
Collapse
|
67
|
Use of FET PET in glioblastoma patients undergoing neurooncological treatment including tumour-treating fields: initial experience. Eur J Nucl Med Mol Imaging 2018; 45:1626-1635. [DOI: 10.1007/s00259-018-3992-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/06/2018] [Indexed: 10/17/2022]
|
68
|
Schröder S, Wenzel B, Deuther-Conrad W, Teodoro R, Kranz M, Scheunemann M, Egerland U, Höfgen N, Briel D, Steinbach J, Brust P. Investigation of an 18F-labelled Imidazopyridotriazine for Molecular Imaging of Cyclic Nucleotide Phosphodiesterase 2A. Molecules 2018; 23:molecules23030556. [PMID: 29498659 PMCID: PMC6017663 DOI: 10.3390/molecules23030556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/15/2018] [Accepted: 02/23/2018] [Indexed: 11/16/2022] Open
Abstract
Specific radioligands for in vivo visualization and quantification of cyclic nucleotide phosphodiesterase 2A (PDE2A) by positron emission tomography (PET) are increasingly gaining interest in brain research. Herein we describe the synthesis, the 18F-labelling as well as the biological evaluation of our latest PDE2A (radio-)ligand 9-(5-Butoxy-2-fluorophenyl)-2-(2-([18F])fluoroethoxy)-7-methylimidazo[5,1-c]pyrido[2,3-e][1,2,4]triazine (([18F])TA5). It is the most potent PDE2A ligand out of our series of imidazopyridotriazine-based derivatives so far (IC50 hPDE2A = 3.0 nM; IC50 hPDE10A > 1000 nM). Radiolabelling was performed in a one-step procedure starting from the corresponding tosylate precursor. In vitro autoradiography on rat and pig brain slices displayed a homogenous and non-specific binding of the radioligand. Investigation of stability in vivo by reversed-phase HPLC (RP-HPLC) and micellar liquid chromatography (MLC) analyses of plasma and brain samples obtained from mice revealed a high fraction of one main radiometabolite. Hence, we concluded that [18F]TA5 is not appropriate for molecular imaging of PDE2A neither in vitro nor in vivo. Our ongoing work is focusing on further structurally modified compounds with enhanced metabolic stability.
Collapse
Affiliation(s)
- Susann Schröder
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
- Correspondence: ; Tel.: +49-341-234-179-4631
| | - Barbara Wenzel
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Winnie Deuther-Conrad
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Rodrigo Teodoro
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Mathias Kranz
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Matthias Scheunemann
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Ute Egerland
- BioCrea GmbH, Radebeul 01445, Germany; (U.E.); (N.H.)
| | | | - Detlef Briel
- Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Faculty of Medicine, Leipzig University, Leipzig 04103, Germany;
| | - Jörg Steinbach
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Peter Brust
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| |
Collapse
|
69
|
Hybrid MR-PET of brain tumours using amino acid PET and chemical exchange saturation transfer MRI. Eur J Nucl Med Mol Imaging 2018; 45:1031-1040. [PMID: 29478081 DOI: 10.1007/s00259-018-3940-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/04/2018] [Indexed: 10/18/2022]
Abstract
PURPOSE PET using radiolabelled amino acids has become a promising tool in the diagnostics of gliomas and brain metastasis. Current research is focused on the evaluation of amide proton transfer (APT) chemical exchange saturation transfer (CEST) MR imaging for brain tumour imaging. In this hybrid MR-PET study, brain tumours were compared using 3D data derived from APT-CEST MRI and amino acid PET using O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET). METHODS Eight patients with gliomas were investigated simultaneously with 18F-FET PET and APT-CEST MRI using a 3-T MR-BrainPET scanner. CEST imaging was based on a steady-state approach using a B1 average power of 1μT. B0 field inhomogeneities were corrected a Prametric images of magnetisation transfer ratio asymmetry (MTRasym) and differences to the extrapolated semi-solid magnetisation transfer reference method, APT# and nuclear Overhauser effect (NOE#), were calculated. Statistical analysis of the tumour-to-brain ratio of the CEST data was performed against PET data using the non-parametric Wilcoxon test. RESULTS A tumour-to-brain ratio derived from APT# and 18F-FET presented no significant differences, and no correlation was found between APT# and 18F-FET PET data. The distance between local hot spot APT# and 18F-FET were different (average 20 ± 13 mm, range 4-45 mm). CONCLUSION For the first time, CEST images were compared with 18F-FET in a simultaneous MR-PET measurement. Imaging findings derived from18F-FET PET and APT CEST MRI seem to provide different biological information. The validation of these imaging findings by histological confirmation is necessary, ideally using stereotactic biopsy.
Collapse
|
70
|
Lerche CW, Kaltsas T, Caldeira L, Scheins J, Rota Kops E, Tellmann L, Pietrzyk U, Herzog H, Shah NJ. PET attenuation correction for rigid MR Tx/Rx coils from 176Lu background activity. Phys Med Biol 2018; 63:035039. [PMID: 29328049 DOI: 10.1088/1361-6560/aaa72a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
One challenge for PET-MR hybrid imaging is the correction for attenuation of the 511 keV annihilation radiation by the required RF transmit and/or RF receive coils. Although there are strategies for building PET transparent Tx/Rx coils, such optimised coils still cause significant attenuation of the annihilation radiation leading to artefacts and biases in the reconstructed activity concentrations. We present a straightforward method to measure the attenuation of Tx/Rx coils in simultaneous MR-PET imaging based on the natural 176Lu background contained in the scintillator of the PET detector without the requirement of an external CT scanner or PET scanner with transmission source. The method was evaluated on a prototype 3T MR-BrainPET produced by Siemens Healthcare GmbH, both with phantom studies and with true emission images from patient/volunteer examinations. Furthermore, the count rate stability of the PET scanner and the x-ray properties of the Tx/Rx head coil were investigated. Even without energy extrapolation from the two dominant γ energies of 176Lu to 511 keV, the presented method for attenuation correction, based on the measurement of 176Lu background attenuation, shows slightly better performance than the coil attenuation correction currently used. The coil attenuation correction currently used is based on an external transmission scan with rotating 68Ge sources acquired on a Siemens ECAT HR + PET scanner. However, the main advantage of the presented approach is its straightforwardness and ready availability without the need for additional accessories.
Collapse
Affiliation(s)
- Christoph W Lerche
- Medical Imaging Physics Department, Institute for Neuroscience and Medicine (INM-4), Forschungszentrum Jülich GmbH, Jülich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
71
|
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
|
72
|
Sun A, Liu X, Tang G. Carbon-11 and Fluorine-18 Labeled Amino Acid Tracers for Positron Emission Tomography Imaging of Tumors. Front Chem 2018; 5:124. [PMID: 29379780 PMCID: PMC5775220 DOI: 10.3389/fchem.2017.00124] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/12/2017] [Indexed: 12/12/2022] Open
Abstract
Tumor cells have an increased nutritional demand for amino acids (AAs) to satisfy their rapid proliferation. Positron-emitting nuclide labeled AAs are interesting probes and are of great importance for imaging tumors using positron emission tomography (PET). Carbon-11 and fluorine-18 labeled AAs include the [1-11C] AAs, labeling alpha-C- AAs, the branched-chain of AAs and N-substituted carbon-11 labeled AAs. These tracers target protein synthesis or amino acid (AA) transport, and their uptake mechanism mainly involves AA transport. AA PET tracers have been widely used in clinical settings to image brain tumors, neuroendocrine tumors, prostate cancer, breast cancer, non-small cell lung cancer (NSCLC) and hepatocellular carcinoma. This review focuses on the fundamental concepts and the uptake mechanism of AAs, AA PET tracers and their clinical applications.
Collapse
Affiliation(s)
- Aixia Sun
- Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiang Liu
- Department of Anesthesiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ganghua Tang
- Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
73
|
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
|
74
|
Langen KJ, Weirich C, Rota Kops E, Kaffanke J, Tellmann L, Scheins J, Neuner I, Stoffels G, Fischer K, Caldeira L, Coenen HH, Shah NJ, Herzog H. High resolution BrainPET combined with simultaneous MRI. Nuklearmedizin 2017; 50:74-82. [DOI: 10.3413/nukmed-0347-10-09] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 01/05/2011] [Indexed: 11/20/2022]
Abstract
SummaryAfter the successful clinical introduction of PET/CT, a novel hybrid imaging technology combining PET with the versatile attributes of MRI is emerging. At the Forschungszentrum Jülich, one of four prototypes available worldwide combining a commercial 3T MRI with a newly developed BrainPET insert has been installed, allowing simultaneous data acquisition with PET and MRI. The BrainPET is equipped with LSO crystals of 2.5 mm width and Avalanche photodiodes (APD) as readout electronics. Here we report on some performance characteristics obtained by phantom studies and also on the initial BrainPET studies on various patients as compared with a conventional HR+ PET-only scanner. Material, methods: The radiotracers [18F]-fluoroethyl- tyrosine (FET), [11C]-flumazenil and [18F]-FP-CIT were applied. Results: Comparing komthe PET data obtained with the BrainPET to those of the HR+ scanner demonstrated the high image quality and the superior resolution capability of the BrainPET. Furthermore, it is shown that various MR images of excellent quality could be acquired simultaneously with BrainPET scans without any relevant artefacts. Discussion, conclusion: Initial experiences with the hybrid MRI/BrainPET indicate a promising basis for further developments of this unique technique allowing simultaneous PET imaging combined with both anatomical and functional MRI.
Collapse
|
75
|
Abstract
Positron emission tomography-computed tomography is a medical imaging method measuring the activity of a radiotracer chosen to accumulate in cancer cells. A recent trend of medical imaging analysis is to account for the radiotracer's pharmacokinetic properties at a voxel (three-dimensional-pixel) level to separate the different tissues. These analyses are closely linked to population pharmacokinetic-pharmacodynamic modelling. Kineticists possess the cultural background to improve medical imaging analysis. This article stresses the common points with population pharmacokinetics and highlights the methodological locks that need to be lifted.
Collapse
|
76
|
Dittmar JO, Kratochwil C, Dittmar A, Welzel T, Habermehl D, Rieken S, Giesel FL, Haberkorn U, Debus J, Combs SE. First intraindividual comparison of contrast-enhanced MRI, FET- and DOTATOC- PET in patients with intracranial meningiomas. Radiat Oncol 2017; 12:169. [PMID: 29110720 PMCID: PMC5674744 DOI: 10.1186/s13014-017-0913-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/27/2017] [Indexed: 11/26/2022] Open
Abstract
Background For irradiation treatment planning of meningiomas the use of PET-scans is well established. The most frequently used tracers are either based on amino acids or the somatostatin receptor ligand DOTATOC. Since up to now no inter-institutionally accepted standard PET-tracer has been defined, the aim of this study was to evaluate the influence of these different types of PET-tracers on the GTV-definition. Methods Twenty-one patients suffering from intracranial meningiomas underwent CT, MRI, FET- and DOTATOC-PET. First, tumour extension was delineated after image-fusion of CT and MRI (GTVCT/MRI). Then distinct GTVs based either on FET- or DOTATOC-PET were contoured and compared with each other as well with GTVCT/MRI. Results Every tumour showed typical enhancement of DOTATOC, but two meningiomas remained FET-negative. The mean relative overlap volume of GTVFET and GTVDOTATOC was only 41.9% and there was a significantly stronger correlation between GTVCT/MRI and GTVDOTATOC than between GTVCT/MRI and GTVFET. Conclusions Further investigations are necessary to clarify the minor conformity of DOTATOC- and FET-PET in meningiomas. Because of the receptor targeting, DOTATOC is known to be more specific for meningiomas and will remain the standard in our institution with the known limitation in areas nearby the pituitary gland.
Collapse
Affiliation(s)
- Jan Oliver Dittmar
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Ismaninger Str. 22, 81675, Munich, Germany.,Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Anne Dittmar
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Thomas Welzel
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Daniel Habermehl
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Ismaninger Str. 22, 81675, Munich, Germany.,Institute for Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764, Oberschleißheim, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany
| | - Stefan Rieken
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Frederik L Giesel
- Department of Nuclear Medicine, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Ismaninger Str. 22, 81675, Munich, Germany.
| |
Collapse
|
77
|
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
|
78
|
Abstract
INTRODUCTION Initial diagnostics and follow-up of gliomas is usually based on contrast-enhanced MRI. However, the capacity of standard MRI to differentiate neoplastic tissue from posttherapeutic effects such as pseudoprogression is limited. Advanced neuroimaging methods may provide relevant additional information, which allow for a more accurate diagnosis especially in clinically equivocal situations. This review article focuses predominantly on PET using radiolabeled amino acids and advanced MRI techniques such as perfusion-weighted imaging (PWI) and summarizes the efforts of these methods regarding the identification of pseudoprogression after glioma therapy. Areas covered: The current literature on pseudoprogression in the field of brain tumors, with a focus on gliomas is summarized. A literature search was performed using the terms 'pseudoprogression', 'temozolomide', 'glioblastoma', 'PET', 'PWI', 'radiochemotherapy', and derivations thereof. Expert commentary: The present literature provides strong evidence that PWI MRI and amino acid PET can be of great value by providing valuable additional diagnostic information in order to overcome the diagnostic challenge of pseudoprogression. Despite various obstacles such as the still limited availability of amino acid PET and the lack of standardization of PWI, the diagnostic improvement probably results in relevant benefits for brain tumor patients and justifies a more widespread use of these diagnostic tools.
Collapse
Affiliation(s)
- Norbert Galldiks
- a Department of Neurology , University of Cologne , Cologne , Germany.,b Institute of Neuroscience and Medicine , Forschungszentrum Jülich , Jülich , Germany.,c Center of Integrated Oncology (CIO) , Universities of Cologne and Bonn , Cologne , Germany
| | - Martin Kocher
- d Department of Radiation Oncology , University of Cologne , Cologne , Germany
| | - Karl-Josef Langen
- b Institute of Neuroscience and Medicine , Forschungszentrum Jülich , Jülich , Germany.,e Department of Nuclear Medicine , University of Aachen , Aachen , Germany
| |
Collapse
|
79
|
Lakshminarayanan N, Kumar A, Roy S, Pawar Y, Chaudhari P, Rajan M. Improved method for preparing Ni(II) complex of (S)-tyrosine Schiff base and its use in the automated synthesis of O-(2′-[ 18 F]fluoroethyl)- l -tyrosine using solid-phase extraction purification. Appl Radiat Isot 2017; 127:122-129. [DOI: 10.1016/j.apradiso.2017.05.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 04/13/2017] [Accepted: 05/21/2017] [Indexed: 11/27/2022]
|
80
|
Comparison of 18F-FET PET and perfusion-weighted MRI for glioma grading: a hybrid PET/MR study. Eur J Nucl Med Mol Imaging 2017; 44:2257-2265. [PMID: 28831534 DOI: 10.1007/s00259-017-3812-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022]
Abstract
PURPOSE Both perfusion-weighted MR imaging (PWI) and O-(2-18F-fluoroethyl)-L-tyrosine PET (18F-FET) provide grading information in cerebral gliomas. The aim of this study was to compare the diagnostic value of 18F-FET PET and PWI for tumor grading in a series of patients with newly diagnosed, untreated gliomas using an integrated PET/MR scanner. METHODS Seventy-two patients with untreated gliomas [22 low-grade gliomas (LGG), and 50 high-grade gliomas (HGG)] were investigated with 18F-FET PET and PWI using a hybrid PET/MR scanner. After visual inspection of PET and PWI maps (rCBV, rCBF, MTT), volumes of interest (VOIs) with a diameter of 16 mm were centered upon the maximum of abnormality in the tumor area in each modality and the contralateral unaffected hemisphere. Mean and maximum tumor-to-brain ratios (TBRmean, TBRmax) were calculated. In addition, Time-to-Peak (TTP) and slopes of time-activity curves were calculated for 18F-FET PET. Diagnostic accuracies of 18F-FET PET and PWI for differentiating low-grade glioma (LGG) from high-grade glioma (HGG) were evaluated by receiver operating characteristic analyses (area under the curve; AUC). RESULTS The diagnostic accuracy of 18F-FET PET and PWI to discriminate LGG from HGG was similar with highest AUC values for TBRmean and TBRmax of 18F-FET PET uptake (0.80, 0.83) and for TBRmean and TBRmax of rCBV (0.80, 0.81). In case of increased signal in the tumor area with both methods (n = 32), local hot-spots were incongruent in 25 patients (78%) with a mean distance of 10.6 ± 9.5 mm. Dynamic FET PET and combination of different parameters did not further improve diagnostic accuracy. CONCLUSIONS Both 18F-FET PET and PWI discriminate LGG from HGG with similar diagnostic performance. Regional abnormalities in the tumor area are usually not congruent indicating that tumor grading by 18F-FET PET and PWI is based on different pathophysiological phenomena.
Collapse
|
81
|
Imperiale A, Boisson F, Kreutter G, Goichot B, Namer IJ, Bachellier P, Laquerriere P, Kessler L, Marchand P, Brasse D. O-(2- 18F-fluoroethyl)-l-tyrosine ( 18F-FET) uptake in insulinoma: first results from a xenograft mouse model and from human. Nucl Med Biol 2017; 53:21-28. [PMID: 28793277 DOI: 10.1016/j.nucmedbio.2017.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Herein we have evaluated the uptake of O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) in insulinoma in comparison with those of 6-18F-fluoro-3,4-dihydroxy-l-phenylalanine (18F-FDOPA) providing first data from both murine xenograft model and one patient with proved endogenous hyperinsulinemic hypoglycemia. METHODS Dynamic 18F-FET and carbidopa-assisted 18F-FDOPA PET were performed on tumor-bearing nude mice after subcutaneous injection of RIN-m5F murine beta cells and on a 30-year-old man with type-1 multiple endocrine neoplasia and hyperinsulinemic hypoglycemia defined by a positive fasting test. RESULTS Seven and three nude mice bearing a RIN-m5F insulinoma xenograft were respectively studied by 18F-FET and 18F-FDOPA μPET. Insulinoma xenograft was detected in all the imaged animals. Xenograft was characterized by an early but moderate increase of 18F-FET uptake followed by a slight decline of uptake intensity during the 20 min dynamic acquisition. Tumoral radiotracer peak intensity and the highest tumor-to-background contrast were reached about 5 minutes after 18F-FET iv. injection (mean SUV: 1.21 ± 0.10). The biodistribution of 18F-FET and 18F-FDOPA and their dynamic tumoral uptake profile and intensity were similar. In the examined patient, 18F-FDOPA and 18F-FET PET/CT showed one concordant focal area of well-defined increased uptake in the pancreatic tail corresponding to 11 mm histologically proved insulinoma. The SUVmax tumor to liver ratio was 1.5, 1.1 for 18F-FDOPA, 1.1, 1 for 18F-FET at early (0-5 min post injection) and delayed (5-20 min post injection) PET/CT acquisition, respectively. Despite the relatively low tumoral uptake intensity, insulinoma was clearly identified due to the low background in the pancreas. At the contrary, no 18F-FDOPA or 18F-FET tumoral uptake was revealed on whole-body PET/CT images performed about 30 min after radiotracer administration. Note of worth, the dynamic uptake pattern of 18F-FET and 18F-FDOPA were similar between human insulinoma and mice xenograft tumor. CONCLUSION 18F-FET PET compared equally to 18F-FDOPA PET in a preclinical RIN-m5F murine model of insulinoma and in one patient with insulinoma-related hypoglycemia. However, in both cases, the tumoral uptake intensity was moderate and the tumor was only visible until 20 min after radiotracer injection. Hence, caution should be taken before asserting the translational relevance of our results in the clinical practices. However, the structural analogies between 18F-FET and 18F-FDOPA as well as the limited pancreatic uptake of 18F-FET in human, encourage evaluating 18F-FET as diagnostic radiotracer for insulinoma detection in further prospective studies involving large cohorts of patients.
Collapse
Affiliation(s)
- Alessio Imperiale
- Biophysics and Nuclear Medicine, Strasbourg University Hospitals, Strasbourg, France; ICube, CNRS/UMR 7357, Strasbourg University, Strasbourg, France; Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France; Université de Strasbourg, CNRS, IPHC, UMR 7178, F-67000, Strasbourg, France.
| | - Frédéric Boisson
- Université de Strasbourg, CNRS, IPHC, UMR 7178, F-67000, Strasbourg, France
| | - Guillaume Kreutter
- Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France; EA7293, Vascular and Tissular Stress in Transplantation, Illkirch, France
| | - Bernard Goichot
- Internal Medicine, Strasbourg University Hospitals, Strasbourg, France
| | - Izzie Jacques Namer
- Biophysics and Nuclear Medicine, Strasbourg University Hospitals, Strasbourg, France; ICube, CNRS/UMR 7357, Strasbourg University, Strasbourg, France; Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France
| | - Philippe Bachellier
- Visceral Surgery and Transplantation, Strasbourg University Hospitals, Strasbourg, France
| | | | - Laurence Kessler
- Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France; EA7293, Vascular and Tissular Stress in Transplantation, Illkirch, France; Diabetology, Strasbourg University Hospitals, Strasbourg, France
| | - Patrice Marchand
- Université de Strasbourg, CNRS, IPHC, UMR 7178, F-67000, Strasbourg, France
| | - David Brasse
- Université de Strasbourg, CNRS, IPHC, UMR 7178, F-67000, Strasbourg, France
| |
Collapse
|
82
|
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
|
83
|
Richard MA, Fouquet JP, Lebel R, Lepage M. Determination of an Optimal Pharmacokinetic Model of 18F-FET for Quantitative Applications in Rat Brain Tumors. J Nucl Med 2017; 58:1278-1284. [PMID: 28765227 DOI: 10.2967/jnumed.116.180612] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 03/16/2017] [Indexed: 02/03/2023] Open
Abstract
O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) is a radiolabeled artificial amino acid used in PET for tumor delineation and grading. The present study compares different kinetic models to determine which are more appropriate for 18F-FET in rats. Methods: Rats were implanted with F98 glioblastoma cells in the right hemisphere and scanned 9-15 d later. PET data were acquired during 50 min after a 1-min bolus of 18F-FET. Arterial blood samples were drawn for arterial input function determination. Two compartmental pharmacokinetic models were tested: the 2-tissue model and the 1-tissue model. Their performance at fitting concentration curves from regions of interest was evaluated using the Akaike information criterion, F test, and residual plots. Graphical models were assessed qualitatively. Results: Metrics indicated that the 2-tissue model was superior to the 1-tissue model for the current dataset. The 2-tissue model allowed adequate decoupling of 18F-FET perfusion and internalization by cells in the different regions of interest. Of the 2 graphical models tested, the Patlak plot provided adequate results for the tumor and brain, whereas the Logan plot was appropriate for muscles. Conclusion: The 2-tissue-compartment model is appropriate to quantify the perfusion and internalization of 18F-FET by cells in various tissues of the rat, whereas graphical models provide a global measure of uptake.
Collapse
Affiliation(s)
- Marie Anne Richard
- Centre d'imagerie moléculaire de Sherbrooke, Département de médecine nucléaire et radiobiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jérémie P Fouquet
- Centre d'imagerie moléculaire de Sherbrooke, Département de médecine nucléaire et radiobiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Réjean Lebel
- Centre d'imagerie moléculaire de Sherbrooke, Département de médecine nucléaire et radiobiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Martin Lepage
- Centre d'imagerie moléculaire de Sherbrooke, Département de médecine nucléaire et radiobiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
84
|
Filss CP, Cicone F, Shah NJ, Galldiks N, Langen KJ. Amino acid PET and MR perfusion imaging in brain tumours. Clin Transl Imaging 2017; 5:209-223. [PMID: 28680873 PMCID: PMC5487907 DOI: 10.1007/s40336-017-0225-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/28/2017] [Indexed: 12/17/2022]
Abstract
Purpose Despite the excellent capacity of the conventional MRI to image brain tumours, problems remain in answering a number of critical diagnostic questions. To overcome these diagnostic shortcomings, PET using radiolabeled amino acids and perfusion-weighted imaging (PWI) are currently under clinical evaluation. The role of amino acid PET and PWI in different diagnostic challenges in brain tumours is controversial. Methods Based on the literature and experience of our centres in correlative imaging with PWI and PET using O-(2-[18F]fluoroethyl)-l-tyrosine or 3,4-dihydroxy-6-[18F]-fluoro-l-phenylalanine, the current role and shortcomings of amino acid PET and PWI in different diagnostic challenges in brain tumours are reviewed. Literature searches were performed on PubMed, and additional literature was retrieved from the reference lists of identified articles. In particular, all studies in which amino acid PET was directly compared with PWI were included. Results PWI is more readily available, but requires substantial expertise and is more sensitive to artifacts than amino acid PET. At initial diagnosis, PWI and amino acid PET can help to define a site for biopsy but amino acid PET appears to be more powerful to define the tumor extent. Both methods are helpful to differentiate progression or recurrence from unspecific posttherapeutic changes. Assessment of therapeutic efficacy can be achieved especially with amino acid PET, while the data with PWI are sparse. Conclusion Both PWI and amino acid PET add valuable diagnostic information to the conventional MRI in the assessment of patients with brain tumours, but further studies are necessary to explore the complementary nature of these two methods.
Collapse
Affiliation(s)
- Christian P Filss
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Aachen, Germany
| | - Francesco Cicone
- Unit of Nuclear Medicine, Department of Surgical and Medical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy.,Nuclear Medicine and Molecular Medicine Department, University Hospital of Lausanne, Lausanne, Switzerland
| | - Nadim Jon Shah
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Aachen, Germany.,JARA-Jülich Aachen Research Alliance, Jülich, Germany.,Monash Institute of Medical Engineering, Department of Electrical and Computer Systems Engineering, and Monash Biomedical Imaging, School of Psychological Sciences, Monash University, Melbourne, VIC Australia
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany.,Center of Integrated Oncology (CIO), University of Cologne and Bonn, Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Aachen, Germany.,JARA-Jülich Aachen Research Alliance, Jülich, Germany
| |
Collapse
|
85
|
Uehara T, Watanabe M, Suzuki H, Furusawa Y, Arano Y. Amino acid transport system - A substrate predicts the therapeutic effects of particle radiotherapy. PLoS One 2017; 12:e0173096. [PMID: 28245294 PMCID: PMC5330493 DOI: 10.1371/journal.pone.0173096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/15/2017] [Indexed: 11/19/2022] Open
Abstract
L-[methyl-11C]Methionine (11C-Met) is useful for estimating the therapeutic efficacy of particle radiotherapy at early stages of the treatment. Given the short half-life of 11C, the development of longer-lived 18F- and 123I-labeled probes that afford diagnostic information similar to 11C-Met, are being sought. Tumor uptake of 11C-Met is involved in many cellular functions such as amino acid transport System-L, protein synthesis, and transmethylation. Among these processes, since the energy-dependent intracellular functions involved with 11C-Met are more reflective of the radiotherapeutic effects, we evaluated the activity of the amino acid transport System-A as an another energy-dependent cellular function in order to estimate radiotherapeutic effects. In this study, using a carbon-ion beam as the radiation source, the activity of System-A was evaluated by a specific System-A substrate, alpha-[1-14C]-methyl-aminoisobutyric acid (14C-MeAIB). Cellular growth and the accumulation of 14C-MeAIB or 14C-Met were evaluated over time in vitro in cultured human salivary gland (HSG) tumor cells (3-Gy) or in vivo in murine xenografts of HSG tumors (6- or 25-Gy) before and after irradiation with the carbon-ion beam. Post 3-Gy irradiation, in vitro accumulation of 14C-Met and 14C-MeAIB decreased over a 5-day period. In xenografts of HSG tumors in mice, tumor re-growth was observed in vivo on day-10 after a 6-Gy irradiation dose, but no re-growth was detected after the 25-Gy irradiation dose. Consistent with the growth results, the in vivo tumor accumulation of 14C-MeAIB did not decrease after the 6-Gy irradiation dose, whereas a significant decrease was observed after the 25-Gy irradiation dose. These results indicate that the activity of energy dependent System-A transporter may reflect the therapeutic efficacy of carbon-ion radiotherapy and suggests that longer half-life radionuclide-labeled probes for System-A may also provide widely available probes to evaluate the effects of particle radiotherapy on tumors at early stage of the treatment.
Collapse
Affiliation(s)
- Tomoya Uehara
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Science, Chiba University, Chiba, Japan
- * E-mail:
| | - Mariko Watanabe
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Science, Chiba University, Chiba, Japan
| | - Hiroyuki Suzuki
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Science, Chiba University, Chiba, Japan
| | - Yoshiya Furusawa
- National Institutes for Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, Chiba, Japan
| | - Yasushi Arano
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Science, Chiba University, Chiba, Japan
| |
Collapse
|
86
|
Nodwell MB, Yang H, Čolović M, Yuan Z, Merkens H, Martin RE, Bénard F, Schaffer P, Britton R. 18F-Fluorination of Unactivated C-H Bonds in Branched Aliphatic Amino Acids: Direct Synthesis of Oncological Positron Emission Tomography Imaging Agents. J Am Chem Soc 2017; 139:3595-3598. [PMID: 28248493 DOI: 10.1021/jacs.6b11533] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A mild and selective photocatalytic C-H 18F-fluorination reaction has been developed that provides direct access to 18F-fluorinated amino acids. The biodistribution and uptake of three 18F-labeled leucine analogues via LAT1 mediated transport in several cancer cell lines is reported. Positron emission tomography imaging of mice bearing PC3 (prostate) or U87 (glioma) xenografts using 5-[18F]-fluorohomoleucine showed high tumor uptake and excellent tumor visualization, highlighting the utility of this strategy for rapid tracer discovery for oncology.
Collapse
Affiliation(s)
- Matthew B Nodwell
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada
| | - Hua Yang
- Life Sciences Division, TRIUMF , Vancouver, British Columbia V6T 2A3, Canada
| | - Milena Čolović
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, British Columbia V5Z 1L3 Canada
| | - Zheliang Yuan
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada.,Life Sciences Division, TRIUMF , Vancouver, British Columbia V6T 2A3, Canada
| | - Helen Merkens
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, British Columbia V5Z 1L3 Canada
| | - Rainer E Martin
- Medicinal Chemistry, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd , Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, British Columbia V5Z 1L3 Canada
| | - Paul Schaffer
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada.,Life Sciences Division, TRIUMF , Vancouver, British Columbia V6T 2A3, Canada
| | - Robert Britton
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada
| |
Collapse
|
87
|
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
|
88
|
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
|
89
|
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
|
90
|
Göttler J, Lukas M, Kluge A, Kaczmarz S, Gempt J, Ringel F, Mustafa M, Meyer B, Zimmer C, Schwaiger M, Förster S, Preibisch C, Pyka T. Intra-lesional spatial correlation of static and dynamic FET-PET parameters with MRI-based cerebral blood volume in patients with untreated glioma. Eur J Nucl Med Mol Imaging 2016; 44:392-397. [PMID: 27913827 DOI: 10.1007/s00259-016-3585-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/22/2016] [Indexed: 11/29/2022]
Abstract
PURPOSE 18F-fluorethyltyrosine-(FET)-PET and MRI-based relative cerebral blood volume (rCBV) have both been used to characterize gliomas. Recently, inter-individual correlations between peak static FET-uptake and rCBV have been reported. Herein, we assess the local intra-lesional relation between FET-PET parameters and rCBV. METHODS Thirty untreated glioma patients (27 high-grade) underwent simultaneous PET/MRI on a 3 T hybrid scanner obtaining structural and dynamic susceptibility contrast sequences. Static FET-uptake and dynamic FET-slope were correlated with rCBV within tumour hotspots across patients and intra-lesionally using a mixed-effects model to account for inter-individual variation. Furthermore, maximal congruency of tumour volumes defined by FET-uptake and rCBV was determined. RESULTS While the inter-individual relationship between peak static FET-uptake and rCBV could be confirmed, our intra-lesional, voxel-wise analysis revealed significant positive correlations (median r = 0.374, p < 0.0001). Similarly, significant inter- and intra-individual correlations were observed between FET-slope and rCBV. However, rCBV explained only 12% of the static and 5% of the dynamic FET-PET variance and maximal overlap of respective tumour volumes was 37% on average. CONCLUSIONS Our results show that the relation between peak values of MR-based rCBV and static FET-uptake can also be observed intra-individually on a voxel basis and also applies to a dynamic FET parameter, possibly determining hotspots of higher biological malignancy. However, just a small part of the FET-PET signal variance is explained by rCBV and tumour volumes determined by the two modalities showed only moderate overlap. These findings indicate that FET-PET and MR-based rCBV provide both congruent and complimentary information on glioma biology.
Collapse
Affiliation(s)
- Jens Göttler
- Department of Neuroradiology, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany. .,TUM Neuroimaging Center (TUM-NIC), Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Mathias Lukas
- Department of Nuclear Medicine, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Anne Kluge
- Department of Neuroradiology, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Stephan Kaczmarz
- Department of Neuroradiology, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Florian Ringel
- Department of Neurosurgery, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Mona Mustafa
- Department of Nuclear Medicine, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Stefan Förster
- TUM Neuroimaging Center (TUM-NIC), Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany.,Department of Nuclear Medicine, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany.,Department of Nuclear Medicine, Klinikum Bayreuth, Preuschwitzer Str. 101, 95445, Bayreuth, Germany
| | - Christine Preibisch
- Department of Neuroradiology, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany.,TUM Neuroimaging Center (TUM-NIC), Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Thomas Pyka
- Department of Nuclear Medicine, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675, Munich, Germany
| |
Collapse
|
91
|
Filp U, Pekošak A, Poot AJ, Windhorst AD. Enantioselective synthesis of carbon-11 labeled l-alanine using phase transfer catalysis of Schiff bases. Tetrahedron 2016. [DOI: 10.1016/j.tet.2016.08.069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
92
|
Abstract
A previous review published in 2012 demonstrated the role of clinical PET for diagnosis and management of brain tumors using mainly FDG, amino acid tracers, and 18F-fluorothymidine. This review provides an update on clinical PET studies, most of which are motivated by prediction of prognosis and planning and monitoring of therapy in gliomas. For FDG, there has been additional evidence supporting late scanning, and combination with 13N ammonia has yielded some promising results. Large neutral amino acid tracers have found widespread applications mostly based on 18F-labeled compounds fluoroethyltyrosine and fluorodopa for targeting biopsies, therapy planning and monitoring, and as outcome markers in clinical trials. 11C-alpha-methyltryptophan (AMT) has been proposed as an alternative to 11C-methionine, and there may also be a role for cyclic amino acid tracers. 18F-fluorothymidine has shown strengths for tumor grading and as an outcome marker. Studies using 18F-fluorocholine (FCH) and 68Ga-labeled compounds are promising but have not yet clearly defined their role. Studies on radiotherapy planning have explored the use of large neutral amino acid tracers to improve the delineation of tumor volume for irradiation and the use of hypoxia markers, in particular 18F-fluoromisonidazole. Many studies employed the combination of PET with advanced multimodal MR imaging methods, mostly demonstrating complementarity and some potential benefits of hybrid PET/MR.
Collapse
Affiliation(s)
- Karl Herholz
- The University of Manchester, Division of Neuroscience and Experimental Psychology Wolfson Molecular Imaging Centre, Manchester, England, United Kingdom.
| |
Collapse
|
93
|
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
|
94
|
Cicone F, Clerico A, Minniti G, Paiano M, Carideo L, Scaringi C, Langen KJ, Scopinaro F. 18F-DOPA Positron Emission Tomography in Medulloblastoma: 2 Case Reports. World Neurosurg 2016; 93:490.e7-490.e11. [DOI: 10.1016/j.wneu.2016.06.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 10/21/2022]
|
95
|
Bolcaen J, Lybaert K, Moerman L, Descamps B, Deblaere K, Boterberg T, Kalala JP, Van den Broecke C, De Vos F, Vanhove C, Goethals I. Kinetic Modeling and Graphical Analysis of 18F-Fluoromethylcholine (FCho), 18F-Fluoroethyltyrosine (FET) and 18F-Fluorodeoxyglucose (FDG) PET for the Fiscrimination between High-Grade Glioma and Radiation Necrosis in Rats. PLoS One 2016; 11:e0161845. [PMID: 27559736 PMCID: PMC4999092 DOI: 10.1371/journal.pone.0161845] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 08/12/2016] [Indexed: 02/07/2023] Open
Abstract
Background Discrimination between glioblastoma (GB) and radiation necrosis (RN) post-irradiation remains challenging but has a large impact on further treatment and prognosis. In this study, the uptake mechanisms of 18F-fluorodeoxyglucose (18F-FDG), 18F-fluoroethyltyrosine (18F-FET) and 18F-fluoromethylcholine (18F-FCho) positron emission tomography (PET) tracers were investigated in a F98 GB and RN rat model applying kinetic modeling (KM) and graphical analysis (GA) to clarify our previous results. Methods Dynamic 18F-FDG (GB n = 6 and RN n = 5), 18F-FET (GB n = 5 and RN n = 5) and 18F-FCho PET (GB n = 5 and RN n = 5) were acquired with continuous arterial blood sampling. Arterial input function (AIF) corrections, KM and GA were performed. Results The influx rate (Ki) of 18F-FDG uptake described by a 2-compartmental model (CM) or using Patlak GA, showed more trapping (k3) in GB (0.07 min-1) compared to RN (0.04 min-1) (p = 0.017). K1 of 18F-FET was significantly higher in GB (0.06 ml/ccm/min) compared to RN (0.02 ml/ccm/min), quantified using a 1-CM and Logan GA (p = 0.036). 18F-FCho was rapidly oxidized complicating data interpretation. Using a 1-CM and Logan GA no clear differences were found to discriminate GB from RN. Conclusions Based on our results we concluded that using KM and GA both 18F-FDG and 18F-FET were able to discriminate GB from RN. Using a 2-CM model more trapping of 18F-FDG was found in GB compared to RN. Secondly, the influx of 18F-FET was higher in GB compared to RN using a 1-CM model. Important correlations were found between SUV and kinetic or graphical measures for 18F-FDG and 18F-FET. 18F-FCho PET did not allow discrimination between GB and RN.
Collapse
Affiliation(s)
- Julie Bolcaen
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
- * E-mail:
| | - Kelly Lybaert
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Lieselotte Moerman
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Benedicte Descamps
- iMinds-IBiTech-MEDISIP, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Karel Deblaere
- Department of Radiology, Ghent University Hospital, Ghent, Belgium
| | - Tom Boterberg
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | | | | | - Filip De Vos
- Department of Radiopharmacy, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- iMinds-IBiTech-MEDISIP, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Ingeborg Goethals
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
96
|
Krakauer M, Kjaer A, Bennedbæk FN. (18)F-FET-PET in Primary Hyperparathyroidism: A Pilot Study. Diagnostics (Basel) 2016; 6:diagnostics6030030. [PMID: 27548229 PMCID: PMC5039564 DOI: 10.3390/diagnostics6030030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 11/30/2022] Open
Abstract
Preoperative localisation of the diseased parathyroid gland(s) in primary hyperparathyroidism (PHP) is a prerequisite for subsequent minimally invasive surgery. Recently, as alternatives to conventional sestamibi parathyroid scintigraphy, the 11C-based positron emission tomography (PET) tracers methionine and choline have shown promise for this purpose. We evaluated the feasibility of using the 18F-based PET tracer fluoroethyl-l-tyrosine (FET), as the longer half-life of 18F makes it logistically more favourable. As a proof-of-concept study, we included two patients with PHP in which dual-isotope parathyroid subtraction single photon emission computed tomography had determined the exact location of the parathyroid adenoma. A dynamic FET PET/CT scan was performed with subsequent visual evaluation and calculation of target-to-background (TBR; parathyroid vs. thyroid). The maximum TBR in the two patients under study was achieved approximately 30 min after the injection of the tracer and was 1.5 and 1.7, respectively. This ratio was too small to allow for confident visualisation of the adenomas. FET PET/CT seems not feasible as a preoperative imaging modality in PHP.
Collapse
Affiliation(s)
- Martin Krakauer
- Department of Clinical Physiology and Nuclear Medicine, Gentofte Hospital, DK-2900 Hellerup, Denmark.
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, DK-2100 Copenhagen, Denmark.
| | - Finn N Bennedbæk
- Department of Endocrinology, Herlev Hospital, DK-2730 Herlev, Denmark.
| |
Collapse
|
97
|
Hutterer M, Ebner Y, Riemenschneider MJ, Willuweit A, McCoy M, Egger B, Schröder M, Wendl C, Hellwig D, Grosse J, Menhart K, Proescholdt M, Fritsch B, Urbach H, Stockhammer G, Roelcke U, Galldiks N, Meyer PT, Langen KJ, Hau P, Trinka E. Epileptic Activity Increases Cerebral Amino Acid Transport Assessed by 18F-Fluoroethyl-l-Tyrosine Amino Acid PET: A Potential Brain Tumor Mimic. J Nucl Med 2016; 58:129-137. [PMID: 27469356 DOI: 10.2967/jnumed.116.176610] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/29/2016] [Indexed: 11/16/2022] Open
Abstract
O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) PET is a well-established method increasingly used for diagnosis, treatment planning, and monitoring in gliomas. Epileptic activity, frequently occurring in glioma patients, can influence MRI findings. Whether seizures also affect 18F-FET PET imaging is currently unknown. The aim of this retrospective analysis was to investigate the brain amino acid metabolism during epileptic seizures by 18F-FET PET and to elucidate the pathophysiologic background. METHODS Ten patients with 11 episodes of serial seizures or status epilepticus, who underwent MRI and 18F-FET PET, were studied. The main diagnosis was glioma World Health Organization grade II-IV (n = 8); 2 patients suffered from nonneoplastic diseases. Immunohistochemical assessment of LAT1/LAT2/CD98 amino acid transporters was performed in seizure-affected cortex (n = 2) and compared with glioma tissues (n = 3). RESULTS All patients exhibited increased seizure-associated strict gyral 18F-FET uptake, which was reversible in follow-up studies or negative shortly before and without any histologic or clinical signs of tumor recurrence. 18F-FET uptake corresponded to structural MRI changes, compatible with cortical vasogenic and cytotoxic edema, partial contrast enhancement, and hyperperfusion. Patients with prolonged postictal symptoms lasting up to 8 wk displayed intensive and widespread (≥ 1 lobe) cortical 18F-FET uptake. LAT1/LAT2/CD98 was strongly expressed in neurons and endothelium of seizure-affected brains and less in reactive astrocytosis. CONCLUSION Seizure activity, in particular status epilepticus, increases cerebral amino acid transport with a strict gyral 18F-FET uptake pattern. Such periictal pseudoprogression represents a potential pitfall of 18F-FET PET and may mimic brain tumor. Our data also indicate a seizure-induced upregulation of neuronal, endothelial, and less astroglial LAT1/LAT2/CD98 amino acid transporter expression.
Collapse
Affiliation(s)
- Markus Hutterer
- Department of Neurology, University of Regensburg Medical School, Regensburg, Germany .,Wilhelm Sander-Neurooncology Unit, University of Regensburg Medical School, Regensburg, Germany.,Department of Neurology and Centre for Cognitive Neuroscience, Christian-Doppler Klinik, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Yvonne Ebner
- Department of Neurology and Centre for Cognitive Neuroscience, Christian-Doppler Klinik, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Markus J Riemenschneider
- Wilhelm Sander-Neurooncology Unit, University of Regensburg Medical School, Regensburg, Germany.,Department of Neuropathology, University of Regensburg Medical School, Regensburg, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
| | - Mark McCoy
- Department of Radiology and Division of Neuroradiology, Christian-Doppler Klinik, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Barbara Egger
- Department of Nuclear Medicine, Landeskrankenhaus Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Michael Schröder
- Department of Neurology, University of Regensburg Medical School, Regensburg, Germany
| | - Christina Wendl
- Department of Radiology and Division of Neuroradiology, University of Regensburg Medical School, Regensburg, Germany
| | - Dirk Hellwig
- Department of Nuclear Medicine, University of Regensburg Medical School, Regensburg, Germany
| | - Jirka Grosse
- Department of Nuclear Medicine, University of Regensburg Medical School, Regensburg, Germany
| | - Karin Menhart
- Department of Nuclear Medicine, University of Regensburg Medical School, Regensburg, Germany
| | - Martin Proescholdt
- Wilhelm Sander-Neurooncology Unit, University of Regensburg Medical School, Regensburg, Germany.,Department of Neurosurgery, University of Regensburg Medical School, Regensburg, Germany
| | - Brita Fritsch
- Department of Neurology, University Hospital Freiburg, Freiburg, Germany
| | - Horst Urbach
- Department of Neuroradiology, University Hospital Freiburg, Freiburg, Germany
| | | | - Ulrich Roelcke
- Department of Neurology and Brain Tumor Center, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, University Hospital Freiburg, Freiburg, Germany; and
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany.,Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Peter Hau
- Department of Neurology, University of Regensburg Medical School, Regensburg, Germany.,Wilhelm Sander-Neurooncology Unit, University of Regensburg Medical School, Regensburg, Germany
| | - Eugen Trinka
- Department of Neurology and Centre for Cognitive Neuroscience, Christian-Doppler Klinik, Paracelsus Medical University Salzburg, Salzburg, Austria
| |
Collapse
|
98
|
Piroth MD, Galldiks N, Pinkawa M, Holy R, Stoffels G, Ermert J, Mottaghy FM, Shah NJ, Langen KJ, Eble MJ. Relapse patterns after radiochemotherapy of glioblastoma with FET PET-guided boost irradiation and simulation to optimize radiation target volume. Radiat Oncol 2016; 11:87. [PMID: 27342976 PMCID: PMC4920983 DOI: 10.1186/s13014-016-0665-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 06/23/2016] [Indexed: 12/14/2022] Open
Abstract
Background O-(2-18 F-fluoroethyl)-L-tyrosine-(FET)-PET may be helpful to improve the definition of radiation target volumes in glioblastomas compared with MRI. We analyzed the relapse patterns in FET-PET after a FET- and MRI-based integrated-boost intensity-modulated radiotherapy (IMRT) of glioblastomas to perform an optimized target volume definition. Methods A relapse pattern analysis was performed in 13 glioblastoma patients treated with radiochemotherapy within a prospective phase-II-study between 2008 and 2009. Radiotherapy was performed as an integrated-boost intensity-modulated radiotherapy (IB-IMRT). The prescribed dose was 72 Gy for the boost target volume, based on baseline FET-PET (FET-1) and 60 Gy for the MRI-based (MRI-1) standard target volume. The single doses were 2.4 and 2.0 Gy, respectively. Location and volume of recurrent tumors in FET-2 and MRI-2 were analyzed related to initial tumor, detected in baseline FET-1. Variable target volumes were created theoretically based on FET-1 to optimally cover recurrent tumor. Results The tumor volume overlap in FET and MRI was poor both at baseline (median 12 %; range 0–32) and at time of recurrence (13 %; 0–100). Recurrent tumor volume in FET-2 was localized to 39 % (12–91) in the initial tumor volume (FET-1). Over the time a shrinking (mean 12 (5–26) ml) and shifting (mean 6 (1–10 mm) of the resection cavity was seen. A simulated target volume based on active tumor in FET-1 with an additional safety margin of 7 mm around the FET-1 volume covered recurrent FET tumor volume (FET-2) significantly better than a corresponding target volume based on contrast enhancement in MRI-1 with a same safety margin of 7 mm (100 % (54–100) versus 85 % (0–100); p < 0.01). A simulated planning target volume (PTV), based on FET-1 and additional 7 mm margin plus 5 mm margin for setup-uncertainties was significantly smaller than the conventional, MR-based PTV applied in this study (median 160 (112–297) ml versus 231 (117–386) ml, p < 0.001). Conclusions In this small study recurrent tumor volume in FET-PET (FET-2) overlapped only to one third with the boost target volume, based on FET-1. The shrinking and shifting of the resection cavity may have an influence considering the limited overlap of initial and relapse tumor volume. A simulated target volume, based on FET-1 with 7 mm margin covered 100 % of relapse volume in median and led to a significantly reduced PTV, compared to MRI-based PTVs. This approach may achieve similar therapeutic efficacy but lower side effects offering a broader window to intensify concomitant systemic treatment focusing distant failures.
Collapse
Affiliation(s)
- Marc D Piroth
- Department of Radiation Oncology, University Hospital RWTH Aachen, Aachen, Germany. .,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Research Center Jülich, Jülich, Germany. .,Department of Radiation Oncology, HELIOS University Hospital Wuppertal, Witten/Herdecke University, Wuppertal, Germany.
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Research Center Jülich, Jülich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany
| | - Michael Pinkawa
- Department of Radiation Oncology, University Hospital RWTH Aachen, Aachen, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Research Center Jülich, Jülich, Germany
| | - Richard Holy
- Department of Radiation Oncology, University Hospital RWTH Aachen, Aachen, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Research Center Jülich, Jülich, Germany.,Department of Radiation Oncology, HELIOS University Hospital Wuppertal, Witten/Herdecke University, Wuppertal, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Research Center Jülich, Jülich, Germany
| | - Johannes Ermert
- Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Research Center Jülich, Jülich, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Research Center Jülich, Jülich, Germany
| | - N Jon Shah
- Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany.,Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Research Center Jülich, Jülich, Germany
| | - Karl-Josef Langen
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany.,Institute of Neuroscience and Medicine, Research Center Jülich, Jülich, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Research Center Jülich, Jülich, Germany
| | - Michael J Eble
- Department of Radiation Oncology, University Hospital RWTH Aachen, Aachen, Germany.,Jülich-Aachen Research Alliance (JARA) - Section JARA-Brain, Research Center Jülich, Jülich, Germany
| |
Collapse
|
99
|
Holzgreve A, Brendel M, Gu S, Carlsen J, Mille E, Böning G, Mastrella G, Unterrainer M, Gildehaus FJ, Rominger A, Bartenstein P, Kälin RE, Glass R, Albert NL. Monitoring of Tumor Growth with [(18)F]-FET PET in a Mouse Model of Glioblastoma: SUV Measurements and Volumetric Approaches. Front Neurosci 2016; 10:260. [PMID: 27378835 PMCID: PMC4906232 DOI: 10.3389/fnins.2016.00260] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/23/2016] [Indexed: 11/15/2022] Open
Abstract
Noninvasive tumor growth monitoring is of particular interest for the evaluation of experimental glioma therapies. This study investigates the potential of positron emission tomography (PET) using O-(2-18F-fluoroethyl)-L-tyrosine ([18F]-FET) to determine tumor growth in a murine glioblastoma (GBM) model—including estimation of the biological tumor volume (BTV), which has hitherto not been investigated in the pre-clinical context. Fifteen GBM-bearing mice (GL261) and six control mice (shams) were investigated during 5 weeks by PET followed by autoradiographic and histological assessments. [18F]-FET PET was quantitated by calculation of maximum and mean standardized uptake values within a universal volume-of-interest (VOI) corrected for healthy background (SUVmax/BG, SUVmean/BG). A partial volume effect correction (PVEC) was applied in comparison to ex vivo autoradiography. BTVs obtained by predefined thresholds for VOI definition (SUV/BG: ≥1.4; ≥1.6; ≥1.8; ≥2.0) were compared to the histologically assessed tumor volume (n = 8). Finally, individual “optimal” thresholds for BTV definition best reflecting the histology were determined. In GBM mice SUVmax/BG and SUVmean/BG clearly increased with time, however at high inter-animal variability. No relevant [18F]-FET uptake was observed in shams. PVEC recovered signal loss of SUVmean/BG assessment in relation to autoradiography. BTV as estimated by predefined thresholds strongly differed from the histology volume. Strikingly, the individual “optimal” thresholds for BTV assessment correlated highly with SUVmax/BG (ρ = 0.97, p < 0.001), allowing SUVmax/BG-based calculation of individual thresholds. The method was verified by a subsequent validation study (n = 15, ρ = 0.88, p < 0.01) leading to extensively higher agreement of BTV estimations when compared to histology in contrast to predefined thresholds. [18F]-FET PET with standard SUV measurements is feasible for glioma imaging in the GBM mouse model. PVEC is beneficial to improve accuracy of [18F]-FET PET SUV quantification. Although SUVmax/BG and SUVmean/BG increase during the disease course, these parameters do not correlate with the respective tumor size. For the first time, we propose a histology-verified method allowing appropriate individual BTV estimation for volumetric in vivo monitoring of tumor growth with [18F]-FET PET and show that standardized thresholds from routine clinical practice seem to be inappropriate for BTV estimation in the GBM mouse model.
Collapse
Affiliation(s)
- Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University of MunichMunich, Germany; Department of Neurosurgery, University Hospital of Munich, Ludwig Maximilians University of MunichMunich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Song Gu
- Department of Neurosurgery, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Janette Carlsen
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Erik Mille
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Guido Böning
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Giorgia Mastrella
- Department of Neurosurgery, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Franz J Gildehaus
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Roland E Kälin
- Department of Neurosurgery, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Rainer Glass
- Department of Neurosurgery, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University of Munich Munich, Germany
| |
Collapse
|
100
|
Persico MG, Buroni FE, Pasi F, Lodola L, Aprile C, Nano R, Hodolic M. (18)F-FET and (18)F-FCH uptake in human glioblastoma T98G cell lines. Radiol Oncol 2016; 50:153-8. [PMID: 27247547 PMCID: PMC4852969 DOI: 10.1515/raon-2016-0022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/18/2016] [Indexed: 11/15/2022] Open
Abstract
Background Despite complex treatment of surgery, radiotherapy and chemotherapy, high grade gliomas often recur. Differentiation between post-treatment changes and recurrence is difficult. 18F-methyl-choline (18F-FCH) is frequently used in staging and detection of recurrent prostate cancer disease as well as some brain tumours; however accumulation in inflammatory tissue limits its specificity. The 18F-ethyl-tyrosine (18F-FET) shows a specific uptake in malignant cells, resulting from increased expression of amino acid transporters or diffusing through the disrupted blood-brain barrier. 18F-FET exhibits lower uptake in machrophages and other inflammatory cells. Aim of this study was to evaluate 18F-FCH and 18F-FET uptake by human glioblastoma T98G cells. Material and methods Human glioblastoma T98G or human dermal fibroblasts cells, seeded at a density to obtain 2 × 105 cells per flask when radioactive tracers were administered, grew adherent to the plastic surface at 37°C in 5% CO2 in complete medium. Equimolar amounts of radiopharmaceuticals were added to cells for different incubation times (20 to 120 minutes) for 18F-FCH and 18F-FET respectively. The cellular radiotracer uptake was determined with a gamma counter. All experiments were carried out in duplicate and repeated three times. The uptake measurements are expressed as the percentage of the administered dose of tracer per 2 × 105 cells. Data (expressed as mean values of % uptake of radiopharmaceuticals) were compared using parametric or non-parametric tests as appropriate. Differences were regarded as statistically significant when p<0.05. Results A significant uptake of 18F-FCH was seen in T98G cells at 60, 90 and 120 minutes. The percentage uptake of 18F-FET in comparison to 18F-FCH was lower by a factor of more than 3, with different kinetic curves.18F-FET showed a more rapid initial uptake up to 40 minutes and 18F-FCH showed a progressive rise reaching a maximum after 90 minutes. Conclusions 18F-FCH and 18F-FET are candidates for neuro-oncological PET imaging. 18F-FET could be the most useful oncological PET marker in the presence of reparative changes after therapy, where the higher affinity of 18F-FCH to inflammatory cells makes it more difficult to discriminate between tumour persistence and non-neoplastic changes. Additional studies on the influence of inflammatory tissue and radionecrotic cellular components on radiopharmaceutical uptake are necessary.
Collapse
Affiliation(s)
- Marco Giovanni Persico
- Department of Oncohaematology, Nuclear Medicine Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Federica Eleonora Buroni
- Department of Oncohaematology, Nuclear Medicine Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Francesca Pasi
- Department of Oncohaematology, Radiotherapy Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Lorenzo Lodola
- Department of Oncohaematology, Nuclear Medicine Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Carlo Aprile
- Department of Oncohaematology, Nuclear Medicine Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Rosanna Nano
- Department of Biology and Biotecnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Marina Hodolic
- Nuclear medicine research department, Iason, Graz, Austria
| |
Collapse
|