1
|
Aamir M, Namoun A, Munir S, Aljohani N, Alanazi MH, Alsahafi Y, Alotibi F. Brain Tumor Detection and Classification Using an Optimized Convolutional Neural Network. Diagnostics (Basel) 2024; 14:1714. [PMID: 39202202 PMCID: PMC11353951 DOI: 10.3390/diagnostics14161714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
Brain tumors are a leading cause of death globally, with numerous types varying in malignancy, and only 12% of adults diagnosed with brain cancer survive beyond five years. This research introduces a hyperparametric convolutional neural network (CNN) model to identify brain tumors, with significant practical implications. By fine-tuning the hyperparameters of the CNN model, we optimize feature extraction and systematically reduce model complexity, thereby enhancing the accuracy of brain tumor diagnosis. The critical hyperparameters include batch size, layer counts, learning rate, activation functions, pooling strategies, padding, and filter size. The hyperparameter-tuned CNN model was trained on three different brain MRI datasets available at Kaggle, producing outstanding performance scores, with an average value of 97% for accuracy, precision, recall, and F1-score. Our optimized model is effective, as demonstrated by our methodical comparisons with state-of-the-art approaches. Our hyperparameter modifications enhanced the model performance and strengthened its capacity for generalization, giving medical practitioners a more accurate and effective tool for making crucial judgments regarding brain tumor diagnosis. Our model is a significant step in the right direction toward trustworthy and accurate medical diagnosis, with practical implications for improving patient outcomes.
Collapse
Affiliation(s)
- Muhammad Aamir
- Department of Computer Science, Sahiwal Campus, COMSATS University Islamabad, Sahiwal 57000, Pakistan; (M.A.); (S.M.)
- Department of Computer Science, Superior University Lahore, Lahore 54000, Pakistan
| | - Abdallah Namoun
- AI Centre, Faculty of Computer and Information Systems, Islamic University of Madinah, Madinah 42351, Saudi Arabia;
| | - Sehrish Munir
- Department of Computer Science, Sahiwal Campus, COMSATS University Islamabad, Sahiwal 57000, Pakistan; (M.A.); (S.M.)
| | - Nasser Aljohani
- AI Centre, Faculty of Computer and Information Systems, Islamic University of Madinah, Madinah 42351, Saudi Arabia;
| | - Meshari Huwaytim Alanazi
- Computer Science Department, College of Sciences, Northern Border University, Arar 73213, Saudi Arabia
| | - Yaser Alsahafi
- School of Information Technology, University of Jeddah, Jeddah 23218, Saudi Arabia;
| | - Faris Alotibi
- College of Computer Science and Engineering, Taibah University, Madinah 42353, Saudi Arabia;
| |
Collapse
|
2
|
Galldiks N, Kaufmann TJ, Vollmuth P, Lohmann P, Smits M, Veronesi MC, Langen KJ, Rudà R, Albert NL, Hattingen E, Law I, Hutterer M, Soffietti R, Vogelbaum MA, Wen PY, Weller M, Tonn JC. Challenges, limitations, and pitfalls of PET and advanced MRI in patients with brain tumors: A report of the PET/RANO group. Neuro Oncol 2024; 26:1181-1194. [PMID: 38466087 PMCID: PMC11226881 DOI: 10.1093/neuonc/noae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Indexed: 03/12/2024] Open
Abstract
Brain tumor diagnostics have significantly evolved with the use of positron emission tomography (PET) and advanced magnetic resonance imaging (MRI) techniques. In addition to anatomical MRI, these modalities may provide valuable information for several clinical applications such as differential diagnosis, delineation of tumor extent, prognostication, differentiation between tumor relapse and treatment-related changes, and the evaluation of response to anticancer therapy. In particular, joint recommendations of the Response Assessment in Neuro-Oncology (RANO) Group, the European Association of Neuro-oncology, and major European and American Nuclear Medicine societies highlighted that the additional clinical value of radiolabeled amino acids compared to anatomical MRI alone is outstanding and that its widespread clinical use should be supported. For advanced MRI and its steadily increasing use in clinical practice, the Standardization Subcommittee of the Jumpstarting Brain Tumor Drug Development Coalition provided more recently an updated acquisition protocol for the widely used dynamic susceptibility contrast perfusion MRI. Besides amino acid PET and perfusion MRI, other PET tracers and advanced MRI techniques (e.g. MR spectroscopy) are of considerable clinical interest and are increasingly integrated into everyday clinical practice. Nevertheless, these modalities have shortcomings which should be considered in clinical routine. This comprehensive review provides an overview of potential challenges, limitations, and pitfalls associated with PET imaging and advanced MRI techniques in patients with gliomas or brain metastases. Despite these issues, PET imaging and advanced MRI techniques continue to play an indispensable role in brain tumor management. Acknowledging and mitigating these challenges through interdisciplinary collaboration, standardized protocols, and continuous innovation will further enhance the utility of these modalities in guiding optimal patient care.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Germany
| | | | - Philipp Vollmuth
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
| | - Marion Smits
- Department of Radiology and Nuclear Medicine and Brain Tumour Center, Erasmus MC, Rotterdam, The Netherlands
| | - Michael C Veronesi
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilians-University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elke Hattingen
- Goethe University, Department of Neuroradiology, University Hospital Frankfurt, Frankfurt, Germany
| | - Ian Law
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Markus Hutterer
- Department of Neurology with Acute Geriatrics, Saint John of God Hospital, Linz, Austria
| | - Riccardo Soffietti
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Michael A Vogelbaum
- Department of Neuro-Oncology and Neurosurgery, Moffit Cancer Center, Tampa, Florida, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, and University Hospital of Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Joerg-Christian Tonn
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurosurgery, University Hospital of Munich (LMU), Munich, Germany
| |
Collapse
|
3
|
Krishnapriya S, Karuna Y. A deep learning model for the localization and extraction of brain tumors from MR images using YOLOv7 and grab cut algorithm. Front Oncol 2024; 14:1347363. [PMID: 38680854 PMCID: PMC11045991 DOI: 10.3389/fonc.2024.1347363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/25/2024] [Indexed: 05/01/2024] Open
Abstract
Introduction Brain tumors are a common disease that affects millions of people worldwide. Considering the severity of brain tumors (BT), it is important to diagnose the disease in its early stages. With advancements in the diagnostic process, Magnetic Resonance Imaging (MRI) has been extensively used in disease detection. However, the accurate identification of BT is a complex task, and conventional techniques are not sufficiently robust to localize and extract tumors in MRI images. Therefore, in this study, we used a deep learning model combined with a segmentation algorithm to localize and extract tumors from MR images. Method This paper presents a Deep Learning (DL)-based You Look Only Once (YOLOv7) model in combination with the Grab Cut algorithm to extract the foreground of the tumor image to enhance the detection process. YOLOv7 is used to localize the tumor region, and the Grab Cut algorithm is used to extract the tumor from the localized region. Results The performance of the YOLOv7 model with and without the Grab Cut algorithm is evaluated. The results show that the proposed approach outperforms other techniques, such as hybrid CNN-SVM, YOLOv5, and YOLOv6, in terms of accuracy, precision, recall, specificity, and F1 score. Discussion Our results show that the proposed technique achieves a high dice score between tumor-extracted images and ground truth images. The findings show that the performance of the YOLOv7 model is improved by the inclusion of the Grab Cut algorithm compared to the performance of the model without the algorithm.
Collapse
Affiliation(s)
| | - Yepuganti Karuna
- School of Electronics Engineering, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
4
|
Kas A, Rozenblum L, Pyatigorskaya N. Clinical Value of Hybrid PET/MR Imaging: Brain Imaging Using PET/MR Imaging. Magn Reson Imaging Clin N Am 2023; 31:591-604. [PMID: 37741643 DOI: 10.1016/j.mric.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2023]
Abstract
Hybrid PET/MR imaging offers a unique opportunity to acquire MR imaging and PET information during a single imaging session. PET/MR imaging has numerous advantages, including enhanced diagnostic accuracy, improved disease characterization, and better treatment planning and monitoring. It enables the immediate integration of anatomic, functional, and metabolic imaging information, allowing for personalized characterization and monitoring of neurologic diseases. This review presents recent advances in PET/MR imaging and highlights advantages in clinical practice for neuro-oncology, epilepsy, and neurodegenerative disorders. PET/MR imaging provides valuable information about brain tumor metabolism, perfusion, and anatomic features, aiding in accurate delineation, treatment response assessment, and prognostication.
Collapse
Affiliation(s)
- Aurélie Kas
- Department of Nuclear Medicine, Pitié-Salpêtrière Hospital, APHP Sorbonne Université, Paris, France; Sorbonne Université, INSERM, CNRS, Laboratoire d'Imagerie Biomédicale, LIB, Paris F-75006, France.
| | - Laura Rozenblum
- Department of Nuclear Medicine, Pitié-Salpêtrière Hospital, APHP Sorbonne Université, Paris, France; Sorbonne Université, INSERM, CNRS, Laboratoire d'Imagerie Biomédicale, LIB, Paris F-75006, France
| | - Nadya Pyatigorskaya
- Neuroradiology Department, Pitié-Salpêtrière Hospital, APHP Sorbonne Université, Paris, France; Sorbonne Université, UMR S 1127, CNRS UMR 722, Institut du Cerveau, Paris, France
| |
Collapse
|
5
|
Hajri R, Nicod-Lalonde M, Hottinger AF, Prior JO, Dunet V. Prediction of Glioma Grade and IDH Status Using 18F-FET PET/CT Dynamic and Multiparametric Texture Analysis. Diagnostics (Basel) 2023; 13:2604. [PMID: 37568967 PMCID: PMC10417545 DOI: 10.3390/diagnostics13152604] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Mutations in isocitrate dehydrogenase (IDH) represent an independent predictor of better survival in patients with gliomas. We aimed to assess grade and IDH mutation status in patients with untreated gliomas, by evaluating the respective value of 18F-FET PET/CT via dynamic and texture analyses. A total of 73 patients (male: 48, median age: 47) who underwent an 18F-FET PET/CT for initial glioma evaluation were retrospectively included. IDH status was available in 61 patients (20 patients with WHO grade 2 gliomas, 41 with grade 3-4 gliomas). Time-activity curve type and 20 parameters obtained from static analysis using LIFEx© v6.30 software were recorded. Respective performance was assessed using receiver operating characteristic curve analysis and stepwise multivariate regression analysis adjusted for patients' age and sex. The time-activity curve type and texture parameters derived from the static parameters showed satisfactory-to-good performance in predicting glioma grade and IDH status. Both time-activity curve type (stepwise OR: 101.6 (95% CI: 5.76-1791), p = 0.002) and NGLDM coarseness (stepwise OR: 2.08 × 1043 (95% CI: 2.76 × 1012-1.57 × 1074), p = 0.006) were independent predictors of glioma grade. No independent predictor of IDH status was found. Dynamic and texture analyses of 18F-FET PET/CT have limited predictive value for IDH status when adjusted for confounding factors. However, they both help predict glioma grade.
Collapse
Affiliation(s)
- Rami Hajri
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland;
| | - Marie Nicod-Lalonde
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; (M.N.-L.); (J.O.P.)
| | - Andreas F. Hottinger
- Department of Neurology, Lausanne University Hospital, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland;
- Lukas Lundin & Family Brain Tumor Research Center, Lausanne University Hospital, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| | - John O. Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; (M.N.-L.); (J.O.P.)
| | - Vincent Dunet
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland;
| |
Collapse
|
6
|
Langen KJ, Galldiks N, Mauler J, Kocher M, Filß CP, Stoffels G, Régio Brambilla C, Stegmayr C, Willuweit A, Worthoff WA, Shah NJ, Lerche C, Mottaghy FM, Lohmann P. Hybrid PET/MRI in Cerebral Glioma: Current Status and Perspectives. Cancers (Basel) 2023; 15:3577. [PMID: 37509252 PMCID: PMC10377176 DOI: 10.3390/cancers15143577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Advanced MRI methods and PET using radiolabelled amino acids provide valuable information, in addition to conventional MR imaging, for brain tumour diagnostics. These methods are particularly helpful in challenging situations such as the differentiation of malignant processes from benign lesions, the identification of non-enhancing glioma subregions, the differentiation of tumour progression from treatment-related changes, and the early assessment of responses to anticancer therapy. The debate over which of the methods is preferable in which situation is ongoing, and has been addressed in numerous studies. Currently, most radiology and nuclear medicine departments perform these examinations independently of each other, leading to multiple examinations for the patient. The advent of hybrid PET/MRI allowed a convergence of the methods, but to date simultaneous imaging has reached little relevance in clinical neuro-oncology. This is partly due to the limited availability of hybrid PET/MRI scanners, but is also due to the fact that PET is a second-line examination in brain tumours. PET is only required in equivocal situations, and the spatial co-registration of PET examinations of the brain to previous MRI is possible without disadvantage. A key factor for the benefit of PET/MRI in neuro-oncology is a multimodal approach that provides decisive improvements in the diagnostics of brain tumours compared with a single modality. This review focuses on studies investigating the diagnostic value of combined amino acid PET and 'advanced' MRI in patients with cerebral gliomas. Available studies suggest that the combination of amino acid PET and advanced MRI improves grading and the histomolecular characterisation of newly diagnosed tumours. Few data are available concerning the delineation of tumour extent. A clear additive diagnostic value of amino acid PET and advanced MRI can be achieved regarding the differentiation of tumour recurrence from treatment-related changes. Here, the PET-guided evaluation of advanced MR methods seems to be helpful. In summary, there is growing evidence that a multimodal approach can achieve decisive improvements in the diagnostics of cerebral gliomas, for which hybrid PET/MRI offers optimal conditions.
Collapse
Affiliation(s)
- Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
- Department of Nuclear Medicine, RWTH Aachen University Hospital, 52074 Aachen, Germany
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Duesseldorf, 53127 Bonn, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Duesseldorf, 53127 Bonn, Germany
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Jörg Mauler
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
| | - Martin Kocher
- Department of Stereotaxy and Functional Neurosurgery, Center for Neurosurgery, Faculty of Medicine, University Hospital Cologne, 50931 Cologne, Germany
| | - Christian Peter Filß
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
- Department of Nuclear Medicine, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
| | - Cláudia Régio Brambilla
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
| | - Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
| | - Wieland Alexander Worthoff
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
| | - Nadim Jon Shah
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
- Department of Neurology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Christoph Lerche
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
| | - Felix Manuel Mottaghy
- Department of Nuclear Medicine, RWTH Aachen University Hospital, 52074 Aachen, Germany
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Duesseldorf, 53127 Bonn, Germany
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-11), Forschungszentrum Juelich, 52425 Juelich, Germany
| |
Collapse
|
7
|
Perera Molligoda Arachchige AS. Neuroimaging with PET/MR: moving beyond 3 T in preclinical systems, when for clinical practice? Clin Transl Imaging 2023; 11:315-319. [DOI: 10.1007/s40336-023-00572-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/11/2023] [Indexed: 03/29/2025]
|
8
|
Sahu A, Mathew R, Ashtekar R, Dasgupta A, Puranik A, Mahajan A, Janu A, Choudhari A, Desai S, Patnam NG, Chatterjee A, Patil V, Menon N, Jain Y, Rangarajan V, Dev I, Epari S, Sahay A, Shetty P, Goda J, Moiyadi A, Gupta T. The complementary role of MRI and FET PET in high-grade gliomas to differentiate recurrence from radionecrosis. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1040998. [PMID: 39355021 PMCID: PMC11440952 DOI: 10.3389/fnume.2023.1040998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 04/11/2023] [Indexed: 10/03/2024]
Abstract
Introduction Conventional magnetic resonance imaging (MRI) has limitations in differentiating tumor recurrence (TR) from radionecrosis (RN) in high-grade gliomas (HGG), which can present with morphologically similar appearances. Multiparametric advanced MR sequences and Positron Emission Tomography (PET) with amino acid tracers can aid in diagnosing tumor metabolism. The role of both modalities on an individual basis and combined performances were investigated in the current study. Materials and Methods Patients with HGG with MRI and PET within three weeks were included in the retrospective analysis. The multiparametric MRI included T1-contrast, T2-weighted sequences, perfusion, diffusion, and spectroscopy. MRI was interpreted by a neuroradiologist without using information from PET imaging. 18F-Fluoroethyl-Tyrosine (FET) uptake was calculated from the areas of maximum enhancement/suspicion, which was assessed by a nuclear medicine physician (having access to MRI to determine tumor-to-white matter ratio over a specific region). A definitive diagnosis of TR or RN was made based on the combination of multidisciplinary joint clinic decisions, histopathological examination, and clinic-radiological follow-up as applicable. Results 62 patients were included in the study between July 2018 and August 2021. The histology during initial diagnosis was glioblastoma, oligodendroglioma, and astrocytoma in 43, 7, and 6 patients, respectively, while in 6, no definitive histological characterization was available. The median time from radiation (RT) was 23 months. 46 and 16 patients had TR and RN recurrence, respectively. Sensitivity, specificity, and accuracy using MRI were 98, 77, and 94%, respectively. Using PET imaging with T/W cut-off of 2.65, sensitivity, specificity, and accuracy were 79, 84, and 80%, respectively. The best results were obtained using both imaging combined with sensitivity, specificity, and accuracy of 98, 100, and 98%, respectively. Conclusion Combined imaging with MRI and FET-PET offers multiparametric assessment of glioma recurrence that is correlative and complimentary, with higher accuracy and clinical value.
Collapse
Affiliation(s)
- Arpita Sahu
- Department of Radiodiagnosis, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Ronny Mathew
- Department of Radiodiagnosis, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Renuka Ashtekar
- Department of Radiodiagnosis, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Archya Dasgupta
- Department of Radiation Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Ameya Puranik
- Department of Nuclear Medicine, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Abhishek Mahajan
- Department of Radiology, The Clatterbridge Cancer Centre NHS Foundation Trust, Pembroke Place, Liverpool, United Kingdom
| | - Amit Janu
- Department of Radiodiagnosis, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Amitkumar Choudhari
- Department of Radiodiagnosis, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Subhash Desai
- Department of Radiodiagnosis, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Nandakumar G. Patnam
- Department of Radiodiagnosis, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Abhishek Chatterjee
- Department of Radiation Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Vijay Patil
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Yash Jain
- Department of Nuclear Medicine, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Indraja Dev
- Department of Nuclear Medicine, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Sridhar Epari
- Department of Pathology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Ayushi Sahay
- Department of Pathology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Prakash Shetty
- Department of Neurosurgery, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Jayant Goda
- Department of Radiation Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Aliasgar Moiyadi
- Department of Neurosurgery, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Tejpal Gupta
- Department of Radiation Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
9
|
Galldiks N, Lohmann P, Fink GR, Langen KJ. Amino Acid PET in Neurooncology. J Nucl Med 2023; 64:693-700. [PMID: 37055222 DOI: 10.2967/jnumed.122.264859] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/10/2023] [Indexed: 04/15/2023] Open
Abstract
For decades, several amino acid PET tracers have been used to optimize diagnostics in patients with brain tumors. In clinical routine, the most important clinical indications for amino acid PET in brain tumor patients are differentiation of neoplasm from nonneoplastic etiologies, delineation of tumor extent for further diagnostic and treatment planning (i.e., diagnostic biopsy, resection, or radiotherapy), differentiation of treatment-related changes such as pseudoprogression or radiation necrosis after radiation or chemoradiation from tumor progression at follow-up, and assessment of response to anticancer therapy, including prediction of patient outcome. This continuing education article addresses the diagnostic value of amino acid PET for patients with either glioblastoma or metastatic brain cancer.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany;
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany; and
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany; and
- Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
10
|
Ramakrishnan D, von Reppert M, Krycia M, Sala M, Mueller S, Aneja S, Nabavizadeh A, Galldiks N, Lohmann P, Raji C, Ikuta I, Memon F, Weinberg BD, Aboian MS. Evolution and implementation of radiographic response criteria in neuro-oncology. Neurooncol Adv 2023; 5:vdad118. [PMID: 37860269 PMCID: PMC10584081 DOI: 10.1093/noajnl/vdad118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
Radiographic response assessment in neuro-oncology is critical in clinical practice and trials. Conventional criteria, such as the MacDonald and response assessment in neuro-oncology (RANO) criteria, rely on bidimensional (2D) measurements of a single tumor cross-section. Although RANO criteria are established for response assessment in clinical trials, there is a critical need to address the complexity of brain tumor treatment response with multiple new approaches being proposed. These include volumetric analysis of tumor compartments, structured MRI reporting systems like the Brain Tumor Reporting and Data System, and standardized approaches to advanced imaging techniques to distinguish tumor response from treatment effects. In this review, we discuss the strengths and limitations of different neuro-oncology response criteria and summarize current research findings on the role of novel response methods in neuro-oncology clinical trials and practice.
Collapse
Affiliation(s)
- Divya Ramakrishnan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Marc von Reppert
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Mark Krycia
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Matthew Sala
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Sabine Mueller
- Department of Neurology, Neurosurgery, and Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Sanjay Aneja
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ali Nabavizadeh
- Department of Radiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Cologne, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-4), Research Center Juelich, Juelich, Germany
| | - Cyrus Raji
- Department of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Ichiro Ikuta
- Department of Radiology, Mayo Clinic, Phoenix, Arizona, USA
| | - Fatima Memon
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Brent D Weinberg
- Department of Radiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mariam S Aboian
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
11
|
Mallum A, Mkhize T, Akudugu JM, Ngwa W, Vorster M. The Role of Positron Emission Tomography and Computed Tomographic (PET/CT) Imaging for Radiation Therapy Planning: A Literature Review. Diagnostics (Basel) 2022; 13:diagnostics13010053. [PMID: 36611345 PMCID: PMC9818506 DOI: 10.3390/diagnostics13010053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/15/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
PET/CT is revolutionising radiotherapy treatment planning in many cancer sites. While its utility has been confirmed in some cancer sites, and is used in routine clinical practice, it is still at an experimental stage in many other cancer sites. This review discusses the utility of PET/CT in cancer sites where the role of PET/CT has been established in cases such as head and neck, cervix, brain, and lung cancers, as well as cancer sites where the role of PET/CT is still under investigation such as uterine, ovarian, and prostate cancers. Finally, the review touches on PET/CT utilisation in Africa.
Collapse
Affiliation(s)
- Abba Mallum
- Department of Radiotherapy and Oncology, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
- Department of Radiotherapy and Oncology, Inkosi Albert Luthuli Central Hospital, Durban 4091, South Africa
- University of Maiduguri Teaching Hospital, Maiduguri 600104, Nigeria
- Correspondence: or
| | - Thokozani Mkhize
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
- Department of Nuclear Medicine, Inkosi Albert Central Hospital, Durban 4091, South Africa
| | - John M. Akudugu
- Division of Radiobiology, Department of Medical Imaging and Clinical Oncology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Wilfred Ngwa
- School of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Brigham and Women’s Hospital, Dana-Farmer Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Mariza Vorster
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
- Department of Nuclear Medicine, Inkosi Albert Central Hospital, Durban 4091, South Africa
| |
Collapse
|
12
|
Application of Metabolic Reprogramming to Cancer Imaging and Diagnosis. Int J Mol Sci 2022; 23:ijms232415831. [PMID: 36555470 PMCID: PMC9782057 DOI: 10.3390/ijms232415831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Cellular metabolism governs the signaling that supports physiological mechanisms and homeostasis in an individual, including neuronal transmission, wound healing, and circadian clock manipulation. Various factors have been linked to abnormal metabolic reprogramming, including gene mutations, epigenetic modifications, altered protein epitopes, and their involvement in the development of disease, including cancer. The presence of multiple distinct hallmarks and the resulting cellular reprogramming process have gradually revealed that these metabolism-related molecules may be able to be used to track or prevent the progression of cancer. Consequently, translational medicines have been developed using metabolic substrates, precursors, and other products depending on their biochemical mechanism of action. It is important to note that these metabolic analogs can also be used for imaging and therapeutic purposes in addition to competing for metabolic functions. In particular, due to their isotopic labeling, these compounds may also be used to localize and visualize tumor cells after uptake. In this review, the current development status, applicability, and limitations of compounds targeting metabolic reprogramming are described, as well as the imaging platforms that are most suitable for each compound and the types of cancer to which they are most appropriate.
Collapse
|
13
|
Goeckeritz J, Cerillo J, Sanghadia C, Hosseini M, Clark A, Pierre K, Lucke-Wold B. Principles of Lung Cancer Metastasis to Brain. JOURNAL OF SKELETON SYSTEM 2022; 1:https://www.mediresonline.org/article/principles-of-lung-cancer-metastasis-to-brain. [PMID: 36745145 PMCID: PMC9893877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lung cancer is a disease associated with significant morbidity and mortality on a global setting. This form of cancer commonly gives raise to metastatic lesions the brain, which can further worsen outcomes. In this focused review, we discuss an overview of lung cancers that metastasize to the brain: known risk factors; means of detection and diagnosis; and options for treatment including a comparison between surgical resection, stereotactic radiosurgery, and whole-brain radiation therapy. These interventions are still being assessed by clinical trials and continue to be modified through evidence-based practice.
Collapse
Affiliation(s)
| | - John Cerillo
- College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL
| | | | | | - Alec Clark
- College of Medicine, University of Central Florida, Orlando, FL
| | - Kevin Pierre
- Department of Radiology, University of Florida, Gainesville, FL
| | | |
Collapse
|
14
|
McCarthy L, Verma G, Hangel G, Neal A, Moffat BA, Stockmann JP, Andronesi OC, Balchandani P, Hadjipanayis CG. Application of 7T MRS to High-Grade Gliomas. AJNR Am J Neuroradiol 2022; 43:1378-1395. [PMID: 35618424 PMCID: PMC9575545 DOI: 10.3174/ajnr.a7502] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/11/2022] [Indexed: 01/26/2023]
Abstract
MRS, including single-voxel spectroscopy and MR spectroscopic imaging, captures metabolites in high-grade gliomas. Emerging evidence indicates that 7T MRS may be more sensitive to aberrant metabolic activity than lower-field strength MRS. However, the literature on the use of 7T MRS to visualize high-grade gliomas has not been summarized. We aimed to identify metabolic information provided by 7T MRS, optimal spectroscopic sequences, and areas for improvement in and new applications for 7T MRS. Literature was found on PubMed using "high-grade glioma," "malignant glioma," "glioblastoma," "anaplastic astrocytoma," "7T," "MR spectroscopy," and "MR spectroscopic imaging." 7T MRS offers higher SNR, modestly improved spatial resolution, and better resolution of overlapping resonances. 7T MRS also yields reduced Cramér-Rao lower bound values. These features help to quantify D-2-hydroxyglutarate in isocitrate dehydrogenase 1 and 2 gliomas and to isolate variable glutamate, increased glutamine, and increased glycine with higher sensitivity and specificity. 7T MRS may better characterize tumor infiltration and treatment effect in high-grade gliomas, though further study is necessary. 7T MRS will benefit from increased sample size; reductions in field inhomogeneity, specific absorption rate, and acquisition time; and advanced editing techniques. These findings suggest that 7T MRS may advance understanding of high-grade glioma metabolism, with reduced Cramér-Rao lower bound values and better measurement of smaller metabolite signals. Nevertheless, 7T is not widely used clinically, and technical improvements are necessary. 7T MRS isolates metabolites that may be valuable therapeutic targets in high-grade gliomas, potentially resulting in wider ranging neuro-oncologic applications.
Collapse
Affiliation(s)
- L McCarthy
- From the Department of Neurosurgery (L.M., C.G.H.), Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York
| | - G Verma
- BioMedical Engineering and Imaging Institute (G.V., P.B.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - G Hangel
- Department of Neurosurgery (G.H.)
- High-field MR Center (G.H.), Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - A Neal
- Department of Medicine (A.N.), Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
- Department of Neurology (A.N.), Royal Melbourne Hospital, Melbourne, Australia
| | - B A Moffat
- The Melbourne Brain Centre Imaging Unit (B.A.M.), Department of Radiology, The University of Melbourne, Melbourne, Australia
| | - J P Stockmann
- A. A. Martinos Center for Biomedical Imaging (J.P.S., O.C.A.), Massachusetts General Hospital, Charlestown, Massachusetts
- Harvard Medical School (J.P.S., O.C.A.), Boston, Massachusetts
| | - O C Andronesi
- A. A. Martinos Center for Biomedical Imaging (J.P.S., O.C.A.), Massachusetts General Hospital, Charlestown, Massachusetts
- Harvard Medical School (J.P.S., O.C.A.), Boston, Massachusetts
| | - P Balchandani
- BioMedical Engineering and Imaging Institute (G.V., P.B.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - C G Hadjipanayis
- From the Department of Neurosurgery (L.M., C.G.H.), Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York
| |
Collapse
|
15
|
Smeraldo A, Ponsiglione AM, Soricelli A, Netti PA, Torino E. Update on the Use of PET/MRI Contrast Agents and Tracers in Brain Oncology: A Systematic Review. Int J Nanomedicine 2022; 17:3343-3359. [PMID: 35937076 PMCID: PMC9346926 DOI: 10.2147/ijn.s362192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
The recent advancements in hybrid positron emission tomography–magnetic resonance imaging systems (PET/MRI) have brought massive value in the investigation of disease processes, in the development of novel treatments, in the monitoring of both therapy response and disease progression, and, not least, in the introduction of new multidisciplinary molecular imaging approaches. While offering potential advantages over PET/CT, the hybrid PET/MRI proved to improve both the image quality and lesion detectability. In particular, it showed to be an effective tool for the study of metabolic information about lesions and pathological conditions affecting the brain, from a better tumor characterization to the analysis of metabolic brain networks. Based on the PRISMA guidelines, this work presents a systematic review on PET/MRI in basic research and clinical differential diagnosis on brain oncology and neurodegenerative disorders. The analysis includes literature works and clinical case studies, with a specific focus on the use of PET tracers and MRI contrast agents, which are usually employed to perform hybrid PET/MRI studies of brain tumors. A systematic literature search for original diagnostic studies is performed using PubMed/MEDLINE, Scopus and Web of Science. Patients, study, and imaging characteristics were extracted from the selected articles. The analysis included acquired data pooling, heterogeneity testing, sensitivity analyses, used tracers, and reported patient outcomes. Our analysis shows that, while PET/MRI for the brain is a promising diagnostic method for early diagnosis, staging and recurrence in patients with brain diseases, a better definition of the role of tracers and imaging agents in both clinical and preclinical hybrid PET/MRI applications is needed and further efforts should be devoted to the standardization of the contrast imaging protocols, also considering the emerging agents and multimodal probes.
Collapse
Affiliation(s)
- Alessio Smeraldo
- Department of Chemical, Materials and Production Engineering, University of Naples “Federico II”, Naples, 80125, Italy
- Interdisciplinary Research Center on Biomaterials, CRIB, Naples, 80125, Italy
- Center for Advanced Biomaterials for Health Care, CABHC, Istituto Italiano di Tecnologia, IIT@CRIB, Naples, 80125, Italy
| | - Alfonso Maria Ponsiglione
- Department of Chemical, Materials and Production Engineering, University of Naples “Federico II”, Naples, 80125, Italy
| | - Andrea Soricelli
- Department of Motor Sciences and Healthiness, University of Naples “Parthenope”, Naples, 80133, Italy
| | - Paolo Antonio Netti
- Department of Chemical, Materials and Production Engineering, University of Naples “Federico II”, Naples, 80125, Italy
- Interdisciplinary Research Center on Biomaterials, CRIB, Naples, 80125, Italy
- Center for Advanced Biomaterials for Health Care, CABHC, Istituto Italiano di Tecnologia, IIT@CRIB, Naples, 80125, Italy
| | - Enza Torino
- Department of Chemical, Materials and Production Engineering, University of Naples “Federico II”, Naples, 80125, Italy
- Interdisciplinary Research Center on Biomaterials, CRIB, Naples, 80125, Italy
- Center for Advanced Biomaterials for Health Care, CABHC, Istituto Italiano di Tecnologia, IIT@CRIB, Naples, 80125, Italy
- Correspondence: Enza Torino, Department of Chemical, Materials and Production Engineering, University of Naples “Federico II”, Piazzale Tecchio 80, Naples, 80125, Italy, Tel +39-328-955-8158, Email
| |
Collapse
|
16
|
Mancini L, Casagranda S, Gautier G, Peter P, Lopez B, Thorne L, McEvoy A, Miserocchi A, Samandouras G, Kitchen N, Brandner S, De Vita E, Torrealdea F, Rega M, Schmitt B, Liebig P, Sanverdi E, Golay X, Bisdas S. CEST MRI provides amide/amine surrogate biomarkers for treatment-naïve glioma sub-typing. Eur J Nucl Med Mol Imaging 2022; 49:2377-2391. [PMID: 35029738 PMCID: PMC9165287 DOI: 10.1007/s00259-022-05676-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/31/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Accurate glioma classification affects patient management and is challenging on non- or low-enhancing gliomas. This study investigated the clinical value of different chemical exchange saturation transfer (CEST) metrics for glioma classification and assessed the diagnostic effect of the presence of abundant fluid in glioma subpopulations. METHODS Forty-five treatment-naïve glioma patients with known isocitrate dehydrogenase (IDH) mutation and 1p/19q codeletion status received CEST MRI (B1rms = 2μT, Tsat = 3.5 s) at 3 T. Magnetization transfer ratio asymmetry and CEST metrics (amides: offset range 3-4 ppm, amines: 1.5-2.5 ppm, amide/amine ratio) were calculated with two models: 'asymmetry-based' (AB) and 'fluid-suppressed' (FS). The presence of T2/FLAIR mismatch was noted. RESULTS IDH-wild type had higher amide/amine ratio than IDH-mutant_1p/19qcodel (p < 0.022). Amide/amine ratio and amine levels differentiated IDH-wild type from IDH-mutant (p < 0.0045) and from IDH-mutant_1p/19qret (p < 0.021). IDH-mutant_1p/19qret had higher amides and amines than IDH-mutant_1p/19qcodel (p < 0.035). IDH-mutant_1p/19qret with AB/FS mismatch had higher amines than IDH-mutant_1p/19qret without AB/FS mismatch ( < 0.016). In IDH-mutant_1p/19qret, the presence of AB/FS mismatch was closely related to the presence of T2/FLAIR mismatch (p = 0.014). CONCLUSIONS CEST-derived biomarkers for amides, amines, and their ratio can help with histomolecular staging in gliomas without intense contrast enhancement. T2/FLAIR mismatch is reflected in the presence of AB/FS CEST mismatch. The AB/FS CEST mismatch identifies glioma subgroups that may have prognostic and clinical relevance.
Collapse
Affiliation(s)
- Laura Mancini
- Box65, Lysholm Department of Neuroradiology, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, 8-11 Queen Square, London, WC1N 3BG, UK.
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK.
| | | | | | | | | | - Lewis Thorne
- Department of Neurosurgery, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Andrew McEvoy
- Department of Neurosurgery, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Anna Miserocchi
- Department of Neurosurgery, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - George Samandouras
- Department of Neurosurgery, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Neil Kitchen
- Department of Neurosurgery, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Sebastian Brandner
- Division of Neuropathology, UCL Queen Square Institute of Neurology, London, UK
- The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Enrico De Vita
- Box65, Lysholm Department of Neuroradiology, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, 8-11 Queen Square, London, WC1N 3BG, UK
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| | - Francisco Torrealdea
- University College Hospital, University College of London Hospitals NHS Foundation Trust, London, UK
| | - Marilena Rega
- University College Hospital, University College of London Hospitals NHS Foundation Trust, London, UK
| | | | | | - Eser Sanverdi
- Box65, Lysholm Department of Neuroradiology, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, 8-11 Queen Square, London, WC1N 3BG, UK
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| | - Xavier Golay
- Box65, Lysholm Department of Neuroradiology, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, 8-11 Queen Square, London, WC1N 3BG, UK
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| | - Sotirios Bisdas
- Box65, Lysholm Department of Neuroradiology, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, 8-11 Queen Square, London, WC1N 3BG, UK
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
17
|
Abstract
Renal cell carcinoma (RCC) is the sixth most common cancer among men and the ninth among women, and its prognosis is closely correlated with metastasis. Targeted therapy and immunotherapy are the main adjuvant treatments for advanced RCC and require early diagnosis, precise assessment, and prediction of the therapeutic responses. Current conventional imaging methods of RCC only provide structural information rather than biological processes. Noninvasive diagnostic tools are therefore needed to image RCC early and accurately at the molecular level. Nuclear medicine imaging combines the high sensitivity of radionuclides with the high resolution of structural imaging to visualize the metabolic processes and specific targets of RCC for more accurate and reliable diagnosis, staging, prognosis prediction, and response assessment. This review summarizes the most recent applications of nuclear medicine receptor imaging and metabolic imaging in RCC and highlights future development perspectives in the field.
Collapse
Affiliation(s)
- Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| |
Collapse
|
18
|
Galldiks N, Angenstein F, Werner JM, Bauer EK, Gutsche R, Fink GR, Langen KJ, Lohmann P. Use of advanced neuroimaging and artificial intelligence in meningiomas. Brain Pathol 2022; 32:e13015. [PMID: 35213083 PMCID: PMC8877736 DOI: 10.1111/bpa.13015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/09/2021] [Accepted: 08/02/2021] [Indexed: 01/04/2023] Open
Abstract
Anatomical cross‐sectional imaging methods such as contrast‐enhanced MRI and CT are the standard for the delineation, treatment planning, and follow‐up of patients with meningioma. Besides, advanced neuroimaging is increasingly used to non‐invasively provide detailed insights into the molecular and metabolic features of meningiomas. These techniques are usually based on MRI, e.g., perfusion‐weighted imaging, diffusion‐weighted imaging, MR spectroscopy, and positron emission tomography. Furthermore, artificial intelligence methods such as radiomics offer the potential to extract quantitative imaging features from routinely acquired anatomical MRI and CT scans and advanced imaging techniques. This allows the linking of imaging phenotypes to meningioma characteristics, e.g., the molecular‐genetic profile. Here, we review several diagnostic applications and future directions of these advanced neuroimaging techniques, including radiomics in preclinical models and patients with meningioma.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Center for Integrated Oncology (CIO), Universities of Aachen, Cologne, Germany
| | - Frank Angenstein
- Functional Neuroimaging Group, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany.,Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany.,Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Jan-Michael Werner
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Elena K Bauer
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Robin Gutsche
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Center for Integrated Oncology (CIO), Universities of Aachen, Cologne, Germany.,Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
19
|
Zhang-Yin JT, Girard A, Bertaux M. What Does PET Imaging Bring to Neuro-Oncology in 2022? A Review. Cancers (Basel) 2022; 14:cancers14040879. [PMID: 35205625 PMCID: PMC8870476 DOI: 10.3390/cancers14040879] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Positron emission tomography (PET) imaging is increasingly used to supplement MRI in the management of patient with brain tumors. In this article, we provide a review of the current place and perspectives of PET imaging for the diagnosis and follow-up of from primary brain tumors such as gliomas, meningiomas and central nervous system lymphomas, as well as brain metastases. Different PET radiotracers targeting different biological processes are used to accurately depict these brain tumors and provide unique metabolic and biologic information. Radiolabeled amino acids such as [18F]FDOPA or [18F]FET are used for imaging of gliomas while both [18F]FDG and amino acids can be used for brain metastases. Meningiomas can be seen with a high contrast using radiolabeled ligands of somatostatin receptors, which they usually carry. Unconventional tracers that allow the study of other biological processes such as cell proliferation, hypoxia, or neo-angiogenesis are currently being studied for brain tumors imaging. Abstract PET imaging is being increasingly used to supplement MRI in the clinical management of brain tumors. The main radiotracers implemented in clinical practice include [18F]FDG, radiolabeled amino acids ([11C]MET, [18F]FDOPA, [18F]FET) and [68Ga]Ga-DOTA-SSTR, targeting glucose metabolism, L-amino-acid transport and somatostatin receptors expression, respectively. This review aims at addressing the current place and perspectives of brain PET imaging for patients who suffer from primary or secondary brain tumors, at diagnosis and during follow-up. A special focus is given to the following: radiolabeled amino acids PET imaging for tumor characterization and follow-up in gliomas; the role of amino acid PET and [18F]FDG PET for detecting brain metastases recurrence; [68Ga]Ga-DOTA-SSTR PET for guiding treatment in meningioma and particularly before targeted radiotherapy.
Collapse
Affiliation(s)
| | - Antoine Girard
- Department of Nuclear Medicine, Centre Eugène Marquis, Université Rennes 1, 35000 Rennes, France
| | - Marc Bertaux
- Department of Nuclear Medicine, Foch Hospital, 92150 Suresnes, France
| |
Collapse
|
20
|
Tillmanns N, Lum AE, Cassinelli G, Merkaj S, Verma T, Zeevi T, Staib L, Subramanian H, Bahar RC, Brim W, Lost J, Jekel L, Brackett A, Payabvash S, Ikuta I, Lin M, Bousabarah K, Johnson MH, Cui J, Malhotra A, Omuro A, Turowski B, Aboian MS. Identifying clinically applicable machine learning algorithms for glioma segmentation: recent advances and discoveries. Neurooncol Adv 2022; 4:vdac093. [PMID: 36071926 PMCID: PMC9446682 DOI: 10.1093/noajnl/vdac093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background While there are innumerable machine learning (ML) research algorithms used for segmentation of gliomas, there is yet to be a US FDA cleared product. The aim of this study is to explore the systemic limitations of research algorithms that have prevented translation from concept to product by a review of the current research literature. Methods We performed a systematic literature review on 4 databases. Of 11 727 articles, 58 articles met the inclusion criteria and were used for data extraction and screening using TRIPOD. Results We found that while many articles were published on ML-based glioma segmentation and report high accuracy results, there were substantial limitations in the methods and results portions of the papers that result in difficulty reproducing the methods and translation into clinical practice. Conclusions In addition, we identified that more than a third of the articles used the same publicly available BRaTS and TCIA datasets and are responsible for the majority of patient data on which ML algorithms were trained, which leads to limited generalizability and potential for overfitting and bias.
Collapse
Affiliation(s)
- Niklas Tillmanns
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- University Dusseldorf, Medical Faculty, Department of Diagnostic and Interventional Radiology, Dusseldorf, Germany
| | - Avery E Lum
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Gabriel Cassinelli
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sara Merkaj
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Tej Verma
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Tal Zeevi
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Lawrence Staib
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Harry Subramanian
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ryan C Bahar
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Waverly Brim
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jan Lost
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Leon Jekel
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Alexandria Brackett
- Harvey Cushing/John Hay Whitney Medical Library, Yale University, New Haven, Connecticut, USA
| | - Sam Payabvash
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ichiro Ikuta
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - MingDe Lin
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Visage Imaging, Inc., San Diego, California, USA
| | | | - Michele H Johnson
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jin Cui
- Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Ajay Malhotra
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Antonio Omuro
- Department of Neurology and Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Bernd Turowski
- University Dusseldorf, Medical Faculty, Department of Diagnostic and Interventional Radiology, Dusseldorf, Germany
| | - Mariam S Aboian
- Brain Tumor Research Group, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
21
|
Cheng X, Ma L. Enzymatic synthesis of fluorinated compounds. Appl Microbiol Biotechnol 2021; 105:8033-8058. [PMID: 34625820 PMCID: PMC8500828 DOI: 10.1007/s00253-021-11608-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 12/31/2022]
Abstract
Fluorinated compounds are widely used in the fields of molecular imaging, pharmaceuticals, and materials. Fluorinated natural products in nature are rare, and the introduction of fluorine atoms into organic compound molecules can give these compounds new functions and make them have better performance. Therefore, the synthesis of fluorides has attracted more and more attention from biologists and chemists. Even so, achieving selective fluorination is still a huge challenge under mild conditions. In this review, the research progress of enzymatic synthesis of fluorinated compounds is summarized since 2015, including cytochrome P450 enzymes, aldolases, fluoroacetyl coenzyme A thioesterases, lipases, transaminases, reductive aminases, purine nucleoside phosphorylases, polyketide synthases, fluoroacetate dehalogenases, tyrosine phenol-lyases, glycosidases, fluorinases, and multienzyme system. Of all enzyme-catalyzed synthesis methods, the direct formation of the C-F bond by fluorinase is the most effective and promising method. The structure and catalytic mechanism of fluorinase are introduced to understand fluorobiochemistry. Furthermore, the distribution, applications, and future development trends of fluorinated compounds are also outlined. Hopefully, this review will help researchers to understand the significance of enzymatic methods for the synthesis of fluorinated compounds and find or create excellent fluoride synthase in future research.Key points• Fluorinated compounds are distributed in plants and microorganisms, and are used in imaging, medicine, materials science.• Enzyme catalysis is essential for the synthesis of fluorinated compounds.• The loop structure of fluorinase is the key to forming the C-F bond.
Collapse
Affiliation(s)
- Xinkuan Cheng
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Laboratory of Metabolic Control Fermentation Technology, College of Biotechnology, Tianjin University of Science & Technology, No. 29, Thirteenth Street, Binhai New District, Tianjin, 300457, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Laboratory of Metabolic Control Fermentation Technology, College of Biotechnology, Tianjin University of Science & Technology, No. 29, Thirteenth Street, Binhai New District, Tianjin, 300457, China.
| |
Collapse
|
22
|
Lim TX, Ahamed M, Reutens DC. The aryl hydrocarbon receptor: A diagnostic and therapeutic target in glioma. Drug Discov Today 2021; 27:422-435. [PMID: 34624509 DOI: 10.1016/j.drudis.2021.09.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 07/29/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
Glioblastoma multiforme (GBM) is a deadly disease; 5-year survival rates have shown little improvement over the past 30 years. In vivo positron emission tomography (PET) imaging is an important method of identifying potential diagnostic and therapeutic molecular targets non-invasively. The aryl hydrocarbon receptor (AhR) is a transcription factor that regulates multiple genes involved in immune response modulation and tumorigenesis. The AhR is an attractive potential drug target and studies have shown that its activation by small molecules can modulate innate and adaptive immunity beneficially and prevent AhR-mediated tumour promotion in several cancer types. In this review, we provide an overview of the role of the AhR in glioma tumorigenesis and highlight its potential as an emerging biomarker for glioma therapies targeting the tumour immune response and PET diagnostics.
Collapse
Affiliation(s)
- Ting Xiang Lim
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Muneer Ahamed
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - David C Reutens
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
23
|
Ma H, Zhao J, Liu S, Xie D, Zhang Z, Nie D, Wen F, Yang Z, Tang G. 18F-Trifluoromethylated D-Cysteine as a Promising New PET Tracer for Glioma Imaging: Comparative Analysis With MRI and Histopathology in Orthotopic C6 Models. Front Oncol 2021; 11:645162. [PMID: 33996562 PMCID: PMC8117348 DOI: 10.3389/fonc.2021.645162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
Comparing MRI and histopathology, this study aims to comprehensively explore the potential application of 18F-trifluoromethylated D-cysteine (S-[18F]CF3-D-CYS) in evaluating glioma by using orthotopic C6 glioma models. Sprague-Dawley (SD) rats (n = 9) were implanted with C6 glioma cells. Tumor growth was monitored every week by multiparameter MRI [including dynamic contrast-enhanced MRI (DCE-MRI)], [18F]FDG, S-[18F]CF3-D-CYS, and [18F]FDOPA PET imaging. Repeated scans of the same rat with the two or three [18F]-labeled radiotracers were investigated. Initial regions of interest were manually delineated on T2WI and set on the same level of PET images, and tumor-to-normal brain uptake ratios (TNRs) were calculated to semiquantitatively assess the tracer accumulation in the tumor. The tumor volume in PET and histopathology was calculated. HE and Ki67 immunohistochemical staining were further performed. The correlations between the uptake of S-[18F]CF3-D-CYS and Ki67 were analyzed. Dynamic S-[18F]CF3-D-CYS PET imaging showed tumor uptake rapidly reached a peak, maintained plateau during 10-30 min after injection, then decreased slowly. Compared with [18F]FDG and [18F]FDOPA PET imaging, S-[18F]CF3-D-CYS PET demonstrated the highest TNRs (P < 0.05). There were no significant differences in the tumor volume measured on S-[18F]CF3-D-CYS PET or HE specimen. Furthermore, our results showed that the uptake of S-[18F]CF3-D-CYS was significantly positively correlated with tumor Ki67, and the poor accumulated S-[18F]CF3-D-CYS was consistent with tumor hemorrhage. There was no significant correlation between the S-[18F]CF3-D-CYS uptakes and the Ktrans values derived from DCE-MRI. In comparison with MRI and histopathology, S-[18F]CF3-D-CYS PET performs well in the diagnosis and evaluation of glioma. S-[18F]CF3-D-CYS PET may serve as a valuable tool in the clinical management of gliomas.
Collapse
Affiliation(s)
- Hui Ma
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Zhao
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shaoyu Liu
- Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Nuclear Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dingxiang Xie
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhanwen Zhang
- Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Nuclear Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dahong Nie
- Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fuhua Wen
- Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiyun Yang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ganghua Tang
- Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Nanfang PET Center, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
24
|
The role of 11C-methionine PET in patients with negative diffusion-weighted magnetic resonance imaging: correlation with histology and molecular biomarkers in operated gliomas. Nucl Med Commun 2021; 41:696-705. [PMID: 32371671 DOI: 10.1097/mnm.0000000000001202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To compare 11C-methionine (11C-METH) PET with diffusion-weighted MRI (DWI-MRI) diagnostic accuracy and prognostic value in patients with glioma candidate to neurosurgery. METHODS We collected and analyzed data from 124 consecutive patients (n = 124) investigated during preoperative work-up. Both visual and semiquantitative parameters were utilized for image analysis. The reference standard was based on histopathology. The median follow-up was 14.3 months. RESULTS Overall, 47 high-grade gliomas (HGG) and 77 low-grade gliomas (LGG) were diagnosed. On visual assessment, sensitivity and specificity for differentiating HGG from LGG were 80.8 and 59.7% for DWI-MRI, versus 95.7 and 41.5% for 11C-METH PET, respectively. On semiquantitative analysis, the sensitivity, specificity, and area under the curve were 78.7, 71.4, and 80.4% for SUVmax, 78.7, 70.1, and 81.1% for SUVratio, and 74.5, 61, and 76.7% for MTB (metabolic tumor burden), respectively. In patients with negative DWI-MRI and IDH-wild type, SUVmax and SUVratio were higher compared to IDH-mutated (P = 0.025 and P = 0.01, respectively). In LGG, patients with 1p/19q codeletion showed higher SUVmax (P = 0.044). In all patients with negative DWI-MRI, median PFS was longer for SUVmax <3.9 (median not reached vs 34.2 months, P = 0.004), SUVratio <2.3 (median not reached vs 21.5 months, P < 0.001), and MTB <3.1 (median not reached vs 45.7 months, P = 0.05). In LGG patients with negative DWI-MRI, only SUVratio <2.3 and MTB <3.1 were associated with longer PFS (P = 0.016 and P = 0.024, respectively). CONCLUSION C-METH PET was found highly sensitive for glioma differentiation and molecular characterization. In DWI-negative patients, PET parameters correlated with molecular profile were associated with clinical outcome.
Collapse
|
25
|
D'Amore F, Grinberg F, Mauler J, Galldiks N, Blazhenets G, Farrher E, Filss C, Stoffels G, Mottaghy FM, Lohmann P, Shah NJ, Langen KJ. Combined 18F-FET PET and diffusion kurtosis MRI in posttreatment glioblastoma: differentiation of true progression from treatment-related changes. Neurooncol Adv 2021; 3:vdab044. [PMID: 34013207 PMCID: PMC8117449 DOI: 10.1093/noajnl/vdab044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Radiological differentiation of tumor progression (TPR) from treatment-related changes (TRC) in pretreated glioblastoma is crucial. This study aimed to explore the diagnostic value of diffusion kurtosis MRI combined with information derived from O-(2-[18F]-fluoroethyl)-l-tyrosine (18F-FET) PET for the differentiation of TPR from TRC in patients with pretreated glioblastoma. Methods Thirty-two patients with histomolecularly defined and pretreated glioblastoma suspected of having TPR were included in this retrospective study. Twenty-one patients were included in the TPR group, and 11 patients in the TRC group, as assessed by neuropathology or clinicoradiological follow-up. Three-dimensional (3D) regions of interest were generated based on increased 18F-FET uptake using a tumor-to-brain ratio of 1.6. Furthermore, diffusion MRI kurtosis maps were obtained from the same regions of interest using co-registered 18F-FET PET images, and advanced histogram analysis of diffusion kurtosis map parameters was applied to generated 3D regions of interest. Diagnostic accuracy was analyzed by receiver operating characteristic curve analysis and combinations of PET and MRI parameters using multivariate logistic regression. Results Parameters derived from diffusion MRI kurtosis maps show high diagnostic accuracy, up to 88%, for differentiating between TPR and TRC. Logistic regression revealed that the highest diagnostic accuracy of 94% (area under the curve, 0.97; sensitivity, 94%; specificity, 91%) was achieved by combining the maximum tumor-to-brain ratio of 18F-FET uptake and diffusion MRI kurtosis metrics. Conclusions The combined use of 18F-FET PET and MRI diffusion kurtosis maps appears to be a promising approach to improve the differentiation of TPR from TRC in pretreated glioblastoma and warrants further investigation.
Collapse
Affiliation(s)
- Francesco D'Amore
- Institute of Neuroscience and Medicine, Research Centre Juelich, Juelich, Germany.,Department of Neuroradiology, Circolo Hospital and Macchi Foundation, Varese, Italy
| | - Farida Grinberg
- Institute of Neuroscience and Medicine, Research Centre Juelich, Juelich, Germany
| | - Jörg Mauler
- Institute of Neuroscience and Medicine, Research Centre Juelich, Juelich, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine, Research Centre Juelich, Juelich, Germany.,Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Duesseldorf, Germany
| | - Ganna Blazhenets
- Institute of Neuroscience and Medicine, Research Centre Juelich, Juelich, Germany.,Department of Nuclear Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ezequiel Farrher
- Institute of Neuroscience and Medicine, Research Centre Juelich, Juelich, Germany
| | - Christian Filss
- Institute of Neuroscience and Medicine, Research Centre Juelich, Juelich, Germany.,Department of Nuclear Medicine, RWTH Aachen University, Aachen, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine, Research Centre Juelich, Juelich, Germany
| | - Felix M Mottaghy
- Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Duesseldorf, Germany.,Department of Nuclear Medicine, RWTH Aachen University, Aachen, Germany.,Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands.,Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine, Research Centre Juelich, Juelich, Germany.,Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nadim Jon Shah
- Institute of Neuroscience and Medicine, Research Centre Juelich, Juelich, Germany.,Department of Neurology, RWTH Aachen University, Aachen, Germany.,JARA-BRAIN-Translational Medicine, Aachen, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Research Centre Juelich, Juelich, Germany.,Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Duesseldorf, Germany.,Department of Nuclear Medicine, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
26
|
Castellano A, Bailo M, Cicone F, Carideo L, Quartuccio N, Mortini P, Falini A, Cascini GL, Minniti G. Advanced Imaging Techniques for Radiotherapy Planning of Gliomas. Cancers (Basel) 2021; 13:cancers13051063. [PMID: 33802292 PMCID: PMC7959155 DOI: 10.3390/cancers13051063] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
The accuracy of target delineation in radiation treatment (RT) planning of cerebral gliomas is crucial to achieve high tumor control, while minimizing treatment-related toxicity. Conventional magnetic resonance imaging (MRI), including contrast-enhanced T1-weighted and fluid-attenuated inversion recovery (FLAIR) sequences, represents the current standard imaging modality for target volume delineation of gliomas. However, conventional sequences have limited capability to discriminate treatment-related changes from viable tumors, owing to the low specificity of increased blood-brain barrier permeability and peritumoral edema. Advanced physiology-based MRI techniques, such as MR spectroscopy, diffusion MRI and perfusion MRI, have been developed for the biological characterization of gliomas and may circumvent these limitations, providing additional metabolic, structural, and hemodynamic information for treatment planning and monitoring. Radionuclide imaging techniques, such as positron emission tomography (PET) with amino acid radiopharmaceuticals, are also increasingly used in the workup of primary brain tumors, and their integration in RT planning is being evaluated in specialized centers. This review focuses on the basic principles and clinical results of advanced MRI and PET imaging techniques that have promise as a complement to RT planning of gliomas.
Collapse
Affiliation(s)
- Antonella Castellano
- Neuroradiology Unit, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (A.C.); (A.F.)
| | - Michele Bailo
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (M.B.); (P.M.)
| | - Francesco Cicone
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, and Nuclear Medicine Unit, University Hospital “Mater Domini”, 88100 Catanzaro, Italy;
- Correspondence: ; Tel.: +39-0-961-369-4155
| | - Luciano Carideo
- National Cancer Institute, G. Pascale Foundation, 80131 Naples, Italy;
| | - Natale Quartuccio
- A.R.N.A.S. Ospedale Civico Di Cristina Benfratelli, 90144 Palermo, Italy;
| | - Pietro Mortini
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (M.B.); (P.M.)
| | - Andrea Falini
- Neuroradiology Unit, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (A.C.); (A.F.)
| | - Giuseppe Lucio Cascini
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, and Nuclear Medicine Unit, University Hospital “Mater Domini”, 88100 Catanzaro, Italy;
| | - Giuseppe Minniti
- Radiation Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, 53100 Siena, Italy;
- IRCCS Neuromed, 86077 Pozzilli (IS), Italy
| |
Collapse
|
27
|
Overcast WB, Davis KM, Ho CY, Hutchins GD, Green MA, Graner BD, Veronesi MC. Advanced imaging techniques for neuro-oncologic tumor diagnosis, with an emphasis on PET-MRI imaging of malignant brain tumors. Curr Oncol Rep 2021; 23:34. [PMID: 33599882 PMCID: PMC7892735 DOI: 10.1007/s11912-021-01020-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW This review will explore the latest in advanced imaging techniques, with a focus on the complementary nature of multiparametric, multimodality imaging using magnetic resonance imaging (MRI) and positron emission tomography (PET). RECENT FINDINGS Advanced MRI techniques including perfusion-weighted imaging (PWI), MR spectroscopy (MRS), diffusion-weighted imaging (DWI), and MR chemical exchange saturation transfer (CEST) offer significant advantages over conventional MR imaging when evaluating tumor extent, predicting grade, and assessing treatment response. PET performed in addition to advanced MRI provides complementary information regarding tumor metabolic properties, particularly when performed simultaneously. 18F-fluoroethyltyrosine (FET) PET improves the specificity of tumor diagnosis and evaluation of post-treatment changes. Incorporation of radiogenomics and machine learning methods further improve advanced imaging. The complementary nature of combining advanced imaging techniques across modalities for brain tumor imaging and incorporating technologies such as radiogenomics has the potential to reshape the landscape in neuro-oncology.
Collapse
Affiliation(s)
- Wynton B. Overcast
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N University Blvd. Room 0663, Indianapolis, IN 46202 USA
| | - Korbin M. Davis
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N University Blvd. Room 0663, Indianapolis, IN 46202 USA
| | - Chang Y. Ho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Goodman Hall, 355 West 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - Gary D. Hutchins
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Research 2 Building (R2), Room E124, 920 W. Walnut Street, Indianapolis, IN 46202-5181 USA
| | - Mark A. Green
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Research 2 Building (R2), Room E124, 920 W. Walnut Street, Indianapolis, IN 46202-5181 USA
| | - Brian D. Graner
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Goodman Hall, 355 West 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - Michael C. Veronesi
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Research 2 Building (R2), Room E174, 920 W. Walnut Street, Indianapolis, IN 46202-5181 USA
| |
Collapse
|
28
|
Comparison of Amino Acid PET to Advanced and Emerging MRI Techniques for Neurooncology Imaging: A Systematic Review of the Recent Studies. Mol Imaging 2021; 2021:8874078. [PMID: 34194287 PMCID: PMC8205602 DOI: 10.1155/2021/8874078] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/23/2020] [Accepted: 11/17/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction Standard neuroimaging protocols for brain tumors have well-known limitations. The clinical use of additional modalities including amino acid PET (aaPET) and advanced MRI (aMRI) techniques (including DWI, PWI, and MRS) is emerging in response to the need for more accurate detection of brain tumors. In this systematic review of the past 2 years of the literature, we discuss the most recent studies that directly compare or combine aaPET and aMRI for brain tumor imaging. Methods A PubMed search was conducted for human studies incorporating both aaPET and aMRI and published between July 2018 and August 2020. Results A total of 22 studies were found in the study period. Recent studies of aaPET with DWI showed a superiority of MET, FET, FDOPA, and AMT PET for detecting tumor, predicting recurrence, diagnosing progression, and predicting survival. Combining modalities further improved performance. Comparisons of aaPET with PWI showed mixed results about spatial correlation. However, both modalities were able to detect high-grade tumors, identify tumor recurrence, differentiate recurrence from treatment effects, and predict survival. aaPET performed better on these measures than PWI, but when combined, they had the strongest results. Studies of aaPET with MRS demonstrated that both modalities have diagnostic potential but MET PET and FDOPA PET performed better than MRS. MRS suffered from some data quality issues that limited analysis in two studies, and, in one study that combined modalities, overall performance actually decreased. Four recent studies compared aaPET with emerging MRI approaches (such as CEST imaging, MR fingerprinting, and SISTINA), but the initial results remain inconclusive. Conclusions aaPET outperformed the aMRI imaging techniques in most recent studies. DWI and PWI added meaningful complementary data, and the combination of aaPET with aMRI yielded the best results in most studies.
Collapse
|
29
|
Reuter G, Moïse M, Roll W, Martin D, Lombard A, Scholtes F, Stummer W, Suero Molina E. Conventional and advanced imaging throughout the cycle of care of gliomas. Neurosurg Rev 2021; 44:2493-2509. [PMID: 33411093 DOI: 10.1007/s10143-020-01448-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 10/22/2022]
Abstract
Although imaging of gliomas has evolved tremendously over the last decades, published techniques and protocols are not always implemented into clinical practice. Furthermore, most of the published literature focuses on specific timepoints in glioma management. This article reviews the current literature on conventional and advanced imaging techniques and chronologically outlines their practical relevance for the clinical management of gliomas throughout the cycle of care. Relevant articles were located through the Pubmed/Medline database and included in this review. Interpretation of conventional and advanced imaging techniques is crucial along the entire process of glioma care, from diagnosis to follow-up. In addition to the described currently existing techniques, we expect deep learning or machine learning approaches to assist each step of glioma management through tumor segmentation, radiogenomics, prognostication, and characterization of pseudoprogression. Thorough knowledge of the specific performance, possibilities, and limitations of each imaging modality is key for their adequate use in glioma management.
Collapse
Affiliation(s)
- Gilles Reuter
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium. .,GIGA-CRC In-vivo Imaging Center, ULiege, Liège, Belgium.
| | - Martin Moïse
- Department of Radiology, University Hospital of Liège, Liège, Belgium
| | - Wolfgang Roll
- Department of Nuclear Medicine, University Hospital of Münster, Münster, Germany
| | - Didier Martin
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium
| | - Arnaud Lombard
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium
| | - Félix Scholtes
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium.,Department of Neuroanatomy, University of Liège, Liège, Belgium
| | - Walter Stummer
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| | - Eric Suero Molina
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| |
Collapse
|
30
|
Solnes LB, Jacobs AH, Coughlin JM, Du Y, Goel R, Hammoud DA, Pomper MG. Central Nervous System Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00088-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
31
|
Aboian M, Barajas R, Shatalov J, Ravanfar V, Bahroos E, Tong E, Taylor JW, Bush NO, Sneed P, Seo Y, Cha S, Hernandez-Pampaloni M. Maximizing the use of batch production of 18F-FDOPA for imaging of brain tumors to increase availability of hybrid PET/MR imaging in clinical setting. Neurooncol Pract 2020; 8:91-97. [PMID: 33664973 DOI: 10.1093/nop/npaa065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Amino acid PET imaging of brain tumors has been shown to play an important role in predicting tumor grade, delineation of tumor margins, and differentiating tumor recurrence from the background of postradiation changes, but is not commonly used in clinical practice because of high cost. We propose that PET/MRI imaging of patients grouped to the day of tracer radiosynthesis will significantly decrease the cost of PET imaging, which will improve patient access to PET. Methods Seventeen patients with either primary brain tumors or metastatic brain tumors were recruited for imaging on 3T PET/MRI and were scanned on 4 separate days in groups of 3 to 5 patients. The first group of consecutively imaged patients contained 3 patients, followed by 2 groups of 5 patients, and a last group of 4 patients. Results For each of the patients, standard of care gadolinium-enhanced MRI and dynamic PET imaging with 18F-FDOPA amino acid tracer was obtained. The total cost savings of scanning 17 patients in batches of 4 as opposed to individual radiosynthesis was 48.5% ($28 321). Semiquantitative analysis of tracer uptake in normal brain were performed with appropriate accumulation and expected subsequent washout. Conclusion Amino acid PET tracers have been shown to play a critical role in the characterization of brain tumors but their adaptation to clinical practice has been limited because of the high cost of PET. Scheduling patient imaging to maximally use the radiosynthesis of imaging tracer significantly reduces the cost of PET and results in increased availability of PET tracer use in neuro-oncology.
Collapse
Affiliation(s)
- Mariam Aboian
- Department of Radiology, Yale University School of Medicine, New Haven, CT
| | - Ramon Barajas
- Department of Radiology, Oregon Health Sciences University
| | - Julia Shatalov
- Department of Radiology, Yale University School of Medicine, New Haven, CT
| | - Vahid Ravanfar
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Emma Bahroos
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Elizabeth Tong
- Department of Radiology, Stanford University, Palo Alto, California
| | - Jennie W Taylor
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California.,Department of Neurology, University of California San Francisco, San Francisco, California
| | - N Oberheim Bush
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Patricia Sneed
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Miguel Hernandez-Pampaloni
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| |
Collapse
|
32
|
Zhou W, Zhou Z, Wen J, Xie F, Zhu Y, Zhang Z, Xiao J, Chen Y, Li M, Guan Y, Hua T. A Nomogram Modeling 11C-MET PET/CT and Clinical Features in Glioma Helps Predict IDH Mutation. Front Oncol 2020; 10:1200. [PMID: 32850348 PMCID: PMC7396495 DOI: 10.3389/fonc.2020.01200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose: We developed a 11C-Methionine positron emission tomography/computed tomography (11C-MET PET/CT)-based nomogram model that uses easy-accessible imaging and clinical features to achieve reliable non-invasive isocitrate dehydrogenase (IDH)-mutant prediction with strong clinical translational capability. Methods: One hundred and ten patients with pathologically proven glioma who underwent pretreatment 11C-MET PET/CT were retrospectively reviewed. IDH genotype was determined by IDH1 R132H immunohistochemistry staining. Maximum, mean and peak tumor-to-normal brain tissue (TNRmax, TNRmean, TNRpeak), metabolic tumor volume (MTV), total lesion methionine uptake (TLMU), and standard deviation of SUV (SUVSD) of the lesions on MET PET images were obtained via a dedicated workstation (Siemens. syngo.via). Univariate and multivariate logistic regression models were used to identify the predictive factors for IDH mutation. Nomogram and calibration plots were further performed. Results: In the entire population, TNRmean, TNRmax, TNRpeak, and SUVSD of IDH-mutant glioma patients were significantly lower than these values of IDH wildtype. Receiver operating characteristic (ROC) analysis suggested SUVSD had the best performance for IDH-mutant discrimination (AUC = 0.731, cut-off ≤ 0.29, p < 0.001). All pairs of the 11C-MET PET metrics showed linear associations by Pearson correlation coefficients between 0.228 and 0.986. Multivariate analyses demonstrated that SUVSD (>0.29 vs. ≤ 0.29 OR: 0.053, p = 0.010), dichotomized brain midline structure involvement (no vs. yes OR: 26.52, p = 0.000) and age (≤ 45 vs. >45 years OR: 3.23, p = 0.023), were associated with a higher incidence of IDH mutation. The nomogram modeling showed good discrimination, with a C-statistics of 0.866 (95% CI: 0.796–0.937) and was well-calibrated. Conclusions:11C-Methionine PET/CT imaging features (SUVSD and the involvement of brain midline structure) can be conveniently used to facilitate the pre-operative prediction of IDH genotype. The nomogram model based on 11C-Methionine PET/CT and clinical age features might be clinically useful in non-invasive IDH mutation status prediction for untreated glioma patients.
Collapse
Affiliation(s)
- Weiyan Zhou
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhirui Zhou
- Department of Radiotherapy, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianbo Wen
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuhua Zhu
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhengwei Zhang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianfei Xiao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yijing Chen
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ming Li
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Tao Hua
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Hybrid PET- MRI is a technique that has the ability to improve diagnostic accuracy in many applications, whereas PET and MRI performed separately often fail to provide accurate responses to clinical questions. Here, we review recent studies and current developments in PET-MRI, focusing on clinical applications. RECENT FINDINGS The combination of PET and MRI imaging methods aims at increasing the potential of each individual modality. Combined methods of image reconstruction and correction of PET-MRI attenuation are being developed, and a number of applications are being introduced into clinical practice. To date, the value of PET-MRI has been demonstrated for the evaluation of brain tumours in epilepsy and neurodegenerative diseases. Continued advances in data analysis regularly improve the efficiency and the potential application of multimodal biomarkers. SUMMARY PET-MRI provides simultaneous of anatomical, functional, biochemical and metabolic information for the personalized characterization and monitoring of neurological diseases. In this review, we show the advantage of the complementarity of different biomarkers obtained using PET-MRI data. We also present the recent advances made in this hybrid imaging modality and its advantages in clinical practice compared with MRI and PET separately.
Collapse
|
34
|
Galldiks N, Lohmann P, Werner JM, Ceccon G, Fink GR, Langen KJ. Molecular imaging and advanced MRI findings following immunotherapy in patients with brain tumors. Expert Rev Anticancer Ther 2019; 20:9-15. [PMID: 31842635 DOI: 10.1080/14737140.2020.1705788] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Currently, immunotherapy using vaccination strategies or oncolytic virus approaches, cell-based immunotherapy, and the blockade of immune checkpoints are under evaluation in patients with brain cancer. Here we summarize clinically significant imaging findings such as treatment-related changes detected by advanced neuroimaging techniques following the most suitable immunotherapy options currently used in neuro-oncology. We, furthermore, provide an overview of how these advanced imaging techniques may help to overcome shortcomings of standard MRI in the assessment and follow-up of patients with brain cancer.Areas covered: The current literature on neuroimaging for immunotherapy in the field of brain tumors, with a focus on gliomas and brain metastases is summarized.Expert commentary: Data suggest that imaging parameters primarily derived from amino acid PET, diffusion- and perfusion-weighted MRI, or MR spectroscopy are particularly helpful for the evaluation of treatment response and provide valuable information for the differentiation of treatment-induced changes from actual brain tumor progression following various immunotherapy approaches.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Jülich, Germany.,Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Jülich, Germany
| | - Jan-Michael Werner
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Garry Ceccon
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Jülich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Jülich, Germany.,Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
35
|
Langen KJ, Heinzel A, Lohmann P, Mottaghy FM, Galldiks N. Advantages and limitations of amino acid PET for tracking therapy response in glioma patients. Expert Rev Neurother 2019; 20:137-146. [DOI: 10.1080/14737175.2020.1704256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Juelich, Juelich, Germany
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany
- Section JARA-Brain, Juelich-Aachen Research Alliance (JARA), Juelich-Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Alexander Heinzel
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany
- Section JARA-Brain, Juelich-Aachen Research Alliance (JARA), Juelich-Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Juelich, Juelich, Germany
| | - Felix M. Mottaghy
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany
- Section JARA-Brain, Juelich-Aachen Research Alliance (JARA), Juelich-Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
- Centre of Integrated Oncology (CIO), Universities of Aachen, Düsseldorf, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Juelich, Juelich, Germany
- Department of Neurology1, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Centre of Integrated Oncology (CIO), Universities of Aachen, Düsseldorf, Germany
| |
Collapse
|
36
|
Metabolic Tumor Volume Response Assessment Using (11)C-Methionine Positron Emission Tomography Identifies Glioblastoma Tumor Subregions That Predict Progression Better Than Baseline or Anatomic Magnetic Resonance Imaging Alone. Adv Radiat Oncol 2019; 5:53-61. [PMID: 32051890 PMCID: PMC7004943 DOI: 10.1016/j.adro.2019.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/05/2019] [Indexed: 02/08/2023] Open
Abstract
Purpose To evaluate whether response assessment of newly diagnosed glioblastoma at 3 months using 11C-methionine-positron emission tomography (MET-PET) is better associated with patient outcome compared with baseline MET-PET or anatomic magnetic resonance imaging alone. Methods and Materials Patients included were participants in a phase I/II trial of dose-escalated chemoradiation based on anatomic magnetic resonance imaging. Automated segmentation of metabolic tumor volume (MTV) was performed at a threshold of 1.5 times mean cerebellar uptake. Progression-free (PFS) and overall survival were estimated with the Kaplan-Meier method and compared with log-rank tests. Multivariate analysis for PFS and overall survival was performed using Cox proportional hazards, and spatial overlap between imaging and recurrence volumes were analyzed. Results Among 37 patients, 15 had gross total resection, of whom 10 (67%) had residual MTV, 16 subtotal resection, and 6 biopsy alone. Median radiation therapy dose was 75 Gy (range, 66-81). Median baseline T1 Gd-enhanced tumor volume (GTV-Gd) was 38.0 cm3 (range, 8.0-81.5). Median pre-CRT MTV was 4.9 cm3 (range, 0-43.8). Among 25 patients with 3-month MET-PET, MTV was only 2.4 cm3 (range, 0.004-18.0) in patients with uptake. Patients with MTV = 0 cm3 at 3 months had superior PFS (18.2 vs 10.1 months, P = .03). On multivariate analysis, larger 3-month MTV (hazard ratio [HR] 2.4, 95% confidence interval [CI], 1.4-4.3, P = .03), persistent MET-PET subvolume (overlap of pre-CRT and 3 month MTV; HR 2.0, 95% CI, 1.2-3.4, P = .06), and increase in MTV (HR 1.8, 95% CI, 1.1-3.1, P = .09) were the only imaging factors significant for worse PFS. GTV-Gd at recurrence encompassed 97% of the persistent MET-PET subvolume (interquartile range 72%-100%), versus 71% (interquartile range 39%-93%) of baseline MTV, 54% of baseline GTV-Gd (18%-87%), and 78% of 3-month MTV (47%-95%). Conclusions The majority of patients with apparent gross total resection of glioblastoma have measurable postoperative MTV. Total and persisting MTV 3 months post-CRT were significant predictors of PFS, and persistent MET-PET subvolume was the strongest predictor for localizing tumor recurrence.
Collapse
|
37
|
Chen C, Ou X, Wang J, Guo W, Ma X. Radiomics-Based Machine Learning in Differentiation Between Glioblastoma and Metastatic Brain Tumors. Front Oncol 2019; 9:806. [PMID: 31508366 PMCID: PMC6714109 DOI: 10.3389/fonc.2019.00806] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/07/2019] [Indexed: 02/05/2023] Open
Abstract
Purpose: To investigative the diagnostic performance of radiomics-based machine learning in differentiating glioblastomas (GBM) from metastatic brain tumors (MBTs). Method: The current study involved 134 patients diagnosed and treated in our institution between April 2014 and December 2018. Radiomics features were extracted from contrast-enhanced T1 weighted imaging (T1C). Thirty diagnostic models were built based on five selection methods and six classification algorithms. The sensitivity, specificity, accuracy, and area under curve (AUC) of each model were calculated, and based on these the optimal model was chosen. Result : Two models represented promising diagnostic performance with AUC of 0.80. The first model was a combination of Distance Correlation as the selection method and Linear Discriminant Analysis (LDA) as the classification algorithm. In the training group, the sensitivity, specificity, accuracy, and AUC were 0.75, 0.85, 0.80, and 0.80, respectively; and in the testing group, the sensitivity, specificity, accuracy, and AUC of the model were 0.69, 0.86, 0.78, and 0.80, respectively. The second model was the Distance Correlation as the selection method and logistic regression (LR) as the classification algorithm, with sensitivity, specificity, accuracy, and AUC of 0.75, 0.85, 0.80, 0.80 in the training group and 0.69, 0.86, 0.78, 0.80 in the testing group. Conclusion: Radiomic-based machine learning has potential to be utilized in differentiating GBM from MBTs.
Collapse
Affiliation(s)
- Chaoyue Chen
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xuejin Ou
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China.,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Wang
- School of Computer Science, Nanjing University of Science and Technology, Nanjing, China
| | - Wen Guo
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
38
|
Tabatabai G, Wakimoto H. Glioblastoma: State of the Art and Future Perspectives. Cancers (Basel) 2019; 11:cancers11081091. [PMID: 31370300 PMCID: PMC6721299 DOI: 10.3390/cancers11081091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Accepted: 01/01/1970] [Indexed: 12/19/2022] Open
Affiliation(s)
- Ghazaleh Tabatabai
- Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for Neuro-Oncology, Comprehensive Cancer Center Tübingen Stuttgart, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School Boston, Boston, MA 02114, USA.
| |
Collapse
|
39
|
Werner JM, Stoffels G, Lichtenstein T, Borggrefe J, Lohmann P, Ceccon G, Shah NJ, Fink GR, Langen KJ, Kabbasch C, Galldiks N. Differentiation of treatment-related changes from tumour progression: a direct comparison between dynamic FET PET and ADC values obtained from DWI MRI. Eur J Nucl Med Mol Imaging 2019; 46:1889-1901. [PMID: 31203420 DOI: 10.1007/s00259-019-04384-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/30/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Following brain cancer treatment, the capacity of anatomical MRI to differentiate neoplastic tissue from treatment-related changes (e.g., pseudoprogression) is limited. This study compared apparent diffusion coefficients (ADC) obtained by diffusion-weighted MRI (DWI) with static and dynamic parameters of O-(2-[18F]fluoroethyl)-L-tyrosine (FET) PET for the differentiation of treatment-related changes from tumour progression. PATIENTS AND METHODS Forty-eight pretreated high-grade glioma patients with anatomical MRI findings suspicious for progression (median time elapsed since last treatment was 16 weeks) were investigated using DWI and dynamic FET PET. Maximum and mean tumour-to-brain ratios (TBRmax, TBRmean) as well as dynamic parameters (time-to-peak and slope values) of FET uptake were calculated. For mean ADC calculation, regions-of-interest analyses were performed on ADC maps calculated from DWI coregistered with the contrast-enhanced MR image. Diagnoses were confirmed neuropathologically (21%) or clinicoradiologically. Diagnostic performance was evaluated using receiver-operating-characteristic analyses or Fisher's exact test for a combinational approach. RESULTS Ten of 48 patients had treatment-related changes (21%). The diagnostic performance of FET PET was significantly higher (threshold for both TBRmax and TBRmean, 1.95; accuracy, 83%; AUC, 0.89 ± 0.05; P < 0.001) than that of ADC values (threshold ADC, 1.09 × 10-3 mm2/s; accuracy, 69%; AUC, 0.73 ± 0.09; P = 0.13). The addition of static FET PET parameters to ADC values increased the latter's accuracy to 89%. The highest accuracy was achieved by combining static and dynamic FET PET parameters (93%). Moreover, in contrast to ADC values, TBRs <1.95 at suspected progression predicted a significantly longer survival (P = 0.01). CONCLUSIONS Data suggest that static and dynamic FET PET provide valuable information concerning the differentiation of early treatment-related changes from tumour progression and outperform ADC measurement for this highly relevant clinical question.
Collapse
Affiliation(s)
- Jan-Michael Werner
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Thorsten Lichtenstein
- Department of Neuroradiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jan Borggrefe
- Department of Neuroradiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Garry Ceccon
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
- Department of Neurology, University Hospital Aachen, Aachen, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| | - Christoph Kabbasch
- Department of Neuroradiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Düsseldorf, Cologne, Germany.
- Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany.
- Department of Neurology, University Hospital Cologne, Kerpener St. 62, 50937, Cologne, Germany.
| |
Collapse
|
40
|
Galldiks N, Lohmann P, Albert NL, Tonn JC, Langen KJ. Current status of PET imaging in neuro-oncology. Neurooncol Adv 2019; 1:vdz010. [PMID: 32642650 PMCID: PMC7324052 DOI: 10.1093/noajnl/vdz010] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Over the past decades, a variety of PET tracers have been used for the evaluation of patients with brain tumors. For clinical routine, the most important clinical indications for PET imaging in patients with brain tumors are the identification of neoplastic tissue including the delineation of tumor extent for the further diagnostic and therapeutic management (ie, biopsy, resection, or radiotherapy planning), the assessment of response to a certain anticancer therapy including its (predictive) effect on the patients’ outcome and the differentiation of treatment-related changes (eg, pseudoprogression and radiation necrosis) from tumor progression at follow-up. To serve medical professionals of all disciplines involved in the diagnosis and care of patients with brain tumors, this review summarizes the value of PET imaging for the latter-mentioned 3 clinically relevant indications in patients with glioma, meningioma, and brain metastases.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig Maximilians-University of Munich, Munich, Germany
| | - Jörg C Tonn
- Department of Neurosurgery, Ludwig Maximilians-University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Germany
| | - Karl-Josef Langen
- Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| |
Collapse
|