1
|
Robert JA, Leclerc A, Ducloie M, Emery E, Agostini D, Vigne J. Contribution of [ 18F]FET PET in the Management of Gliomas, from Diagnosis to Follow-Up: A Review. Pharmaceuticals (Basel) 2024; 17:1228. [PMID: 39338390 PMCID: PMC11435125 DOI: 10.3390/ph17091228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/14/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Gliomas, the most common type of primary malignant brain tumors in adults, pose significant challenges in diagnosis and management due to their heterogeneity and potential aggressiveness. This review evaluates the utility of O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) positron emission tomography (PET), a promising imaging modality, to enhance the clinical management of gliomas. We reviewed 82 studies involving 4657 patients, focusing on the application of [18F]FET in several key areas: diagnosis, grading, identification of IDH status and presence of oligodendroglial component, guided resection or biopsy, detection of residual tumor, guided radiotherapy, detection of malignant transformation in low-grade glioma, differentiation of recurrence versus treatment-related changes and prognostic factors, and treatment response evaluation. Our findings confirm that [18F]FET helps delineate tumor tissue, improves diagnostic accuracy, and aids in therapeutic decision-making by providing crucial insights into tumor metabolism. This review underscores the need for standardized parameters and further multicentric studies to solidify the role of [18F]FET PET in routine clinical practice. By offering a comprehensive overview of current research and practical implications, this paper highlights the added value of [18F]FET PET in improving management of glioma patients from diagnosis to follow-up.
Collapse
Affiliation(s)
- Jade Apolline Robert
- CHU de Caen Normandie, UNICAEN, Department of Nuclear Medicine, Normandie Université, 14000 Caen, France; (J.A.R.)
| | - Arthur Leclerc
- Department of Neurosurgery, Caen University Hospital, 14000 Caen, France
- Caen Normandie University, ISTCT UMR6030, GIP Cyceron, 14000 Caen, France
| | - Mathilde Ducloie
- Department of Neurology, Caen University Hospital, 14000 Caen, France
- Centre François Baclesse, Department of Neurology, 14000 Caen, France
| | - Evelyne Emery
- Department of Neurosurgery, Caen University Hospital, 14000 Caen, France
| | - Denis Agostini
- CHU de Caen Normandie, UNICAEN, Department of Nuclear Medicine, Normandie Université, 14000 Caen, France; (J.A.R.)
| | - Jonathan Vigne
- CHU de Caen Normandie, UNICAEN, Department of Nuclear Medicine, Normandie Université, 14000 Caen, France; (J.A.R.)
- CHU de Caen Normandie, UNICAEN Department of Pharmacy, Normandie Université, 14000 Caen, France
- Centre Cyceron, Institut Blood and Brain @ Caen-Normandie, Normandie Université, UNICAEN, INSERM U1237, PhIND, 14000 Caen, France
| |
Collapse
|
2
|
Xiong M, Chen Z, Zhou C, Yang X, Hu W, Jiang Y, Zheng R, Fan W, Mou Y, Lin X. PSMA PET/MR is a New Imaging Option for Identifying Glioma Recurrence and Predicting Prognosis. Recent Pat Anticancer Drug Discov 2024; 19:383-395. [PMID: 38214322 DOI: 10.2174/1574892818666230519150401] [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: 03/04/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Glioma is characterized by a high recurrence rate, while the results of the traditional imaging methods (including magnetic resonance imaging, MRI) to distinguish recurrence from treatment-related changes (TRCs) are poor. Prostate-specific membrane antigen (PSMA) (US10815200B2, Deutsches Krebsforschungszentrum, German Cancer Research Center) is a type II transmembrane glycoprotein overexpressed in glioma vascular endothelium, and it is a promising target for imaging and therapy. OBJECTIVE The study aimed to assess the performance of PSMA positron emission tomography/ magnetic resonance (PET/MR) for diagnosing recurrence and predicting prognosis in glioma patients. MATERIALS AND METHODS Patients suspected of glioma recurrence who underwent 18F-PSMA-1007 PET/MR were prospectively enrolled. Eight metabolic parameters and fifteen texture features of the lesion were extracted from PSMA PET/MR. The ability of PSMA PET/MR to diagnose glioma recurrence was investigated and compared with conventional MRI. The diagnostic agreement was assessed using Cohen κ scores and the predictive parameters of PSMA PET/MR were obtained. Kaplan-Meier method and Cox proportional hazard model were used to analyze recurrence- free survival (RFS) and overall survival (OS). Finally, the expression of PSMA was analyzed by immunohistochemistry (IHC). RESULTS Nineteen patients with a mean age of 48.11±15.72 were assessed. The maximum tumorto- parotid ratio (TPRmax) and texture features extracted from PET and T1-weighted contrast enhancement (T1-CE) MR showed differences between recurrence and TRCs (all p <0.05). PSMA PET/MR and conventional MRI exhibited comparable power in diagnosing recurrence with specificity and PPV of 100%. The interobserver concordance was fair between the two modalities (κ = 0.542, p = 0.072). The optimal cutoffs of metabolic parameters, including standardized uptake value (SUV, SUVmax, SUVmean, and SUVpeak) and TPRmax for predicting recurrence were 3.35, 1.73, 1.99, and 0.17 respectively, with the area under the curve (AUC) ranging from 0.767 to 0.817 (all p <0.05). In grade 4 glioblastoma (GBM) patients, SUVmax, SUVmean, SUVpeak, TBRmax, TBRmean, and TPRmax showed improved performance of AUC (0.833-0.867, p <0.05). Patients with SUVmax, SUVmean, or SUVpeak more than the cutoff value had significantly shorter RFS (all p <0.05). In addition, patients with SUVmean, SUVpeak, or TPRmax more than the cutoff value had significantly shorter OS (all p <0.05). PSMA expression of glioma vascular endothelium was observed in ten (10/11, 90.9%) patients with moderate-to-high levels in all GBM cases (n = 6/6, 100%). CONCLUSION This primitive study shows multiparameter PSMA PET/MR to be useful in identifying glioma (especially GBM) recurrence by providing excellent tumor background comparison, tumor heterogeneity, recurrence prediction and prognosis information, although it did not improve the diagnostic performance compared to conventional MRI. Further and larger studies are required to define its potential clinical application in this setting.
Collapse
Affiliation(s)
- Min Xiong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhenghe Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chao Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaochun Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wanming Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yongluo Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rongliang Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei Fan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yonggao Mou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoping Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
3
|
Teske N, Teske NC, Niyazi M, Belka C, Thon N, Tonn JC, Forbrig R, Karschnia P. Frequency and Prognostic Relevance of Volumetric MRI Changes in Contrast- and Non-Contrast-Enhancing Tumor Compartments between Surgery and Radiotherapy of IDHwt Glioblastoma. Cancers (Basel) 2023; 15:cancers15061745. [PMID: 36980633 PMCID: PMC10046652 DOI: 10.3390/cancers15061745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/23/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
In newly diagnosed IDH-wildtype glioblastoma, the frequency and prognostic relevance of tumor regrowth between resection and the initiation of adjuvant radiochemotherapy are unclear. In this retrospective single-center study we included 64 consecutive cases, for whom magnetic resonance imaging (MRI) was available for both the volumetric assessment of the extent of resection immediately after surgery as well as the volumetric target delineation before the initiation of adjuvant radiochemotherapy (time interval: 15.5 ± 1.9 days). Overall, a median new contrast-enhancement volume was seen in 21/64 individuals (33%, 1.5 ± 1.5 cm3), and new non-contrast lesion volume in 18/64 patients (28%, 5.0 ± 2.3 cm3). A multidisciplinary in-depth review revealed that new contrast-enhancement was either due to (I) the progression of contrast-enhancing tumor remnants in 6/21 patients or (II) distant contrast-enhancing foci or breakdown of the blood–brain barrier in previously non-contrast-enhancing tumor remnants in 5/21 patients, whereas it was unspecific or due to ischemia in 10/21 patients. For non-contrast-enhancing lesions, three of eighteen had progression of non-contrast-enhancing tumor remnants and fifteen of eighteen had unspecific changes or changes due to ischemia. There was no significant association between findings consistent with tumor regrowth and a less favorable outcome (overall survival: 14 vs. 19 months; p = 0.423). These findings support the rationale that analysis of the postsurgical remaining tumor-volume for prognostic stratification should be carried out on immediate postoperative MRI (<72 h), as unspecific changes are common. However, tumor regrowth including distant foci may occur in a subset of IDH-wildtype glioblastoma patients diagnosed per WHO 2021 classification. Thus, MRI imaging prior to radiotherapy should be obtained to adjust radiotherapy planning accordingly.
Collapse
Affiliation(s)
- Nico Teske
- Department of Neurosurgery, Munich University Hospital, LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
- Correspondence: (N.T.); (P.K.); Tel.: +49-(0)89-4400-711361 (N.T.); +49-(0)89-4400-711365 (P.K.); Fax: +49-(0)89-4400-72592 (N.T. & P.K.)
| | - Nina C. Teske
- Department of Neurosurgery, Munich University Hospital, LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Maximilian Niyazi
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
- Department of Radiation Oncology, Munich University Hospital, LMU Munich, 81377 Munich, Germany
- Bavarian Center for Cancer Research (BZKF), 91054 Erlangen, Germany
| | - Claus Belka
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
- Department of Radiation Oncology, Munich University Hospital, LMU Munich, 81377 Munich, Germany
- Bavarian Center for Cancer Research (BZKF), 91054 Erlangen, Germany
| | - Niklas Thon
- Department of Neurosurgery, Munich University Hospital, LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Joerg-Christian Tonn
- Department of Neurosurgery, Munich University Hospital, LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Robert Forbrig
- Institute of Neuroradiology, Munich University Hospital, LMU Munich, 81377 Munich, Germany
| | - Philipp Karschnia
- Department of Neurosurgery, Munich University Hospital, LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
- Correspondence: (N.T.); (P.K.); Tel.: +49-(0)89-4400-711361 (N.T.); +49-(0)89-4400-711365 (P.K.); Fax: +49-(0)89-4400-72592 (N.T. & P.K.)
| |
Collapse
|
4
|
Soni N, Ora M, Jena A, Rana P, Mangla R, Ellika S, Almast J, Puri S, Meyers SP. Amino Acid Tracer PET MRI in Glioma Management: What a Neuroradiologist Needs to Know. AJNR Am J Neuroradiol 2023; 44:236-246. [PMID: 36657945 PMCID: PMC10187808 DOI: 10.3174/ajnr.a7762] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/21/2022] [Indexed: 01/21/2023]
Abstract
PET with amino acid tracers provides additional insight beyond MR imaging into the biology of gliomas that can be used for initial diagnosis, delineation of tumor margins, planning of surgical and radiation therapy, assessment of residual tumor, and evaluation of posttreatment response. Hybrid PET MR imaging allows the simultaneous acquisition of various PET and MR imaging parameters in a single investigation with reduced scanning time and improved anatomic localization. This review aimed to provide neuroradiologists with a concise overview of the various amino acid tracers and a practical understanding of the clinical applications of amino acid PET MR imaging in glioma management. Future perspectives in newer advances, novel radiotracers, radiomics, and cost-effectiveness are also outlined.
Collapse
Affiliation(s)
- N Soni
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - M Ora
- Sanjay Gandhi Postgraduate Institute of Medical Sciences (M.O.), Lucknow, Uttar Pradesh, India
| | - A Jena
- Indraprastha Apollo Hospital (A.J., P.R.), New Delhi, India
| | - P Rana
- Indraprastha Apollo Hospital (A.J., P.R.), New Delhi, India
| | - R Mangla
- Upstate University Hospital (R.M.), Syracuse, New York
| | - S Ellika
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - J Almast
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - S Puri
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - S P Meyers
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| |
Collapse
|
5
|
Tosic L, Thoma M, Voglis S, Hofer AS, Bektas D, Pangalu A, Regli L, Germans MR. Evaluation of patient STress level caused by radiological Investigations in early Postoperative phase After CRANIOtomy (IPAST-CRANIO): protocol of a Swiss prospective cohort study. BMJ Open 2022; 12:e061452. [PMID: 36130762 PMCID: PMC9494566 DOI: 10.1136/bmjopen-2022-061452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Postoperative imaging after neurosurgical interventions is usually performed in the first 72 hours after surgery to provide an accurate assessment of postoperative resection status. Patient frequently report that early postoperative examination after craniotomy for tumour and vascular procedures is associated with distress, exertion, nausea and pain. Delayed postoperative imaging (between 36 and 72 hours postoperatively) may have an advantage regarding psychological and physical stress compared with early imaging. The goal of this study is to evaluate and determine the optimal time frame for postoperative imaging with MRI and CT in terms of medical and neuroradiological implications and patient's subjective stress level. METHODS AND ANALYSIS Data will be prospectively collected from all patients aged 18-80 years who receive postoperative MRI or CT imaging following a craniotomy for resection of a cerebral tumour (benign and malignant) or vascular surgery. Participants have to complete questionnaires containing visual analogue scores (VAS) for headache and nausea, Body Part Discomfort score and a single question addressing subjective preference of timing of postoperative imaging after craniotomy. The primary endpoint of the study is the difference in subjective stress due to imaging studies after craniotomy, measured just before and after postoperative MRI or CT with the above-mentioned instruments. Subjective stress is defined as a combination of the scores VAS pain, VAS nausea and 0.5* Body Part Discomfort core.This study determines whether proper timing of postoperative imaging can improve patient satisfaction and reduce pain, stress and discomfort caused by postoperative imaging. Factors causing additional postoperative stress are likely responsible for delayed recovery of neurosurgical patients. ETHICS AND DISSEMINATION The institutional review board (Kantonale Ethikkommission Zürich) approved this study on 4 August 2020 under case number BASEC 2020-01590. The authors are planning to publish the data of this study in a peer-reviewed paper. After database closure, the data will be exported to the local data repository (Zurich Open Repository and Archive) of the University of Zurich. The sponsor (LR) and the project leader (MR.G) will make the final decision on the publication of the results. The data that support the findings of this study are available on request from the corresponding author LT. The data are not publicly available due to privacy/ethical restrictions. TRIAL REGISTRATION NUMBER NCT05112575; ClinicalTrials.gov.
Collapse
Affiliation(s)
- Lazar Tosic
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Marco Thoma
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Stefanos Voglis
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Anna Sophie Hofer
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Delal Bektas
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Athina Pangalu
- Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Menno R Germans
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
García Vicente AM, Pena Pardo FJ, Amo-Salas M, Villena Martín M, López Menéndez C, Soriano Castrejón ÁM, Pérez-Beteta J. Prognostic Potential of Postoperative 18F-Fluorocholine PET/CT in Patients With High-Grade Glioma. Clinical Validation of FuMeGA Postoperative PET Criteria. Clin Nucl Med 2022; 47:480-487. [PMID: 35426853 DOI: 10.1097/rlu.0000000000004127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of this study was to assess the prognostic performance of postoperative 18F-fluorocholine PET/CT in patients with high-grade glioma (HGG). METHODS Patients with HGG who underwent preoperative and postoperative 18F-fluorocholine PET/CT were prospectively enrolled in the study. Postoperative MRI was classified as complete versus incomplete resection. Postoperative 18F-fluorocholine PET/CT was classified as negative (complete) or positive for metabolic residual tumor (incomplete resection) using a 5-point score system. The correlation of positive locations on PET/CT with the sites of subsequent tumor recurrence was evaluated. The concordance of postoperative imaging techniques (Cohen κ) and their relation with progression-free survival and overall survival were assessed using Kaplan-Meier method and Cox regression analysis. RESULTS Fifty-one studies, belonging to 47 patients, were assessed. Four patients underwent 2 postoperative 18F-fluorocholine PET/CT scans as they needed a second tumor resection for recurrence. In the follow-up, 42 patients progressed, and 37 died. Concordance between postoperative PET/CT and MRI assessment was poor. Resection grade on MRI did not show any significant association with prognosis. In multivariate analysis, only age and postoperative PET/CT showed significant association with progression-free survival (hazard ratio [HR], 1.03 [1.01-1.06, P = 0.006] and 1.88 [0.96-3.71, P = 0.067], respectively) and overall survival (HR, 1.04 [1.01-1.07, P = 0.004] and 2.63 [1.22-5.68, P = 0.014], respectively). Postoperative positive 18F-fluorocholine PET/CT locations correlated with the sites of subsequent tumor recurrence in 81.82% of cases. CONCLUSION Postoperative 18F-fluorocholine PET/CT seems superior to postoperative MRI in the outcome prediction of patients with HGG, outperforming it in the identification of the most probable location of tumor recurrence.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Julián Pérez-Beteta
- Mathematical Oncology Laboratory (MôLAB), Castilla-La Mancha University, Ciudad Real, Spain
| |
Collapse
|
7
|
Beinat C, Patel CB, Haywood T, Murty S, Naya L, Castillo JB, Reyes ST, Phillips M, Buccino P, Shen B, Park JH, Koran MEI, Alam IS, James ML, Holley D, Halbert K, Gandhi H, He JQ, Granucci M, Johnson E, Liu DD, Uchida N, Sinha R, Chu P, Born DE, Warnock GI, Weissman I, Hayden-Gephart M, Khalighi M, Massoud TF, Iagaru A, Davidzon G, Thomas R, Nagpal S, Recht LD, Gambhir SS. A Clinical PET Imaging Tracer ([ 18F]DASA-23) to Monitor Pyruvate Kinase M2-Induced Glycolytic Reprogramming in Glioblastoma. Clin Cancer Res 2021; 27:6467-6478. [PMID: 34475101 PMCID: PMC8639752 DOI: 10.1158/1078-0432.ccr-21-0544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/15/2021] [Accepted: 08/30/2021] [Indexed: 01/10/2023]
Abstract
PURPOSE Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, a key process of cancer metabolism. PKM2 is preferentially expressed by glioblastoma (GBM) cells with minimal expression in healthy brain. We describe the development, validation, and translation of a novel PET tracer to study PKM2 in GBM. We evaluated 1-((2-fluoro-6-[18F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA-23) in cell culture, mouse models of GBM, healthy human volunteers, and patients with GBM. EXPERIMENTAL DESIGN [18F]DASA-23 was synthesized with a molar activity of 100.47 ± 29.58 GBq/μmol and radiochemical purity >95%. We performed initial testing of [18F]DASA-23 in GBM cell culture and human GBM xenografts implanted orthotopically into mice. Next, we produced [18F]DASA-23 under FDA oversight, and evaluated it in healthy volunteers and a pilot cohort of patients with glioma. RESULTS In mouse imaging studies, [18F]DASA-23 clearly delineated the U87 GBM from surrounding healthy brain tissue and had a tumor-to-brain ratio of 3.6 ± 0.5. In human volunteers, [18F]DASA-23 crossed the intact blood-brain barrier and was rapidly cleared. In patients with GBM, [18F]DASA-23 successfully outlined tumors visible on contrast-enhanced MRI. The uptake of [18F]DASA-23 was markedly elevated in GBMs compared with normal brain, and it identified a metabolic nonresponder within 1 week of treatment initiation. CONCLUSIONS We developed and translated [18F]DASA-23 as a new tracer that demonstrated the visualization of aberrantly expressed PKM2 for the first time in human subjects. These results warrant further clinical evaluation of [18F]DASA-23 to assess its utility for imaging therapy-induced normalization of aberrant cancer metabolism.
Collapse
Affiliation(s)
- Corinne Beinat
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California.
| | - Chirag B Patel
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Tom Haywood
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Surya Murty
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Lewis Naya
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Jessa B Castillo
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Samantha T Reyes
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Megan Phillips
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Pablo Buccino
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Bin Shen
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Jun Hyung Park
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Mary Ellen I Koran
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Israt S Alam
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Michelle L James
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Dawn Holley
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Kim Halbert
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Harsh Gandhi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Joy Q He
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Monica Granucci
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Eli Johnson
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Daniel Dan Liu
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Nobuko Uchida
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Rahul Sinha
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Pauline Chu
- Stanford Human Research Histology Core, Stanford University School of Medicine, Stanford, California
| | - Donald E Born
- Department of Pathology, Neuropathology, Stanford University School of Medicine, Stanford, California
| | | | - Irving Weissman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Melanie Hayden-Gephart
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Mehdi Khalighi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Tarik F Massoud
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
- Division of Neuroimaging and Neurointervention, Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Guido Davidzon
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Reena Thomas
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Seema Nagpal
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Lawrence D Recht
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California.
| | - Sanjiv Sam Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
- Departments of Bioengineering and Materials Science & Engineering, Stanford University, Stanford, California
| |
Collapse
|
8
|
Ort J, Hamou HA, Kernbach JM, Hakvoort K, Blume C, Lohmann P, Galldiks N, Heiland DH, Mottaghy FM, Clusmann H, Neuloh G, Langen KJ, Delev D. 18F-FET-PET-guided gross total resection improves overall survival in patients with WHO grade III/IV glioma: moving towards a multimodal imaging-guided resection. J Neurooncol 2021; 155:71-80. [PMID: 34599479 PMCID: PMC8545732 DOI: 10.1007/s11060-021-03844-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/08/2021] [Indexed: 11/15/2022]
Abstract
Purpose PET using radiolabeled amino acid [18F]-fluoro-ethyl-L-tyrosine (FET-PET) is a well-established imaging modality for glioma diagnostics. The biological tumor volume (BTV) as depicted by FET-PET often differs in volume and location from tumor volume of contrast enhancement (CE) in MRI. Our aim was to investigate whether a gross total resection of BTVs defined as < 1 cm3 of residual BTV (PET GTR) correlates with better oncological outcome. Methods We retrospectively analyzed imaging and survival data from patients with primary and recurrent WHO grade III or IV gliomas who underwent FET-PET before surgical resection. Tumor overlap between FET-PET and CE was evaluated. Completeness of FET-PET resection (PET GTR) was calculated after superimposition and semi-automated segmentation of pre-operative FET-PET and postoperative MRI imaging. Survival analysis was performed using the Kaplan–Meier method and the log-rank test. Results From 30 included patients, PET GTR was achieved in 20 patients. Patients with PET GTR showed improved median OS with 19.3 compared to 13.7 months for patients with residual FET uptake (p = 0.007; HR 0.3; 95% CI 0.12–0.76). This finding remained as independent prognostic factor after performing multivariate analysis (HR 0.19, 95% CI 0.06–0.62, p = 0.006). Other survival influencing factors such as age, IDH-mutation, MGMT promotor status, and adjuvant treatment modalities were equally distributed between both groups. Conclusion Our results suggest that PET GTR improves the OS in patients with WHO grade III or IV gliomas. A multimodal imaging approach including FET-PET for surgical planning in newly diagnosed and recurrent tumors may improve the oncological outcome in glioma patients. Supplementary Information The online version contains supplementary material available at 10.1007/s11060-021-03844-1.
Collapse
Affiliation(s)
- Jonas Ort
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany. .,NAILA-Neurosurgical Artificial Intelligence Laboratory Aachen, Aachen, Germany. .,Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany.
| | - Hussam Aldin Hamou
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Julius M Kernbach
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.,NAILA-Neurosurgical Artificial Intelligence Laboratory Aachen, Aachen, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Karlijn Hakvoort
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.,NAILA-Neurosurgical Artificial Intelligence Laboratory Aachen, Aachen, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Christian Blume
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany.,Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany.,Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Dieter Henrik Heiland
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, Freiburg University, Freiburg, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.,JARA-Juelich Aachen Research Alliance, Juelich, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Hans Clusmann
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Georg Neuloh
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.,JARA-Juelich Aachen Research Alliance, Juelich, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Daniel Delev
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.,NAILA-Neurosurgical Artificial Intelligence Laboratory Aachen, Aachen, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| |
Collapse
|
9
|
Advanced Imaging and Computational Techniques for the Diagnostic and Prognostic Assessment of Malignant Gliomas. Cancer J 2021; 27:344-352. [PMID: 34570448 DOI: 10.1097/ppo.0000000000000545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ABSTRACT Advanced imaging techniques provide a powerful tool to assess the intratumoral and intertumoral heterogeneity of gliomas. Advances in the molecular understanding of glioma subgroups may allow improved diagnostic assessment combining imaging and molecular tumor features, with enhanced prognostic utility and implications for patient treatment. In this article, a comprehensive overview of the physiologic basis for conventional and advanced imaging techniques is presented, and clinical applications before and after treatment are discussed. An introduction to the principles of radiomics and the advanced integration of imaging, clinical outcomes, and genomic data highlights the future potential for this field of research to better stratify and select patients for standard as well as investigational therapies.
Collapse
|
10
|
Tatkovic A, McBean R, Perkins E, Wong D. 18 F-FET PET maximum standard uptake value and WHO tumour classification grade in glioma. J Med Imaging Radiat Oncol 2021; 66:332-336. [PMID: 34510758 DOI: 10.1111/1754-9485.13322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/09/2021] [Accepted: 08/22/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND In the area of oncology, molecular imaging techniques are becoming increasingly utilised. In neuro-oncology imaging, 18 Fluoro-O-(2) fluoroethyl-L-tyrosine (18 F-FET) is one of the molecular tracers used in positron-emission tomography (PET). Here, we investigated the correlation between maximum standard uptake value (SUV) of 18 F-FET PET and histologically determined World Health Organization (WHO) grade in glioma. PATIENTS AND METHODS This was a retrospective review of all 18 F-FET PET studies conducted between August 2014 and August 2019. Review was conducted to identify imaging studies performed on patients who had a glioma with histopathology results from surgical resection or biopsy available. RESULTS A total of 31 18 F-FET PET studies of histologically confirmed glioma were included. WHO grades ranged from II-IV. A positive correlation between maximum SUV uptake on 18 F-FET PET and WHO grade was observed. CONCLUSIONS There was a correlation identified between WHO glioma grade and maximum SUV on 18 F-FET PET. Further studies are recommended to explore this relationship.
Collapse
Affiliation(s)
- Annaleis Tatkovic
- I-MED Radiology, The Wesley Hospital, Auchenflower, Queensland, Australia
| | - Rhiannon McBean
- I-MED Radiology, The Wesley Hospital, Auchenflower, Queensland, Australia
| | - Ebony Perkins
- I-MED Radiology, The Wesley Hospital, Auchenflower, Queensland, Australia
| | - David Wong
- I-MED Radiology, The Wesley Hospital, Auchenflower, Queensland, Australia
| |
Collapse
|
11
|
Sadaghiani MS, Sheikhbahaei S, Rowe SP, Pomper MG, Solnes LB. Cellular and Molecular Imaging with SPECT and PET in Brain Tumors. Radiol Clin North Am 2021; 59:363-375. [PMID: 33926683 DOI: 10.1016/j.rcl.2021.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This review highlights the 2 major molecular imaging modalities that are used in clinics, namely single-photon emission computed tomography (SPECT) and positron emission tomography (PET), and their added value in management of patients with brain tumors. There are a variety of SPECT and PET radiotracers that can allow imaging of different molecular processes. Those radiotracers target specific molecular features of tumors, resulting in improved specificity of these agents. Potential applications include staging of brain tumors and evaluating post-therapeutic changes.
Collapse
Affiliation(s)
- Mohammad S Sadaghiani
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3150, Baltimore, MD 21287, USA
| | - Sara Sheikhbahaei
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3150, Baltimore, MD 21287, USA
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3150, Baltimore, MD 21287, USA
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3150, Baltimore, MD 21287, USA
| | - Lilja B Solnes
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3150, Baltimore, MD 21287, USA.
| |
Collapse
|
12
|
Booth TC, Thompson G, Bulbeck H, Boele F, Buckley C, Cardoso J, Dos Santos Canas L, Jenkinson D, Ashkan K, Kreindler J, Huskens N, Luis A, McBain C, Mills SJ, Modat M, Morley N, Murphy C, Ourselin S, Pennington M, Powell J, Summers D, Waldman AD, Watts C, Williams M, Grant R, Jenkinson MD. A Position Statement on the Utility of Interval Imaging in Standard of Care Brain Tumour Management: Defining the Evidence Gap and Opportunities for Future Research. Front Oncol 2021; 11:620070. [PMID: 33634034 PMCID: PMC7900557 DOI: 10.3389/fonc.2021.620070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIV E To summarise current evidence for the utility of interval imaging in monitoring disease in adult brain tumours, and to develop a position for future evidence gathering while incorporating the application of data science and health economics. METHODS Experts in 'interval imaging' (imaging at pre-planned time-points to assess tumour status); data science; health economics, trial management of adult brain tumours, and patient representatives convened in London, UK. The current evidence on the use of interval imaging for monitoring brain tumours was reviewed. To improve the evidence that interval imaging has a role in disease management, we discussed specific themes of data science, health economics, statistical considerations, patient and carer perspectives, and multi-centre study design. Suggestions for future studies aimed at filling knowledge gaps were discussed. RESULTS Meningioma and glioma were identified as priorities for interval imaging utility analysis. The "monitoring biomarkers" most commonly used in adult brain tumour patients were standard structural MRI features. Interval imaging was commonly scheduled to provide reported imaging prior to planned, regular clinic visits. There is limited evidence relating interval imaging in the absence of clinical deterioration to management change that alters morbidity, mortality, quality of life, or resource use. Progression-free survival is confounded as an outcome measure when using structural MRI in glioma. Uncertainty from imaging causes distress for some patients and their caregivers, while for others it provides an important indicator of disease activity. Any study design that changes imaging regimens should consider the potential for influencing current or planned therapeutic trials, ensure that opportunity costs are measured, and capture indirect benefits and added value. CONCLUSION Evidence for the value, and therefore utility, of regular interval imaging is currently lacking. Ongoing collaborative efforts will improve trial design and generate the evidence to optimise monitoring imaging biomarkers in standard of care brain tumour management.
Collapse
Affiliation(s)
- Thomas C. Booth
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
- Department of Neuroradiology, King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | - Gerard Thompson
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Florien Boele
- Leeds Institute of Medical Research at St James’s, St James’s University Hospital, Leeds, United Kingdom
- Faculty of Medicine and Health, Leeds Institute of Health Sciences, University of Leeds, Leeds, United Kingdom
| | | | - Jorge Cardoso
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
| | - Liane Dos Santos Canas
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
| | | | - Keyoumars Ashkan
- Department of Neurosurgery, King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | | | - Nicky Huskens
- The Tessa Jowell Brain Cancer Mission, London, United Kingdom
| | - Aysha Luis
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Catherine McBain
- Department of Oncology, Christie Hospital NHS Foundation Trust, Manchester, United Kingdom
| | - Samantha J. Mills
- Department of Neuroradiology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| | - Marc Modat
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
| | - Nick Morley
- Department of Radiology, Wales Research and Diagnostic PET Imaging Centre, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Caroline Murphy
- King’s College Trials Unit, King’s College London, London, United Kingdom
| | - Sebastian Ourselin
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
| | - Mark Pennington
- King’s Health Economics, King’s College London, London, United Kingdom
| | - James Powell
- Department of Oncology, Velindre Cancer Centre, Cardiff, United Kingdom
| | - David Summers
- Department of Neuroradiology, Western General Hospital, Edinburgh, United Kingdom
| | - Adam D. Waldman
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Colin Watts
- Birmingham Brain Cancer Program, University of Birmingham, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Matthew Williams
- Department of Neuro-oncology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Robin Grant
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael D. Jenkinson
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
13
|
Stegmayr C, Stoffels G, Filß C, Heinzel A, Lohmann P, Willuweit A, Ermert J, Coenen HH, Mottaghy FM, Galldiks N, Langen KJ. Current trends in the use of O-(2-[ 18F]fluoroethyl)-L-tyrosine ([ 18F]FET) in neurooncology. Nucl Med Biol 2021; 92:78-84. [PMID: 32113820 DOI: 10.1016/j.nucmedbio.2020.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/16/2020] [Indexed: 12/14/2022]
Abstract
The diagnostic potential of PET using the amino acid analogue O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) in brain tumor diagnostics has been proven in many studies during the last two decades and is still the subject of multiple studies every year. In addition to standard magnetic resonance imaging (MRI), positron emission tomography (PET) using [18F]FET provides important diagnostic data concerning brain tumor delineation, therapy planning, treatment monitoring, and improved differentiation between treatment-related changes and tumor recurrence. The pharmacokinetics, uptake mechanisms and metabolism have been well described in various preclinical studies. The accumulation of [18F]FET in most benign lesions and healthy brain tissue has been shown to be low, thus providing a high contrast between tumor tissue and benign tissue alterations. Based on logistic advantages of F-18 labelling and convincing clinical results, [18F]FET has widely replaced short lived amino acid tracers such as L-[11C]methyl-methionine ([11C]MET) in many centers across Western Europe. This review summarizes the basic knowledge on [18F]FET and its contribution to the care of patients with brain tumors. In particular, recent studies about specificity, possible pitfalls, and the utility of [18F]FET PET in tumor grading and prognostication regarding the revised WHO classification of brain tumors are addressed.
Collapse
Affiliation(s)
- Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Christian Filß
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany
| | - Alexander Heinzel
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany; Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Johannes Ermert
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Heinz H Coenen
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Felix M Mottaghy
- Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany; Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Germany; Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Duesseldorf, Germany; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Duesseldorf, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany; Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Germany; Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Duesseldorf, Germany.
| |
Collapse
|
14
|
Booth TC, Luis A, Brazil L, Thompson G, Daniel RA, Shuaib H, Ashkan K, Pandey A. Glioblastoma post-operative imaging in neuro-oncology: current UK practice (GIN CUP study). Eur Radiol 2020; 31:2933-2943. [PMID: 33151394 PMCID: PMC8043861 DOI: 10.1007/s00330-020-07387-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/13/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVES MRI remains the preferred imaging investigation for glioblastoma. Appropriate and timely neuroimaging in the follow-up period is considered to be important in making management decisions. There is a paucity of evidence-based information in current UK, European and international guidelines regarding the optimal timing and type of neuroimaging following initial neurosurgical treatment. This study assessed the current imaging practices amongst UK neuro-oncology centres, thus providing baseline data and informing future practice. METHODS The lead neuro-oncologist, neuroradiologist and neurosurgeon from every UK neuro-oncology centre were invited to complete an online survey. Participants were asked about current and ideal imaging practices following initial treatment. RESULTS Ninety-two participants from all 31 neuro-oncology centres completed the survey (100% response rate). Most centres routinely performed an early post-operative MRI (87%, 27/31), whereas only a third performed a pre-radiotherapy MRI (32%, 10/31). The number and timing of scans routinely performed during adjuvant TMZ treatment varied widely between centres. At the end of the adjuvant period, most centres performed an MRI (71%, 22/31), followed by monitoring scans at 3 monthly intervals (81%, 25/31). Additional short-interval imaging was carried out in cases of possible pseudoprogression in most centres (71%, 22/31). Routine use of advanced imaging was infrequent; however, the addition of advanced sequences was the most popular suggestion for ideal imaging practice, followed by changes in the timing of EPMRI. CONCLUSION Variations in neuroimaging practices exist after initial glioblastoma treatment within the UK. Multicentre, longitudinal, prospective trials are needed to define the optimal imaging schedule for assessment. KEY POINTS • Variations in imaging practices exist in the frequency, timing and type of interval neuroimaging after initial treatment of glioblastoma within the UK. • Large, multicentre, longitudinal, prospective trials are needed to define the optimal imaging schedule for assessment.
Collapse
Affiliation(s)
- Thomas C Booth
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, SE1 7EH, UK. .,Department of Neuroradiology Ruskin Wing, King's College Hospital NHS Foundation Trust, London, SE5 9RS, UK.
| | - Aysha Luis
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, SE1 7EH, UK.,Department of Neuroradiology, National Hospital For Neurology and Neurosrgery, London, WC1N 3BG, UK
| | - Lucy Brazil
- Department of Oncology, Guy's and St Thomas' NHS Foundation Trust, London, SE1 7EH, UK
| | - Gerry Thompson
- Centre for Clinical Brain Sciences, Edinburgh, EH16 4SB, UK
| | - Rachel A Daniel
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, SE1 7EH, UK
| | - Haris Shuaib
- Department of Medical Physics, Guy's & St. Thomas' NHS Foundation Trust, London, SE1 7EH, UK.,Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK
| | - Keyoumars Ashkan
- Department of Neurosurgery, King's College Hospital NHS Foundation Trust, London, SE5 9RS, UK
| | - Anmol Pandey
- Faculty of Life Sciences and Medicine, King's College London Strand, London, WC2R 2LS, UK
| |
Collapse
|
15
|
Verburg N, de Witt Hamer PC. State-of-the-art imaging for glioma surgery. Neurosurg Rev 2020; 44:1331-1343. [PMID: 32607869 PMCID: PMC8121714 DOI: 10.1007/s10143-020-01337-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/25/2020] [Accepted: 06/15/2020] [Indexed: 11/29/2022]
Abstract
Diffuse gliomas are infiltrative primary brain tumors with a poor prognosis despite multimodal treatment. Maximum safe resection is recommended whenever feasible. The extent of resection (EOR) is positively correlated with survival. Identification of glioma tissue during surgery is difficult due to its diffuse nature. Therefore, glioma resection is imaging-guided, making the choice for imaging technique an important aspect of glioma surgery. The current standard for resection guidance in non-enhancing gliomas is T2 weighted or T2w-fluid attenuation inversion recovery magnetic resonance imaging (MRI), and in enhancing gliomas T1-weighted MRI with a gadolinium-based contrast agent. Other MRI sequences, like magnetic resonance spectroscopy, imaging modalities, such as positron emission tomography, as well as intraoperative imaging techniques, including the use of fluorescence, are also available for the guidance of glioma resection. The neurosurgeon’s goal is to find the balance between maximizing the EOR and preserving brain functions since surgery-induced neurological deficits result in lower quality of life and shortened survival. This requires localization of important brain functions and white matter tracts to aid the pre-operative planning and surgical decision-making. Visualization of brain functions and white matter tracts is possible with functional MRI, diffusion tensor imaging, magnetoencephalography, and navigated transcranial magnetic stimulation. In this review, we discuss the current available imaging techniques for the guidance of glioma resection and the localization of brain functions and white matter tracts.
Collapse
Affiliation(s)
- Niels Verburg
- Department of Neurosurgery and Cancer Center Amsterdam, Amsterdam UMC location VU University Medical Center, Amsterdam, The Netherlands. .,Division of Neurosurgery, Department of Clinical Neurosciences, Cambridge Brain Tumor Imaging Laboratory, University of Cambridge, Addenbrooke's Hospital, Hill Rd, Cambridge, CB2 0QQ, UK.
| | - Philip C de Witt Hamer
- Department of Neurosurgery and Cancer Center Amsterdam, Amsterdam UMC location VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Stegmayr C, Willuweit A, Lohmann P, Langen KJ. O-(2-[18F]-Fluoroethyl)-L-Tyrosine (FET) in Neurooncology: A Review of Experimental Results. Curr Radiopharm 2020; 12:201-210. [PMID: 30636621 DOI: 10.2174/1874471012666190111111046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 11/22/2022]
Abstract
In recent years, PET using radiolabelled amino acids has gained considerable interest as an additional tool besides MRI to improve the diagnosis of cerebral gliomas and brain metastases. A very successful tracer in this field is O-(2-[18F]fluoroethyl)-L-tyrosine (FET) which in recent years has replaced short-lived tracers such as [11C]-methyl-L-methionine in many neuro-oncological centers in Western Europe. FET can be produced with high efficiency and distributed in a satellite concept like 2- [18F]fluoro-2-deoxy-D-glucose. Many clinical studies have demonstrated that FET PET provides important diagnostic information regarding the delineation of cerebral gliomas for therapy planning, an improved differentiation of tumor recurrence from treatment-related changes and sensitive treatment monitoring. In parallel, a considerable number of experimental studies have investigated the uptake mechanisms of FET on the cellular level and the behavior of the tracer in various benign lesions in order to clarify the specificity of FET uptake for tumor tissue. Further studies have explored the effects of treatment related tissue alterations on tracer uptake such as surgery, radiation and drug therapy. Finally, the role of blood-brain barrier integrity for FET uptake which presents an important aspect for PET tracers targeting neoplastic lesions in the brain has been investigated in several studies. Based on a literature research regarding experimental FET studies and corresponding clinical applications this article summarizes the knowledge on the uptake behavior of FET, which has been collected in more than 30 experimental studies during the last two decades and discusses the role of these results in the clinical context.
Collapse
Affiliation(s)
- Carina Stegmayr
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany.,Department of Nuclear Medicine, University of Aachen, Aachen, Germany.,Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Juelich, Germany
| |
Collapse
|
17
|
Human biodistribution and radiation dosimetry of [ 18F]DASA-23, a PET probe targeting pyruvate kinase M2. Eur J Nucl Med Mol Imaging 2020; 47:2123-2130. [PMID: 31938892 DOI: 10.1007/s00259-020-04687-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE To assess the safety, biodistribution, and radiation dosimetry of the novel positron emission tomography (PET) radiopharmaceutical 1-((2-fluoro-6-[[18F]]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA-23) in healthy volunteers. METHODS We recruited 5 healthy volunteers who provided a written informed consent. Volunteers were injected with 295.0 ± 8.2 MBq of [18F]DASA-23 intravenously. Immediately following injection, a dynamic scan of the brain was acquired for 15 min. This was followed by serial whole-body PET/MRI scans acquired up to 3 h post-injection. Blood samples were collected at regular intervals, and vital signs monitored pre- and post-radiotracer administration. Regions of interest were drawn around multiple organs, time-activity curves were calculated, and organ uptake and dosimetry were estimated with OLINDA/EXM (version 1.1) software. RESULTS All subjects tolerated the PET/MRI examination, without adverse reactions to [18F]DASA-23. [18F]DASA-23 passively crossed the blood-brain barrier, followed by rapid clearance from the brain. High accumulation of [18F]DASA-23 was noted in organs such as the gallbladder, liver, small intestine, and urinary bladder, suggesting hepatobiliary and urinary clearance. The effective dose of [18F]DASA-23 was 23.5 ± 5.8 μSv/MBq. CONCLUSION We successfully completed a pilot first-in-human study of [18F]DASA-23. Our results indicate that [18F]DASA-23 can be used safely in humans to evaluate pyruvate kinase M2 levels. Ongoing studies are evaluating the ability of [18F]DASA-23 to visualize intracranial malignancies, NCT03539731. TRIAL REGISTRATION ClinicalTrials.gov , NCT03539731 (registered 28 May 2018).
Collapse
|
18
|
Geisler S, Stegmayr C, Niemitz N, Lohmann P, Rapp M, Stoffels G, Willuweit A, Galldiks N, Filss C, Sabel MC, Coenen HH, Shah NJ, Langen KJ. Treatment-Related Uptake of O-(2-18F-Fluoroethyl)-l-Tyrosine and l-[Methyl-3H]-Methionine After Tumor Resection in Rat Glioma Models. J Nucl Med 2019; 60:1373-1379. [DOI: 10.2967/jnumed.119.225680] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/20/2019] [Indexed: 12/20/2022] Open
|
19
|
Abstract
PURPOSE OF REVIEW The aim of this study was to give an update on the emerging role of PET using radiolabelled amino acids in the diagnostic workup and management of patients with cerebral gliomas and brain metastases. RECENT FINDINGS Numerous studies have demonstrated the potential of PET using radiolabelled amino acids for differential diagnosis of brain tumours, delineation of tumour extent for treatment planning and biopsy guidance, differentiation between tumour progression and recurrence versus treatment-related changes, and for monitoring of therapy. The Response Assessment in Neuro-Oncology (RANO) working group - an international effort to develop new standardized response criteria for clinical trials in brain tumours - has recently recommended the use of amino acid PET imaging for brain tumour management in addition to MRI at every stage of disease. With the introduction of F-18 labelled amino acids, a broader clinical application has become possible, but is still hampered by the lack of regulatory approval and of reimbursement in many countries. SUMMARY PET using radiolabelled amino acids is a rapidly evolving method that can significantly enhance the diagnostic value of MRI in brain tumours. Current developments suggest that this imaging technique will become an indispensable tool in neuro-oncological centres in the near future.
Collapse
|
20
|
Can Early Postoperative O-(2- 18FFluoroethyl)-l-Tyrosine Positron Emission Tomography After Resection of Glioblastoma Predict the Location of Later Tumor Recurrence? World Neurosurg 2018; 121:e467-e474. [PMID: 30267942 DOI: 10.1016/j.wneu.2018.09.139] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Glioblastoma inevitably recurs despite aggressive therapy. Therefore, it would be helpful to predict the location of tumor recurrence from postoperative imaging to customize further treatment. O-(2-18Ffluoroethyl)-l-tyrosine (FET) positron emission tomography (PET) might be a helpful technique, because tumor tissue can be differentiated from normal brain tissue with high specificity. METHODS Thirty-two consecutive patients with perioperative and follow-up imaging data available were included. On postoperative FET-PET, the tumor/normal brain (TTB) ratio around the resection cavity borders was measured. Increased TTB ratios were recorded and anatomically correlated with the site of later tumor recurrence. On postoperative magnetic resonance imaging (MRI), residual contrast-enhancing tumor correlated with the site of later tumor recurrence. RESULTS Location of progression was predictable using MRI alone in 42% of patients by residual tumor on postoperative MRI. FET-PET was predictive in 25 patients by a clear hot spot at the site of later tumor recurrence. In 3 patients, it was partially predictive and in 4 was not predictive of the tumor recurrence location. One patient without any tracer uptake was recurrence free at the last follow-up examination. In contrast to the postoperative MRI results, tumor recurrence was found in 79% at a site of elevated TTB ratio on postoperative FET-PET. Therefore, the predictability of the tumor recurrence location using postoperative FET-PET was greater than that with MRI, and all cases predictable using MRI could have been predicted using FET-PET. CONCLUSIONS Postoperative FET-PET can be helpful for planning subsequent therapy, such as repeat resection or radiotherapy, because tumor recurrence can be predicted with relatively greater sensitivity than with MRI alone.
Collapse
|
21
|
Is It Possible to Establish the Extent of Resection of Glioblastoma With 18F-Fluorocholine PET/CT? Clin Nucl Med 2018; 43:e269-e273. [DOI: 10.1097/rlu.0000000000002153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Haider AS, Graffeo CS, Perry A, Carlstrom LP, Burns TC. Spontaneous Resolution of a Confounding Insular Lesion. Cureus 2018. [PMID: 29541573 PMCID: PMC5844597 DOI: 10.7759/cureus.2053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Insular gliomas were previously considered inoperable lesions and were typically treated via biopsy, chemotherapy and/or radiation, if not observation alone. Stereotactic biopsies of low grade insular gliomas can underestimate tumor grade or fail to establish malignancy. Moreover, the survival advantages of maximal safe resection for insular lesions are increasingly being recognized. As such, early surgical resection is increasingly being performed. As with most lesions, a differential diagnosis exists for apparent insular gliomas, with definitive diagnosis generally obtained upon resection. We report an illuminating case that presented similarly to an insular glioma undergoing malignant transformation, but resolved spontaneously following a nondiagnostic biopsy. A 53-year-old female patient presented with aphasia and dizziness, followed by syncope and a 30-minute loss-of-consciousness. Imaging findings included a 12 mm region of contrast enhancement and central necrosis within a larger 3.5 cm expansile, T2-hyperintense lesion involving most of the left insula and extending into the anterior left temporal lobe. Imaging was felt most consistent with high-grade glioma. Stereotactic biopsy demonstrated nonspecific gliosis without definitive neoplastic tissue. A systemic workup was unrevealing, and thus an open procedure was subsequently planned in the intraoperative magnetic resonance imaging (MRI) suite. Preoperative MRI demonstrated interval resolution of the original lesion, despite profound T2 hyperintensity along the needle tract; thus, the planned resection was aborted. Subsequent imaging and systemic studies failed to establish a definitive infectious, neoplastic, autoimmune, or other diagnosis. However, poor dentition, history of a recent dental procedure, and the tiny central focus of diffusion restriction on the index MRI rendered abscess the most parsimonious explanation. On follow-up imaging, the lesion was noted to have further resolved without intervention. Our case illustrates the complexity of managing insular lesions and highlights the potential for alternate pathologies that can mimic insular glioma. Additionally, it provides a humbling reminder that, even in the presence of seemingly pathognomonic imaging findings, a differential diagnosis of insular lesions must be thoughtfully considered in patient counseling and presurgical planning.
Collapse
|
23
|
Imaging of amino acid transport in brain tumours: Positron emission tomography with O-(2-[ 18 F]fluoroethyl)- L -tyrosine (FET). Methods 2017; 130:124-134. [DOI: 10.1016/j.ymeth.2017.05.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/08/2017] [Accepted: 05/21/2017] [Indexed: 01/01/2023] Open
|
24
|
Ladefoged CN, Andersen FL, Kjær A, Højgaard L, Law I. RESOLUTE PET/MRI Attenuation Correction for O-(2- 18F-fluoroethyl)-L-tyrosine (FET) in Brain Tumor Patients with Metal Implants. Front Neurosci 2017; 11:453. [PMID: 28848379 PMCID: PMC5554515 DOI: 10.3389/fnins.2017.00453] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/25/2017] [Indexed: 01/06/2023] Open
Abstract
Aim: Positron emission tomography (PET) imaging is a useful tool for assisting in correct differentiation of tumor progression from reactive changes, and the radiolabeled amino acid analog tracer O-(2-18F-fluoroethyl)-L-tyrosine (FET)-PET is amongst the most frequently used. The FET-PET images need to be quantitatively correct in order to be used clinically, which require accurate attenuation correction (AC) in PET/MRI. The aim of this study was to evaluate the use of the subject-specific MR-derived AC method RESOLUTE in post-operative brain tumor patients. Methods: We analyzed 51 post-operative brain tumor patients (68 examinations, 200 MBq [18F]-FET) investigated in a PET/MRI scanner. MR-AC maps were acquired using: (1) the Dixon water fat separation sequence, (2) the ultra short echo time (UTE) sequences, (3) calculated using our new RESOLUTE methodology, and (4) a same day low-dose CT used as reference “gold standard.” For each subject and each AC method the tumor was delineated by isocontouring tracer uptake above a tumor(T)-to-brain background (B) activity ratio of 1.6. We measured B, tumor mean and maximal activity (TMEAN, TMAX), biological tumor volume (BTV), and calculated the clinical metrics TMEAN/B and TMAX/B. Results: When using RESOLUTE 5/68 studies did not meet our predefined acceptance criteria of TMAX/B difference to CT-AC < ±0.1 or 5%, TMEAN/B < ±0.05 or 5%, and BTV < ±2 mL or 10%. In total, 46/68 studies failed our acceptance criteria using Dixon, and 26/68 using UTE. The 95% limits of agreement for TMAX/B was for RESOLUTE (−3%; 4%), Dixon (−9%; 16%), and UTE (−7%; 10%). The absolute error when measuring BTV was 0.7 ± 1.9 mL (N.S) with RESOLUTE, 5.3 ± 10 mL using Dixon, and 1.7 ± 3.7 mL using UTE. RESOLUTE performed best in the identification of the location of peak activity and in brain tumor follow-up monitoring using clinical FET PET metrics. Conclusions: Overall, we found RESOLUTE to be the AC method that most robustly reproduced the CT-AC clinical metrics per se, during follow-up, and when interpreted into defined clinical use cut-off criteria and into the patient history. RESOLUTE is especially suitable for brain tumor patients, as these often present with distorted anatomy where other methods based on atlas/template information might fail.
Collapse
Affiliation(s)
- Claes N Ladefoged
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of CopenhagenCopenhagen, Denmark
| | - Flemming L Andersen
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of CopenhagenCopenhagen, Denmark
| | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of CopenhagenCopenhagen, Denmark
| | - Liselotte Højgaard
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of CopenhagenCopenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of CopenhagenCopenhagen, Denmark
| |
Collapse
|
25
|
Xu W, Gao L, Shao A, Zheng J, Zhang J. The performance of 11C-Methionine PET in the differential diagnosis of glioma recurrence. Oncotarget 2017; 8:91030-91039. [PMID: 29207622 PMCID: PMC5710903 DOI: 10.18632/oncotarget.19024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/20/2017] [Indexed: 01/02/2023] Open
Abstract
Despite the advancement of neuroimaging techniques, it often remains a diagnostic challenge to distinguish recurrent glioma from lesions representing treatment effect. Preliminary reports suggest that 11C-methionine Positron emission tomography (PET) can assist in diagnosing true glioma recurrence. We present here a meta-analysis to assess the accuracy of 11C-methionine PET in identifying recurrent glioma in patients who had undergone prior therapy. A comprehensive search of the PubMed, Embase and Chinese Biomedical (CBM) databases yielded 23 eligible articles comprising 29 studies listed prior to November 20, 2016, representing 891 patients. In this report, we assess the methodological quality of each article individually and perform a meta-analysis to obtain the summary diagnostic accuracy of 11C-methionine PET in correctly identifying recurrent glioma. The pooled sensitivity and specificity are 0.88 (95% CI: 0.85, 0.91) and 0.85 (95% CI: 0.80, 0.89), respectively, with an area under the curve (AUC) for the summary receiver-operating characteristic curve (SROC) of 0.9352. We conclude that 11C-methionine PET has excellent diagnostic performance for differentiating glioma recurrence from treatment effect.
Collapse
Affiliation(s)
- Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liansheng Gao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingwei Zheng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
26
|
Galldiks N, Law I, Pope WB, Arbizu J, Langen KJ. The use of amino acid PET and conventional MRI for monitoring of brain tumor therapy. Neuroimage Clin 2016; 13:386-394. [PMID: 28116231 PMCID: PMC5226808 DOI: 10.1016/j.nicl.2016.12.020] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/09/2016] [Accepted: 12/16/2016] [Indexed: 12/03/2022]
Abstract
Routine diagnostics and treatment monitoring of brain tumors is usually based on contrast-enhanced MRI. However, the capacity of conventional MRI to differentiate tumor tissue from posttherapeutic effects following neurosurgical resection, chemoradiation, alkylating chemotherapy, radiosurgery, and/or immunotherapy may be limited. Metabolic imaging using PET can provide relevant additional information on tumor metabolism, which allows for more accurate diagnostics especially in clinically equivocal situations. This review article focuses predominantly on the amino acid PET tracers 11C-methyl-l-methionine (MET), O-(2-[18F]fluoroethyl)-l-tyrosine (FET) and 3,4-dihydroxy-6-[18F]-fluoro-l-phenylalanine (FDOPA) and summarizes investigations regarding monitoring of brain tumor therapy.
Collapse
Affiliation(s)
- Norbert Galldiks
- Dept. of Neurology, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
- Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany
| | - Ian Law
- Dept.of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Whitney B. Pope
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Javier Arbizu
- Dept. of Nuclear Medicine, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
- Dept. of Nuclear Medicine, University of Aachen, Aachen, Germany
| |
Collapse
|
27
|
Abstract
A previous review published in 2012 demonstrated the role of clinical PET for diagnosis and management of brain tumors using mainly FDG, amino acid tracers, and 18F-fluorothymidine. This review provides an update on clinical PET studies, most of which are motivated by prediction of prognosis and planning and monitoring of therapy in gliomas. For FDG, there has been additional evidence supporting late scanning, and combination with 13N ammonia has yielded some promising results. Large neutral amino acid tracers have found widespread applications mostly based on 18F-labeled compounds fluoroethyltyrosine and fluorodopa for targeting biopsies, therapy planning and monitoring, and as outcome markers in clinical trials. 11C-alpha-methyltryptophan (AMT) has been proposed as an alternative to 11C-methionine, and there may also be a role for cyclic amino acid tracers. 18F-fluorothymidine has shown strengths for tumor grading and as an outcome marker. Studies using 18F-fluorocholine (FCH) and 68Ga-labeled compounds are promising but have not yet clearly defined their role. Studies on radiotherapy planning have explored the use of large neutral amino acid tracers to improve the delineation of tumor volume for irradiation and the use of hypoxia markers, in particular 18F-fluoromisonidazole. Many studies employed the combination of PET with advanced multimodal MR imaging methods, mostly demonstrating complementarity and some potential benefits of hybrid PET/MR.
Collapse
Affiliation(s)
- Karl Herholz
- The University of Manchester, Division of Neuroscience and Experimental Psychology Wolfson Molecular Imaging Centre, Manchester, England, United Kingdom.
| |
Collapse
|
28
|
Buchmann N, Kläsner B, Gempt J, Bauer JS, Pyka T, Delbridge C, Meyer B, Krause BJ, Ringel F. (18)F-Fluoroethyl-l-Thyrosine Positron Emission Tomography to Delineate Tumor Residuals After Glioblastoma Resection: A Comparison with Standard Postoperative Magnetic Resonance Imaging. World Neurosurg 2016; 89:420-6. [PMID: 26893043 DOI: 10.1016/j.wneu.2016.02.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 02/05/2016] [Accepted: 02/06/2016] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Complete resection of contrast-enhancing tumor is an important prognostic factor in glioblastoma therapy. The current clinical standard for control of resection is magnetic resonance imaging (MRI). (18)F-Fluoroethyl-l-thyrosine (FET) is a positron emission tomography (PET) radiopharmaceutical applicable for widespread use because of its long half-life radionuclide. We assessed the sensitivity of postoperative MRI versus FET-PET to detect residual tumor and the impact of the time interval between resection and FET-PET. METHODS MRI and FET-PET were performed preoperatively and postoperatively in 62 patients undergoing 63 operations. FET-PET was performed in 43 cases within 72 hours after resection and in 20 cases >72 hours after resection. Detection and measurement of volume of residual tumors were compared. Correlations between residual tumor detection and timing of PET after resection and recurrence were examined. RESULTS Complete resection was confirmed by both imaging modalities in 44% of cases, and residual tumor was detected consistently in 37% of cases. FET-PET detected residual tumor in 14% of cases in which MRI showed no residual tumor. MRI showed residual tumors in 5% of cases that were not identified by PET. Average PET-based residual tumor volume was higher than MRI-based volume (3.99 cm(3) vs. 1.59 cm(3)). Detection of and difference in volume of residual tumor were not correlated with timing of PET after resection or recurrence status. CONCLUSIONS Postoperative FET-PET revealed residual tumor with higher sensitivity than MRI and showed larger tumor volumes. In this series, performing PET >72 hours after resection did not influence the results of PET. We recommend FET-PET as a helpful adjunct in addition to MRI for postoperative assessment of residual tumor.
Collapse
Affiliation(s)
- Niels Buchmann
- Department of Neurosurgery, Klinikum rechts der Isar, Technische Universität München, München, Germany.
| | - Benjamin Kläsner
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, München, Germany; Department of Nuclear Medicine, Klinikum Konstanz, Konstanz, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Jan Stefan Bauer
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Thomas Pyka
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Claire Delbridge
- Division of Neuropathology, Institute of Pathology, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Bernd Joachim Krause
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, München, Germany; Department of Nuclear Medicine, Universitätsklinikum Rostock, Rostock, Germany
| | - Florian Ringel
- Department of Neurosurgery, Klinikum rechts der Isar, Technische Universität München, München, Germany
| |
Collapse
|