1
|
Bhattacharya K, Rastogi S, Mahajan A. Post-treatment imaging of gliomas: challenging the existing dogmas. Clin Radiol 2024; 79:e376-e392. [PMID: 38123395 DOI: 10.1016/j.crad.2023.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 10/23/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Gliomas are the commonest malignant central nervous system tumours in adults and imaging is the cornerstone of diagnosis, treatment, and post-treatment follow-up of these patients. With the ever-evolving treatment strategies post-treatment imaging and interpretation in glioma remains challenging, more so with the advent of anti-angiogenic drugs and immunotherapy, which can significantly alter the appearance in this setting, thus making interpretation of routine imaging findings such as contrast enhancement, oedema, and mass effect difficult to interpret. This review details the various methods of management of glioma including the upcoming novel therapies and their impact on imaging findings, with a comprehensive description of the imaging findings in conventional and advanced imaging techniques. A systematic appraisal for the existing and emerging techniques of imaging in these settings and their clinical application including various response assessment guidelines and artificial intelligence based response assessment will also be discussed.
Collapse
Affiliation(s)
- K Bhattacharya
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - S Rastogi
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - A Mahajan
- Department of imaging, The Clatterbridge Cancer Centre, NHS Foundation Trust, Pembroke Place, Liverpool L7 8YA, UK; University of Liverpool, Liverpool L69 3BX, UK.
| |
Collapse
|
2
|
Xiaoxue T, Yinzhong W, Meng Q, Lu X, Lei J. Diagnostic value of PET with different radiotracers and MRI for recurrent glioma: a Bayesian network meta-analysis. BMJ Open 2023; 13:e062555. [PMID: 36863738 PMCID: PMC9990663 DOI: 10.1136/bmjopen-2022-062555] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 01/09/2023] [Indexed: 03/04/2023] Open
Abstract
OBJECTIVE The purpose of this study was to evaluate the diagnostic accuracy of 6 different imaging modalities for differentiating glioma recurrence from postradiotherapy changes by performing a network meta-analysis (NMA) using direct comparison studies with 2 or more imaging techniques. DATA SOURCES PubMed, Scopus, EMBASE, the Web of Science and the Cochrane Library were searched from inception to August 2021. The Confidence In Network Meta-Analysis (CINeMA) tool was used to evaluate the quality of the included studies with the criterion for study inclusion being direct comparison using 2 or more imaging modalities. DATA EXTRACTION AND SYNTHESIS The consistency was evaluated by examining the agreement between direct and indirect effects. NMA was performed and the surface under the the cumulative ranking curve (SUCRA) values was obtained to calculate the probability of each imaging modality being the most effective diagnostic method. The CINeMA tool was used to evaluate the quality of the included studies. MAIN OUTCOMES AND MEASURES Direct comparison, inconsistency test, NMA and SUCRA values. RESULTS A total of 8853 potentially relevant articles were retrieved and 15 articles met the inclusion criteria. 18F-FET showed the highest SUCRA values for sensitivity, specificity, positive predictive value and accuracy, followed by 18F-FDOPA. The quality of the included evidence is classified as moderate. CONCLUSION AND RELEVANCE This review indicates that 18F-FET and 18F-FDOPA may have greater diagnostic value for glioma recurrence relative to other imaging modalities (Grading of Recommendations, Assessment, Development and Evaluations B). PROSPERO REGISTRATION NUMBER CRD42021293075.
Collapse
Affiliation(s)
- Tian Xiaoxue
- Department of Nuclear Medicine, the Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Wang Yinzhong
- Department of Radiology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Qi Meng
- Department of Radiology, No.2 Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xingru Lu
- Department of Radiology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Junqiang Lei
- Department of Radiology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
3
|
Wiggins RH, Hoffman JM, Fine GC, Covington MF, Salem AE, Koppula BR, Morton KA. PET-CT in Clinical Adult Oncology-V. Head and Neck and Neuro Oncology. Cancers (Basel) 2022; 14:cancers14112726. [PMID: 35681709 PMCID: PMC9179458 DOI: 10.3390/cancers14112726] [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: 04/29/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Positron emission tomography (PET), typically combined with computed tomography (CT) has become a critical advanced imaging technique in oncology. With PET-CT, a radioactive molecule (radiotracer) is injected in the bloodstream and localizes to sites of tumor because of specific cellular features of the tumor that accumulate the targeting radiotracer. The CT scan, performed at the same time, provides information to facilitate attenuation correction, so that radioactivity from deep or dense structures can be better visualized, but with head and neck malignancies it is critical to provide correlating detailed anatomic imaging. PET-CT has a variety of applications in oncology, including staging, therapeutic response assessment, restaging, and surveillance. This series of six review articles provides an overview of the value, applications, and imaging and interpretive strategies of PET-CT in the more common adult malignancies. The fifth report in this series provides a review of PET-CT imaging in head and neck and neuro oncology. Abstract PET-CT is an advanced imaging modality with many oncologic applications, including staging, assessment of response to therapy, restaging, and longitudinal surveillance for recurrence. The goal of this series of six review articles is to provide practical information to providers and imaging professionals regarding the best use of PET-CT for specific oncologic indications, and the potential pitfalls and nuances that characterize these applications. In addition, key tumor-specific clinical information and representative PET-CT images are provided to outline the role that PET-CT plays in the management of oncology patients. Hundreds of different types of tumors exist, both pediatric and adult. A discussion of the role of FDG PET for all of these is beyond the scope of this review. Rather, this series of articles focuses on the most common adult malignancies that may be encountered in clinical practice. It also focuses on FDA-approved and clinically available radiopharmaceuticals, rather than research tracers or those requiring a local cyclotron. The fifth review article in this series focuses on PET-CT imaging in head and neck tumors, as well as brain tumors. Common normal variants, key anatomic features, and benign mimics of these tumors are reviewed. The goal of this review article is to provide the imaging professional with guidance in the interpretation of PET-CT for the more common head and neck malignancies and neuro oncology, and to inform the referring providers so that they can have realistic expectations of the value and limitations of PET-CT for the specific type of tumor being addressed.
Collapse
Affiliation(s)
- Richard H. Wiggins
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
| | - John M. Hoffman
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
| | - Gabriel C. Fine
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
| | - Matthew F. Covington
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
| | - Ahmed Ebada Salem
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
- Department of Radiodiagnosis and Intervention, Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Bhasker R. Koppula
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
| | - Kathryn A. Morton
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA; (R.H.W.); (J.M.H.); (G.C.F.); (M.F.C.); (A.E.S.); (B.R.K.)
- Intermountain Healthcare Hospitals, Summit Physician Specialists, Murray, UT 84123, USA
- Correspondence: ; Tel.: +1-801-581-7553
| |
Collapse
|
4
|
Tong J, Wang C, Liu H. Temporal information guided dynamic dual-tracer PET signal separation network. Med Phys 2022; 49:4585-4598. [PMID: 35396705 DOI: 10.1002/mp.15566] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 02/21/2021] [Accepted: 01/24/2022] [Indexed: 11/09/2022] Open
Abstract
PURPOSE The difficulty of dynamic dual-tracer positron emission tomography (PET) technology is to separate the complete single-tracer information from mixed dual-tracer. Traditional methods cannot separate single injection single-scan dynamic dual-tracer PET images. In this paper, we propose a deep learning framework based on gated recurrent unit (GRU) network and evaluate its performance with simulation experiments and realistic monkey data. METHODS The proposed single-scan dynamic dual-tracer PET image separation network consists of three parts, including encoder, separation and decoder module. Encoder part is to map the mixed time activity curves (TACs) from the low-dimensional space to the high-dimensional space to get mixed weight vector matrix. Separation part is to capture the temporal information of mixed weight vector matrix using bi-directional GRU (bi-GRU) layer to obtain the single-tracer masks, and the decoding part remaps the high-dimensional single-tracer weight vector matrix to the low-dimensional space to obtain two separated single tracers. RESULTS In the simulation experiments under different tracers, phantoms, noise levels, arterial input function (AIF) and k-parameter with Gaussian random, compared to the stacked auto encoder (SAE) network and traditional background subtraction method, GRU-based network has better performance with low bias and mean squared error (MSE). In the realistic study, the image results of GRU network have higher mean structural similarity (MSSIM), and peak signal to noise ratio (PSNR). CONCLUSIONS This study demonstrates the feasibility of temporal information guided neural network in single-injection single-scan dynamic dual-tracer PET images separation. The GRU-based network uses TAC temporal information without AIFs to make the separation results more robust and accurate, which significantly outperforms state-of-the-art method qualitatively and quantitatively. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Junyi Tong
- State Key Laboratory of Modern Optical Instrumentation, Zhejiang University, Hangzhou, 310027, China
| | - Chunxia Wang
- State Key Laboratory of Modern Optical Instrumentation, Zhejiang University, Hangzhou, 310027, China
| | - Huafeng Liu
- State Key Laboratory of Modern Optical Instrumentation, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
5
|
Diao W, Su D, Cao Y, Jia Z. The diagnostic accuracy of O-(2-18F-fluoroethyl)-L-tyrosine parameters for the differentiation of brain tumour progression from treatment-related changes. Nucl Med Commun 2022; 43:350-358. [PMID: 35102078 DOI: 10.1097/mnm.0000000000001524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND 18F-fluoro-ethyl-tyrosine (18F-FET) is recommended to distinguish brain tumours post-therapeutic true progression (including recurrent and metastatic brain tumours) and treatment-related change (TRC). However, many parameters of 18F-FET can be used for this differential diagnosis. Our purpose was to investigate the diagnostic accuracy of various 18F-FET parameters to differentiate true progression from TRC. METHODS We performed a literature search using the following databases: the PubMed, Embase and Web of Science databases up to 29 November 2020. We included studies that reported the diagnostic test results of 18F-FET to distinguish true progression from TRC. The Quality Assessment of Diagnostic Accuracy Studies-2 tool was used to evaluate the quality of the included studies. The diagnostic accuracy of various parameters was pooled using a random-effects model. RESULTS We included 17 eligible studies (nine parameters). For static parameters of 18F-FET, the maximum and mean tumour-to-brain ratios (TBRmax and TBRmean) showed similar pooled sensitivities of 82% [95% confidence interval (CI), 80-85%) and 82% (95% CI, 78-85%), respectively. Among the three kinetic parameters (slope, time to peak and kinetic pattern), the kinetic pattern presented the optimal diagnostic value with a pooled sensitivity of 81% (95% CI, 75-86%). When combining the static and kinetic parameters, the diagnostic performance of 18F-FET was significantly improved, with a pooled sensitivity of 90% (95% CI, 84-94%) in the combination of TBR and kinetic patterns. CONCLUSIONS 18F-FET static parameters alone showed a comparably high sensitivity in the differentiation between brain tumour true progression and TRC. Combining static and kinetic parameters provided improved diagnostic performance.
Collapse
Affiliation(s)
- Wei Diao
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, Peoples Republic of China
| | | | | | | |
Collapse
|
6
|
Guglielmo P, Quartuccio N, Rossetti V, Celli M, Alongi P, Boero M, Arnone G, Baldari S, Matteucci F, Laudicella R. [ 18F] Fluorothymidine Positron Emission Tomography Imaging in Primary Brain Tumours: A Systematic Review. Curr Med Imaging 2022; 18:363-371. [PMID: 34533446 DOI: 10.2174/1573405617666210917123012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE This review aimed to summarize the available literature on the clinical application of [18F] FLT PET imaging in primary brain tumours. METHODS A comprehensive search strategy based on Pubmed/Medline, Scopus, Web of Science, Cochrane Library, Google Scholar, and the Embase databases was carried on using the following search string: ('3` Fluorothymidine'/exp OR 'FLT' OR '[81F]-FLT' OR '[18F] Fluorothymidine') AND ('pet'/exp OR 'pet' OR 'positron emission tomography') AND ('glioma'/exp OR 'glioma' OR 'brain tumour'/exp OR 'brain tumour'). The search was updated till March 2021 and only articles in English and studies investigating the clinical applications of [18F] FLT PET and PET/CT in primary brain tumours were considered eligible for inclusion. RESULTS The literature search ultimately yielded 52 studies included in the systematic review, with main results as follows: a) the uptake of [18F] FLT may guide stereotactic biopsy but does not discriminate between grade II and III glioma. b) [18F] FLT uptake and texture parameters correlate with overall survival (OS) in newly diagnosed gliomas. c) In patients with recurrent glioma, proliferative volume (PV) and tumour-to-normal brain (T/N) uptake ratio are independent predictors of survival. d) Patients demonstrating response to therapy at [18F] FLT PET scan show longer OS compared to non-responders. e) [18F] FLT PET demonstrated good performance in discriminating tumour recurrence from radionecrosis. However, controversial results exist in comparative literature examining the performance of [18F] FLT vs. other radiotracers in the assessment of recurrence. CONCLUSION [18F] FLT PET imaging has demonstrated potential benefits for grading, diagnostic and prognostic purposes, despite the small sample size studies due to the relatively low availability of the radiotracer.
Collapse
Affiliation(s)
| | - Natale Quartuccio
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico Di Cristina Benfratelli, Italy
| | - Virginia Rossetti
- Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Monica Celli
- Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Pierpaolo Alongi
- Nuclear Medicine Unit, Fondazione Istituto G. Giglio, Ct. da Pietra Pollastra-pisciotto, Cefalù. Italy
| | - Michele Boero
- Nuclear Medicine Unit, AO Brotzu, 09134 Cagliari, Italy
| | - Gaspare Arnone
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico Di Cristina Benfratelli, Italy
| | - Sergio Baldari
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy
| | - Federica Matteucci
- Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
7
|
Jia H, Xie T. Tracers progress for positron emission tomography imaging of glial-related disease. J Biomed Res 2022; 36:321-335. [PMID: 36131689 PMCID: PMC9548440 DOI: 10.7555/jbr.36.20220017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Glial cells play an essential part in the neuron system. They can not only serve as structural blocks in the human brain but also participate in many biological processes. Extensive studies have shown that astrocytes and microglia play an important role in neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, as well as glioma, epilepsy, ischemic stroke, and infections. Positron emission tomography is a functional imaging technique providing molecular-level information before anatomic changes are visible and has been widely used in many above-mentioned diseases. In this review, we focus on the positron emission tomography tracers used in pathologies related to glial cells, such as glioma, Alzheimer's disease, and neuroinflammation.
Collapse
Affiliation(s)
- Haoran Jia
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Tianwu Xie
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
- Tianwu Xie, Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China. Tel: +86-21-64048363, E-mail:
| |
Collapse
|
8
|
Binneboese A, Covington MF, Horn KP, Archibald ZG, Boucher KM, Morton KA, Hoffman JM. Correlation between FDG-PET uptake and survival in patients with primary brain tumors. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2021; 11:196-206. [PMID: 34234998 PMCID: PMC8255218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/07/2021] [Indexed: 06/13/2023]
Abstract
This study evaluates F-18 fluorodeoxyglucose positron emission tomography (FDG-PET) semi-quantitative analysis as biomarker of tumor aggressiveness and predictor of survival in patients with primary brain tumors. Semi-quantitative analyses (SUVmax, SUVmean) were derived from FDG PET images in 78 patients with suspected recurrence of primary brain tumors based on MRI. SUVmax and the ratio of lesion SUVmax to the SUVmean of contralateral white matter (SUVmax/WM) were measured. A one-way Analysis of Variance (ANOVA), Kaplan-Meier analyses and the log rank test for evaluating statistical significance were utilized. There was statistical significance for time between FDG-PET and patient death. There was a significant difference with respect to FDG-PET time to death between patients with glioblastoma and patients with anaplastic oligodendroglioma, oligodendroglioma, and other histological subtypes. There is significant correlation with SUVmax/WM and patient survival following FDG-PET when a cut-point ratio of 1.90 is used. A 1.90 cut-point ratio of SUVmax/WM was associated with a difference in survival. GBM was associated with a significant difference in terms of reduced survival following FDG PET compared to most other histological sub-types. These results may inform current treatment and counseling strategies for patients with primary brain tumors.
Collapse
Affiliation(s)
- Adam Binneboese
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, University of UtahSalt Lake, UT, USA
- Huntsman Cancer Institute, University of UtahSalt Lake, UT, USA
| | - Matthew F Covington
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, University of UtahSalt Lake, UT, USA
- Huntsman Cancer Institute, University of UtahSalt Lake, UT, USA
- Department of Radiology and Imaging Sciences, University of UtahSalt Lake, UT, USA
| | - Kevin P Horn
- Division of Nuclear Medicine, Department of Radiology, University of WashingtonSeattle, WA, USA
| | | | - Kenneth M Boucher
- Huntsman Cancer Institute, University of UtahSalt Lake, UT, USA
- Department of Internal Medicine, University of UtahSalt Lake, UT, USA
| | - Kathryn A Morton
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, University of UtahSalt Lake, UT, USA
- Huntsman Cancer Institute, University of UtahSalt Lake, UT, USA
- Department of Radiology and Imaging Sciences, University of UtahSalt Lake, UT, USA
| | - John M Hoffman
- Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, University of UtahSalt Lake, UT, USA
- Huntsman Cancer Institute, University of UtahSalt Lake, UT, USA
- Department of Radiology and Imaging Sciences, University of UtahSalt Lake, UT, USA
| |
Collapse
|
9
|
Vellayappan BA, McGranahan T, Graber J, Taylor L, Venur V, Ellenbogen R, Sloan AE, Redmond KJ, Foote M, Chao ST, Suh JH, Chang EL, Sahgal A, Lo SS. Radiation Necrosis from Stereotactic Radiosurgery-How Do We Mitigate? Curr Treat Options Oncol 2021; 22:57. [PMID: 34097171 DOI: 10.1007/s11864-021-00854-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 12/12/2022]
Abstract
OPINION STATEMENT Intracranial stereotactic radiosurgery (SRS) is an effective and convenient treatment for many brain conditions. Data regarding safety come mostly from retrospective single institutional studies and a small number of prospective studies. Variations in target delineation, treatment delivery, imaging follow-up protocols and dose prescription limit the interpretation of this data. There has been much clinical focus on radiation necrosis (RN) in particular, as it is being increasingly recognized on follow-up imaging. Symptomatic RN may be treated with medical therapy (such as corticosteroids and bevacizumab) with surgical resection being reserved for refractory patients. Nevertheless, RN remains a challenging condition to manage, and therefore upfront patient selection for SRS remains critical to provide complication-free control. Mitigation strategies need to be considered in situations where the baseline risk of RN is expected to be high-such as large target volume or re-irradiation. These may involve reduction in the prescribed dose or hypofractionated stereotactic radiation therapy (HSRT). Recently published guidelines and international meta-analysis report the benefit of HSRT in larger lesions, without compromising control rates. However, careful attention to planning parameters and SRS techniques still need to be adhered, even with HSRT. In cases where the risk is deemed to be high despite mitigation, a combination approach of surgery with or without post-operative radiation should be considered.
Collapse
Affiliation(s)
- Balamurugan A Vellayappan
- Department of Radiation oncology, National University Cancer Institute, 1E Kent Ridge Road, Level 7 Tower block, Singapore, 119228, Singapore.
| | - Tresa McGranahan
- Department of Neurology, Alvord Brain Tumor Center, University of Washington, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Jerome Graber
- Department of Neurology, Alvord Brain Tumor Center, University of Washington, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Lynne Taylor
- Department of Neurology, Alvord Brain Tumor Center, University of Washington, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Vyshak Venur
- Department of Neurology, Alvord Brain Tumor Center, University of Washington, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Richard Ellenbogen
- Department of Neurology, Alvord Brain Tumor Center, University of Washington, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Andrew E Sloan
- Department of Neurological Surgery, Seidman Cancer Center and University Hospitals of Cleveland, Case Western Reserve University, Cleveland, OH, USA
| | - Kristin J Redmond
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University, Baltimore, MD, USA
| | - Matthew Foote
- Department of Radiation Oncology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Samuel T Chao
- Department of Radiation Oncology, Rose Ella Burkhardt Brain Tumor and Neuro-oncology Center, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - John H Suh
- Department of Radiation Oncology, Rose Ella Burkhardt Brain Tumor and Neuro-oncology Center, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eric L Chang
- Department of Radiation Oncology, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA
| | - Arjun Sahgal
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Simon S Lo
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
10
|
Li C, Yi C, Chen Y, Xi S, Guo C, Yang Q, Wang J, Sai K, Zhang J, Ke C, Chen F, Lv Y, Zhang X, Chen Z. Identify glioma recurrence and treatment effects with triple-tracer PET/CT. BMC Med Imaging 2021; 21:92. [PMID: 34059015 PMCID: PMC8165792 DOI: 10.1186/s12880-021-00624-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/24/2021] [Indexed: 02/16/2023] Open
Abstract
Background Differential diagnosis of tumour recurrence (TuR) from treatment effects (TrE), mostly induced by radiotherapy and chemotherapy, is still difficult by using conventional computed tomography (CT) or magnetic resonance (MR) imaging. We have investigated the diagnostic performance of PET/CT with 3 tracers, 13N-NH3, 18F-FDOPA, and 18F-FDG, to identify TuR and TrE in glioma patients following treatment. Methods Forty-three patients with MR-suspected recurrent glioma were included. The maximum and mean standardized uptake values (SUVmax and SUVmean) of the lesion and the lesion-to-normal grey-matter cortex uptake (L/G) ratio were obtained from each tracer PET/CT. TuR or TrE was determined by histopathology or clinical MR follow-up for at least 6 months. Results In this cohort, 34 patients were confirmed to have TuR, and 9 patients met the diagnostic standard of TrE. The SUVmax and SUVmean of 13N-NH3 and 18F-FDOPA PET/CT at TuR lesions were significantly higher compared with normal brain tissue (13N-NH3 0.696 ± 0.558, 0.625 ± 0.507 vs 0.486 ± 0.413; 18F-FDOPA 0.455 ± 0.518, 0.415 ± 0.477 vs 0.194 ± 0.203; both P < 0.01), but there was no significant difference in 18F-FDG (6.918 ± 3.190, 6.016 ± 2.807 vs 6.356 ± 3.104, P = 0.290 and 0.493). L/G ratios of 13N-NH3 and 18F-FDOPA were significantly higher in TuR than in TrE group (13N-NH3, 1.573 ± 0.099 vs 1.025 ± 0.128, P = 0.008; 18F-FDOPA, 2.729 ± 0.131 vs 1.514 ± 0.141, P < 0.001). The sensitivity, specificity and AUC (area under the curve) by ROC (receiver operating characteristic) analysis were 57.7%, 100% and 0.803, for 13N-NH3; 84.6%, 100% and 0.938, for 18F-FDOPA; and 80.8%, 100%, and 0.952, for the combination, respectively. Conclusion Our results suggest that although multiple tracer PET/CT may improve differential diagnosis efficacy, for glioma TuR from TrE, 18F-FDOPA PET-CT is the most reliable. The combination of 18F-FDOPA and 13N-NH3 does not increase the diagnostic efficiency, while 18F-FDG is not worthy for differential diagnosis of glioma TuR and TrE.
Collapse
Affiliation(s)
- Cong Li
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Chang Yi
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yingshen Chen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Shaoyan Xi
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Chengcheng Guo
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Qunying Yang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Jian Wang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Ke Sai
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Ji Zhang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Chao Ke
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Fanfan Chen
- Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Yanchun Lv
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Xiangsong Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Zhongping Chen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
| |
Collapse
|
11
|
Cui M, Zorrilla-Veloz RI, Hu J, Guan B, Ma X. Diagnostic Accuracy of PET for Differentiating True Glioma Progression From Post Treatment-Related Changes: A Systematic Review and Meta-Analysis. Front Neurol 2021; 12:671867. [PMID: 34093419 PMCID: PMC8173157 DOI: 10.3389/fneur.2021.671867] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/24/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose: To evaluate the diagnostic accuracy of PET with different radiotracers and parameters in differentiating between true glioma progression (TPR) and post treatment-related change (PTRC). Methods: Studies on using PET to differentiate between TPR and PTRC were screened from the PubMed and Embase databases. By following the PRISMA checklist, the quality assessment of included studies was performed, the true positive and negative values (TP and TN), false positive and negative values (FP and FN), and general characteristics of all the included studies were extracted. Results of PET consistent with reference standard were defined as TP or TN. The pooled sensitivity (Sen), specificity (Spe), and hierarchical summary receiver operating characteristic curves (HSROC) were generated to evaluate the diagnostic accuracy. Results: The 33 included studies had 1,734 patients with 1,811 lesions suspected of glioma recurrence. Fifteen studies tested the accuracy of 18F-FET PET, 12 tested 18F-FDG PET, seven tested 11C-MET PET, and three tested 18F-DOPA PET. 18F-FET PET showed a pooled Sen and Spe of 0.88 (95% CI: 0.80, 0.93) and 0.78 (0.69, 0.85), respectively. In the subgroup analysis of FET-PET, diagnostic accuracy of high-grade gliomas (HGGs) was higher than that of mixed-grade gliomas (P interaction = 0.04). 18F-FDG PET showed a pooled Sen and Spe of 0.78 (95% CI: 0.71, 0.83) and 0.87 (0.80, 0.92), the Spe of the HGGs group was lower than that of the low-grade gliomas group (0.82 vs. 0.90, P = 0.02). 11C-MET PET had a pooled Sen and Spe of 0.92 (95% CI: 0.83, 0.96) and 0.78 (0.69, 0.86). 18F-DOPA PET had a pooled Sen and Spe of 0.85 (95% CI: 0.80, 0.89) and 0.70 (0.60, 0.79). FET-PET combined with MRI had a pooled Sen and Spe of 0.88 (95% CI: 0.78, 0.94) and 0.76 (0.57, 0.88). Multi-parameters analysis of FET-PET had pooled Sen and Spe values of 0.88 (95% CI: 0.81, 0.92) and 0.79 (0.63, 0.89). Conclusion: PET has a moderate diagnostic accuracy in differentiating between TPR and PTRC. The high Sen of amino acid PET and high Spe of FDG-PET suggest that the combination of commonly used FET-PET and FDG-PET may be more accurate and promising, especially for low-grade glioma.
Collapse
Affiliation(s)
- Meng Cui
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Neurosurgery, The First Medical Centre of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Rocío Isabel Zorrilla-Veloz
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- The University of Texas MD Anderson Cancer Centre UT Health Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- The University of Texas MD Anderson Cancer Centre UT Health Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Bing Guan
- Department of Health Economics, The First Medical Centre of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaodong Ma
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Neurosurgery, The First Medical Centre of Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
12
|
Strauss SB, Meng A, Ebani EJ, Chiang GC. Imaging Glioblastoma Posttreatment: Progression, Pseudoprogression, Pseudoresponse, Radiation Necrosis. Neuroimaging Clin N Am 2021; 31:103-120. [PMID: 33220823 DOI: 10.1016/j.nic.2020.09.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Radiographic monitoring of posttreatment glioblastoma is important for clinical trials and determining next steps in management. Evaluation for tumor progression is confounded by the presence of treatment-related radiographic changes, making a definitive determination less straight-forward. The purpose of this article was to describe imaging tools available for assessing treatment response in glioblastoma, as well as to highlight the definitions, pathophysiology, and imaging features typical of true progression, pseudoprogression, pseudoresponse, and radiation necrosis.
Collapse
Affiliation(s)
- Sara B Strauss
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Alicia Meng
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Edward J Ebani
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Gloria C Chiang
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA.
| |
Collapse
|
13
|
Le Fèvre C, Constans JM, Chambrelant I, Antoni D, Bund C, Leroy-Freschini B, Schott R, Cebula H, Noël G. Pseudoprogression versus true progression in glioblastoma patients: A multiapproach literature review. Part 2 - Radiological features and metric markers. Crit Rev Oncol Hematol 2021; 159:103230. [PMID: 33515701 DOI: 10.1016/j.critrevonc.2021.103230] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/10/2021] [Accepted: 01/16/2021] [Indexed: 12/28/2022] Open
Abstract
After chemoradiotherapy for glioblastoma, pseudoprogression can occur and must be distinguished from true progression to correctly manage glioblastoma treatment and follow-up. Conventional treatment response assessment is evaluated via conventional MRI (contrast-enhanced T1-weighted and T2/FLAIR), which is unreliable. The emergence of advanced MRI techniques, MR spectroscopy, and PET tracers has improved pseudoprogression diagnostic accuracy. This review presents a literature review of the different imaging techniques and potential imaging biomarkers to differentiate pseudoprogression from true progression.
Collapse
Affiliation(s)
- Clara Le Fèvre
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Jean-Marc Constans
- Department of Radiology, Amiens-Picardie University Hospital, 1 rond-point du Professeur Christian Cabrol, 80054, Amiens Cedex 1, France.
| | - Isabelle Chambrelant
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Delphine Antoni
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Caroline Bund
- Department of Nuclear Medicine, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Benjamin Leroy-Freschini
- Department of Nuclear Medicine, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Roland Schott
- Departement of Medical Oncology, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Hélène Cebula
- Departement of Neurosurgery, Hautepierre University Hospital, 1, avenue Molière, 67200, Strasbourg, France.
| | - Georges Noël
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| |
Collapse
|
14
|
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
|
15
|
Role of Radiological Intervention in Brain Tumor: A Meta-Analysis. Int Surg 2020. [DOI: 10.9738/intsurg-d-20-00014.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background
This meta-analysis highlights the diagnostic efficacy of computed tomography (CT), computed tomography angiography (CTA), magnetic resonance image (MRI), as well as magnetic resonance spectroscopy (MRS). This paper assesses the detection of the primary outcome comprising choline/creatine ratio, relative cerebral blood volume (rCBV), as well as choline/N-acetyl aspartate. Cochrane, Medline, ScienceDirect, Google Scholar, and EMBASE databases were searched for extracting the relevant studies.
Methods
A sample of 12 studies on radiologic assessment of brain tumors was selected.
Results
The evidence provides that the heterogeneity exists concerning the CBV of 311.623, I2 = 96.12%, with a significance value of P < 0.001. The pooled difference showed rCBV mean (as 2.18, 95% confidence interval = 0.85 to 3.50) substantially enhances lesion.
Conclusion
The study concluded that radiological interventions, particularly the combination of MRS and MRI, help in the brain patient's precise diagnosis and treatment.
Collapse
|
16
|
Quartuccio N, Laudicella R, Vento A, Pignata S, Mattoli MV, Filice R, Comis AD, Arnone A, Baldari S, Cabria M, Cistaro A. The Additional Value of 18F-FDG PET and MRI in Patients with Glioma: A Review of the Literature from 2015 to 2020. Diagnostics (Basel) 2020; 10:357. [PMID: 32486075 PMCID: PMC7345880 DOI: 10.3390/diagnostics10060357] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/15/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023] Open
Abstract
AIM Beyond brain computed tomography (CT) scan, Magnetic Resonance Imaging (MRI) and Positron Emission Tomography (PET) hold paramount importance in neuro-oncology. The aim of this narrative review is to discuss the literature from 2015 to 2020, showing advantages or complementary information of fluorine-18 fluorodeoxyglucose (18F-FDG) PET imaging to the anatomical and functional data offered by MRI in patients with glioma. METHODS A comprehensive Pubmed/MEDLINE literature search was performed to retrieve original studies, with a minimum of 10 glioma patients, published from 2015 until the end of April 2020, on the use of 18F-FDG PET in conjunction with MRI. RESULTS Twenty-two articles were selected. Combined use of the two modalities improves the accuracy in predicting prognosis, planning treatments, and evaluating recurrence. CONCLUSION According to the recent literature, 18F-FDG PET provides different and complementary information to MRI and may enhance performance in the whole management of gliomas. Therefore, integrated PET/MRI may be particularly useful in gliomas, since it could provide accurate morphological and metabolic information in one-shoot examination and improve the diagnostic value compared to each of procedures.
Collapse
Affiliation(s)
- Natale Quartuccio
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico, Di Cristina e Benfratelli, 90127 Palermo, Italy; (N.Q.); (A.A.)
- Committee of AIMN Pediatric Study Group, 20159 Milan, Italy
| | - Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (R.L.); (A.V.); (S.P.); (R.F.); (A.D.C.); (S.B.)
- AIMN -Italian Association of Nuclear Medicine- Young Members Working Group, 20159 Milan, Italy
| | - Antonio Vento
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (R.L.); (A.V.); (S.P.); (R.F.); (A.D.C.); (S.B.)
| | - Salvatore Pignata
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (R.L.); (A.V.); (S.P.); (R.F.); (A.D.C.); (S.B.)
| | - Maria Vittoria Mattoli
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University, 66100 Chieti, Italy;
| | - Rossella Filice
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (R.L.); (A.V.); (S.P.); (R.F.); (A.D.C.); (S.B.)
| | - Alessio Danilo Comis
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (R.L.); (A.V.); (S.P.); (R.F.); (A.D.C.); (S.B.)
| | - Annachiara Arnone
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico, Di Cristina e Benfratelli, 90127 Palermo, Italy; (N.Q.); (A.A.)
| | - Sergio Baldari
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (R.L.); (A.V.); (S.P.); (R.F.); (A.D.C.); (S.B.)
| | - Manlio Cabria
- Nuclear Medicine Department, Ente Ospedaliero Ospedali Galliera, Italy, Mura delle Cappuccine, 14, 16128 Genova, Italy;
| | - Angelina Cistaro
- Nuclear Medicine Department, Ente Ospedaliero Ospedali Galliera, Italy, Mura delle Cappuccine, 14, 16128 Genova, Italy;
- Committee of AIMN Neuroimaging Study Group, 20159 Milan, Italy
- Coordinator of AIMN Paediatric Study Group, 20159 Milan, Italy
| |
Collapse
|
17
|
Lv Y, Zhou J, Lv X, Tian L, He H, Liu Z, Wu Y, Han L, Sun M, Yang Y, Guo C, Li C, Zhang R, Xie C, Chen Y, Chen Z. Dual-energy spectral CT quantitative parameters for the differentiation of Glioma recurrence from treatment-related changes: a preliminary study. BMC Med Imaging 2020; 20:5. [PMID: 31948400 PMCID: PMC6966828 DOI: 10.1186/s12880-019-0406-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 12/24/2019] [Indexed: 11/18/2022] Open
Abstract
Background Differentiating glioma recurrence from treatment-related changes can be challenging on conventional imaging. We evaluated the efficacy of quantitative parameters measured by dual-energy spectral computed tomographic (CT) for this differentiation. Methods Twenty-eight patients were examined by dual-energy spectral CT. The effective and normalized atomic number (Zeff and Zeff-N, respectively); spectral Hounsfield unit curve (λHU) slope; and iodine and normalized iodine concentration (IC and ICN, respectively) in the post-treatment enhanced areas were calculated. Pathological results or clinicoradiologic follow-up of ≥2 months were used for final diagnosis. Nonparametric and t-tests were used to compare quantitative parameters between glioma recurrence and treatment-related changes. Sensitivity, specificity, positive and negative predictive values (PPV and NPV, respectively), and accuracy were calculated using receiver operating characteristic (ROC) curves. Predictive probabilities were used to generate ROC curves to determine the diagnostic value. Results Examination of pre-contrast λHU, Zeff, Zeff-N, IC, ICN, and venous phase ICN showed no significant differences in quantitative parameters (P > 0.05). Venous phase λHU, Zeff, Zeff-N, and IC in glioma recurrence were higher than in treatment-related changes (P < 0.001). The optimal venous phase threshold was 1.03, 7.75, 1.04, and 2.85 mg/cm3, achieving 66.7, 91.7, 83.3, and 91.7% sensitivity; 100.0, 77.8, 88.9, and 77.8% specificity; 100.0, 73.3, 83.3, and 73.3% PPV; 81.8, 93.3, 88.9, and 93.3% NPV; and 86.7, 83.3, 86.7, and 83.3% accuracy, respectively. The respective areas under the curve (AUCs) were 0.912, 0.912, 0.931, and 0.910 in glioma recurrence and treatment-related changes. Conclusions Glioma recurrence could be potentially differentiated from treatment-related changes based on quantitative values measured by dual-energy spectral CT imaging.
Collapse
Affiliation(s)
- Yanchun Lv
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Jian Zhou
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Xiaofei Lv
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Li Tian
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Haoqiang He
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhigang Liu
- Department of head and neck oncology, Phase 1 clinical trial ward, The cancer center of the fifth affiliated hospital of Sun Yat-sen University, Zhuhai, 519001, China
| | - Yi Wu
- Department of Radiology, Shantou Central Hospital, No.114 Waima Road, Shantou, 515041, Guangdong, China
| | - Lujun Han
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Meili Sun
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yadi Yang
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Chengcheng Guo
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Cong Li
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Rong Zhang
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Chuanmiao Xie
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yinsheng Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China.
| | - Zhongping Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
18
|
Moreau A, Febvey O, Mognetti T, Frappaz D, Kryza D. Contribution of Different Positron Emission Tomography Tracers in Glioma Management: Focus on Glioblastoma. Front Oncol 2019; 9:1134. [PMID: 31737567 PMCID: PMC6839136 DOI: 10.3389/fonc.2019.01134] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022] Open
Abstract
Although rare, glioblastomas account for the majority of primary brain lesions, with a dreadful prognosis. Magnetic resonance imaging (MRI) is currently the imaging method providing the higher resolution. However, it does not always succeed in distinguishing recurrences from non-specific temozolomide, have been shown to improve -related changes caused by the combination of radiotherapy, chemotherapy, and targeted therapy, also called pseudoprogression. Strenuous attempts to overcome this issue is highly required for these patients with a short life expectancy for both ethical and economic reasons. Additional reliable information may be obtained from positron emission tomography (PET) imaging. The development of this technique, along with the emerging of new classes of tracers, can help in the diagnosis, prognosis, and assessment of therapies. We reviewed the current data about the commonly used tracers, such as 18F-fluorodeoxyglucose (18F-FDG) and radiolabeled amino acids, as well as different PET tracers recently investigated, to report their strengths, limitations, and relevance in glioblastoma management.
Collapse
Affiliation(s)
| | | | | | | | - David Kryza
- UNIV Lyon - Université Claude Bernard Lyon 1, LAGEPP UMR 5007 CNRS Villeurbanne, Villeurbanne, France
- Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
19
|
The Molecular Effects of Ionizing Radiations on Brain Cells: Radiation Necrosis vs. Tumor Recurrence. Diagnostics (Basel) 2019; 9:diagnostics9040127. [PMID: 31554255 PMCID: PMC6963489 DOI: 10.3390/diagnostics9040127] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/13/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022] Open
Abstract
The central nervous system (CNS) is generally resistant to the effects of radiation, but higher doses, such as those related to radiation therapy, can cause both acute and long-term brain damage. The most important results is a decline in cognitive function that follows, in most cases, cerebral radionecrosis. The essence of radio-induced brain damage is multifactorial, being linked to total administered dose, dose per fraction, tumor volume, duration of irradiation and dependent on complex interactions between multiple brain cell types. Cognitive impairment has been described following brain radiotherapy, but the mechanisms leading to this adverse event remain mostly unknown. In the event of a brain tumor, on follow-up radiological imaging often cannot clearly distinguish between recurrence and necrosis, while, especially in patients that underwent radiation therapy (RT) post-surgery, positron emission tomography (PET) functional imaging, is able to differentiate tumors from reactive phenomena. More recently, efforts have been done to combine both morphological and functional data in a single exam and acquisition thanks to the co-registration of PET/MRI. The future of PET imaging to differentiate between radionecrosis and tumor recurrence could be represented by a third-generation PET tracer already used to reveal the spatial extent of brain inflammation. The aim of the following review is to analyze the effect of ionizing radiations on CNS with specific regard to effect of radiotherapy, focusing the attention on the mechanism underling the radionecrosis and the brain damage, and show the role of nuclear medicine techniques to distinguish necrosis from recurrence and to early detect of cognitive decline after treatment.
Collapse
|
20
|
de Zwart PL, van Dijken BR, Holtman GA, Stormezand GN, Dierckx RA, Jan van Laar P, van der Hoorn A. Diagnostic Accuracy of PET Tracers for the Differentiation of Tumor Progression from Treatment-Related Changes in High-Grade Glioma: A Systematic Review and Metaanalysis. J Nucl Med 2019; 61:498-504. [DOI: 10.2967/jnumed.119.233809] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
|
21
|
Strauss SB, Meng A, Ebani EJ, Chiang GC. Imaging Glioblastoma Posttreatment: Progression, Pseudoprogression, Pseudoresponse, Radiation Necrosis. Radiol Clin North Am 2019; 57:1199-1216. [PMID: 31582045 DOI: 10.1016/j.rcl.2019.07.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Radiographic monitoring of posttreatment glioblastoma is important for clinical trials and determining next steps in management. Evaluation for tumor progression is confounded by the presence of treatment-related radiographic changes, making a definitive determination less straight-forward. The purpose of this article was to describe imaging tools available for assessing treatment response in glioblastoma, as well as to highlight the definitions, pathophysiology, and imaging features typical of true progression, pseudoprogression, pseudoresponse, and radiation necrosis.
Collapse
Affiliation(s)
- Sara B Strauss
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Alicia Meng
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Edward J Ebani
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Gloria C Chiang
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA.
| |
Collapse
|
22
|
Zikou A, Sioka C, Alexiou GA, Fotopoulos A, Voulgaris S, Argyropoulou MI. Radiation Necrosis, Pseudoprogression, Pseudoresponse, and Tumor Recurrence: Imaging Challenges for the Evaluation of Treated Gliomas. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:6828396. [PMID: 30627060 PMCID: PMC6305027 DOI: 10.1155/2018/6828396] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/15/2018] [Indexed: 01/16/2023]
Abstract
Glioblastoma (GBM) is the most common primary malignant type of brain neoplasm in adults and carries a dismal prognosis. The current standard of care for GBM is surgical excision followed by radiation therapy (RT) with concurrent and adjuvant temozolomide-based chemotherapy (TMZ) by six additional cycles. In addition, antiangiogenic therapy with an antivascular endothelial growth factor (VEGF) agent has been used for recurrent glioblastoma. Over the last years, new posttreatment entities such as pseudoprogression and pseudoresponse have been recognized, apart from radiation necrosis. This review article focuses on the role of different imaging techniques such as conventional magnetic resonance imaging (MRI), diffusion-weighted imaging (DWI), diffusion tensor imaging (DTI), dynamic contrast enhancement (DCE-MRI) and dynamic susceptibility contrast (DSE-MRI) perfusion, magnetic resonance spectroscopy (MRS), and PET/SPECT in differentiation of such treatment-related changes from tumor recurrence.
Collapse
Affiliation(s)
- Anastasia Zikou
- Department of Radiology, University Hospital of Ioannina, Ioannina, Greece
| | - Chrissa Sioka
- Department of Nuclear Medicine, University Hospital of Ioannina, Ioannina, Greece
| | - George A. Alexiou
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| | - Andreas Fotopoulos
- Department of Nuclear Medicine, University Hospital of Ioannina, Ioannina, Greece
| | - Spyridon Voulgaris
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| | | |
Collapse
|
23
|
Serial FLT PET imaging to discriminate between true progression and pseudoprogression in patients with newly diagnosed glioblastoma: a long-term follow-up study. Eur J Nucl Med Mol Imaging 2018; 45:2404-2412. [PMID: 30032322 PMCID: PMC6208814 DOI: 10.1007/s00259-018-4090-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/09/2018] [Indexed: 12/23/2022]
Abstract
Purpose Response evaluation in patients with glioblastoma after chemoradiotherapy is challenging due to progressive, contrast-enhancing lesions on MRI that do not reflect true tumour progression. In this study, we prospectively evaluated the ability of the PET tracer 18F-fluorothymidine (FLT), a tracer reflecting proliferative activity, to discriminate between true progression and pseudoprogression in newly diagnosed glioblastoma patients treated with chemoradiotherapy. Methods FLT PET and MRI scans were performed before and 4 weeks after chemoradiotherapy. MRI scans were also performed after three cycles of adjuvant temozolomide. Pseudoprogression was defined as progressive disease on MRI after chemoradiotherapy with stabilisation or reduction of contrast-enhanced lesions after three cycles of temozolomide, and was compared with the disease course during long-term follow-up. Changes in maximum standardized uptake value (SUVmax) and tumour-to-normal uptake ratios were calculated for FLT and are presented as the mean SUVmax for multiple lesions. Results Between 2009 and 2012, 30 patients were included. Of 24 evaluable patients, 7 showed pseudoprogression and 7 had true progression as defined by MRI response. FLT PET parameters did not significantly differ between patients with true progression and pseudoprogression defined by MRI. The correlation between change in SUVmax and survival (p = 0.059) almost reached the standard level of statistical significance. Lower baseline FLT PET uptake was significantly correlated with improved survival (p = 0.022). Conclusion Baseline FLT uptake appears to be predictive of overall survival. Furthermore, changes in SUVmax over time showed a tendency to be associated with improved survival. However, further studies are necessary to investigate the ability of FLT PET imaging to discriminate between true progression and pseudoprogression in patients with glioblastoma.
Collapse
|
24
|
Donabedian PL, Kossatz S, Engelbach JA, Jannetti SA, Carney B, Young RJ, Weber WA, Garbow JR, Reiner T. Discriminating radiation injury from recurrent tumor with [ 18F]PARPi and amino acid PET in mouse models. EJNMMI Res 2018; 8:59. [PMID: 29974335 PMCID: PMC6031550 DOI: 10.1186/s13550-018-0399-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/15/2018] [Indexed: 01/20/2023] Open
Abstract
Background Radiation injury can be indistinguishable from recurrent tumor on standard imaging. Current protocols for this differential diagnosis require one or more follow-up imaging studies, long dynamic acquisitions, or complex image post-processing; despite much research, the inability to confidently distinguish between these two entities continues to pose a significant dilemma for the treating clinician. Using mouse models of both glioblastoma and radiation necrosis, we tested the potential of poly(ADP-ribose) polymerase (PARP)-targeted PET imaging with [18F]PARPi to better discriminate radiation injury from tumor. Results In mice with experimental radiation necrosis, lesion uptake on [18F]PARPi-PET was similar to contralateral uptake (1.02 ± 0.26 lesion/contralateral %IA/ccmax ratio), while [18F]FET-PET clearly delineated the contrast-enhancing region on MR (2.12 ± 0.16 lesion/contralateral %IA/ccmax ratio). In mice with focal intracranial U251 xenografts, tumor visualization on PARPi-PET was superior to FET-PET, and lesion-to-contralateral activity ratios (max/max, p = 0.034) were higher on PARPi-PET than on FET-PET. Conclusions A murine model of radiation necrosis does not demonstrate [18F]PARPi avidity, and [18F]PARPi-PET is better than [18F]FET-PET in distinguishing radiation injury from brain tumor. [18F]PARPi-PET can be used for discrimination between recurrent tumor and radiation injury within a single, static imaging session, which may be of value to resolve a common dilemma in neuro-oncology. Electronic supplementary material The online version of this article (10.1186/s13550-018-0399-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Patrick L Donabedian
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - John A Engelbach
- Department of Radiology, Washington University, St. Louis, MO, USA
| | - Stephen A Jannetti
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Department of Chemistry, Hunter College of the City University of New York, New York, NY, USA.,Ph.D. Program in Biochemistry, Graduate Center of the City University of New York, New York, NY, USA
| | - Brandon Carney
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Department of Chemistry, Hunter College of the City University of New York, New York, NY, USA.,Ph.D. Program in Chemistry, Graduate Center of the City University of New York, New York, NY, USA
| | - Robert J Young
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Wolfgang A Weber
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Department of Nuclear Medicine, Technical University Munich, Munich, Germany
| | - Joel R Garbow
- Department of Radiology, Washington University, St. Louis, MO, USA.,Alvin J. Siteman Cancer Center, Washington University, St. Louis, MO, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA. .,Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
25
|
Buder-Bakhaya K, Hassel JC. Biomarkers for Clinical Benefit of Immune Checkpoint Inhibitor Treatment-A Review From the Melanoma Perspective and Beyond. Front Immunol 2018; 9:1474. [PMID: 30002656 PMCID: PMC6031714 DOI: 10.3389/fimmu.2018.01474] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/13/2018] [Indexed: 12/26/2022] Open
Abstract
Background Immune checkpoint inhibition (ICI) with anti-CTLA-4 and/or anti-PD-1 antibodies is standard treatment for metastatic melanoma. Anti-PD-1 (pembrolizumab, nivolumab) and anti-PD-L1 antibodies (atezolizumab, durvalumab, and avelumab) have been approved for treatment of several other advanced malignancies, including non-small-cell lung cancer (NSCLC); renal cell, and urothelial carcinoma; head and neck cancer; gastric, hepatocellular, and Merkel-cell carcinoma; and classical Hodgkin lymphoma. In some of these malignancies approval was based on the detection of biomarkers such as PD-L1 expression or high microsatellite instability. Methods We review the current status of prognostic and predictive biomarkers used in ICI for melanoma and other malignancies. We include clinical, tissue, blood, and stool biomarkers, as well as imaging biomarkers. Results Several biomarkers have been studied in ICI for metastatic melanoma. In clinical practice, pre-treatment tumor burden measured by means of imaging and serum lactate dehydrogenase level is already being used to estimate the likelihood of effective ICI treatment. In peripheral blood, the number of different immune cell types, such as lymphocytes, neutrophils, and eosinophils, as well as different soluble factors, have been correlated with clinical outcome. For intra-tumoral biomarkers, expression of the PD-1 ligand PD-L1 has been found to be of some predictive value for anti-PD-1-directed therapy for NSCLC and melanoma. A high mutational load, particularly when accompanied by neoantigens, seems to facilitate immune response and correlates with patient survival for all entities treated by use of ICI. Tumor microenvironment also seems to be of major importance. Interestingly, even the gut microbiome has been found to correlate with response to ICI, most likely through immuno-stimulatory effects of distinct bacteria. New imaging biomarkers, e.g., for PET, and magnetic resonance imaging are also being investigated, and results suggest they will make early prediction of patient response possible. Conclusion Several promising results are available regarding possible biomarkers for response to ICI, which need to be validated in large clinical trials. A better understanding of how ICI works will enable the development of biomarkers that can predict the response of individual patients.
Collapse
Affiliation(s)
- Kristina Buder-Bakhaya
- Section of Dermatooncology, Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica C Hassel
- Section of Dermatooncology, Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
26
|
Parapharyngeal neuroglial heterotopia appearing as high uptake on 18F-FDG PET: case report and literature review of radiographical findings. Acta Neurochir (Wien) 2018; 160:801-809. [PMID: 29197937 DOI: 10.1007/s00701-017-3403-x] [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: 08/30/2017] [Accepted: 11/13/2017] [Indexed: 10/18/2022]
Abstract
Parapharyngeal neuroglial heterotopia is a rare entity, and the specific radiographical findings are unclear. We present a case of parapharyngeal neuroglial heterotopia examined with proton magnetic resonance spectroscopy (1H-MRS) and 18F-fluorodesoxyglucose positron emission tomography (18F-FDG PET). Our neonate patient presented with neck mass and polyhydramnios during gestation. Computed tomography and magnetic resonance imaging demonstrated the morphological characteristics, but failed to establish the diagnosis. 1H-MRS showed a non-malignant pattern, but 18F-FDG PET demonstrated high glucose metabolism. Complete resection was achieved and the histopathological diagnosis was neuroglial heterotopia. Assessment of biological activity may be useful for both preoperative diagnosis and postoperative evaluation of residual lesions.
Collapse
|
27
|
Nguyen NC, Yee MK, Tuchayi AM, Kirkwood JM, Tawbi H, Mountz JM. Targeted Therapy and Immunotherapy Response Assessment with F-18 Fluorothymidine Positron-Emission Tomography/Magnetic Resonance Imaging in Melanoma Brain Metastasis: A Pilot Study. Front Oncol 2018; 8:18. [PMID: 29520339 PMCID: PMC5827168 DOI: 10.3389/fonc.2018.00018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/17/2018] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION This pilot study aimed at exploring the utility of the proliferation tracer F-18 fluorothymidine (FLT) and positron-emission tomography (PET)/magnetic resonance imaging (MRI) (FLT-PET/MRI) for early treatment monitoring in patients with melanoma brain metastasis (MBM) who undergo targeted therapy or immunotherapy. MATERIAL AND METHODS Patients with newly diagnosed MBM underwent baseline and follow-up FLT-PET/MRI scans at 3-4 weeks of targeted therapy or immunotherapy. Up to six measurable brain lesions ≥1.0 cm per subject, as identified on T1-weighted post-gadolinium images, were included for quantitative analyses. The maximum SUV of each lesion was divided by the mean SUV of the pons to obtain the SUV ratio (SUVR). RESULTS Five enrolled subjects underwent the baseline FLT-PET/MRI study in which the MBM showed a median size of 1.7 cm (range 1.0-2.9) and increased metabolic activity with SUVR of 9.9 (range 3.2-18.4). However, only two subjects (cases #1 and #2) returned for a follow-up scan. At baseline, a total of 22 lesions were analyzed in all five subjects, which showed a median size of 1.7 cm (range 1.0-2.9) and median SUVR of 9.9 (range 3.2-18.4). At follow-up, case #1 was a 55-year-old man who received targeted BRAF inhibitor and MEK inhibitor therapy with dabrafenib and trametinib. Fused PET/MRI data of six measured lesions demonstrated a significant reduction in MBM proliferative activity (median -68%; range -38 to -77%) and size (median -23%; range -4 to -55%) at three weeks of therapy. Nevertheless, the subject eventually progressed and died 13 months after therapy initiation. Case #2 was a 36-year-old man who received immunotherapy with nivolumab and ipilimumab. The five measured MBM lesions showed a mixed response at both proliferative and morphologic imaging at 1-month follow-up. Some lesions demonstrated interval decrease while others interval increase in proliferative activity with a median -44% (range -77 to +68%). On MRI, the size change was +7% (range -64 to +50%). The therapy was switched to dabrafenib and trametinib, which led to a partial response. The patient is still alive 16 months following therapy initiation. CONCLUSION The five cases presented show the potential benefit of hybrid FLT-PET/MRI for the diagnosis of MBM and treatment monitoring of targeted therapy and immunotherapy. However, further studies are required to assess their complementary role in distinguishing true progression from pseudoprogression.
Collapse
Affiliation(s)
- Nghi C. Nguyen
- Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Melissa K. Yee
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Abuzar M. Tuchayi
- Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - John M. Kirkwood
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Hussein Tawbi
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James M. Mountz
- Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
28
|
Li H, Deng L, Bai HX, Sun J, Cao Y, Tao Y, States LJ, Farwell MD, Zhang P, Xiao B, Yang L. Diagnostic Accuracy of Amino Acid and FDG-PET in Differentiating Brain Metastasis Recurrence from Radionecrosis after Radiotherapy: A Systematic Review and Meta-Analysis. AJNR Am J Neuroradiol 2018; 39:280-288. [PMID: 29242363 DOI: 10.3174/ajnr.a5472] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/19/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Current studies that analyze the usefulness of amino acid and FDG-PET in distinguishing brain metastasis recurrence and radionecrosis after radiation therapy are limited by small cohort size. PURPOSE Our aim was to assess the diagnostic accuracy of amino acid and FDG-PET in differentiating brain metastasis recurrence from radionecrosis after radiation therapy. DATA SOURCES Studies were retrieved from PubMed, Embase, and the Cochrane Library. STUDY SELECTION Fifteen studies were included from the literature. Each study used PET to differentiate radiation necrosis from tumor recurrence in contrast-enhancing lesions on follow-up brain MR imaging after treating brain metastasis with radiation therapy. DATA ANALYSIS Data were analyzed with a bivariate random-effects model. Sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio were pooled, and a summary receiver operating characteristic curve was fit to the data. DATA SYNTHESIS The overall pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio of PET were 0.85, 0.88, 7.0, 0.17, and 40, respectively. The area under the receiver operating characteristic curve was 0.93. On subgroup analysis of different tracers, amino acid and FDG-PET had similar diagnostic accuracy. Meta-regression analysis demonstrated that the method of quantification based on patient, lesion, or PET scan (based on lesion versus not, P = .07) contributed to the heterogeneity. LIMITATIONS Our study was limited by small sample size, and 60% of the included studies were of retrospective design. CONCLUSIONS Amino acid and FDG-PET had good diagnostic accuracy in differentiating brain metastasis recurrence from radionecrosis after radiation therapy.
Collapse
Affiliation(s)
- H Li
- From the Department of Neurology (H.L., L.D., L.Y.), Second Xiangya Hospital of Central South University, Changsha, Hunan Province, People's Republic of China
| | - L Deng
- From the Department of Neurology (H.L., L.D., L.Y.), Second Xiangya Hospital of Central South University, Changsha, Hunan Province, People's Republic of China
| | - H X Bai
- Departments of Radiology (H.X.B., J.S., M.D.F.)
| | - J Sun
- Departments of Radiology (H.X.B., J.S., M.D.F.)
| | - Y Cao
- Cancer Research Institute (Y.C., Y.T.), Central South University, Changsha, Hunan Province, People's Republic of China
| | - Y Tao
- Cancer Research Institute (Y.C., Y.T.), Central South University, Changsha, Hunan Province, People's Republic of China
| | - L J States
- Department of Radiology (L.J.S.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - M D Farwell
- Departments of Radiology (H.X.B., J.S., M.D.F.)
| | - P Zhang
- Pathology (P.Z.), Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - B Xiao
- Department of Neurology (B.X.), Xiangya Hospital of Central South University, Changsha, Hunan Province, People's Republic of China.
| | - L Yang
- From the Department of Neurology (H.L., L.D., L.Y.), Second Xiangya Hospital of Central South University, Changsha, Hunan Province, People's Republic of China
| |
Collapse
|
29
|
Deuschl C, Kirchner J, Poeppel TD, Schaarschmidt B, Kebir S, El Hindy N, Hense J, Quick HH, Glas M, Herrmann K, Umutlu L, Moenninghoff C, Radbruch A, Forsting M, Schlamann M. 11C-MET PET/MRI for detection of recurrent glioma. Eur J Nucl Med Mol Imaging 2017; 45:593-601. [PMID: 29282517 DOI: 10.1007/s00259-017-3916-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 12/11/2017] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Radiological assessment of brain tumors is widely based on the Radiology Assessment of Neuro-Oncology (RANO) criteria that consider non-specific T1 and T2 weighted images. Limitation of the RANO criteria is that they do not include metabolic imaging techniques that have been reported to be helpful to differentiate treatment related changes from true tumor progression. In the current study, we assessed if the combined use of MRI and PET with hybrid 11C-MET PET/MRI can improve diagnostic accuracy and diagnostic confidence of the readers to differentiate treatment related changes from true progression in recurrent glioma. METHODS Fifty consecutive patients with histopathologically proven glioma were prospectively enrolled for a hybrid 11C-MET PET/MRI to differentiate recurrent glioma from treatment induced changes. Sole MRI data were analyzed based on RANO. Sole PET data and in a third evaluation hybrid 11C-MET-PET/MRI data were assessed for metabolic respectively metabolic and morphologic glioma recurrence. Diagnostic performance and diagnostic confidence of the reader were calculated for the different modalities, and the McNemar test and Mann-Whitney U Test were applied for statistical analysis. RESULTS Hybrid 11C-MET PET/MRI was successfully performed in all 50 patients. Glioma recurrence was diagnosed in 35 of the 50 patients (70%). Sensitivity and specificity were calculated for MRI (86.11% and 71.43%), for 11C-MET PET (96.77% and 73.68%), and for hybrid 11C-MET-PET/MRI (97.14% and 93.33%). For diagnostic accuracy hybrid 11C-MET-PET/MRI (96%) showed significantly higher values than MRI alone (82%), whereas no significant difference was found for 11C-MET PET (88%). Furthermore, by rating on a five-point Likert scale significantly higher scores were found for diagnostic confidence when comparing 11C-MET PET/MRI (4.26 ± 0,777) to either PET alone (3.44 ± 0.705) or MRI alone (3.56 ± 0.733). CONCLUSION This feasibility study showed that hybrid PET/MRI might strengthen RANO classification by adding metabolic information to conventional MRI information. Future studies should evaluate the clinical utility of the combined use of 11C-MET PET/MRI in larger patient cohorts.
Collapse
Affiliation(s)
- C Deuschl
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany.
- Erwin L. Hahn Institute for Magnetic Resonance Imaging, University of Duisburg-Essen, Essen, Germany.
| | - J Kirchner
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Duesseldorf, Duesseldorf, Germany
| | - T D Poeppel
- Clinic for Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - B Schaarschmidt
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Duesseldorf, Duesseldorf, Germany
| | - S Kebir
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, Essen, Germany
| | - N El Hindy
- Department of Neurosurgery, University Hospital Essen, Essen, Germany
| | - J Hense
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - H H Quick
- Erwin L. Hahn Institute for Magnetic Resonance Imaging, University of Duisburg-Essen, Essen, Germany
- High Field and Hybrid MR Imaging, University Hospital Essen, Essen, Germany
| | - M Glas
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, Essen, Germany
| | - K Herrmann
- Clinic for Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - L Umutlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - C Moenninghoff
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - A Radbruch
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - M Forsting
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - M Schlamann
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
30
|
Nikaki A, Angelidis G, Efthimiadou R, Tsougos I, Valotassiou V, Fountas K, Prasopoulos V, Georgoulias P. 18F-fluorothymidine PET imaging in gliomas: an update. Ann Nucl Med 2017; 31:495-505. [PMID: 28612247 PMCID: PMC5517561 DOI: 10.1007/s12149-017-1183-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/31/2017] [Indexed: 01/18/2023]
Abstract
Brain neoplasms constitute a group of tumors with discrete differentiation grades, and therefore, course of disease and prognosis. Magnetic resonance imaging (MRI) remains the gold standard method for the investigation of central nervous system tumors. However, MRI suffers certain limitations, especially if radiation therapy or chemotherapy has been previously applied. On the other hand, given the development of newer radiopharmaceuticals, positron emission tomography (PET) aims to a better investigation of brain tumors, assisting in the clinical management of the patients. In the present review, the potential contribution of radiolabeled fluorothymidine (FLT) imaging for the evaluation of brain tumors will be discussed. In particular, we will present the role of FLT-PET imaging in the depiction of well and poorly differentiated lesions, the assessment of patient prognosis and treatment response, and the recognition of disease recurrence. Moreover, related semi-quantitative and kinetic parameters will be discussed.
Collapse
Affiliation(s)
- Alexandra Nikaki
- Department of Clinical Physiology, KHSHP, 20 Ahvenistontie Str., 13530, Hämeenlinna, Finland.,Department of Nuclear Medicine, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - George Angelidis
- Department of Nuclear Medicine, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - Roxani Efthimiadou
- PET/CT Department, Hygeia Hospital, 4 Erythrou Stavrou Str., 15123, Athens, Greece
| | - Ioannis Tsougos
- Department of Nuclear Medicine, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - Varvara Valotassiou
- Department of Nuclear Medicine, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - Konstantinos Fountas
- Department of Neurosurgery, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - Vasileios Prasopoulos
- PET/CT Department, Hygeia Hospital, 4 Erythrou Stavrou Str., 15123, Athens, Greece.,Department of Nuclear Medicine, Hygeia Hospital, 4 Erythrou Stavrou Str., 15123, Athens, Greece
| | - Panagiotis Georgoulias
- Department of Nuclear Medicine, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece.
| |
Collapse
|
31
|
Yano H, Shinoda J, Iwama T. Clinical Utility of Positron Emission Tomography in Patients with Malignant Glioma. Neurol Med Chir (Tokyo) 2017; 57:312-320. [PMID: 28458384 PMCID: PMC5566704 DOI: 10.2176/nmc.ra.2016-0312] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Positron emission tomography (PET) is being increasingly utilized for the management of brain tumors. Herein, we primarily review our previous studies on the use of PET in glioma that utilize three types of tracers: 11C-methionine (MET), 11C-choline, and 18F-fluorodeoxyglucose. These studies included aspects such as tumor behavior, diagnosis, grade of malignancy, spread and invasion, viability, and genetic deletions; moreover, they also evaluated PET as a tool for planning radiation therapy (RT) and determining its outcome. MET-PET in particular is considered to be the most informative for diagnosis and therapeutic decision-making for glioma patients; it is therefore considered crucial for brain tumor therapy. MET-PET is expected to be widely used for brain tumor patients going forward.
Collapse
Affiliation(s)
- Hirohito Yano
- Department of Neurosurgery, Gifu University Graduate School of Medicine
| | - Jun Shinoda
- Chubu Medical Center for Prolonged Traumatic Brain Dysfunction, Department of Neurosurgery, Kizawa Memorial Hospital
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine
| |
Collapse
|
32
|
Furtado AD, Ceschin R, Blüml S, Mason G, Jakacki RI, Okada H, Pollack IF, Panigrahy A. Neuroimaging of Peptide-based Vaccine Therapy in Pediatric Brain Tumors: Initial Experience. Neuroimaging Clin N Am 2017; 27:155-166. [PMID: 27889021 DOI: 10.1016/j.nic.2016.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The potential benefits of peptide-based immunotherapy for pediatric brain tumors are under investigation. Treatment-related heterogeneity has resulted in radiographic challenges, including pseudoprogression. Conventional MR imaging has limitations in assessment of different forms of treatment-related heterogeneity, particularly regarding distinguishing true tumor progression from efficacious treatment responses. Advanced neuroimaging techniques, including diffusion magnetic resonance (MR), perfusion MR, and MR spectroscopy, may add value in the assessment of treatment-related heterogeneity. Observations suggest that recent delineation of specific response criteria for immunotherapy of adult brain tumors is likely relevant to the pediatric population and further validation in multicenter pediatric brain tumor peptide-based vaccine studies is warranted.
Collapse
Affiliation(s)
- Andre D Furtado
- Department of Radiology, University of Pittsburgh, 3600 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Rafael Ceschin
- Department of Radiology, University of Pittsburgh, 3600 Forbes Avenue, Pittsburgh, PA 15213, USA; Department of Bioinformatics, University of Pittsburgh, 5607 Baum Boulevard, Suite 500, Pittsburgh, PA 15206, USA
| | - Stefan Blüml
- Department of Radiology, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90007, USA
| | - Gary Mason
- Department of Pediatrics, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Regina I Jakacki
- Department of Pediatrics, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Hideho Okada
- Department of Neurosurgery, University of California, San Francisco, 505 Parnassus Avenue, M-779, San Francisco, CA 94143, USA
| | - Ian F Pollack
- Department of Neurosurgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Children's Hospital of Pittsburgh, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Ashok Panigrahy
- Department of Radiology, University of Pittsburgh, 3600 Forbes Avenue, Pittsburgh, PA 15213, USA.
| |
Collapse
|
33
|
Jena A, Taneja S, Jha A, Damesha NK, Negi P, Jadhav GK, Verma SM, Sogani SK. Multiparametric Evaluation in Differentiating Glioma Recurrence from Treatment-Induced Necrosis Using Simultaneous 18F-FDG-PET/MRI: A Single-Institution Retrospective Study. AJNR Am J Neuroradiol 2017; 38:899-907. [PMID: 28341716 DOI: 10.3174/ajnr.a5124] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/21/2016] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Differentiating glioma recurrence from treatment-induced necrosis can be a challenge on conventional imaging. This study aimed to assess the diagnostic performance of each functional MR imaging and PET parameter derived by using simultaneous FDG-PET/MR imaging individually and in combination in the evaluation of suspected glioma recurrence. MATERIALS AND METHODS Thirty-five treated glioma patients with 41 enhancing lesions (World Health Organization grade II = 9, III = 13, IV = 19) on MR imaging after an operation followed by radiation therapy and/or chemotherapy formed part of this study. Using PET/MR imaging, we calculated the normalized mean relative CBV, mean ADC, Cho/Cr, and maximum and mean target-to-background ratios. Statistical analysis was performed to determine the diagnostic performance of each parameter by receiver operating characteristic analysis individually and in combination with multivariate receiver operating characteristic analysis for the detection of glioma recurrence. Histopathology or clinicoradiologic follow-up was considered the criterion standard. RESULTS Of 35 patients, 25 (30 lesions) were classified as having a recurrence and 10 (11 lesions) patients as having treatment-induced necrosis. Parameters like rCBVmean (mean relative CBV), ADCmean, Cho/Cr, and maximum and mean target-to-background ratios were statistically significant in the detection of recurrent lesions with an accuracy of 77.5%, 78.0%, 90.9%, 87.8%, and 87.8%, respectively. On multivariate receiver operating characteristic analysis, the combination of all 3 MR imaging parameters resulted in an area under the curve of 0.913 ± 0.053. Furthermore, an area under the curve of 0.935 ± 0.046 was obtained when MR imaging parameters (ADCmean and Cho/Cr) were combined with the PET parameter (mean target-to-background ratio), demonstrating an increase in diagnostic accuracy. CONCLUSIONS Simultaneous PET/MR imaging with FDG offers correlative and synergistic multiparametric assessment of glioma recurrence with increased accuracy and clinical utility.
Collapse
Affiliation(s)
- A Jena
- From the PET SUITE (A. Jena, S.T., A. Jha, P.N.)
| | - S Taneja
- From the PET SUITE (A. Jena, S.T., A. Jha, P.N.)
| | - A Jha
- From the PET SUITE (A. Jena, S.T., A. Jha, P.N.)
| | - N K Damesha
- Neurosurgery (N.K.D., S.K.S.), Indraprastha Apollo Hospitals, Sarita Vihar, New Delhi, India
| | - P Negi
- From the PET SUITE (A. Jena, S.T., A. Jha, P.N.)
| | - G K Jadhav
- Departments of Molecular Imaging and Nuclear Medicine, Radiation Oncology (G.K.J., S.M.V.)
| | - S M Verma
- Departments of Molecular Imaging and Nuclear Medicine, Radiation Oncology (G.K.J., S.M.V.)
| | - S K Sogani
- Neurosurgery (N.K.D., S.K.S.), Indraprastha Apollo Hospitals, Sarita Vihar, New Delhi, India
| |
Collapse
|
34
|
Wan B, Wang S, Tu M, Wu B, Han P, Xu H. The diagnostic performance of perfusion MRI for differentiating glioma recurrence from pseudoprogression: A meta-analysis. Medicine (Baltimore) 2017; 96:e6333. [PMID: 28296759 PMCID: PMC5369914 DOI: 10.1097/md.0000000000006333] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND The purpose of this meta-analysis was to evaluate the diagnostic accuracy of perfusion magnetic resonance imaging (MRI) as a method for differentiating glioma recurrence from pseudoprogression. METHODS The PubMed, Embase, Cochrane Library, and Chinese Biomedical databases were searched comprehensively for relevant studies up to August 3, 2016 according to specific inclusion and exclusion criteria. The quality of the included studies was assessed according to the quality assessment of diagnostic accuracy studies (QUADAS-2). After performing heterogeneity and threshold effect tests, pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio were calculated. Publication bias was evaluated visually by a funnel plot and quantitatively using Deek funnel plot asymmetry test. The area under the summary receiver operating characteristic curve was calculated to demonstrate the diagnostic performance of perfusion MRI. RESULTS Eleven studies covering 416 patients and 418 lesions were included in this meta-analysis. The pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio were 0.88 (95% confidence interval [CI] 0.84-0.92), 0.77 (95% CI 0.69-0.84), 3.93 (95% CI 2.83-5.46), 0.16 (95% CI 0.11-0.22), and 27.17 (95% CI 14.96-49.35), respectively. The area under the summary receiver operating characteristic curve was 0.8899. There was no notable publication bias. Sensitivity analysis showed that the meta-analysis results were stable and credible. CONCLUSION While perfusion MRI is not the ideal diagnostic method for differentiating glioma recurrence from pseudoprogression, it could improve diagnostic accuracy. Therefore, further research on combining perfusion MRI with other imaging modalities is warranted.
Collapse
Affiliation(s)
- Bing Wan
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology
| | - Siqi Wang
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology
| | - Mengqi Tu
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology
| | - Bo Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ping Han
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology
| | - Haibo Xu
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
35
|
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
|
36
|
Chuang MT, Liu YS, Tsai YS, Chen YC, Wang CK. Differentiating Radiation-Induced Necrosis from Recurrent Brain Tumor Using MR Perfusion and Spectroscopy: A Meta-Analysis. PLoS One 2016; 11:e0141438. [PMID: 26741961 PMCID: PMC4712150 DOI: 10.1371/journal.pone.0141438] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 08/16/2015] [Indexed: 01/03/2023] Open
Abstract
Purpose This meta-analysis examined roles of several metabolites in differentiating recurrent tumor from necrosis in patients with brain tumors using MR perfusion and spectroscopy. Methods Medline, Cochrane, EMBASE, and Google Scholar were searched for studies using perfusion MRI and/or MR spectroscopy published up to March 4, 2015 which differentiated between recurrent tumor vs. necrosis in patients with primary brain tumors or brain metastasis. Only two-armed, prospective or retrospective studies were included. A meta-analysis was performed on the difference in relative cerebral blood volume (rCBV), ratios of choline/creatine (Cho/Cr) and/or choline/N-acetyl aspartate (Cho/NAA) between participants undergoing MRI evaluation. A χ2-based test of homogeneity was performed using Cochran’s Q statistic and I2. Results Of 397 patients in 13 studies who were analyzed, the majority had tumor recurrence. As there was evidence of heterogeneity among 10 of the studies which used rCBV for evaluation (Q statistic = 31.634, I2 = 97.11%, P < 0.0001) a random-effects analysis was applied. The pooled difference in means (2.18, 95%CI = 0.85 to 3.50) indicated that the average rCBV in a contrast-enhancing lesion was significantly higher in tumor recurrence compared with radiation injury (P = 0.001). Based on a fixed-effect model of analysis encompassing the six studies which used Cho/Cr ratios for evaluation (Q statistic = 8.388, I2 = 40.39%, P = 0.137), the pooled difference in means (0.77, 95%CI = 0.57 to 0.98) of the average Cho/Cr ratio was significantly higher in tumor recurrence than in tumor necrosis (P = 0.001). There was significant difference in ratios of Cho to NAA between recurrent tumor and necrosis (1.02, 95%CI = 0.03 to 2.00, P = 0.044). Conclusions MR spectroscopy and MR perfusion using Cho/NAA and Cho/Cr ratios and rCBV may increase the accuracy of differentiating necrosis from recurrent tumor in patients with primary brain tumors or metastases.
Collapse
Affiliation(s)
- Ming-Tsung Chuang
- Department of Diagnostic Radiology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Yi-Sheng Liu
- Department of Diagnostic Radiology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Yi-Shan Tsai
- Department of Diagnostic Radiology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Ying-Chen Chen
- Department of Diagnostic Radiology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chien-Kuo Wang
- Department of Diagnostic Radiology, National Cheng Kung University Hospital, Tainan, Taiwan
- * E-mail:
| |
Collapse
|
37
|
Jeraj R, Bradshaw T, Simončič U. Molecular Imaging to Plan Radiotherapy and Evaluate Its Efficacy. J Nucl Med 2015; 56:1752-65. [PMID: 26383148 DOI: 10.2967/jnumed.114.141424] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 09/08/2015] [Indexed: 12/25/2022] Open
Abstract
Molecular imaging plays a central role in the management of radiation oncology patients. Specific uses of imaging, particularly to plan radiotherapy and assess its efficacy, require an additional level of reproducibility and image quality beyond what is required for diagnostic imaging. Specific requirements include proper patient preparation, adequate technologist training, careful imaging protocol design, reliable scanner technology, reproducible software algorithms, and reliable data analysis methods. As uncertainty in target definition is arguably the greatest challenge facing radiation oncology, the greatest impact that molecular imaging can have may be in the reduction of interobserver variability in target volume delineation and in providing greater conformity between target volume boundaries and true tumor boundaries. Several automatic and semiautomatic contouring methods based on molecular imaging are available but still need sufficient validation to be widely adopted. Biologically conformal radiotherapy (dose painting) based on molecular imaging-assessed tumor heterogeneity is being investigated, but many challenges remain to fully exploring its potential. Molecular imaging also plays increasingly important roles in both early (during treatment) and late (after treatment) response assessment as both a predictive and a prognostic tool. Because of potentially confounding effects of radiation-induced inflammation, treatment response assessment requires careful interpretation. Although molecular imaging is already strongly embedded in radiotherapy, the path to widespread and all-inclusive use is still long. The lack of solid clinical evidence is the main impediment to broader use. Recommendations for practicing physicians are still rather scarce. (18)F-FDG PET/CT remains the main molecular imaging modality in radiation oncology applications. Although other molecular imaging options (e.g., proliferation imaging) are becoming more common, their widespread use is limited by lack of tracer availability and inadequate reimbursement models. With the increasing presence of molecular imaging in radiation oncology, special emphasis should be placed on adequate training of radiation oncology personnel to understand the potential, and particularly the limitations, of quantitative molecular imaging applications. Similarly, radiologists and nuclear medicine specialists should be sensitized to the special need of the radiation oncologist in terms of quantification and reproducibility. Furthermore, strong collaboration between radiation oncology, nuclear medicine/radiology, and medical physics teams is necessary, as optimal and safe use of molecular imaging can be ensured only within appropriate interdisciplinary teams.
Collapse
Affiliation(s)
- Robert Jeraj
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin; and Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
| | - Tyler Bradshaw
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin; and
| | - Urban Simončič
- Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
38
|
Feng D, Cortese G, Baumgartner R. A comparison of confidence/credible interval methods for the area under the ROC curve for continuous diagnostic tests with small sample size. Stat Methods Med Res 2015; 26:2603-2621. [PMID: 26323286 DOI: 10.1177/0962280215602040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The receiver operating characteristic (ROC) curve is frequently used as a measure of accuracy of continuous markers in diagnostic tests. The area under the ROC curve (AUC) is arguably the most widely used summary index for the ROC curve. Although the small sample size scenario is common in medical tests, a comprehensive study of small sample size properties of various methods for the construction of the confidence/credible interval (CI) for the AUC has been by and large missing in the literature. In this paper, we describe and compare 29 non-parametric and parametric methods for the construction of the CI for the AUC when the number of available observations is small. The methods considered include not only those that have been widely adopted, but also those that have been less frequently mentioned or, to our knowledge, never applied to the AUC context. To compare different methods, we carried out a simulation study with data generated from binormal models with equal and unequal variances and from exponential models with various parameters and with equal and unequal small sample sizes. We found that the larger the true AUC value and the smaller the sample size, the larger the discrepancy among the results of different approaches. When the model is correctly specified, the parametric approaches tend to outperform the non-parametric ones. Moreover, in the non-parametric domain, we found that a method based on the Mann-Whitney statistic is in general superior to the others. We further elucidate potential issues and provide possible solutions to along with general guidance on the CI construction for the AUC when the sample size is small. Finally, we illustrate the utility of different methods through real life examples.
Collapse
Affiliation(s)
- Dai Feng
- 1 Biometrics Research, Merck Research Lab, NJ, USA
| | - Giuliana Cortese
- 2 Department of Statistical Sciences, University of Padova, Italy
| | | |
Collapse
|
39
|
Kohutek ZA, Yamada Y, Chan TA, Brennan CW, Tabar V, Gutin PH, Yang TJ, Rosenblum MK, Ballangrud Å, Young RJ, Zhang Z, Beal K. Long-term risk of radionecrosis and imaging changes after stereotactic radiosurgery for brain metastases. J Neurooncol 2015; 125:149-56. [PMID: 26307446 DOI: 10.1007/s11060-015-1881-3] [Citation(s) in RCA: 214] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 08/10/2015] [Indexed: 10/23/2022]
Abstract
Radionecrosis is a well-characterized effect of stereotactic radiosurgery (SRS) and is occasionally associated with serious neurologic sequelae. Here, we investigated the incidence of and clinical variables associated with the development of radionecrosis and related radiographic changes after SRS for brain metastases in a cohort of patients with long-term follow up. 271 brain metastases treated with single-fraction linear accelerator-based SRS were analyzed. Radionecrosis was diagnosed either pathologically or radiographically. Univariate and multivariate Cox regression was performed to determine the association between radionecrosis and clinical factors available prior to treatment planning. After median follow up of 17.2 months, radionecrosis was observed in 70 (25.8%) lesions, including 47 (17.3%) symptomatic cases. 22 of 70 cases (31.4%) were diagnosed pathologically and 48 (68.6%) were diagnosed radiographically. The actuarial incidence of radionecrosis was 5.2% at 6 months, 17.2% at 12 months and 34.0% at 24 months. On univariate analysis, radionecrosis was associated with maximum tumor diameter (HR 3.55, p < 0.001), prior whole brain radiotherapy (HR 2.21, p = 0.004), prescription dose (HR 0.56, p = 0.02) and histology other than non-small cell lung, breast or melanoma (HR 1.85, p = 0.04). On multivariate analysis, only maximum tumor diameter (HR 3.10, p < 0.001) was associated with radionecrosis risk. This data demonstrates that with close imaging follow-up, radionecrosis after single-fraction SRS for brain metastases is not uncommon. Maximum tumor diameter on pre-treatment MR imaging can provide a reliable estimate of radionecrosis risk prior to treatment planning, with the greatest risk among tumors measuring >1 cm.
Collapse
Affiliation(s)
- Zachary A Kohutek
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Ave., New York, NY, 10065, USA
| | - Yoshiya Yamada
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Ave., New York, NY, 10065, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Ave., New York, NY, 10065, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cameron W Brennan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Viviane Tabar
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Philip H Gutin
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - T Jonathan Yang
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Ave., New York, NY, 10065, USA
| | - Marc K Rosenblum
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Åse Ballangrud
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert J Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhigang Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kathryn Beal
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Ave., New York, NY, 10065, USA.
| |
Collapse
|
40
|
A meta-analysis comparing 18F-FLT PET with 18F-FDG PET for assessment of brain tumor recurrence. Nucl Med Commun 2015; 36:695-701. [DOI: 10.1097/mnm.0000000000000302] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
41
|
Peck M, Pollack HA, Friesen A, Muzi M, Shoner SC, Shankland EG, Fink JR, Armstrong JO, Link JM, Krohn KA. Applications of PET imaging with the proliferation marker [18F]-FLT. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2015; 59:95-104. [PMID: 25737423 PMCID: PMC4415691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
[18F]-3'-fluoro-3'-deoxythymidine (FLT) is a nucleoside-analog imaging agent for quantifying cellular proliferation that was first reported in 1998. It accumulates during the S-phase of the cell cycle through the action of cytosolic thymidine kinase, TK1. Since TK1 is primarily expressed in dividing cells, FLT uptake is essentially limited to dividing cells. Thus FLT is an effective measure of cell proliferation. FLT uptake has been shown to correlate with the more classic proliferation marker, the monoclonal antibody to Ki-67. Increased cellular proliferation is known to correlate with worse outcome in many cancers. However, the Ki-67 binding assay is performed on a sampled preparation, ex vivo, whereas FLT can be quantitatively measured in vivo using positron emission tomography (PET). FLT is an effective and quantitative marker of cell proliferation, and therefore a useful prognostic predictor in the setting of neoplastic disease. This review summarizes clinical studies from 2011 forward that used FLT-PET to assess tumor response to therapy. The paper focuses on our recommendations for a standardized clinical trial protocol and components of a report so multi center studies can be effectively conducted, and different studies can be compared. For example, since FLT is glucuronidated by the liver, and the metabolite is not transported into the cell, the plasma fraction of FLT can be significantly changed by treatment with particular drugs that deplete this enzyme, including some chemotherapy agents and pain medications. Therefore, the plasma level of metabolites should be measured to assure FLT uptake kinetics can be accurately calculated. This is important because the flux constant (KFLT) is a more accurate measure of proliferation and, by inference, a better discriminator of tumor recurrence than standardized uptake value (SUVFLT). This will allow FLT imaging to be a specific and clinically relevant prognostic predictor in the treatment of neoplastic disease.
Collapse
Affiliation(s)
- M Peck
- Stanford University, Stanford, CA, USA -
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kim MM, Lawrence TS, Cao Y. Advances in Magnetic Resonance and Positron Emission Tomography Imaging: Assessing Response in the Treatment of Low-Grade Glioma. Semin Radiat Oncol 2015; 25:172-80. [PMID: 26050587 DOI: 10.1016/j.semradonc.2015.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Following combined-modality therapy for the treatment of low-grade gliomas, the assessment of treatment response and the evaluation of disease progression are uniformly challenging. In this article, we review existing response criteria, and discuss the limitations of conventional magnetic resonance imaging to distinguish between progression and treatment effect. We review the data on advanced imaging techniques including positron emission tomography and functional magnetic resonance imaging, which may enhance the interpretation of posttreatment changes, and enable the earlier assessment of the efficacy and toxicity of therapy in these patients with prolonged survival.
Collapse
Affiliation(s)
- Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.
| | | | - Yue Cao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI; Department of Radiology, University of Michigan, Ann Arbor, MI; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| |
Collapse
|
43
|
Wang X, Hu X, Xie P, Li W, Li X, Ma L. Comparison of magnetic resonance spectroscopy and positron emission tomography in detection of tumor recurrence in posttreatment of glioma: A diagnostic meta-analysis. Asia Pac J Clin Oncol 2014; 11:97-105. [PMID: 24783970 DOI: 10.1111/ajco.12202] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2014] [Indexed: 11/27/2022]
Abstract
It is important to distinguish between tumor recurrence and treatment effects in posttreatment patients with high-grade gliomas. Several imaging modalities have been reported in differentiating between tumor recurrence and treatment effects. However, there were no consistent conclusions between different studies. We performed a meta-analysis of 23 studies that compared the diagnostic values of fluorine-18-fluorodeoxyglucose ((18)F-FDG) and (11)C-methionine ((11)C-MET) PET (positron emission tomography) or PET/CT (computed tomography) and magnetic resonance spectroscopy (MRS) in predicting tumor recurrence of gliomas. The pooled estimated sensitivity, specificity, positive likelihood ratios, negative likelihood ratios and summary receiver operating characteristic curves of (18)F-FDG and (11)C-MET PET or PET/CT and MRS in detecting tumor recurrence were calculated. In conclusion, MRS is highly sensitive in the detection of tumor recurrence in glioma.(18)F-FDG PET or PET/CT is highly specific in recurrence diagnosis. (11)C-MET does not have noticeable advantage over (18)F-FDG. The current evidence shows no statistical difference between MRS and PET on the accuracy.
Collapse
Affiliation(s)
- Xiaoyue Wang
- Department of Surgery, Shandong Cancer Hospital & Institute, Jinan, Shandong, China
| | | | | | | | | | | |
Collapse
|
44
|
The role of imaging in the management of progressive glioblastoma : a systematic review and evidence-based clinical practice guideline. J Neurooncol 2014; 118:435-60. [PMID: 24715656 DOI: 10.1007/s11060-013-1330-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 12/27/2013] [Indexed: 10/25/2022]
Abstract
QUESTION Which imaging techniques most accurately differentiate true tumor progression from pseudo-progression or treatment related changes in patients with previously diagnosed glioblastoma? TARGET POPULATION These recommendations apply to adults with previously diagnosed glioblastoma who are suspected of experiencing progression of the neoplastic process. RECOMMENDATIONS LEVEL II Magnetic resonance imaging with and without gadolinium enhancement is recommended as the imaging surveillance method to detect the progression of previously diagnosed glioblastoma. LEVEL II Magnetic resonance spectroscopy is recommended as a diagnostic method to differentiate true tumor progression from treatment-related imaging changes or pseudo-progression in patients with suspected progressive glioblastoma. LEVEL III The routine use of positron emission tomography to identify progression of glioblastoma is not recommended. LEVEL III Single-photon emission computed tomography imaging is recommended as a diagnostic method to differentiate true tumor progression from treatment-related imaging changes or pseudo-progression in patients with suspected progressive glioblastoma.
Collapse
|
45
|
Imani F, Boada FE, Lieberman FS, Davis DK, Mountz JM. Molecular and metabolic pattern classification for detection of brain glioma progression. Eur J Radiol 2013; 83:e100-5. [PMID: 24321226 DOI: 10.1016/j.ejrad.2013.06.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 06/06/2013] [Accepted: 06/07/2013] [Indexed: 01/01/2023]
Abstract
OBJECTIVES The ability to differentiate between brain tumor progression and radiation therapy induced necrosis is critical for appropriate patient management. In order to improve the differential diagnosis, we combined fluorine-18 2-fluoro-deoxyglucose positron emission tomography ((18)F-FDG PET), proton magnetic resonance spectroscopy ((1)H MRS) and histological data to develop a multi-parametric machine-learning model. METHODS We enrolled twelve post-therapy patients with grade 2 and 3 gliomas that were suspicious of tumor progression. All patients underwent (18)F-FDG PET and (1)H MRS. Maximal standardized uptake value (SUVmax) of the tumors and reference regions were obtained. Multiple 2D maps of choline (Cho), creatine (Cr), and N-acetylaspartate (NAA) of the tumors were generated. A support vector machine (SVM) learning model was established to take imaging biomarkers and histological data as input vectors. A combination of clinical follow-up and multiple sequential MRI studies served as the basis for assessing the clinical outcome. All vector combinations were evaluated for diagnostic accuracy and cross validation. The optimal cutoff value of individual parameters was calculated using Receiver operating characteristic (ROC) plots. RESULTS The SVM and ROC analyses both demonstrated that SUVmax of the lesion was the most significant single diagnostic parameter (75% accuracy) followed by Cho concentration (67% accuracy). SVM analysis of all paired parameters showed SUVmax and Cho concentration in combination could achieve 83% accuracy. SUVmax of the lesion paired with SUVmax of the white matter as well as the tumor Cho paired with the tumor Cr both showed 83% accuracy. These were the most significant paired diagnostic parameters of either modality. Combining all four parameters did not improve the results. However, addition of two more parameters, Cho and Cr of brain parenchyma contralateral to the tumor, increased the accuracy to 92%. CONCLUSION This study suggests that SVM models may improve detection of glioma progression more accurately than single parametric imaging methods. RESEARCH SUPPORT National Cancer Institute, Cancer Center Support Grant Supplement Award, Imaging Response Assessment Teams.
Collapse
Affiliation(s)
- Farzin Imani
- Department of Radiology, University of Pittsburgh Medical Center, PA, United States.
| | - Fernando E Boada
- Department of Radiology, University of Pittsburgh Medical Center, PA, United States
| | - Frank S Lieberman
- Department of Neurology, University of Pittsburgh Medical Center, PA, United States
| | - Denise K Davis
- Department of Radiology, University of Pittsburgh Medical Center, PA, United States
| | - James M Mountz
- Department of Radiology, University of Pittsburgh Medical Center, PA, United States
| |
Collapse
|
46
|
Diagnostic Challenges of Radiation Necrosis. Can J Neurol Sci 2013; 40:763-4. [DOI: 10.1017/s0317167100015845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
47
|
Kadrmas DJ, Hoffman JM. Methodology for quantitative rapid multi-tracer PET tumor characterizations. Am J Cancer Res 2013; 3:757-73. [PMID: 24312149 PMCID: PMC3840410 DOI: 10.7150/thno.5201] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 04/15/2013] [Indexed: 01/18/2023] Open
Abstract
Positron emission tomography (PET) can image a wide variety of functional and physiological parameters in vivo using different radiotracers. As more is learned about the molecular basis for disease and treatment, the potential value of molecular imaging for characterizing and monitoring disease status has increased. Characterizing multiple aspects of tumor physiology by imaging multiple PET tracers in a single patient provides additional complementary information, and there is a significant body of literature supporting the potential value of multi-tracer PET imaging in oncology. However, imaging multiple PET tracers in a single patient presents a number of challenges. A number of techniques are under development for rapidly imaging multiple PET tracers in a single scan, where signal-recovery processing algorithms are employed to recover various imaging endpoints for each tracer. Dynamic imaging is generally used with tracer injections staggered in time, and kinetic constraints are utilized to estimate each tracers' contribution to the multi-tracer imaging signal. This article summarizes past and ongoing work in multi-tracer PET tumor imaging, and then organizes and describes the main algorithmic approaches for achieving multi-tracer PET signal-recovery. While significant advances have been made, the complexity of the approach necessitates protocol design, optimization, and testing for each particular tracer combination and application. Rapid multi-tracer PET techniques have great potential for both research and clinical cancer imaging applications, and continued research in this area is warranted.
Collapse
|
48
|
Jacobson O, Chen X. Interrogating tumor metabolism and tumor microenvironments using molecular positron emission tomography imaging. Theranostic approaches to improve therapeutics. Pharmacol Rev 2013; 65:1214-56. [PMID: 24064460 PMCID: PMC3799232 DOI: 10.1124/pr.113.007625] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Positron emission tomography (PET) is a noninvasive molecular imaging technology that is becoming increasingly important for the measurement of physiologic, biochemical, and pharmacological functions at cellular and molecular levels in patients with cancer. Formation, development, and aggressiveness of tumor involve a number of molecular pathways, including intrinsic tumor cell mutations and extrinsic interaction between tumor cells and the microenvironment. Currently, evaluation of these processes is mainly through biopsy, which is invasive and limited to the site of biopsy. Ongoing research on specific target molecules of the tumor and its microenvironment for PET imaging is showing great potential. To date, the use of PET for diagnosing local recurrence and metastatic sites of various cancers and evaluation of treatment response is mainly based on [(18)F]fluorodeoxyglucose ([(18)F]FDG), which measures glucose metabolism. However, [(18)F]FDG is not a target-specific PET tracer and does not give enough insight into tumor biology and/or its vulnerability to potential treatments. Hence, there is an increasing need for the development of selective biologic radiotracers that will yield specific biochemical information and allow for noninvasive molecular imaging. The possibility of cancer-associated targets for imaging will provide the opportunity to use PET for diagnosis and therapy response monitoring (theranostics) and thus personalized medicine. This article will focus on the review of non-[(18)F]FDG PET tracers for specific tumor biology processes and their preclinical and clinical applications.
Collapse
Affiliation(s)
- Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD.
| | | |
Collapse
|
49
|
Zhao C, Zhang Y, Wang J. A meta-analysis on the diagnostic performance of (18)F-FDG and (11)C-methionine PET for differentiating brain tumors. AJNR Am J Neuroradiol 2013; 35:1058-65. [PMID: 24029389 DOI: 10.3174/ajnr.a3718] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SUMMARY (18)F-FDG-PET has been widely used in patients with brain tumors. However, the reported sensitivity and specificity of (18)F-FDG-PET for brain tumor differentiation varied greatly. We performed this meta-analysis to systematically assess the diagnostic performance of (18)F-FDG-PET in differentiating brain tumors. The diagnostic performance of (11)C-methionine PET was assessed for comparison. Relevant studies were searched in PubMed/MEDLINE, Scopus, and China National Knowledge Infrastructure (until February 2013). The methodologic quality of eligible studies was evaluated, and a meta-analysis was performed to obtain the combined diagnostic performance of (18)F-FDG and (11)C-methionine PET with a bivariate model. Thirty eligible studies, including 5 studies with both (18)F-FDG and (11)C-methionine PET data were enrolled. Pooled sensitivity, pooled specificity, and area under the receiver operating characteristic curve of (18)F-FDG-PET (n = 24) for differentiating brain tumors were 0.71 (95% CI, 0.63-0.78), 0.77 (95% CI, 0.67-0.85), and 0.80. Heterogeneity was found among (18)F-FDG studies. Subsequent subgroup analysis revealed that the disease status was a statistically significant source of the heterogeneity and that the sensitivity in the patients with recurrent brain tumor was markedly higher than those with suspected primary brain tumors. Pooled sensitivity, pooled specificity, and area under the receiver operating characteristic of (11)C-methionine PET (n = 11) were 0.91 (95% CI, 0.85-0.94), 0.86 (95% CI, 0.78-0.92), and 0.94. No significant statistical heterogeneity was found among (11)C-methionine studies. This meta-analysis suggested that (18)F-FDG-PET has limited diagnostic performance in brain tumor differentiation, though its performance may vary according to the status of brain tumor, whereas (11)C-methionine PET has excellent diagnostic accuracy in brain tumor differentiation.
Collapse
Affiliation(s)
- C Zhao
- From the Department of Nuclear Medicine (C.Z., J.W.), Hangzhou First People's Hospital, Hangzhou Cancer Hospital, Hangzhou, China
| | - Y Zhang
- Department of Nuclear Medicine (Y.Z.), Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| | - J Wang
- From the Department of Nuclear Medicine (C.Z., J.W.), Hangzhou First People's Hospital, Hangzhou Cancer Hospital, Hangzhou, China
| |
Collapse
|
50
|
Abstract
Several new tracers are being developed for use with PET to assess pathways that are altered in cancers, including energy use, cellular signaling, transport, and proliferation. Because increased proliferation is a hallmark of many cancers, several tracers have been tested to track the DNA synthesis pathway. Thymidine, which is incorporated into DNA but not RNA, has been used in laboratory studies to measure tumor growth. Because thymidine labeled with (11)C undergoes rapid biologic degradation and has a short physical half-life, tracers labeled with (18)F have been preferred in PET imaging. One such tracer is (18)F-labeled 3'-deoxy-3'-fluorothymidine ((18)F-FLT). (18)F-FLT is trapped after phosphorylation by thymidine kinase 1, whose expression is increased in replicating cells. Several studies on breast, lung, and brain tumors have demonstrated that retention of (18)F-FLT correlated with tumor proliferation. Although (18)F-FLT has been used to image and stage several tumor types, the standardized uptake value is generally lower than that obtained with (18)F-FDG. (18)F-FLT can be used to image many areas of the body, but background uptake is high in the liver, marrow, and renal system, limiting use in these organs. (18)F-FLT PET imaging has primarily been studied in the assessment of treatment response. Rapid declines in (18)F-FLT retention within days to weeks have been demonstrated in several tumor types treated with cytotoxic drugs, targeted agents, and radiotherapy. Further work is ongoing to validate this approach and determine its utility in the development of new drugs and in the clinical evaluation of standard treatment approaches.
Collapse
Affiliation(s)
- Omid S Tehrani
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan 48201, USA
| | | |
Collapse
|