1
|
Rogers S, Gross M, Ermis E, Cosgun G, Baumert BG, Mader T, Schroeder C, Lomax N, Alonso S, Ademaj A, Lazeroms T, Lee SY, Mayinger M, Mamot C, Schwyzer L, Schubert GA, Riesterer O. Re-irradiation for recurrent glioblastoma: a pattern of care analysis. BMC Neurol 2024; 24:462. [PMID: 39587462 PMCID: PMC11590342 DOI: 10.1186/s12883-024-03954-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND 90% of glioblastomas (GBM) relapse within two years of diagnosis. In contrast to the initial setting, there is no standard management for recurrent disease and options include hypofractionated stereotactic re-irradiation (re-mHSRT). The aims of this study were to investigate re-mHSRT practice in Swiss neuro-oncology centres. METHODS A survey of 18 questions regarding re-irradiation for GBM was created and distributed electronically (SurveyMonkey, USA) to 11 radiation oncologists in Switzerland specialising in brain tumours. We evaluated the clinical outcomes of a multicentre series of patients treated with an established re-mHSRT schedule to benchmark these against the literature and investigated the radiological patterns of relapse after re-mHSRT. RESULTS 8 of 11 (73%) radiation oncologists responded to the survey and re-irradiation practice was heterogeneous. The 10 × 3.5 Gy schedule (RTOG 1205, BRIOChe trials) was used by 5/8 respondents and 47/50 patients with recurrent GBM treated with re-mHSRT with this schedule in daily practice were included in the analysis. The median time to re-mHSRT following completion of adjuvant RT was 23.3 (7-224) months. The median PTV at re-mHSRT was 22.0 cm3 (0.9-190). Combined CTV + PTV margins ranged from 0 to 10 mm and median prescription isodose was 80% (67-100). 14/47 (30%) patients received temozolomide and four (8.5%) continued bevacizumab concomitantly. On multivariable analysis, concomitant systemic therapy predicted for progression-free survival (PFS), HR 2.87 (95% CI 1-03-7.96), p = 0.042. Median PFS following re-mHSRT was 6.6 (0.2-92.5) months and 26/47 patients (55%) received subsequent systemic therapy. The median overall survival (OS) following recurrence was 11.8 months (1.5-92.5), similar to the 10.8 months in the literature with the same schedule. The six-month OS rate was 37/47 (79%), which compares well with the 73% reported in a meta-analysis of 50 publications employing various schedules. In a subgroup analysis of 36/47 (79%) patients with MR follow-up after re-mHSRT, 8/36 (22%) had no radiological evidence of tumour progression at a median follow-up of 9.4 months. 21/28 (75%) radiological relapses were in-field, two were marginal and five were out of field. CONCLUSIONS Re-mHSRT with 10 × 3.5 Gy can achieve local control in selected patients with recurrent GBM.
Collapse
Affiliation(s)
- Susanne Rogers
- Radiation Oncology Center Mittelland, Kantonsspital Aarau, Aarau, Switzerland.
| | - Markus Gross
- Department of Radiation Oncology, University Hospital Basel, Basel, Switzerland
| | - Ekin Ermis
- Department of Radiation Oncology, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Gizem Cosgun
- Department of Radiation Oncology, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Brigitta G Baumert
- Department of Radiation Oncology, Kantonsspital Graubünden, Chur, Switzerland
| | - Thomas Mader
- Department of Radiation Oncology, Kantonsspital Graubünden, Chur, Switzerland
| | - Christina Schroeder
- Department of Radiation Oncology, Kantonsspital Winterthur, Winterthur, Switzerland
- Department of Neurology, Kantonsspital Aarau, Aarau, Switzerland
| | - Nicoletta Lomax
- Radiation Oncology Center Mittelland, Kantonsspital Aarau, Aarau, Switzerland
| | - Sara Alonso
- Radiation Oncology Center Mittelland, Kantonsspital Aarau, Aarau, Switzerland
| | - Adela Ademaj
- Radiation Oncology Center Mittelland, Kantonsspital Aarau, Aarau, Switzerland
| | - Tessa Lazeroms
- Radiation Oncology Center Mittelland, Kantonsspital Aarau, Aarau, Switzerland
| | - Seok-Yun Lee
- Department of Medical Oncology, Kantonsspital Aarau, Aarau, Switzerland
| | - Michael Mayinger
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Christoph Mamot
- Department of Medical Oncology, Kantonsspital Aarau, Aarau, Switzerland
| | - Lucia Schwyzer
- Department of Neurosurgery, Kantonsspital Aarau, Aarau, Switzerland
| | - Gerrit A Schubert
- Department of Neurosurgery, Kantonsspital Aarau, Aarau, Switzerland
- Department of Neurosurgery, RWTH Aachen University, Aachen, Germany
| | - Oliver Riesterer
- Radiation Oncology Center Mittelland, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
2
|
Beddok A, Orlhac F, Rozenblum L, Calugaru V, Créhange G, Dercle L, Nioche C, Thariat J, Marin T, El Fakhri G, Buvat I. Radiomics-driven personalized radiotherapy for primary and recurrent tumors: A general review with a focus on reirradiation. Cancer Radiother 2024; 28:597-602. [PMID: 39406602 DOI: 10.1016/j.canrad.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 11/03/2024]
Abstract
PURPOSE This review systematically investigates the role of radiomics in radiotherapy, with a particular emphasis on the use of quantitative imaging biomarkers for predicting clinical outcomes, assessing toxicity, and optimizing treatment planning. While the review encompasses various applications of radiomics in radiotherapy, it particularly highlights its potential for guiding reirradiation of recurrent cancers. METHODS A systematic review was conducted based on a Medline search with the search engine PubMed using the keywords "radiomics or radiomic" and "radiotherapy or reirradiation". Out of 189 abstracts reviewed, 147 original articles were included in the analysis. These studies were categorized by tumor localization, imaging modality, study objectives, and performance metrics, with a particular emphasis on the inclusion of external validation and adherence to a standardized radiomics pipeline. RESULTS The review identified 14 tumor localizations, with the majority of studies focusing on lung (33 studies), head and neck (27 studies), and brain (15 studies) cancers. CT was the most frequently employed imaging modality (77 studies) for radiomics, followed by MRI (46 studies) and PET (13 studies). The overall AUC across all studies, primarily focused on predicting the risk of recurrence (94 studies) or toxicity (41 studies), was 0.80 (SD=0.08). However, only 24 studies (16.3%) included external validation, with a slightly lower AUC compared to those without it. For studies using CT versus MRI or PET, both had a median AUC of 0.79, with IQRs of 0.73-0.86 for CT and 0.76-0.855 for MRI/PET, showing no significant differences in performance. Five studies involving reirradiation reported a median AUC of 0.81 (IQR: 0.73-0.825). CONCLUSION Radiomics demonstrates considerable potential in personalizing radiotherapy by improving treatment precision through better outcome prediction and treatment planning. However, its clinical adoption is hindered by the lack of external validation and variability in study designs. Future research should focus on implementing rigorous validation methods and standardizing imaging protocols to enhance the reliability and generalizability of radiomics in clinical radiotherapy, with particular attention to its application in reirradiation.
Collapse
Affiliation(s)
- Arnaud Beddok
- Department of Radiation Oncology, institut Godinot, Reims, France; Université de Reims Champagne-Ardenne, Crestic, Reims, France; Yale PET Center, Department of Radiology & Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA.
| | - Fanny Orlhac
- Institut Curie, université PSL, université Paris Saclay, Inserm Lito U1288, Orsay, France
| | - Laura Rozenblum
- Yale PET Center, Department of Radiology & Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Nuclear Medicine, hôpitaux universitaires la Pitié Salpêtrière-Charles-Foix, AP-HP, Sorbonne université, 75013 Paris, France
| | - Valentin Calugaru
- Institut Curie, université PSL, université Paris Saclay, Inserm Lito U1288, Orsay, France; Department of Radiation Oncology, institut Curie, université PSL, Paris, France
| | - Gilles Créhange
- Institut Curie, université PSL, université Paris Saclay, Inserm Lito U1288, Orsay, France; Department of Radiation Oncology, institut Curie, université PSL, Paris, France
| | - Laurent Dercle
- Department of Radiology, New York-Presbyterian Hospital, Columbia University, Vagelos College of Physicians and Surgeons, New York, USA
| | - Christophe Nioche
- Institut Curie, université PSL, université Paris Saclay, Inserm Lito U1288, Orsay, France
| | - Juliette Thariat
- Department of Radiation Oncology, centre François-Baclesse, Caen, France
| | - Thibault Marin
- Yale PET Center, Department of Radiology & Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Georges El Fakhri
- Yale PET Center, Department of Radiology & Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Irène Buvat
- Institut Curie, université PSL, université Paris Saclay, Inserm Lito U1288, Orsay, France
| |
Collapse
|
3
|
Robert JA, Leclerc A, Ducloie M, Emery E, Agostini D, Vigne J. Contribution of [ 18F]FET PET in the Management of Gliomas, from Diagnosis to Follow-Up: A Review. Pharmaceuticals (Basel) 2024; 17:1228. [PMID: 39338390 PMCID: PMC11435125 DOI: 10.3390/ph17091228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/14/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Gliomas, the most common type of primary malignant brain tumors in adults, pose significant challenges in diagnosis and management due to their heterogeneity and potential aggressiveness. This review evaluates the utility of O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) positron emission tomography (PET), a promising imaging modality, to enhance the clinical management of gliomas. We reviewed 82 studies involving 4657 patients, focusing on the application of [18F]FET in several key areas: diagnosis, grading, identification of IDH status and presence of oligodendroglial component, guided resection or biopsy, detection of residual tumor, guided radiotherapy, detection of malignant transformation in low-grade glioma, differentiation of recurrence versus treatment-related changes and prognostic factors, and treatment response evaluation. Our findings confirm that [18F]FET helps delineate tumor tissue, improves diagnostic accuracy, and aids in therapeutic decision-making by providing crucial insights into tumor metabolism. This review underscores the need for standardized parameters and further multicentric studies to solidify the role of [18F]FET PET in routine clinical practice. By offering a comprehensive overview of current research and practical implications, this paper highlights the added value of [18F]FET PET in improving management of glioma patients from diagnosis to follow-up.
Collapse
Affiliation(s)
- Jade Apolline Robert
- CHU de Caen Normandie, UNICAEN, Department of Nuclear Medicine, Normandie Université, 14000 Caen, France; (J.A.R.)
| | - Arthur Leclerc
- Department of Neurosurgery, Caen University Hospital, 14000 Caen, France
- Caen Normandie University, ISTCT UMR6030, GIP Cyceron, 14000 Caen, France
| | - Mathilde Ducloie
- Department of Neurology, Caen University Hospital, 14000 Caen, France
- Centre François Baclesse, Department of Neurology, 14000 Caen, France
| | - Evelyne Emery
- Department of Neurosurgery, Caen University Hospital, 14000 Caen, France
| | - Denis Agostini
- CHU de Caen Normandie, UNICAEN, Department of Nuclear Medicine, Normandie Université, 14000 Caen, France; (J.A.R.)
| | - Jonathan Vigne
- CHU de Caen Normandie, UNICAEN, Department of Nuclear Medicine, Normandie Université, 14000 Caen, France; (J.A.R.)
- CHU de Caen Normandie, UNICAEN Department of Pharmacy, Normandie Université, 14000 Caen, France
- Centre Cyceron, Institut Blood and Brain @ Caen-Normandie, Normandie Université, UNICAEN, INSERM U1237, PhIND, 14000 Caen, France
| |
Collapse
|
4
|
Huma C, Hawon L, Sarisha J, Erdal T, Kevin C, Valentina KA. Advances in the field of developing biomarkers for re-irradiation: a how-to guide to small, powerful data sets and artificial intelligence. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2024; 9:3-16. [PMID: 38550554 PMCID: PMC10972602 DOI: 10.1080/23808993.2024.2325936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/28/2024] [Indexed: 04/01/2024]
Abstract
Introduction Patient selection remains challenging as the clinical use of re-irradiation (re-RT) increases. Re-RT data is limited to retrospective studies and small prospective single-institution reports, resulting in small, heterogenous data sets. Validated prognostic and predictive biomarkers are derived from large-volume studies with long-term follow-up. This review aims to examine existing re-RT publications and available data sets and discuss strategies using artificial intelligence (AI) to approach small data sets to optimize the use of re-RT data. Methods Re-RT publications were identified where associated public data was present. The existing literature on small data sets to identify biomarkers was also explored. Results Publications with associated public data were identified, with glioma and nasopharyngeal cancers emerging as the most common tumor sites where the use of re-RT was the primary management approach. Existing and emerging AI strategies have been used to approach small data sets including data generation, augmentation, discovery, and transfer learning. Conclusions Further data is needed to generate adaptive frameworks, improve the collection of specimens for molecular analysis, and improve the interpretability of results in re-RT data.
Collapse
Affiliation(s)
- Chaudhry Huma
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| | - Lee Hawon
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| | - Jagasia Sarisha
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| | - Tasci Erdal
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| | - Camphausen Kevin
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| | - Krauze Andra Valentina
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| |
Collapse
|
5
|
Eisazadeh R, Shahbazi-Akbari M, Mirshahvalad SA, Pirich C, Beheshti M. Application of Artificial Intelligence in Oncologic Molecular PET-Imaging: A Narrative Review on Beyond [ 18F]F-FDG Tracers Part II. [ 18F]F-FLT, [ 18F]F-FET, [ 11C]C-MET and Other Less-Commonly Used Radiotracers. Semin Nucl Med 2024; 54:293-301. [PMID: 38331629 DOI: 10.1053/j.semnuclmed.2024.01.002] [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: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 02/10/2024]
Abstract
Following the previous part of the narrative review on artificial intelligence (AI) applications in positron emission tomography (PET) using tracers rather than 18F-fluorodeoxyglucose ([18F]F-FDG), in this part we review the impact of PET-derived radiomics data on the diagnostic performance of other PET radiotracers, 18F-O-(2-fluoroethyl)-L-tyrosine ([18F]F-FET), 18F-Fluorothymidine ([18F]F-FLT) and 11C-Methionine ([11C]C-MET). [18F]F-FET-PET, using an artificial amino acid taken up into upregulated tumoral cells, showed potential in lesion detection and tumor characterization, especially with its ability to reflect glioma heterogeneity. [18F]F-FET-PET-derived textural features appeared to have the potential to reveal considerable information for accurate delineation for guiding biopsy and treatment, differentiate between low-grade and high-grade glioma and related wild-type genotypes, and distinguish pseudoprogression from true progression. In addition, models built using clinical parameters and [18F]F-FET-PET-derived radiomics features showed acceptable results for survival stratification of glioblastoma patients. [18F]F-FLT-PET-based characteristics also showed potential in evaluating glioma patients, correlating with Ki-67 and patient prognosis. AI-based PET-volumetry using this radiotracer as a proliferation marker also revealed promising preliminary results in terms of guide-targeting bone marrow-preserving adaptive radiation therapy. Similar to [18F]F-FET, the other amino acid tracer which reflects cellular proliferation, [11C]C-MET, has also shown acceptable performance in predicting tumor grade, distinguishing brain tumor recurrence from radiation necrosis, and treatment monitoring by PET-derived radiomics models. In addition, PET-derived radiomics features of various radiotracers such as [18F]F-DOPA, [18F]F-FACBC, [18F]F-NaF, [68Ga]Ga-CXCR-4 and [18F]F-FMISO may also provide useful information for tumor characterization and predict of disease outcome. In conclusion, AI using tracers beyond [18F]F-FDG could improve the diagnostic performance of PET-imaging for specific indications and help clinicians in their daily routine by providing features that are often not detectable by the naked eye.
Collapse
Affiliation(s)
- Roya Eisazadeh
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Malihe Shahbazi-Akbari
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Research center for Nuclear Medicine, Department of Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirshahvalad
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Research center for Nuclear Medicine, Department of Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran; Joint Department of Medical Imaging, University Medical Imaging Toronto (UMIT), University Health Network, Mount Sinai Hospital & Women's College Hospital; University of Toronto, Toronto, Ontario, Canada
| | - Christian Pirich
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
6
|
Ahrari S, Zaragori T, Zinsz A, Oster J, Imbert L, Verger A. Application of PET imaging delta radiomics for predicting progression-free survival in rare high-grade glioma. Sci Rep 2024; 14:3256. [PMID: 38332004 PMCID: PMC10853227 DOI: 10.1038/s41598-024-53693-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 02/03/2024] [Indexed: 02/10/2024] Open
Abstract
This study assesses the feasibility of using a sample-efficient model to investigate radiomics changes over time for predicting progression-free survival in rare diseases. Eighteen high-grade glioma patients underwent two L-3,4-dihydroxy-6-[18F]-fluoro-phenylalanine positron emission tomography (PET) dynamic scans: the first during treatment and the second at temozolomide chemotherapy discontinuation. Radiomics features from static/dynamic parametric images, alongside conventional features, were extracted. After excluding highly correlated features, 16 different models were trained by combining various feature selection methods and time-to-event survival algorithms. Performance was assessed using cross-validation. To evaluate model robustness, an additional dataset including 35 patients with a single PET scan at therapy discontinuation was used. Model performance was compared with a strategy extracting informative features from the set of 35 patients and applying them to the 18 patients with 2 PET scans. Delta-absolute radiomics achieved the highest performance when the pipeline was directly applied to the 18-patient subset (support vector machine (SVM) and recursive feature elimination (RFE): C-index = 0.783 [0.744-0.818]). This result remained consistent when transferring informative features from 35 patients (SVM + RFE: C-index = 0.751 [0.716-0.784], p = 0.06). In addition, it significantly outperformed delta-absolute conventional (C-index = 0.584 [0.548-0.620], p < 0.001) and single-time-point radiomics features (C-index = 0.546 [0.512-0.580], p < 0.001), highlighting the considerable potential of delta radiomics in rare cancer cohorts.
Collapse
Affiliation(s)
- Shamimeh Ahrari
- Imagerie Adaptative Diagnostique et Interventionnelle, Institut National de la Santé et de la Recherche Médicale U1254, Université de Lorraine, 54000, Nancy, France
- Nancyclotep Imaging Platform, Université de Lorraine, 54000, Nancy, France
| | - Timothée Zaragori
- Imagerie Adaptative Diagnostique et Interventionnelle, Institut National de la Santé et de la Recherche Médicale U1254, Université de Lorraine, 54000, Nancy, France
- Nancyclotep Imaging Platform, Université de Lorraine, 54000, Nancy, France
| | - Adeline Zinsz
- Department of Nuclear Medicine, Centre Hospitalier Régional Universitaire de Nancy, 54000, Nancy, France
| | - Julien Oster
- Imagerie Adaptative Diagnostique et Interventionnelle, Institut National de la Santé et de la Recherche Médicale U1254, Université de Lorraine, 54000, Nancy, France
| | - Laetitia Imbert
- Imagerie Adaptative Diagnostique et Interventionnelle, Institut National de la Santé et de la Recherche Médicale U1254, Université de Lorraine, 54000, Nancy, France
- Nancyclotep Imaging Platform, Université de Lorraine, 54000, Nancy, France
- Department of Nuclear Medicine, Centre Hospitalier Régional Universitaire de Nancy, 54000, Nancy, France
| | - Antoine Verger
- Imagerie Adaptative Diagnostique et Interventionnelle, Institut National de la Santé et de la Recherche Médicale U1254, Université de Lorraine, 54000, Nancy, France.
- Nancyclotep Imaging Platform, Université de Lorraine, 54000, Nancy, France.
- Department of Nuclear Medicine, Centre Hospitalier Régional Universitaire de Nancy, 54000, Nancy, France.
| |
Collapse
|
7
|
Zhao Z, Song Z, Wang Z, Zhang F, Ding Z, Fan T. Advances in Molecular Pathology, Diagnosis and Treatment of Spinal Cord Astrocytomas. Technol Cancer Res Treat 2024; 23:15330338241262483. [PMID: 39043042 PMCID: PMC11271101 DOI: 10.1177/15330338241262483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 07/25/2024] Open
Abstract
Spinal cord astrocytoma (SCA) is a rare subtype of astrocytoma, posing challenges in diagnosis and treatment. Low-grade SCA can achieve long-term survival solely through surgery, while high-grade has a disappointing prognosis even with comprehensive treatment. Diagnostic criteria and standard treatment of intracranial astrocytoma have shown obvious limitations in SCA. Research on the molecular mechanism in SCA is lagging far behind that on intracranial astrocytoma. In recent years, huge breakthroughs have been made in molecular pathology of astrocytoma, and novel techniques have emerged, including DNA methylation analysis and radiomics. These advances are now making it possible to provide a precise diagnosis and develop corresponding treatment strategies in SCA. Our aim is to review the current status of diagnosis and treatment of SCA, and summarize the latest research advancement, including tumor subtype, molecular characteristics, diagnostic technology, and potential therapy strategies, thus deepening our understanding of this uncommon tumor type and providing guidance for accurate diagnosis and treatment.
Collapse
Affiliation(s)
- Zijun Zhao
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Zihan Song
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zairan Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fan Zhang
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Ze Ding
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Tao Fan
- Spine Center, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Manzarbeitia-Arroba B, Hodolic M, Pichler R, Osipova O, Soriano-Castrejón ÁM, García-Vicente AM. 18F-Fluoroethyl-L Tyrosine Positron Emission Tomography Radiomics in the Differentiation of Treatment-Related Changes from Disease Progression in Patients with Glioblastoma. Cancers (Basel) 2023; 16:195. [PMID: 38201621 PMCID: PMC10778283 DOI: 10.3390/cancers16010195] [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: 10/27/2023] [Revised: 12/10/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
The follow-up of glioma patients after therapeutic intervention remains a challenging topic, as therapy-related changes can emulate true progression in contrast-enhanced magnetic resonance imaging. 18F-fluoroethyl-tyrosine (18F-FET) is a radiopharmaceutical that accumulates in glioma cells due to an increased expression of L-amino acid transporters and, contrary to gadolinium, does not depend on blood-brain barrier disruption to reach tumoral cells. It has demonstrated a high diagnostic value in the differentiation of tumoral viability and pseudoprogression or any other therapy-related changes, especially when combining traditional visual analysis with modern radiomics. In this review, we aim to cover the potential role of 18F-FET positron emission tomography in everyday clinical practice when applied to the follow-up of patients after the first therapeutical intervention, early response evaluation, and the differential diagnosis between therapy-related changes and progression.
Collapse
Affiliation(s)
| | - Marina Hodolic
- Nuclear Medicine Department, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic;
| | - Robert Pichler
- Institute of Nuclear Medicine Kepler University Hospital—Neuromed Campus, 4020 Linz, Austria; (R.P.); (O.O.)
| | - Olga Osipova
- Institute of Nuclear Medicine Kepler University Hospital—Neuromed Campus, 4020 Linz, Austria; (R.P.); (O.O.)
| | | | - Ana María García-Vicente
- Nuclear Medicine Department, University Hospital of Toledo, 45007 Toledo, Spain; (B.M.-A.); (Á.M.S.-C.)
| |
Collapse
|
9
|
Rogasch JMM, Shi K, Kersting D, Seifert R. Methodological evaluation of original articles on radiomics and machine learning for outcome prediction based on positron emission tomography (PET). Nuklearmedizin 2023; 62:361-369. [PMID: 37995708 PMCID: PMC10667066 DOI: 10.1055/a-2198-0545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023]
Abstract
AIM Despite a vast number of articles on radiomics and machine learning in positron emission tomography (PET) imaging, clinical applicability remains limited, partly owing to poor methodological quality. We therefore systematically investigated the methodology described in publications on radiomics and machine learning for PET-based outcome prediction. METHODS A systematic search for original articles was run on PubMed. All articles were rated according to 17 criteria proposed by the authors. Criteria with >2 rating categories were binarized into "adequate" or "inadequate". The association between the number of "adequate" criteria per article and the date of publication was examined. RESULTS One hundred articles were identified (published between 07/2017 and 09/2023). The median proportion of articles per criterion that were rated "adequate" was 65% (range: 23-98%). Nineteen articles (19%) mentioned neither a test cohort nor cross-validation to separate training from testing. The median number of criteria with an "adequate" rating per article was 12.5 out of 17 (range, 4-17), and this did not increase with later dates of publication (Spearman's rho, 0.094; p = 0.35). In 22 articles (22%), less than half of the items were rated "adequate". Only 8% of articles published the source code, and 10% made the dataset openly available. CONCLUSION Among the articles investigated, methodological weaknesses have been identified, and the degree of compliance with recommendations on methodological quality and reporting shows potential for improvement. Better adherence to established guidelines could increase the clinical significance of radiomics and machine learning for PET-based outcome prediction and finally lead to the widespread use in routine clinical practice.
Collapse
Affiliation(s)
- Julian Manuel Michael Rogasch
- Department of Nuclear Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital University Hospital Bern, Bern, Switzerland
| | - David Kersting
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
10
|
Hajianfar G, Haddadi Avval A, Hosseini SA, Nazari M, Oveisi M, Shiri I, Zaidi H. Time-to-event overall survival prediction in glioblastoma multiforme patients using magnetic resonance imaging radiomics. LA RADIOLOGIA MEDICA 2023; 128:1521-1534. [PMID: 37751102 PMCID: PMC10700216 DOI: 10.1007/s11547-023-01725-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 09/05/2023] [Indexed: 09/27/2023]
Abstract
PURPOSE Glioblastoma Multiforme (GBM) represents the predominant aggressive primary tumor of the brain with short overall survival (OS) time. We aim to assess the potential of radiomic features in predicting the time-to-event OS of patients with GBM using machine learning (ML) algorithms. MATERIALS AND METHODS One hundred nineteen patients with GBM, who had T1-weighted contrast-enhanced and T2-FLAIR MRI sequences, along with clinical data and survival time, were enrolled. Image preprocessing methods included 64 bin discretization, Laplacian of Gaussian (LOG) filters with three Sigma values and eight variations of Wavelet Transform. Images were then segmented, followed by the extraction of 1212 radiomic features. Seven feature selection (FS) methods and six time-to-event ML algorithms were utilized. The combination of preprocessing, FS, and ML algorithms (12 × 7 × 6 = 504 models) was evaluated by multivariate analysis. RESULTS Our multivariate analysis showed that the best prognostic FS/ML combinations are the Mutual Information (MI)/Cox Boost, MI/Generalized Linear Model Boosting (GLMB) and MI/Generalized Linear Model Network (GLMN), all of which were done via the LOG (Sigma = 1 mm) preprocessing method (C-index = 0.77). The LOG filter with Sigma = 1 mm preprocessing method, MI, GLMB and GLMN achieved significantly higher C-indices than other preprocessing, FS, and ML methods (all p values < 0.05, mean C-indices of 0.65, 0.70, and 0.64, respectively). CONCLUSION ML algorithms are capable of predicting the time-to-event OS of patients using MRI-based radiomic and clinical features. MRI-based radiomics analysis in combination with clinical variables might appear promising in assisting clinicians in the survival prediction of patients with GBM. Further research is needed to establish the applicability of radiomics in the management of GBM in the clinic.
Collapse
Affiliation(s)
- Ghasem Hajianfar
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, 1211, Geneva, Switzerland
| | | | - Seyyed Ali Hosseini
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Montréal, QC, Canada
| | - Mostafa Nazari
- Department of Medical Physics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Oveisi
- Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, 1211, Geneva, Switzerland
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, 1211, Geneva, Switzerland.
- Geneva University Neurocenter, Geneva University, Geneva, Switzerland.
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.
- Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
11
|
Chen C, Du X, Yang L, Liu H, Li Z, Gou Z, Qi J. Research on application of radiomics in glioma: a bibliometric and visual analysis. Front Oncol 2023; 13:1083080. [PMID: 37771434 PMCID: PMC10523166 DOI: 10.3389/fonc.2023.1083080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 08/16/2023] [Indexed: 09/30/2023] Open
Abstract
Background With the continuous development of medical imaging informatics technology, radiomics has become a new and evolving field in medical applications. Radiomics aims to be an aid to support clinical decision making by extracting quantitative features from medical images and has a very wide range of applications. The purpose of this study was to perform a bibliometric and visual analysis of scientific results and research trends in the research application of radiomics in glioma. Methods We searched the Web of Science Core Collection (WOScc) for publications related to glioma radiomics. A bibliometric and visual analysis of online publications in this field related to countries/regions, authors, journals, references and keywords was performed using CiteSpace and R software. Results A total of 587 relevant literature published from 2012 to September 2022 were retrieved in WOScc, and finally a total of 484 publications were obtained according to the filtering criteria, including 393 (81.20%) articles and 91 (18.80%) reviews. The number of relevant publications increases year by year. The highest number of publications was from the USA (171 articles, 35.33%) and China (170 articles, 35.12%). The research institution with the highest number of publications was Chinese Acad Sci (24), followed by Univ Penn (22) and Fudan Univ (21). WANG Y (27) had the most publications, followed by LI Y (22), and WANG J (20). Among the 555 co-cited authors, LOUIS DN (207) and KICKINGEREDER P (207) were the most cited authors. FRONTIERS IN ONCOLOGY (42) was the most published journal and NEURO-ONCOLOGY (412) was the most co-cited journal. The most frequent keywords in all publications included glioblastoma (187), survival (136), classification (131), magnetic resonance imaging (113), machine learning (100), tumor (82), and feature (79), central nervous system (66), IDH (57), and radiomics (55). Cluster analysis was performed on the basis of keyword co-occurrence, and a total of 16 clusters were formed, indicating that these directions are the current hotspots of radiomics research applications in glioma and may be the future directions of continuous development. Conclusion In the past decade, radiomics has received much attention in the medical field and has been widely used in clinical research applications. Cooperation and communication between countries/regions need to be enhanced in future research to promote the development of radiomics in the field of medicine. In addition, the application of radiomics has improved the accuracy of pre-treatment diagnosis, efficacy prediction and prognosis assessment of glioma and helped to promote the development into precision medicine, the future still faces many challenges.
Collapse
Affiliation(s)
- Chunbao Chen
- Department of Neurosurgery, Afiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xue Du
- Department of Oncology, The People's Hospital of Hechuan, Chongqing, China
- Department of Oncology, North Sichuan Medical College, Nanchong, China
| | - Lu Yang
- Department of Oncology, Suining Central Hospital, Suining, China
| | - Hongjun Liu
- Department of Neurosurgery, Afiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhou Li
- Department of Neurosurgery, Nanchong Central Hospital, The Afiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhangyang Gou
- Department of Neurosurgery, Afiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jian Qi
- Department of Neurosurgery, Afiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
12
|
Tan D, Mohamad Salleh SA, Manan HA, Yahya N. Delta-radiomics-based models for toxicity prediction in radiotherapy: A systematic review and meta-analysis. J Med Imaging Radiat Oncol 2023; 67:564-579. [PMID: 37309680 DOI: 10.1111/1754-9485.13546] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/28/2023] [Indexed: 06/14/2023]
Abstract
INTRODUCTION Delta-radiomics models are potentially able to improve the treatment assessment than single-time point features. The purpose of this study is to systematically synthesize the performance of delta-radiomics-based models for radiotherapy (RT)-induced toxicity. METHODS A literature search was performed following the PRISMA guidelines. Systematic searches were performed in PubMed, Scopus, Cochrane and Embase databases in October 2022. Retrospective and prospective studies on the delta-radiomics model for RT-induced toxicity were included based on predefined PICOS criteria. A random-effect meta-analysis of AUC was performed on the performance of delta-radiomics models, and a comparison with non-delta radiomics models was included. RESULTS Of the 563 articles retrieved, 13 selected studies of RT-treated patients on different types of cancer (HNC = 571, NPC = 186, NSCLC = 165, oesophagus = 106, prostate = 33, OPC = 21) were eligible for inclusion in the systematic review. Included studies show that morphological and dosimetric features may improve the predictive model performance for the selected toxicity. Four studies that reported both delta and non-delta radiomics features with AUC were included in the meta-analysis. The AUC random effects estimate for delta and non-delta radiomics models were 0.80 and 0.78 with heterogeneity, I2 of 73% and 27% respectively. CONCLUSION Delta-radiomics-based models were found to be promising predictors of predefined end points. Future studies should consider using standardized methods and radiomics features and external validation to the reviewed delta-radiomics model.
Collapse
Affiliation(s)
- Daryl Tan
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | - Hanani Abdul Manan
- Functional Image Processing Laboratory, Department of Radiology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Noorazrul Yahya
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Marmorino F, Faggioni L, Rossini D, Gabelloni M, Goddi A, Ferrer L, Conca V, Vargas J, Biagiarelli F, Daniel F, Carullo M, Vetere G, Granetto C, Boccaccio C, Cioni D, Antonuzzo L, Bergamo F, Pietrantonio F, Cremolini C, Neri E. The prognostic value of radiomic features in liver-limited metastatic colorectal cancer patients from the TRIBE2 study. Future Oncol 2023; 19:1601-1611. [PMID: 37577810 DOI: 10.2217/fon-2023-0406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023] Open
Abstract
Aims: Evaluating the prognostic role of radiomic features in liver-limited metastatic colorectal cancer treated with first-line therapy at baseline and best response among patients undergoing resection. Patients & methods: Among patients enrolled in TRIBE2 (NCT02339116), the association of clinical and radiomic data, extracted by SOPHiA-DDM™ with progression-free and overall survival (OS) in the overall population and with disease-free survival/postresection OS in those undergoing resection was investigated. Results: Among 98 patients, radiomic parameters improved the prediction accuracy of our model for OS (area under the curve: 0.83; sensitivity: 0.85; specificity: 0.73; accuracy: 0.78), but not progression-free survival. Of 46 resected patients, small-distance high gray-level emphasis was associated with shorter disease-free survival and high gray-level zone emphasis/higher kurtosis with shorter postresection OS. Conclusion: Radiomic features should be implemented as tools of outcome prediction for liver-limited metastatic colorectal cancer.
Collapse
Affiliation(s)
- Federica Marmorino
- Unit of Oncology, University Hospital of Pisa, Pisa, Italy & Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Lorenzo Faggioni
- Academic Radiology, Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Daniele Rossini
- Unit of Oncology, University Hospital of Pisa, Pisa, Italy & Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Michela Gabelloni
- Academic Radiology, Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Antonio Goddi
- Academic Radiology, Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Loïc Ferrer
- SOPHiA GENETICS, Multimodal Research team, Cité de la Photonique, 11 avenue de Canteranne, 33600, PESSAC, France
| | - Veronica Conca
- Unit of Oncology, University Hospital of Pisa, Pisa, Italy & Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Jennifer Vargas
- SOPHiA GENETICS, Multimodal Research team, Cité de la Photonique, 11 avenue de Canteranne, 33600, PESSAC, France
| | | | - Francesca Daniel
- Oncology Unit 1, Veneto Institute of Oncology IOV - IRCCS, 35128, Padova, Italy
| | - Martina Carullo
- Unit of Oncology, University Hospital of Pisa, Pisa, Italy & Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Guglielmo Vetere
- Unit of Oncology, University Hospital of Pisa, Pisa, Italy & Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Cristina Granetto
- SC Oncologia AO S. Croce & Carle, University Teaching Hospital, Via A. Carle 25, 12100, Cuneo, Italy
| | - Chiara Boccaccio
- Unit of Oncology, University Hospital of Pisa, Pisa, Italy & Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Dania Cioni
- Academic Radiology, Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Department of Experimental & Clinical Medicine, University of Florence, Viale Pieraccini 6, 50139, Firenze, Italy
| | - Francesca Bergamo
- Oncology Unit 1, Veneto Institute of Oncology IOV - IRCCS, 35128, Padova, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milano, Italy
| | - Chiara Cremolini
- Unit of Oncology, University Hospital of Pisa, Pisa, Italy & Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Emanuele Neri
- Academic Radiology, Department of Translational Research & New Technologies in Medicine & Surgery, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| |
Collapse
|
14
|
Cruz N, Herculano-Carvalho M, Roque D, Faria CC, Cascão R, Ferreira HA, Reis CP, Matela N. Highlighted Advances in Therapies for Difficult-To-Treat Brain Tumours Such as Glioblastoma. Pharmaceutics 2023; 15:pharmaceutics15030928. [PMID: 36986790 PMCID: PMC10054750 DOI: 10.3390/pharmaceutics15030928] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/25/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023] Open
Abstract
Glioblastoma multiforme (GBM) remains a challenging disease, as it is the most common and deadly brain tumour in adults and has no curative solution and an overall short survival time. This incurability and short survival time means that, despite its rarity (average incidence of 3.2 per 100,000 persons), there has been an increased effort to try to treat this disease. Standard of care in newly diagnosed glioblastoma is maximal tumour resection followed by initial concomitant radiotherapy and temozolomide (TMZ) and then further chemotherapy with TMZ. Imaging techniques are key not only to diagnose the extent of the affected tissue but also for surgery planning and even for intraoperative use. Eligible patients may combine TMZ with tumour treating fields (TTF) therapy, which delivers low-intensity and intermediate-frequency electric fields to arrest tumour growth. Nonetheless, the blood–brain barrier (BBB) and systemic side effects are obstacles to successful chemotherapy in GBM; thus, more targeted, custom therapies such as immunotherapy and nanotechnological drug delivery systems have been undergoing research with varying degrees of success. This review proposes an overview of the pathophysiology, possible treatments, and the most (not all) representative examples of the latest advancements.
Collapse
Affiliation(s)
- Nuno Cruz
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- iMED.ULisboa, Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Manuel Herculano-Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), 1649-028 Lisboa, Portugal
| | - Diogo Roque
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), 1649-028 Lisboa, Portugal
| | - Cláudia C. Faria
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), 1649-028 Lisboa, Portugal
| | - Rita Cascão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Hugo Alexandre Ferreira
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Catarina Pinto Reis
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- iMED.ULisboa, Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Correspondence: (C.P.R.); (N.M.); Tel.: +351-217-946-400 (ext. 14244) (C.P.R.); Fax: +351-217-946-470 (C.P.R.)
| | - Nuno Matela
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- Correspondence: (C.P.R.); (N.M.); Tel.: +351-217-946-400 (ext. 14244) (C.P.R.); Fax: +351-217-946-470 (C.P.R.)
| |
Collapse
|
15
|
Soni N, Ora M, Jena A, Rana P, Mangla R, Ellika S, Almast J, Puri S, Meyers SP. Amino Acid Tracer PET MRI in Glioma Management: What a Neuroradiologist Needs to Know. AJNR Am J Neuroradiol 2023; 44:236-246. [PMID: 36657945 PMCID: PMC10187808 DOI: 10.3174/ajnr.a7762] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/21/2022] [Indexed: 01/21/2023]
Abstract
PET with amino acid tracers provides additional insight beyond MR imaging into the biology of gliomas that can be used for initial diagnosis, delineation of tumor margins, planning of surgical and radiation therapy, assessment of residual tumor, and evaluation of posttreatment response. Hybrid PET MR imaging allows the simultaneous acquisition of various PET and MR imaging parameters in a single investigation with reduced scanning time and improved anatomic localization. This review aimed to provide neuroradiologists with a concise overview of the various amino acid tracers and a practical understanding of the clinical applications of amino acid PET MR imaging in glioma management. Future perspectives in newer advances, novel radiotracers, radiomics, and cost-effectiveness are also outlined.
Collapse
Affiliation(s)
- N Soni
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - M Ora
- Sanjay Gandhi Postgraduate Institute of Medical Sciences (M.O.), Lucknow, Uttar Pradesh, India
| | - A Jena
- Indraprastha Apollo Hospital (A.J., P.R.), New Delhi, India
| | - P Rana
- Indraprastha Apollo Hospital (A.J., P.R.), New Delhi, India
| | - R Mangla
- Upstate University Hospital (R.M.), Syracuse, New York
| | - S Ellika
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - J Almast
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - S Puri
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| | - S P Meyers
- From the University of Rochester Medical Center (N.S., S.E., J.A., S.P., S.M.), Rochester, New York
| |
Collapse
|
16
|
Danilov G, Kalayeva D, Vikhrova N, Konakova T, Zagorodnova A, Popova A, Postnov A, Shugay S, Pronin I. Radiomics in Determining Tumor-to-Normal Brain SUV Ratio Based on 11C-Methionine PET/CT in Glioblastoma. Sovrem Tekhnologii Med 2023; 15:5-11. [PMID: 37388754 PMCID: PMC10306961 DOI: 10.17691/stm2023.15.1.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Indexed: 12/10/2023] Open
Abstract
UNLABELLED Modern methodology of PET/CT quantitative analysis in patients with glioblastomas is not strictly standardized in clinic settings and does not exclude the influence of the human factor. Methods of radiomics may facilitate unification, and improve objectivity and efficiency of the medical image analysis. The aim of the study is to evaluate the potential of radiomics in the analysis of PET/CT glioblastoma images identifying the relationship between the radiomic features and the 11С-methionine tumor-to-normal brain uptake ratio (TNR) determined by an expert in routine. MATERIALS AND METHODS PET/CT data (2018-2020) from 40 patients (average age was 55±12 years; 77.5% were males) with a histologically confirmed diagnosis of "glioblastoma" were included in the analysis. TNR was calculated as a ratio of the standardized uptake value of 11C-methionine measured in the tumor and intact tissue. Calculation of radiomic features for each PET was performed in the specified volumetric region of interest, capturing the tumor with the surrounding tissues. The relationship between TNR and the radiomic features was determined using the linear regression model. Predictors were included in the model following correlation analysis and LASSO regularization. The experiment with machine learning was repeated 300 times, splitting the training (70%) and test (30%) subsets randomly. The model quality metrics and predictor significance obtained in 300 tests were summarized. RESULTS Of 412 PET/CT radiomic parameters significantly correlated with TNR (p<0.05), the regularization procedure left no more than 30 in each model (the median number of predictors was 9 [7; 13]). The experiment has demonstrated a non-random linear correlation (the Spearman correlation coefficient was 0.58 [0.43; 0.74]) between TNR and separate radiomic features, primarily fractal dimensions, characterizing the geometrical properties of the image. CONCLUSION Radiomics enabled an objective determination of PET/CT image texture features reflecting the biological activity of glioblastomas. Despite the existing limitations in the application, the first results provide a good perspective of these methods in neurooncology.
Collapse
Affiliation(s)
- G.V. Danilov
- Scientific Board Secretary, Head of the Laboratory of Biomedical Informatics and Artificial Intelligence; N.N. Burdenko National Medical Research Center for Neurosurgery, Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - D.B. Kalayeva
- Medical Physicist; N.N. Burdenko National Medical Research Center for Neurosurgery, Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia; PhD Student; National Research Nuclear University MEPhI, 31 Kashirskoe Shosse, Moscow, 115409, Russia
| | - N.B. Vikhrova
- Radiologist; N.N. Burdenko National Medical Research Center for Neurosurgery, Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - T.A. Konakova
- PhD Student; N.N. Burdenko National Medical Research Center for Neurosurgery, Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - A.I. Zagorodnova
- Student; Pirogov Russian National Research Medical University, 1 Ostrovitianova St., Moscow, 117997, Russia
| | - A.A. Popova
- Student; Pirogov Russian National Research Medical University, 1 Ostrovitianova St., Moscow, 117997, Russia
| | - A.A. Postnov
- Researcher; N.N. Burdenko National Medical Research Center for Neurosurgery, Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia; Associate Professor; National Research Nuclear University MEPhI, 31 Kashirskoe Shosse, Moscow, 115409, Russia; Researcher; Lebedev Physical Institute of the Russian Academy of Sciences, 53 Leninskiy Prospect, Moscow, 119991, Russia; Head of the Project; Research Institute of Technical Physics and Automation, 46 Varshavskoye Shosse, Moscow, 115230, Russia
| | - S.V. Shugay
- Professor, Academician of the Russian Academy of Sciences, Deputy Director for Research, Head of the Unit for X-ray and Radioisotopic Methods of Diagnosis; N.N. Burdenko National Medical Research Center for Neurosurgery, Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - I.N. Pronin
- Pathologist; N.N. Burdenko National Medical Research Center for Neurosurgery, Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| |
Collapse
|
17
|
Popp I, Oehlke O, Nieder C, Grosu AL. Brain Gliomas of Adulthood. TARGET VOLUME DEFINITION IN RADIATION ONCOLOGY 2023:1-20. [DOI: 10.1007/978-3-031-45489-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
18
|
Wiltgen T, Fleischmann DF, Kaiser L, Holzgreve A, Corradini S, Landry G, Ingrisch M, Popp I, Grosu AL, Unterrainer M, Bartenstein P, Parodi K, Belka C, Albert N, Niyazi M, Riboldi M. 18F-FET PET radiomics-based survival prediction in glioblastoma patients receiving radio(chemo)therapy. Radiat Oncol 2022; 17:198. [DOI: 10.1186/s13014-022-02164-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 10/07/2022] [Indexed: 12/04/2022] Open
Abstract
Abstract
Background
Quantitative image analysis based on radiomic feature extraction is an emerging field for survival prediction in oncological patients. 18F-Fluorethyltyrosine positron emission tomography (18F-FET PET) provides important diagnostic and grading information for brain tumors, but data on its use in survival prediction is scarce. In this study, we aim at investigating survival prediction based on multiple radiomic features in glioblastoma patients undergoing radio(chemo)therapy.
Methods
A dataset of 37 patients with glioblastoma (WHO grade 4) receiving radio(chemo)therapy was analyzed. Radiomic features were extracted from pre-treatment 18F-FET PET images, following intensity rebinning with a fixed bin width. Principal component analysis (PCA) was applied for variable selection, aiming at the identification of the most relevant features in survival prediction. Random forest classification and prediction algorithms were optimized on an initial set of 25 patients. Testing of the implemented algorithms was carried out in different scenarios, which included additional 12 patients whose images were acquired with a different scanner to check the reproducibility in prediction results.
Results
First order intensity variations and shape features were predominant in the selection of most important radiomic signatures for survival prediction in the available dataset. The major axis length of the 18F-FET-PET volume at tumor to background ratio (TBR) 1.4 and 1.6 correlated significantly with reduced probability of survival. Additional radiomic features were identified as potential survival predictors in the PTV region, showing 76% accuracy in independent testing for both classification and regression.
Conclusions
18F-FET PET prior to radiation provides relevant information for survival prediction in glioblastoma patients. Based on our preliminary analysis, radiomic features in the PTV can be considered a robust dataset for survival prediction.
Collapse
|
19
|
Radiomic and Volumetric Measurements as Clinical Trial Endpoints—A Comprehensive Review. Cancers (Basel) 2022; 14:cancers14205076. [PMID: 36291865 PMCID: PMC9599928 DOI: 10.3390/cancers14205076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary The extraction of quantitative data from standard-of-care imaging modalities offers opportunities to improve the relevance and salience of imaging biomarkers used in drug development. This review aims to identify the challenges and opportunities for discovering new imaging-based biomarkers based on radiomic and volumetric assessment in the single-site solid tumor sites: breast cancer, rectal cancer, lung cancer and glioblastoma. Developing approaches to harmonize three essential areas: segmentation, validation and data sharing may expedite regulatory approval and adoption of novel cancer imaging biomarkers. Abstract Clinical trials for oncology drug development have long relied on surrogate outcome biomarkers that assess changes in tumor burden to accelerate drug registration (i.e., Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1) criteria). Drug-induced reduction in tumor size represents an imperfect surrogate marker for drug activity and yet a radiologically determined objective response rate is a widely used endpoint for Phase 2 trials. With the addition of therapies targeting complex biological systems such as immune system and DNA damage repair pathways, incorporation of integrative response and outcome biomarkers may add more predictive value. We performed a review of the relevant literature in four representative tumor types (breast cancer, rectal cancer, lung cancer and glioblastoma) to assess the preparedness of volumetric and radiomics metrics as clinical trial endpoints. We identified three key areas—segmentation, validation and data sharing strategies—where concerted efforts are required to enable progress of volumetric- and radiomics-based clinical trial endpoints for wider clinical implementation.
Collapse
|
20
|
Martin P, Holloway L, Metcalfe P, Koh ES, Brighi C. Challenges in Glioblastoma Radiomics and the Path to Clinical Implementation. Cancers (Basel) 2022; 14:3897. [PMID: 36010891 PMCID: PMC9406186 DOI: 10.3390/cancers14163897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/04/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Radiomics is a field of medical imaging analysis that focuses on the extraction of many quantitative imaging features related to shape, intensity and texture. These features are incorporated into models designed to predict important clinical or biological endpoints for patients. Attention for radiomics research has recently grown dramatically due to the increased use of imaging and the availability of large, publicly available imaging datasets. Glioblastoma multiforme (GBM) patients stand to benefit from this emerging research field as radiomics has the potential to assess the biological heterogeneity of the tumour, which contributes significantly to the inefficacy of current standard of care therapy. Radiomics models still require further development before they are implemented clinically in GBM patient management. Challenges relating to the standardisation of the radiomics process and the validation of radiomic models impede the progress of research towards clinical implementation. In this manuscript, we review the current state of radiomics in GBM, and we highlight the barriers to clinical implementation and discuss future validation studies needed to advance radiomics models towards clinical application.
Collapse
Affiliation(s)
- Philip Martin
- Centre for Medical and Radiation Physics, School of Physics, University of Wollongong, Wollongong, NSW 2522, Australia
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
| | - Lois Holloway
- Centre for Medical and Radiation Physics, School of Physics, University of Wollongong, Wollongong, NSW 2522, Australia
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
- Liverpool and Macarthur Cancer Therapy Centres, Liverpool, NSW 2170, Australia
- South Western Sydney Clinical Campus, School of Medicine, University of New South Wales, Liverpool, NSW 2170, Australia
| | - Peter Metcalfe
- Centre for Medical and Radiation Physics, School of Physics, University of Wollongong, Wollongong, NSW 2522, Australia
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
| | - Eng-Siew Koh
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
- Liverpool and Macarthur Cancer Therapy Centres, Liverpool, NSW 2170, Australia
- South Western Sydney Clinical Campus, School of Medicine, University of New South Wales, Liverpool, NSW 2170, Australia
| | - Caterina Brighi
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
- ACRF Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
21
|
Jian A, Liu S, Di Ieva A. Artificial Intelligence for Survival Prediction in Brain Tumors on Neuroimaging. Neurosurgery 2022; 91:8-26. [PMID: 35348129 DOI: 10.1227/neu.0000000000001938] [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] [Received: 06/30/2021] [Accepted: 01/08/2022] [Indexed: 12/30/2022] Open
Abstract
Survival prediction of patients affected by brain tumors provides essential information to guide surgical planning, adjuvant treatment selection, and patient counseling. Current reliance on clinical factors, such as Karnofsky Performance Status Scale, and simplistic radiological characteristics are, however, inadequate for survival prediction in tumors such as glioma that demonstrate molecular and clinical heterogeneity with variable survival outcomes. Advances in the domain of artificial intelligence have afforded powerful tools to capture a large number of hidden high-dimensional imaging features that reflect abundant information about tumor structure and physiology. Here, we provide an overview of current literature that apply computational analysis tools such as radiomics and machine learning methods to the pipeline of image preprocessing, tumor segmentation, feature extraction, and construction of classifiers to establish survival prediction models based on neuroimaging. We also discuss challenges relating to the development and evaluation of such models and explore ethical issues surrounding the future use of machine learning predictions.
Collapse
Affiliation(s)
- Anne Jian
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Royal Melbourne Hospital, Melbourne, Australia
| | - Sidong Liu
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Centre for Health Informatics, Australian Institute of Health Innovation, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Antonio Di Ieva
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
22
|
Morland D, Triumbari EKA, Boldrini L, Gatta R, Pizzuto D, Annunziata S. Radiomics in Oncological PET Imaging: A Systematic Review-Part 1, Supradiaphragmatic Cancers. Diagnostics (Basel) 2022; 12:1329. [PMID: 35741138 PMCID: PMC9221970 DOI: 10.3390/diagnostics12061329] [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: 05/04/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/10/2022] Open
Abstract
Radiomics is an upcoming field in nuclear oncology, both promising and technically challenging. To summarize the already undertaken work on supradiaphragmatic neoplasia and assess its quality, we performed a literature search in the PubMed database up to 18 February 2022. Inclusion criteria were: studies based on human data; at least one specified tumor type; supradiaphragmatic malignancy; performing radiomics on PET imaging. Exclusion criteria were: studies only based on phantom or animal data; technical articles without a clinically oriented question; fewer than 30 patients in the training cohort. A review database containing PMID, year of publication, cancer type, and quality criteria (number of patients, retrospective or prospective nature, independent validation cohort) was constructed. A total of 220 studies met the inclusion criteria. Among them, 119 (54.1%) studies included more than 100 patients, 21 studies (9.5%) were based on prospectively acquired data, and 91 (41.4%) used an independent validation set. Most studies focused on prognostic and treatment response objectives. Because the textural parameters and methods employed are very different from one article to another, it is complicated to aggregate and compare articles. New contributions and radiomics guidelines tend to help improving quality of the reported studies over the years.
Collapse
Affiliation(s)
- David Morland
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
- Service de Médecine Nucléaire, Institut Godinot, 51100 Reims, France
- Laboratoire de Biophysique, UFR de Médecine, Université de Reims Champagne-Ardenne, 51100 Reims, France
- CReSTIC (Centre de Recherche en Sciences et Technologies de l’Information et de la Communication), EA 3804, Université de Reims Champagne-Ardenne, 51100 Reims, France
| | - Elizabeth Katherine Anna Triumbari
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Luca Boldrini
- Radiotherapy Unit, Radiomics, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (L.B.); (R.G.)
| | - Roberto Gatta
- Radiotherapy Unit, Radiomics, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (L.B.); (R.G.)
- Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
- Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Daniele Pizzuto
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Salvatore Annunziata
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
| |
Collapse
|
23
|
PET Imaging in Neuro-Oncology: An Update and Overview of a Rapidly Growing Area. Cancers (Basel) 2022; 14:cancers14051103. [PMID: 35267411 PMCID: PMC8909369 DOI: 10.3390/cancers14051103] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/08/2022] [Accepted: 02/19/2022] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Positron emission tomography (PET) is a functional imaging technique which plays an increasingly important role in the management of brain tumors. Owing different radiotracers, PET allows to image different metabolic aspects of the brain tumors. This review outlines currently available PET radiotracers and their respective indications in neuro-oncology. It specifically focuses on the investigation of gliomas, meningiomas, primary central nervous system lymphomas as well as brain metastases. Recent advances in the production of PET radiotracers, image analyses and translational applications to peptide radionuclide receptor therapy, which allow to treat brain tumors with radiotracers, are also discussed. The objective of this review is to provide a comprehensive overview of PET imaging’s potential in neuro-oncology as an adjunct to brain magnetic resonance imaging (MRI). Abstract PET plays an increasingly important role in the management of brain tumors. This review outlines currently available PET radiotracers and their respective indications. It specifically focuses on 18F-FDG, amino acid and somatostatin receptor radiotracers, for imaging gliomas, meningiomas, primary central nervous system lymphomas as well as brain metastases. Recent advances in radiopharmaceuticals, image analyses and translational applications to therapy are also discussed. The objective of this review is to provide a comprehensive overview of PET imaging’s potential in neuro-oncology as an adjunct to brain MRI for all medical professionals implicated in brain tumor diagnosis and care.
Collapse
|
24
|
Ahrari S, Zaragori T, Rozenblum L, Oster J, Imbert L, Kas A, Verger A. Relevance of Dynamic 18F-DOPA PET Radiomics for Differentiation of High-Grade Glioma Progression from Treatment-Related Changes. Biomedicines 2021; 9:biomedicines9121924. [PMID: 34944740 PMCID: PMC8698938 DOI: 10.3390/biomedicines9121924] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 12/22/2022] Open
Abstract
This study evaluates the relevance of 18F-DOPA PET static and dynamic radiomics for differentiation of high-grade glioma (HGG) progression from treatment-related changes (TRC) by comparing diagnostic performances to the current PET imaging standard of care. Eighty-five patients with histologically confirmed HGG and investigated by dynamic 18F-FDOPA PET in two institutions were retrospectively selected. ElasticNet logistic regression, Random Forest and XGBoost machine models were trained with different sets of features-radiomics extracted from static tumor-to-background-ratio (TBR) parametric images, radiomics extracted from time-to-peak (TTP) parametric images, as well as combination of both-in order to discriminate glioma progression from TRC at 6 months from the PET scan. Diagnostic performances of the models were compared to a logistic regression model with TBRmean ± clinical features used as reference. Training was performed on data from the first center, while external validation was performed on data from the second center. Best radiomics models showed only slightly better performances than the reference model (respective AUCs of 0.834 vs. 0.792, p < 0.001). Our current results show similar findings at the multicentric level using different machine learning models and report a marginal additional value for TBR static and TTP dynamic radiomics over the classical analysis based on TBR values.
Collapse
Affiliation(s)
- Shamimeh Ahrari
- Université de Lorraine, IADI, INSERM, UMR 1254, F-54000 Nancy, France; (S.A.); (T.Z.); (J.O.); (L.I.)
| | - Timothée Zaragori
- Université de Lorraine, IADI, INSERM, UMR 1254, F-54000 Nancy, France; (S.A.); (T.Z.); (J.O.); (L.I.)
| | - Laura Rozenblum
- Sorbonne Université, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Service de Médecine Nucléaire and LIB, INSERM U1146, F-75013 Paris, France; (L.R.); (A.K.)
| | - Julien Oster
- Université de Lorraine, IADI, INSERM, UMR 1254, F-54000 Nancy, France; (S.A.); (T.Z.); (J.O.); (L.I.)
| | - Laëtitia Imbert
- Université de Lorraine, IADI, INSERM, UMR 1254, F-54000 Nancy, France; (S.A.); (T.Z.); (J.O.); (L.I.)
- Department of Nuclear Medicine & Nancyclotep Imaging Platform, Université de Lorraine, CHRU-Nancy, F-54000 Nancy, France
| | - Aurélie Kas
- Sorbonne Université, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Service de Médecine Nucléaire and LIB, INSERM U1146, F-75013 Paris, France; (L.R.); (A.K.)
| | - Antoine Verger
- Université de Lorraine, IADI, INSERM, UMR 1254, F-54000 Nancy, France; (S.A.); (T.Z.); (J.O.); (L.I.)
- Department of Nuclear Medicine & Nancyclotep Imaging Platform, Université de Lorraine, CHRU-Nancy, F-54000 Nancy, France
- Correspondence:
| |
Collapse
|
25
|
Li Z, Kaiser L, Holzgreve A, Ruf VC, Suchorska B, Wenter V, Quach S, Herms J, Bartenstein P, Tonn JC, Unterrainer M, Albert NL. Prediction of TERTp-mutation status in IDH-wildtype high-grade gliomas using pre-treatment dynamic [ 18F]FET PET radiomics. Eur J Nucl Med Mol Imaging 2021; 48:4415-4425. [PMID: 34490493 PMCID: PMC8566644 DOI: 10.1007/s00259-021-05526-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/05/2021] [Indexed: 12/22/2022]
Abstract
Purpose To evaluate radiomic features extracted from standard static images (20–40 min p.i.), early summation images (5–15 min p.i.), and dynamic [18F]FET PET images for the prediction of TERTp-mutation status in patients with IDH-wildtype high-grade glioma. Methods A total of 159 patients (median age 60.2 years, range 19–82 years) with newly diagnosed IDH-wildtype diffuse astrocytic glioma (WHO grade III or IV) and dynamic [18F]FET PET prior to surgical intervention were enrolled and divided into a training (n = 112) and a testing cohort (n = 47) randomly. First-order, shape, and texture radiomic features were extracted from standard static (20–40 min summation images; TBR20–40), early static (5–15 min summation images; TBR5–15), and dynamic (time-to-peak; TTP) images, respectively. Recursive feature elimination was used for feature selection by 10-fold cross-validation in the training cohort after normalization, and logistic regression models were generated using the radiomic features extracted from each image to differentiate TERTp-mutation status. The areas under the ROC curve (AUC), accuracy, sensitivity, specificity, and positive and negative predictive value were calculated to illustrate diagnostic power in both the training and testing cohort. Results The TTP model comprised nine selected features and achieved highest predictability of TERTp-mutation with an AUC of 0.82 (95% confidence interval 0.71–0.92) and sensitivity of 92.1% in the independent testing cohort. Weak predictive capability was obtained in the TBR5–15 model, with an AUC of 0.61 (95% CI 0.42–0.80) in the testing cohort, while no predictive power was observed in the TBR20–40 model. Conclusions Radiomics based on TTP images extracted from dynamic [18F]FET PET can predict the TERTp-mutation status of IDH-wildtype diffuse astrocytic high-grade gliomas with high accuracy preoperatively. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05526-6.
Collapse
Affiliation(s)
- Zhicong Li
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Viktoria C Ruf
- Center for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Bogdana Suchorska
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
- Department of Neurosurgery, Sana Hospital, Duisburg, Germany
| | - Vera Wenter
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jörg-Christian Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcus Unterrainer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
26
|
Repeatability of image features extracted from FET PET in application to post-surgical glioblastoma assessment. Phys Eng Sci Med 2021; 44:1131-1140. [PMID: 34436751 DOI: 10.1007/s13246-021-01049-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/18/2021] [Indexed: 11/27/2022]
Abstract
Positron emission tomography (PET) imaging using the amino acid tracer O-[2-(18F)fluoroethyl]-L-tyrosine (FET) has gained increased popularity within the past decade in the management of glioblastoma (GBM). Radiomics features extracted from FET PET images may be sensitive to variations when imaging at multiple time points. It is therefore necessary to assess feature robustness to test-retest imaging. Eight patients with histologically confirmed GBM that had undergone post-surgical test-retest FET PET imaging were recruited. In total, 1578 radiomic features were extracted from biological tumour volumes (BTVs) delineated using a semi-automatic contouring method. Feature repeatability was assessed using the intraclass correlation coefficient (ICC). The effect of both bin width and filter choice on feature repeatability was also investigated. 59/106 (55.7%) features from the original image and 843/1472 (57.3%) features from filtered images had an ICC ≥ 0.85. Shape and first order features were most stable. Choice of bin width showed minimal impact on features defined as stable. The Laplacian of Gaussian (LoG, σ = 5 mm) and Wavelet filters (HLL and LHL) significantly improved feature repeatability (p ≪ 0.0001, p = 0.003, p = 0.002, respectively). Correlation of textural features with tumour volume was reported for transparency. FET PET radiomic features extracted from post-surgical images of GBM patients that are robust to test-retest imaging were identified. An investigation with a larger dataset is warranted to validate the findings in this study.
Collapse
|
27
|
Carles M, Fechter T, Grosu AL, Sörensen A, Thomann B, Stoian RG, Wiedenmann N, Rühle A, Zamboglou C, Ruf J, Martí-Bonmatí L, Baltas D, Mix M, Nicolay NH. 18F-FMISO-PET Hypoxia Monitoring for Head-and-Neck Cancer Patients: Radiomics Analyses Predict the Outcome of Chemo-Radiotherapy. Cancers (Basel) 2021; 13:3449. [PMID: 34298663 PMCID: PMC8303992 DOI: 10.3390/cancers13143449] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022] Open
Abstract
Tumor hypoxia is associated with radiation resistance and can be longitudinally monitored by 18F-fluoromisonidazole (18F-FMISO)-PET/CT. Our study aimed at evaluating radiomics dynamics of 18F-FMISO-hypoxia imaging during chemo-radiotherapy (CRT) as predictors for treatment outcome in head-and-neck squamous cell carcinoma (HNSCC) patients. We prospectively recruited 35 HNSCC patients undergoing definitive CRT and longitudinal 18F-FMISO-PET/CT scans at weeks 0, 2 and 5 (W0/W2/W5). Patients were classified based on peritherapeutic variations of the hypoxic sub-volume (HSV) size (increasing/stable/decreasing) and location (geographically-static/geographically-dynamic) by a new objective classification parameter (CP) accounting for spatial overlap. Additionally, 130 radiomic features (RF) were extracted from HSV at W0, and their variations during CRT were quantified by relative deviations (∆RF). Prediction of treatment outcome was considered statistically relevant after being corrected for multiple testing and confirmed for the two 18F-FMISO-PET/CT time-points and for a validation cohort. HSV decreased in 64% of patients at W2 and in 80% at W5. CP distinguished earlier disease progression (geographically-dynamic) from later disease progression (geographically-static) in both time-points and cohorts. The texture feature low grey-level zone emphasis predicted local recurrence with AUCW2 = 0.82 and AUCW5 = 0.81 in initial cohort (N = 25) and AUCW2 = 0.79 and AUCW5 = 0.80 in validation cohort. Radiomics analysis of 18F-FMISO-derived hypoxia dynamics was able to predict outcome of HNSCC patients after CRT.
Collapse
Affiliation(s)
- Montserrat Carles
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- La Fe Health Research Institute, Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), 46026 Valencia, Spain;
| | - Tobias Fechter
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
| | - Anca L. Grosu
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Arnd Sörensen
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Benedikt Thomann
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
| | - Raluca G. Stoian
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Nicole Wiedenmann
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexander Rühle
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Constantinos Zamboglou
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Juri Ruf
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Luis Martí-Bonmatí
- La Fe Health Research Institute, Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), 46026 Valencia, Spain;
| | - Dimos Baltas
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
| | - Michael Mix
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Nuclear Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Nils H. Nicolay
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|