Skip to main content
Advertisement

Main menu

  • Home
  • Content
    • Current Issue
    • Accepted Manuscripts
    • Article Preview
    • Past Issue Archive
    • AJNR Case Collection
    • Case of the Week Archive
    • Classic Case Archive
    • Case of the Month Archive
  • Special Collections
    • Spinal CSF Leak Articles (Jan 2020-June 2024)
    • 2024 AJNR Journal Awards
    • Most Impactful AJNR Articles
  • Multimedia
    • AJNR Podcast
    • AJNR Scantastics
    • Video Articles
  • For Authors
    • Submit a Manuscript
    • Author Policies
    • Fast publishing of Accepted Manuscripts
    • Graphical Abstract Preparation
    • Manuscript Submission Guidelines
    • Imaging Protocol Submission
    • Submit a Case for the Case Collection
  • About Us
    • About AJNR
    • Editorial Board
  • More
    • Become a Reviewer/Academy of Reviewers
    • Subscribers
    • Permissions
    • Alerts
    • Feedback
    • Advertisers
    • ASNR Home
  • Other Publications
    • ajnr

User menu

  • Alerts
  • Log in

Search

  • Advanced search
American Journal of Neuroradiology
American Journal of Neuroradiology

American Journal of Neuroradiology

ASHNR American Society of Functional Neuroradiology ASHNR American Society of Pediatric Neuroradiology ASSR
  • Alerts
  • Log in

Advanced Search

  • Home
  • Content
    • Current Issue
    • Accepted Manuscripts
    • Article Preview
    • Past Issue Archive
    • AJNR Case Collection
    • Case of the Week Archive
    • Classic Case Archive
    • Case of the Month Archive
  • Special Collections
    • Spinal CSF Leak Articles (Jan 2020-June 2024)
    • 2024 AJNR Journal Awards
    • Most Impactful AJNR Articles
  • Multimedia
    • AJNR Podcast
    • AJNR Scantastics
    • Video Articles
  • For Authors
    • Submit a Manuscript
    • Author Policies
    • Fast publishing of Accepted Manuscripts
    • Graphical Abstract Preparation
    • Manuscript Submission Guidelines
    • Imaging Protocol Submission
    • Submit a Case for the Case Collection
  • About Us
    • About AJNR
    • Editorial Board
  • More
    • Become a Reviewer/Academy of Reviewers
    • Subscribers
    • Permissions
    • Alerts
    • Feedback
    • Advertisers
    • ASNR Home
  • Follow AJNR on Twitter
  • Visit AJNR on Facebook
  • Follow AJNR on Instagram
  • Join AJNR on LinkedIn
  • RSS Feeds

Welcome to the new AJNR, Updated Hall of Fame, and more. Read the full announcements.


AJNR is seeking candidates for the position of Associate Section Editor, AJNR Case Collection. Read the full announcement.

 

Getting new auth cookie, if you see this message a lot, tell someone!
Review ArticleAdult Brain
Open Access

Evolving Role and Translation of Radiomics and Radiogenomics in Adult and Pediatric Neuro-Oncology

M. Ak, S.A. Toll, K.Z. Hein, R.R. Colen and S. Khatua
American Journal of Neuroradiology June 2022, 43 (6) 792-801; DOI: https://doi.org/10.3174/ajnr.A7297
M. Ak
aFrom the Department of Radiology (M.A., R.R.C.), University of Pittsburgh, Pittsburgh, Pennsylvania
bHillman Cancer Center (M.A., R.R.C.), University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for M. Ak
S.A. Toll
cDepartment of Hematology-Oncology (S.A.T.), Children's Hospital of Michigan, Detroit, Michigan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for S.A. Toll
K.Z. Hein
dDepartment of Leukemia (K.Z.H.), The University of Texas MD Anderson Cancer Center, Houston, Texas
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for K.Z. Hein
R.R. Colen
aFrom the Department of Radiology (M.A., R.R.C.), University of Pittsburgh, Pittsburgh, Pennsylvania
bHillman Cancer Center (M.A., R.R.C.), University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for R.R. Colen
S. Khatua
eDepartment of Pediatric Hematology-Oncology (S.K.), Mayo Clinic, Rochester, Minnesota.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for S. Khatua
  • Article
  • Figures & Data
  • Supplemental
  • Info & Metrics
  • Responses
  • References
  • PDF
Loading

Abstract

SUMMARY: Exponential technologic advancements in imaging, high-performance computing, and artificial intelligence, in addition to increasing access to vast amounts of diverse data, have revolutionized the role of imaging in medicine. Radiomics is defined as a high-throughput feature-extraction method that unlocks microscale quantitative data hidden within standard-of-care medical imaging. Radiogenomics is defined as the linkage between imaging and genomics information. Multiple radiomics and radiogenomics studies performed on conventional and advanced neuro-oncology image modalities show that they have the potential to differentiate pseudoprogression from true progression, classify tumor subgroups, and predict recurrence, survival, and mutation status with high accuracy. In this article, we outline the technical steps involved in radiomics and radiogenomics analyses with the use of artificial intelligence methods and review current applications in adult and pediatric neuro-oncology.

ABBREVIATIONS:

AI
artificial intelligence
AUC
area under the curve
DL
deep learning
GBM
glioblastoma
ML
machine learning
SUV
standardized uptake value

In parallel to the growing understanding of the impact of genomics and epigenomics, marked advances in clinical imaging and allied computational technologies have occurred during the past few years. These have facilitated enhanced diagnosis, molecular stratification, and targeted therapeutic clinical trials. Radiomics and radiogenomics are rapidly growing fields in imaging and, since their early inception, have been explored in the field of neuro-oncology.1 Radiomics is a high-throughput computational process that unlocks microscale quantitative data hidden within conventional imaging, not otherwise visualized by the naked human eye; radiogenomics is the linkage between imaging and genomics data.2,3 With use of radiomics analysis, a patient's scans are converted into mineable quantitative data to which machine learning (ML) techniques can be applied for integrative analysis. This process has enabled the identification of quantitative imaging markers and signature models that are reflective of microscopic tumor biology, which has led to enhanced, biologically relevant classification, tumor grading, survival prediction, and treatment response in adult and pediatric brain tumors.4⇓⇓⇓-8 Although studies of radiomics and radiogenomics have been performed in adult brain tumors, their application and use in pediatric neuro-oncology are evolving.5,9,10 Initial studies showed promising ability to distinguish posterior fossa tumors like ependymoma and pilocytic astrocytoma.9 Recent studies using a combination of radiomics and ML were able to distinguish pathologic subtypes of pediatric brain tumors.10 This article reviews the basics of radiomics, radiogenomics, and their clinical application; their evolving role in adult neuro-oncology; and their recent translation and application in pediatric neuro-oncology.

Search Criteria

Details of this review article were based on a PubMed search of categories of radiomics, adult brain tumors, and pediatric neuro-oncology. The search was limited to malignant brain tumors (excluding neurologic and benign lesions of the brain).

Technologic Methods and Computational Process

Overview of Radiomics Workflow.

The radiomics processes can be structured into multiple phases, and they are the same in pediatric and adult patients: 1) image acquisition and data selection, 2) image registration and segmentation, 3) image preprocessing, 4) radiomics feature extraction, and 5) feature selection and modeling to predict the outcomes of interest (Fig 1).

FIG 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIG 1.

Radiomics pipeline for brain tumors. Upper line: Segmentation of the 3 imaging phenotypes: necrosis (left), enhancing tumor (middle), and edema/invasion (right). Middle and lower lines: Radiomics feature extraction, normalization, and volume-dependent feature generation are followed by feature selection and predictive modeling for outcomes.

Image Acquisition and Data Selection.

Image acquisition and data selection are considered the initial steps in the workflow. For radiomics and radiogenomics studies in neuro-oncology, MR imaging and PET are generally used.

Image Registration and Segmentation.

Image registration refers to the spatial alignment of the various imaging sequences into the same geometric/anatomic space (Fig 2A).11 Tumor segmentation refers to contour delineation of the volumes of interest (3D) or ROIs (2D) (Fig 3). Manual segmentation or semiautomated and automated segmentation with superimposed manual segmentation is the criterion standard. Complete manual segmentation is tedious and time-consuming and requires contouring of the tumor section by section; furthermore, it is prone to interobserver and intraobserver variability.12 Semiautomated and automated segmentation methods have been explored in various studies to reduce manual labor and improve consistency and reproducibility to overcome these challenges.12⇓-14 Although promising, semiautomated and automated methods are not entirely accurate and typically require editing by a radiologist to ensure more precise borders.13

FIG 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIG 2.

A, Registration of T1-postcontrast MR imaging and FLAIR MR imaging. B, Skull stripping.

FIG 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIG 3.

Segmentation and 3D volume extraction of a diffuse intrinsic pontine glioma.

Image Preprocessing.

Skull stripping: Because high-resolution structural images include a significant amount of nonbrain tissue such as bone, skin, and eyeballs, these can affect the radiomics analysis (Fig 2B). The FSL Brain Extraction Tool (http://fsl.fmrib.ox.ac.uk/fsl/fslwiki/BET) and Robust Brain Extraction (https://www.nitrc.org/projects/robex) are the commonly used algorithms for skull stripping.15 Intensity normalization: During MR imaging acquisition, the use of different scanner types and scanning parameters can result in large intensity variations. These variations can weaken the performance of subsequent radiomics analysis. It is an essential process of mapping intensities to a standard reference scale in MR imaging to account for variations between patients and longitudinal studies and to increase radiomics reproducibility. The algorithm generated by Nyúl and Udupa,16 which involves the matching of histograms, is the most commonly used normalization method. Noise reduction: Noise suggests that the pixels in the image show distinct intensity values instead of actual pixel values that are achieved from the image.17 Rician noise decreases the quality of MR imaging and makes quantitative feature extraction difficult.17 Gaussian convolution and neighborhood filter are the most commonly used noise-reduction algorithms.17 Bias field correction: The bias field signal is nonuniform low-frequency intensities that corrupt MR imaging.18 The bias field is a potential confounder for radiomics analysis, and it should be corrected. The N4 bias field correction algorithm is the most commonly used approach for inhomogeneity correction.18

Radiomics Feature Extraction.

After segmentation and preprocessing, extraction of radiomics features is then performed. Radiomics features can be categorized as first-order, second-order, and high-order features. First-order features describe the distribution of individual voxel values without concern for spatial associations between neighboring voxels.17 Second-order features calculate the statistical interrelationships between adjacent voxels. Second-order features give a measure of the spatial alignment of the voxel intensities and intralesion heterogeneity. There are multiple different ways of quantifying spatial association among pixels: Gray-level co-occurrence matrix is a most commonly used second-order statistical texture-analysis technique, which analyzes the spatial relationship among pixels and defines how frequently voxel pairs are present in different directions.17 Gray-level run length matrix, another commonly used second-order feature, quantifies consequent voxels that have the same gray-level value along a fixed direction.17 High-order features involve performing filters or mathematic transforms to the images before the feature-extraction process.19 These features aim to detect repetitive or nonrepetitive patterns, remove noise, or highlight details. The wavelet transform–based method is a very popular approach in which the image is decomposed in multiple scales and used for texture analysis.19 Laplacian transforms of the Gaussian filter identify areas of rapid intensity change (edges) in images.20,21

Artificial Intelligence Applied to Imaging: Feature Selection and Statistical Modeling

Machine Learning.

ML is a branch of artificial intelligence (AI) in which computers are given the capability of learning like humans by feeding data and information without being explicitly programmed (Fig 4).22 Fundamental to ML is that classification, regression, prediction, clustering, and association models are provided. ML methods are categorized into supervised, unsupervised, and semi-supervised learning.

FIG 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIG 4.

AI, ML, and DL.

Supervised Machine Learning.

In supervised learning, a model is able to predict target clinical outcomes with the help of a labeled clinical dataset.22 The supervised learning model includes 2 steps: training and testing. Model training consists of inputs paired with the corresponding outputs to train the model. During training, the algorithms search for patterns in the data that relate to outcomes. Model testing is the process of predictive performance evaluation of the trained model on the test dataset. Support vector machine,8 random forests,8 and eXtreme Gradient Boosting (XGBoost; https://xgboost.ai/)23,24 are the most commonly used supervised machine learning methods in radiomics.

Unsupervised Machine Learning.

Unsupervised learning methods can classify radiomic features into subgroups using clustering algorithms such as k-means clustering,25 fuzzy clustering,26 or consensus clustering.27 Next, the relationships of the various features within their groups are compared. Subsequently, the capability of various subgroups to predict clinical output can be compared; however, labeled data are not used for the initial model development. Radiomics features can also be used to provide new subclasses that may more closely align with the underlying biology of CNS tumors by using unsupervised machine learning methods.

Deep Learning.

Deep learning (DL), also known as deep neural network, is a subfield of machine learning in AI that has multiple neural-like networks (Fig 5).28 The artificial neural networks are capable of learning from data that are unstructured or unlabeled. The most significant difference between DL and conventional ML algorithms is that DL does not need any human interference such as radiomics, feature extraction, and segmentation to learn the connection between the input and the corresponding output. Some of the known deep learning algorithms are the recurrent neural network, restricted Boltzmann machine, and convolutional neural network.28 The convolutional neural network is most commonly used and is popular in radiomics studies.28

FIG 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIG 5.

A deep learning–based model for predicting outcomes.

Feature Selection.

Feature selection is the process of selecting a subset of the most relevant and significant discriminatory features associated with the specific outcome. These are then used for building radiomics models. Feature selection helps in clarifying the data by revealing, in an individual, the important features and how they are related to each other and to the image itself. There are several commonly used methods for radiomics feature selection such as Least Absolute Shrinkage and Selection Operator,8 Minimum Redundancy Maximum Relevance,24,29 and Generalized Linear Models with Elastic Net Penalties.30

Modeling.

Radiomics uses prediction models for predicting outcomes, including clinical or demographic, genomic, survival, response, and resistance models.31⇓⇓⇓-35 AI, specifically ML and DL, provides several approaches to achieve this aim.31 The support vector machine uses a hyperplane, which distinctly classifies the data points into 2 classes.31 Generalizability and the possibility of achieving probabilistic outputs are its advantages.31 XGBoost is a decision tree–based ensemble algorithm that uses a gradient boosting.31,36 It enables cross-validation, regularization, missing-value imputation, and flexibility.36 The choice of modeling technique has been demonstrated to affect prediction performance in a radiomics study.29

Validation.

The most effective approach for validation is the use of prospective independent external validation. In cases for which no such prospective or independent external data are available, internal validation techniques can be performed. K-fold, leave-one-out cross-validation, and hold-out are the most commonly used internal validation approaches.37

Clinical Applications of Radiomics in Neuro-Oncology: Adults

Most research in radiomics was initially performed in lung cancer.38 Radiogenomics, however, was initially performed in adult brain cancer, specifically glioblastoma (GBM).39 Since then, numerous studies have focused on the use of radiomics and radiogenomics in various adult CNS tumors (Online Supplemental Data).4,7,40⇓⇓⇓⇓⇓⇓-47 With increasing computing speed and the availability of computerized algorithms, large numbers of radiomics features can be extracted rapidly from MRIs and used to generate specific signature models. These have aided in noninvasively identifying histologic and molecular profiles of tumors,48,49 predicting response,35 differentiating pseudoprogression from true progressive disease,24 and delineating oncogenic markers in the microenvironment of brain tumors.50

Radiomics as a Diagnostic Marker.

The most recent 2016 World Health Organization classification demonstrated the importance of integrating phenotype-genotype characteristics of CNS tumors, enabling newer subclassifications of tumor groups.51 This landmark article led to seminal studies44,52,53 that showed that brain tumors could be directly related to a specific set of genomics, helping to preoperatively predict tumor genotype. GBM is the most common malignant brain tumor in adults, with poor survival outcome and a median survival of 15 months.54 Discernment of the intratumoral genetic heterogeneity of GBMs has important implications in optimizing targeted therapy to improve survival and in understanding the mechanisms of therapeutic resistance.54,55 GBMs have multiple genetically distinct clonal populations driving oncogenesis and thus have distinct therapeutic sensitivities and habitats of resistance.54 Biopsy and surgery cannot capture such spatially extensive tumor heterogeneity because both comprise samples that represent a small and static molecular “snapshot” of the entire tumor, which is very heterogeneous; furthermore, these procedures are invasive.

Radiomics and radiogenomics have been shown to potentially complement biopsy by capturing regional genetic heterogeneity and by noninvasively evaluating various driver genes and prognostic markers at diagnosis.43,44,47,56⇓-58 Recent studies have shown that radiomics models can preoperatively predict O6-methylguanine-DNA methyltransferase (MGMT) methylation, epidermal growth factor (EGFR) amplification, and EGFR variant III status in GBM.48,58⇓-60 Zhang et al46 demonstrated that ML algorithms generated from preoperative MR imaging and clinical features of 120 patients with grade III and IV gliomas predicted isocitrate dehydrogenase (IDH) 1/2 status with accuracies of 86% and 89% in the training and validation cohorts, respectively. Similarly, Chang et al61 showed the feasibility of a DL, convolutional neural network approach for the classification of genetic mutations of both low- and high-grade gliomas. They used 259 patients with either low- or high-grade gliomas to classify IDH1 mutation, 1p/19q codeletion, and MGMT methylation status. Their classification demonstrated that the convolutional neural network is capable of learning significant imaging components without prior feature selection or human-directed training with high accuracy: IDH1 mutation status, 94%; 1p/19q codeletion, 92%; and MGMT methylation status, 83%.

In a recent study, Hu et al50 built a radiomics model to preoperatively identify 6 important driver genes (EGFR, PDGFRA, PTEN, CDKN2A, RB1, and TP53) in primary GBM. The authors evaluated 48 biopsies from the regions of enhancing and nonenhancing parenchyma of 13 patients with GBM and identified significant imaging correlations for these 6 genes. They achieved accuracies ranging from 68% to 87.5% for PDGFRA, EGFR, CDKN2A, PTEN, and RB1 prediction models, whereas the accuracy for the TP53 prediction model was 37%. Similarly, Zinn et al62 showed that there are distinct MR imaging radiomics features associated with the TP53-PTEN-EGFR mutational landscape and that radiomics is approaching the complexity of whole-genome microarray expression data. Additionally, recent studies demonstrated that MR imaging–based radiomics models predicted 1p/19q codeletion status in histopathologically diagnosed gliomas.43,47 The latter is a predictor of better prognosis and a durable response to therapy.63 These results provide proof of concept and reaffirm that genomics-based biomarkers can be correlated and/or predicted noninvasively by radiomics, facilitating the evaluation of intratumoral genetic heterogeneity; and if prospectively validated, can be translated to the clinic as a noninvasive, cost-effective genomic test approach and advance individualized patient management.

Tumor Type and Tumor Grading Prediction.

Noninvasive diagnosis of brain tumors is clinically challenging but necessary because various treatment strategies are needed depending on the tumor type. Conventional or advanced MR imaging techniques are often challenging in their ability to differentiate GBM from metastatic brain tumors and lymphoma. All these tumors can show enhancement on T1-weighted images with gadolinium and hyperintensity on FLAIR.64,65 Although advanced MR imaging (MR spectroscopy and PWI) has shown promising results in the evaluation of brain tumors, further work is needed for a transition to the clinic as part of routine brain tumor management.64,65 Radiomics-based ML techniques have demonstrated potential in differentiating GBM from metastatic brain tumors.66 Another study showed that a radiomics model generated by features obtained from diffusion MR imaging yielded a better diagnostic performance than a radiomics model created by features acquired from conventional MR imaging in differentiating atypical primary CNS lymphoma, which often mimics GBM.67 Similarly, Kong et al68 showed that radiomics features obtained from the PET images have the potential to distinguish primary CNS lymphoma from GBM. They generated 3 groups of maps: a standardized uptake value (SUV) map, an SUV map calibrated with the normal contralateral cortex activity, and an SUV map calibrated with the normal brain mean activity; a total of 107 radiomics features were extracted from each SUV map. Their areas under the curve (AUCs) for differentiating CNS lymphoma and GBM ranged from 0.644 to 0.999. In addition, a recently published study determined the capability of radiomics analysis to identify the primary origin of brain metastases despite neuropathologic procedures and imaging evaluations often failing to identify the primary tumor site and leading to delayed diagnosis and treatment.69 However, further studies are needed to validate and reproduce these findings.

High-grade gliomas, including GBMs, are biologically elusive, due to their genetic heterogeneity and complex imaging phenotypes.54,55 Previous studies have shown that radiomics can predict the pathologic behavior of gliomas, histologic/molecular subtypes/grading, and the proliferative index.49,70,71 This finding could facilitate the presurgical evaluation of tumor behavior, determine the extent of surgical resection, and enable optimal clinical treatment decision-making in patients with various grades of gliomas. Histologic grade and the Ki-67 labeling index are used to help predict biologic behavior and prognosis of gliomas.72 A recently published study has demonstrated that radiomics features based on T2 FLAIR images have been shown to preoperatively correlate and predict multiple immunohistochemical features of gliomas such as Ki-67, S-100, vimentin, and CD34 expression.49 These insights are expected to enhance the personalized treatment of patients with gliomas.49 Furthermore, radiomics-based ML techniques have shown potential in differentiating GBM from anaplastic oligodendroglioma.42 Although the AUCs of the models used were >0.900, more studies in a larger cohort of patients are required to further validate these revelations.

Risk Stratification and Prognostication of Tumors.

The 2016 World Health Organization classification considers the genomics heterogeneity of diffuse gliomas, necessitating effective risk stratification for optimizing therapy.51 The invasiveness of existing methods to identify histopathologic/molecular profiles and the difficulty of replicating results demonstrated the need to identify noninvasive methods to complement and solidify the diagnostic armamentarium. Development of radiomics profiles and comprehensive analysis of radiogenomics phenotypes using multiomics molecular approaches have been successful in risk stratification and survival estimation of diffuse gliomas.6,32,33,35,41,73,74 A study using MR imaging radiomics features of patients with de novo GBM determined the imaging signatures associated with poor prognosis.75

In another cohort of 82 patients with de novo GBM, analysis of 5 texture features from preoperative MR images predicted molecular subtypes and 12-month survival status (overall survival at 12 months indicating whether the patient was alive).32 Another radiomics study evaluating CBV and ADC in 119 patients with newly diagnosed GBM demonstrated that radiomics-based classification allows noninvasive prediction of survival and stratification of patients with GBM with better accuracy than that determined with established conventional clinical (age and Karnofsky Performance Scale score) or radiologic risk models (Gaussian normalized relative CBV and ADC).6 These methods and tools could guide planning of surgical resection, define radiation treatment margins, and optimize the intensity of chemotherapy. The evolving and molecular profile of lower-grade gliomas, including the favorable outcome associated with IDH1 and 1p/19q codeletion mutation status, now partially explains the heterogeneous survival outcomes; recent studies have shown that radiomics phenotyping using ML techniques can identify these genomic markers43,47 and predict overall survival with better accuracy than with the use of nonimaging markers (clinical features and genomic data).41 Identification of the ATRX mutation or loss of ATRX expression is a potential biomarker in gliomas and has been associated with favorable survival outcome; in a recently published article, the authors showed that radiomics could identify this genotype at diagnosis.56 This could be an important addition to the noninvasive detection of pivotal biomarkers such as IDH1 and MGMT in gliomas.44,46,47,58,59

Predicting Treatment-Related Surrogate End Points.

A few studies have robustly defined the biologic heterogeneity of the peritumoral brain zone and its interaction with the microenvironment in brain tumors.55 This heterogeneity contributes to therapeutic resistance and poor survival in infiltrating tumors such as GBM.55 Nearly 90% of recurrences in GBM occur in the peritumoral brain zone, which is usually the nonenhancing component.55 Interrogating 10 radiomics peritumoral features from the peritumoral brain zone on routine preoperative MR imaging in patients with GBM was predictive of long-term (>18 months) versus short-term (<7 months) survival.33 In addition, a model generated by 6 imaging features for CD3 infiltration prediction in GBM achieved high accuracies of 97.1% and 76.5% in the training and testing sets, respectively.76 The role of radiomics is being used increasingly to improve characterization and understanding of the tumor microenvironment.70,76 This will have important clinical relevance in optimizing a targeted therapeutic approach.77

Bevacizumab has been widely used to treat patients with recurrent GBM; however, the data show a lack of sustained long-term efficacy.78 The therapeutic responses, furthermore, vary substantially in patients.78 However, quantitative imaging biomarkers that use radiomics to help identify patients who will benefit from bevacizumab treatment before initiation of therapy have been researched.35

With the use of immunotherapeutics in brain tumors, transient increases in tumor size and/or new inflammatory lesions often appear. These changes, known as pseudoprogression, typically stabilize or decrease with the continuation of treatment, and differentiating them from progressive disease is often difficult and delayed.79 Conventional imaging using MR perfusion studies has been used frequently, though specificity and sensitivity remain suboptimal.80,81 A multicenter study using an MR perfusion–based radiomics model was able to distinguish pseudoprogression from progressive disease with the AUC, sensitivity, and specificity of 90%, 93%, and 89%, respectively.24 These studies highlight the capability of radiomics to address this important clinical distinction—an unmet need in neuroimmunotherapy.

Evolving Era of Radiomics in Pediatric Neuro-Oncology

Pediatric CNS tumors have special characteristics in tissues, morphology, molecular subtype, and texture compared with their adult counterparts.82 With the emerging biologic differences between adult and pediatric brain tumors, the need for improved characterization of CNS tumors in children is imperative. Although radiomics and radiogenomics are now used in the adult clinical arena, their translation into clinical practice in pediatric neuro-oncology is still in its infancy (Online Supplemental Data). Pediatric brain tumors are the most common solid tumors in children.83 Unlike adults, in whom we have seen increased use of noninvasive neuroimaging using multimodal MR imaging combining multiple sequences to enhance tissue characterization, few studies to date have been reported in pediatrics.5,9,84⇓⇓-87 Similar noninvasive imaging studies are needed in parallel with the evolving molecular era in pediatric brain tumors, facilitating improved tumor classification (histologic and molecular), grading, survival prediction, and treatment response.

Earlier studies evaluated the role and potential of MR imaging texture analysis in capturing quantitative information of different pediatric brain tumors.88 A recent multicenter study was performed within a supervised classification framework on clinical MR imaging, and a support vector machine was trained with 3D textural attributes obtained from conventional MR imaging.85 The developed model was very successful in capturing transferable tumor information, and this study supported the use of 3D texture analysis on conventional MR imaging to aid in the diagnostic classification of various pediatric brain tumors. A recently published study84 using a combination of radiomics and ML approaches on 3D multimodal MR imaging was able to build a radiomics model with multivariable logistic regression. These models could differentiate pediatric ependymoma and medulloblastoma (common malignant brain tumors of the posterior fossa in children) with an AUC of 0.91. Similarly, Zhou et al87 demonstrated the capability of radiomics in differentiating the types of pediatric posterior fossa tumors (medulloblastoma, ependymoma, and pilocytic astrocytoma) with an accuracy of 0.85. The success of this trend needs to be evaluated further in prospective studies.

Pediatric medulloblastoma has been the most biologically interrogated malignant CNS tumor in children.89 Its molecular subtypes with distinct clinical and prognostic differences now reaffirm the need to tailor therapy accordingly.89 A recent study5 provided proof-of-concept results for the application of radiomics and ML approaches for prediction of distinct medulloblastoma subgroups. The reproducibility of this finding would be of immense clinical significance, with noninvasive ability to diagnose and biologically subtype these neoplasms. Another research group published that model based on texture extracted from conventional MR imaging to preoperatively differentiate ependymoma and pilocytic astrocytoma and achieved high sensitivity, specificity, and AUC.9 This could be an invaluable tool because often these lesions occur in the eloquent areas of the brain both at diagnosis and recurrence, precluding surgical intervention.

DNA methylation now identifies various molecular subtypes of aggressive pediatric CNS tumors, and genomics analysis was able to decipher the intrinsic tumor heterogeneity.90 Noninvasive imaging that uses radiomics to correlate with these genomic aberrations would be a clinically significant addition to diagnosing these neoplasms and facilitating the design of optimal targeted therapy.

Diffuse midline gliomas in children are aggressive brain tumors with 10% overall survival at 18 months. MR imaging–based texture analysis in 32 children with diffuse midline gliomas was able to stratify patients into poor and good prognostic groups, with a median survival of 7.5 months versus 17.5 months, respectively.74 Diffuse midline gliomas with more homogeneous texture on diagnostic MR imaging were associated with a worse prognosis.74 These findings should be further explored and correlated with the evolving molecular subtypes, providing valuable insight into prognosis and tailored therapy for these tumors. Profiling an optimal decision tree model with the use of ADC histogram analysis and structural MR imaging findings provided the ability to differentiate 7 histopathologic subtypes of pediatric CNS tumors, with a nearly 90% accurate classification.34 This decision tree model could preoperatively distinguish, with precision, 2 biologically different tumors, pilocytic astrocytomas and atypical teratoid/rhabdoid tumor, in comparison with neuroradiologists. Similar prospective studies evaluating the ability of this model to correlate with the diverse genomic aberrations would be a breakthrough in the management of targeted therapy for these tumors at diagnosis and recurrence. A major success of targeted therapy in pediatric CNS tumors has been the use of BRAF and Mek inhibitors in low-grade gliomas. Currently, a biopsy is needed to identify these molecular aberrations, often not possible due to the eloquent location of these neoplasms. A recent study showed the ability of radiomics-based prediction of BRAF status in pediatric low-grade gliomas before a biopsy, which could have significant implications in clinical practice.91

Immunology.

Use of immunotherapeutic strategies, including adoptive cell therapy, vaccine therapy, and checkpoint inhibitors, in pediatric brain tumors has been rapidly evolving.92 However, therapeutic efficacy is undermined by the low mutational burden seen in these tumors, precluding targeted therapy. Reviews of immunotherapy in pediatric brain tumors show some preliminary promising results, which need to be further explored. In a recently published study of natural killer cell infusion in children with recurrent medulloblastoma and ependymoma, a high radiomic performance was achieved; accuracy, sensitivity, and specificity were 100%, though it did not attain statistical significance, likely due to the low number of patients.86,92,93 A few studies in adults have shown the ability of radiomics models to differentiate pseudoprogression from early tumor progression in infiltrative tumors such as GBM.24 The ability to make these distinctions in pediatric neuro-oncology is important and needs to be pursued.

Limitations

Although radiomics has shown its potential for diagnostic, prognostic, and predictive purposes in multiple neuro-oncology studies, the translation of radiomics into the clinical settings is slow because the field is facing several challenges. Most important, the repeatability, variability, and reproducibility of radiomics are still issues and often depend on the imaging sequence used, technique, size of the image, image quality, software used for feature extraction and reconstruction, as well as motion artifacts and segmentation, which can lead to greater interobserver variations in addition to the time-consuming nature of manual segmentation.

DL-based radiomics may overcome these issues because the features are extracted automatically from the huge data, and a subset of best-performing features is automatically identified. The most important pitfall of DL-based radiomics is the high association between the images and the clinical output data. Thus, in contrast to feature-based radiomics, large datasets are needed to identify the most relevant and robust features to overcome this challenge. DL-based radiomics models must validate their reliability in prospective large studies.

Another important limitation to the translation of radiomics models in clinical management is the interpretability of the radiomics features and models. Radiomics is perceived as a “black box,” ie, it is very difficult to clinically interpret the developed predictions. As an emerging technique, explainable AI is better understood, trusted, and the results are more efficiently interpreted by human-users, in contrast to the “black box” concept in ML.94

Conclusions

In this review, we outlined the principles of radiomics analysis in a step-by-step approach and described how large amounts of imaging data can be interrogated and analyzed in patients with brain tumors. We believe that soon, with cumulative developments in AI and increasing public access to large data bases, radiomics can be expected to play an important role in clinical decision-making by answering many clinical questions of diagnostic and therapeutic relevance. However, many limitations and challenges exist, and much work needs to be done to robustly translate radiomics into the clinical arena.

Footnotes

  • Rivka R. Colen and Soumen Khatua are senior co-authors.

  • This work was funded, in part, by startup funding from The University of Pittsburgh Hillman Cancer Center (R.R.C.) and the Hillman Cancer Center's National Cancer Institute Cancer Center Support Grant No. P30CA047904 (R.R.C.).

Indicates open access to non-subscribers at www.ajnr.org

References

  1. 1.↵
    1. Zhou M,
    2. Scott J,
    3. Chaudhury B, et al
    . Radiomics in brain tumor: image assessment, quantitative feature descriptors, and machine-learning approaches. AJNR Am J Neuroradiol 2018;39:208–16 doi:10.3174/ajnr.A5391 pmid:28982791
    Abstract/FREE Full Text
  2. 2.↵
    1. Colen RR,
    2. Rolfo C,
    3. Ak M, et al
    . Radiomics analysis for predicting pembrolizumab response in patients with advanced rare cancers. J Immunother Cancer 2021;9:e001752 doi:10.1136/jitc-2020-001752 pmid:33849924
    Abstract/FREE Full Text
  3. 3.↵
    1. Lambin P,
    2. Rios-Velazquez E,
    3. Leijenaar R, et al
    . Radiomics: extracting more information from medical images using advanced feature analysis. Eur J Cancer 2012;48:441–46 doi:10.1016/j.ejca.2011.11.036 pmid:22257792
    CrossRefPubMed
  4. 4.↵
    1. Habib A,
    2. Jovanovich N,
    3. Hoppe M, et al
    . MRI-based radiomics and radiogenomics in the management of low-grade gliomas: evaluating the evidence for a paradigm shift. J Clin Med 2021;10:1411 doi:10.3390/jcm10071411
    CrossRef
  5. 5.↵
    1. Iv M,
    2. Zhou M,
    3. Shpanskaya K, et al
    . MR imaging-based radiomic signatures of distinct molecular subgroups of medulloblastoma. AJNR Am J Neuroradiol 2019;40:154–61 doi:10.3174/ajnr.A5899 pmid:30523141
    Abstract/FREE Full Text
  6. 6.↵
    1. Kickingereder P,
    2. Burth S,
    3. Wick A, et al
    . Radiomic profiling of glioblastoma: identifying an imaging predictor of patient survival with improved performance over established clinical and radiologic risk models. Radiology 2016;280:880–89 doi:10.1148/radiol.2016160845 pmid:27326665
    CrossRefPubMed
  7. 7.↵
    1. Lin P,
    2. Peng YT,
    3. Gao RZ, et al
    . Radiomic profiles in diffuse glioma reveal distinct subtypes with prognostic value. J Cancer Res Clin Oncol 2020;146:1253–62 doi:10.1007/s00432-020-03153-6 pmid:32065261
    CrossRefPubMed
  8. 8.↵
    1. Nakamoto T,
    2. Takahashi W,
    3. Haga A, et al
    . Prediction of malignant glioma grades using contrast-enhanced T1-weighted and T2-weighted magnetic resonance images based on a radiomic analysis. Sci Rep 2019;9:19411 doi:10.1038/s41598-019-55922-0 pmid:31857632
    CrossRefPubMed
  9. 9.↵
    1. Li M,
    2. Wang H,
    3. Shang Z, et al
    . Ependymoma and pilocytic astrocytoma: Differentiation using radiomics approach based on machine learning. J Clin Neurosci 2020;78:175–80 doi:10.1016/j.jocn.2020.04.080 pmid:32336636
    CrossRefPubMed
  10. 10.↵
    1. Rodriguez Gutierrez D,
    2. Awwad A,
    3. Meijer L, et al
    . Metrics and textural features of MRI diffusion to improve classification of pediatric posterior fossa tumors. AJNR Am J Neuroradiol 2014;35:1009–15 doi:10.3174/ajnr.A3784 pmid:24309122
    Abstract/FREE Full Text
  11. 11.↵
    1. Visser M,
    2. Petr J,
    3. Muller DMJ, et al
    . Accurate MR image registration to anatomical reference space for diffuse glioma. Front Neurosci 2020;14:585 doi:10.3389/fnins.2020.00585 pmid:32581699
    CrossRefPubMed
  12. 12.↵
    1. Akkus Z,
    2. Sedlar J,
    3. Coufalova L, et al
    . Semi-automated segmentation of pre-operative low grade gliomas in magnetic resonance imaging. Cancer Imaging 2015;15:12 doi:10.1186/s40644-015-0047-z pmid:26268363
    CrossRefPubMed
  13. 13.↵
    1. Hayes K,
    2. Buist R,
    3. Vincent TJ, et al
    . Comparison of manual and semi-automated segmentation methods to evaluate hippocampus volume in APP and PS1 transgenic mice obtained via in vivo magnetic resonance imaging. J Neurosci Methods 2014;221:103–11 doi:10.1016/j.jneumeth.2013.09.014 pmid:24091139
    CrossRefPubMed
  14. 14.↵
    1. Zhao Z,
    2. Yang G,
    3. Lin Y, et al
    . Automated glioma detection and segmentation using graphical models. PLoS One 2018;13:e0200745 doi:10.1371/journal.pone.0200745 pmid:30130371
    CrossRefPubMed
  15. 15.↵
    1. Kalavathi P,
    2. Prasath VB
    . Methods on skull stripping of MRI head scan images: a review. J Digit Imaging 2016;29:365–79 doi:10.1007/s10278-015-9847-8 pmid:26628083
    CrossRefPubMed
  16. 16.↵
    1. Nyúl LG,
    2. Udupa JK
    . On standardizing the MR image intensity scale. Magn Reson Med 1999;42:1072–81 doi:10.1002/(SICI)1522-2594(199912)42:6<1072::AID-MRM11>3.0.CO;2-M pmid:10571928
    CrossRefPubMed
  17. 17.↵
    1. Buades A,
    2. Coll B,
    3. Morel JM
    . A review of image denoising algorithms, with a new one. Multiscale Model Simul 2005;4:490–530 doi:10.1137/040616024
    CrossRef
  18. 18.↵
    1. Vovk U,
    2. Pernus F,
    3. Likar B
    . A review of methods for correction of intensity inhomogeneity in MRI. IEEE Trans Med Imaging 2007;26:405–21 doi:10.1109/TMI.2006.891486 pmid:17354645
    CrossRefPubMed
  19. 19.↵
    1. Cattell R,
    2. Chen S,
    3. Huang C
    . Robustness of radiomic features in magnetic resonance imaging: review and a phantom study. Vis Comput Ind Biomed Art 2019;2:19 doi:10.1186/s42492-019-0025-6 pmid:32240418
    CrossRefPubMed
  20. 20.↵
    1. Chaurasia K,
    2. Garg PA
    . Brief Review on Texture Analysis Methods. American Society of Science and Engineering; 2013
  21. 21.↵
    1. Parekh V,
    2. Jacobs MA
    . Radiomics: a new application from established techniques. Expert Rev Precis Med Drug Dev 2016;1:207–26 doi:10.1080/23808993.2016.1164013 pmid:28042608
    CrossRefPubMed
  22. 22.↵
    1. Deo RC
    . Machine learning in medicine. Circulation 2015;132:1920–30 doi:10.1161/CIRCULATIONAHA.115.001593 pmid:26572668
    Abstract/FREE Full Text
  23. 23.↵
    1. Colen RR,
    2. Ologun GO,
    3. Zinn P, et al
    . Radiomic signatures to predict response to targeted therapy and immune checkpoint blockade in melanoma patients (pts) on neoadjuvant therapy. J Clin Oncol 2020;38:10067 doi:10.1200/JCO.2020.38.15_suppl.10067
    CrossRef
  24. 24.↵
    1. Elshafeey N,
    2. Kotrotsou A,
    3. Hassan A, et al
    . Multicenter study demonstrates radiomic features derived from magnetic resonance perfusion images identify pseudoprogression in glioblastoma. Nat Commun 2019;10:3170 doi:10.1038/s41467-019-11007-0 pmid:31320621
    CrossRefPubMed
  25. 25.↵
    1. Rathore S,
    2. Akbari H,
    3. Rozycki M, et al
    . Radiomic MRI signature reveals three distinct subtypes of glioblastoma with different clinical and molecular characteristics, offering prognostic value beyond IDH1. Sci Rep 2018;8:5087 doi:10.1038/s41598-018-22739-2 pmid:29572492
    CrossRefPubMed
  26. 26.↵
    1. Sompong C,
    2. Wongthanavasu S
    . Brain tumor segmentation using cellular automata-based fuzzy c-means. In: Proceedings of the International Joint Conference on Computer Science and Software Engineering, Khon Kaen, Thailand. July 13-15;2016:1–6 doi:10.1109/JCSSE.2016.7748902
    CrossRef
  27. 27.↵
    1. Kong DS,
    2. Kim J,
    3. Ryu G, et al
    . Quantitative radiomic profiling of glioblastoma represents transcriptomic expression. Oncotarget 2018;9:6336–45 doi:10.18632/oncotarget.23975 pmid:29464076
    CrossRefPubMed
  28. 28.↵
    1. Parekh VS,
    2. Jacobs MA
    . Deep learning and radiomics in precision medicine. Expert Rev Precis Med Drug Dev 2019;4:59–72 doi:10.1080/23808993.2019.1585805 pmid:31080889
    CrossRefPubMed
  29. 29.↵
    1. Parmar C,
    2. Grossmann P,
    3. Bussink J, et al
    . Machine learning methods for quantitative radiomic biomarkers. Sci Rep 2015;5:13087 doi:10.1038/srep13087 pmid:26278466
    CrossRefPubMed
  30. 30.↵
    1. Sasaki T,
    2. Kinoshita M,
    3. Fujita K, et al
    . Radiomics and MGMT promoter methylation for prognostication of newly diagnosed glioblastoma. Sci Rep 2019;9:14435 doi:10.1038/s41598-019-50849-y pmid:31594994
    CrossRefPubMed
  31. 31.↵
    1. Sun P,
    2. Wang D,
    3. Mok VC, et al
    . Comparison of feature selection methods and machine learning classifiers for radiomics analysis in glioma grading. IEEE Access 2019;7:102010–20 doi:10.1109/ACCESS.2019.2928975
    CrossRef
  32. 32.↵
    1. Yang D,
    2. Rao G,
    3. Martinez J, et al
    . Evaluation of tumor-derived MRI-texture features for discrimination of molecular subtypes and prediction of 12-month survival status in glioblastoma. Med Phys 2015;42:6725–35 doi:10.1118/1.4934373 pmid:26520762
    CrossRefPubMed
  33. 33.↵
    1. Prasanna P,
    2. Patel J,
    3. Partovi S, et al
    . Radiomic features from the peritumoral brain parenchyma on treatment-naive multi-parametric MR imaging predict long versus short-term survival in glioblastoma multiforme: preliminary findings. Eur Radiol 2017;27:4188–97 doi:10.1007/s00330-016-4637-3 pmid:27778090
    CrossRefPubMed
  34. 34.↵
    1. Payabvash S,
    2. Aboian M,
    3. Tihan T, et al
    . Machine learning decision tree models for differentiation of posterior fossa tumors using diffusion histogram analysis and structural MRI findings. Front Oncol 2020;10:71 doi:10.3389/fonc.2020.00071 pmid:32117728
    CrossRefPubMed
  35. 35.↵
    1. Kickingereder P,
    2. Gotz M,
    3. Muschelli J, et al
    . Large-scale radiomic profiling of recurrent glioblastoma identifies an imaging predictor for stratifying anti-angiogenic treatment response. Clin Cancer Res 2016;22:5765–71 doi:10.1158/1078-0432.CCR-16-0702 pmid:27803067
    Abstract/FREE Full Text
  36. 36.↵
    1. Chen T,
    2. Guestrin C
    . XGBoost: a scalable tree boosting system. In: Proceedings of the 22nd ACM SIGKDD International Conference on Knowledge Discovery and Data Mining. Association for Computing Machinery; 2016:785–94
  37. 37.↵
    1. Koçak B,
    2. Durmaz EŞ,
    3. Ateş E, et al
    . Radiomics with artificial intelligence: a practical guide for beginners. Diagn Interv Radiol 2019;25:485–95 doi:10.5152/dir.2019.19321 pmid:31650960
    CrossRefPubMed
  38. 38.↵
    1. Kumar V,
    2. Gu Y,
    3. Basu S, et al
    . Radiomics: the process and the challenges. Magn Reson Imaging 2012;30:1234–48 doi:10.1016/j.mri.2012.06.010 pmid:22898692
    CrossRefPubMed
  39. 39.↵
    1. Zinn PO,
    2. Majadan B,
    3. Sathyan P, et al
    . Radiogenomic mapping of edema/cellular invasion MRI-phenotypes in glioblastoma multiforme. PLoS One 2011;6:e25451 doi:10.1371/journal.pone.0025451 pmid:21998659
    CrossRefPubMed
  40. 40.↵
    1. Cho HH,
    2. Lee SH,
    3. Kim J, et al
    . Classification of the glioma grading using radiomics analysis. PeerJ 2018;6:e5982 doi:10.7717/peerj.5982 pmid:30498643
    CrossRefPubMed
  41. 41.↵
    1. Choi YS,
    2. Ahn SS,
    3. Chang JH, et al
    . Machine learning and radiomic phenotyping of lower grade gliomas: improving survival prediction. Eur Radiol 2020;30:3834–42 doi:10.1007/s00330-020-06737-5 pmid:32162004
    CrossRefPubMed
  42. 42.↵
    1. Fan Y,
    2. Chen C,
    3. Zhao F, et al
    . Radiomics-based machine learning technology enables better differentiation between glioblastoma and anaplastic oligodendroglioma. Front Oncol 2019;9:1164 doi:10.3389/fonc.2019.01164 pmid:31750250
    CrossRefPubMed
  43. 43.↵
    1. Han Y,
    2. Xie Z,
    3. Zang Y, et al
    . Non-invasive genotype prediction of chromosome 1p/19q co-deletion by development and validation of an MRI-based radiomics signature in lower-grade gliomas. J Neurooncol 2018;140:297–306 doi:10.1007/s11060-018-2953-y pmid:30097822
    CrossRefPubMed
  44. 44.↵
    1. Lohmann P,
    2. Lerche C,
    3. Bauer EK, et al
    . Predicting IDH genotype in gliomas using FET PET radiomics. Sci Rep 2018;8:13328 doi:10.1038/s41598-018-31806-7 pmid:30190592
    CrossRefPubMed
  45. 45.↵
    1. Su C,
    2. Jiang J,
    3. Zhang S, et al
    . Radiomics based on multicontrast MRI can precisely differentiate among glioma subtypes and predict tumour-proliferative behaviour. Eur Radiol 2019;29:1986 doi:10.1007/s00330-018-5704-8 pmid:30315419
    CrossRefPubMed
  46. 46.↵
    1. Zhang B,
    2. Chang K,
    3. Ramkissoon S, et al
    . Multimodal MRI features predict isocitrate dehydrogenase genotype in high-grade gliomas. Neuro Oncol 2017;19:109–17 doi:10.1093/neuonc/now121 pmid:27353503
    CrossRefPubMed
  47. 47.↵
    1. Zhou H,
    2. Chang K,
    3. Bai HX, et al
    . Machine learning reveals multimodal MRI patterns predictive of isocitrate dehydrogenase and 1p/19q status in diffuse low- and high-grade gliomas. J Neurooncol 2019;142:299–307 doi:10.1007/s11060-019-03096-0 pmid:30661193
    CrossRefPubMed
  48. 48.↵
    1. Kickingereder P,
    2. Bonekamp D,
    3. Nowosielski M, et al
    . Radiogenomics of glioblastoma: machine learning-based classification of molecular characteristics by using multiparametric and multiregional MR imaging features. Radiology 2016;281:907–18 doi:10.1148/radiol.2016161382 pmid:27636026
    CrossRefPubMed
  49. 49.↵
    1. Li J,
    2. Liu S,
    3. Qin Y, et al
    . High-order radiomics features based on T2 FLAIR MRI predict multiple glioma immunohistochemical features: a more precise and personalized gliomas management. PLoS One 2020;15:e0227703 doi:10.1371/journal.pone.0227703 pmid:31968004
    CrossRefPubMed
  50. 50.↵
    1. Hu LS,
    2. Ning S,
    3. Eschbacher JM, et al
    . Radiogenomics to characterize regional genetic heterogeneity in glioblastoma. Neuro Oncol 2017;19:128–37 doi:10.1093/neuonc/now135 pmid:27502248
    CrossRefPubMed
  51. 51.↵
    1. Colen RR,
    2. Hassan I,
    3. Elshafeey N, et al
    . Shedding light on the 2016 World Health Organization Classification of Tumors of the Central Nervous System in the era of radiomics and radiogenomics. Magn Reson Imaging Clin N Am 2016;24:741–49 doi:10.1016/j.mric.2016.07.001 pmid:27742114
    CrossRefPubMed
  52. 52.↵
    1. Gutman DA,
    2. Cooper LA,
    3. Hwang SN, et al
    . MR imaging predictors of molecular profile and survival: multi-institutional study of the TCGA glioblastoma data set. Radiology 2013;267:560–69 doi:10.1148/radiol.13120118 pmid:23392431
    CrossRefPubMed
  53. 53.↵
    1. Zinn PO,
    2. Singh SK,
    3. Kotrotsou A, et al
    . A co-clinical radiogenomic validation study: conserved magnetic resonance radiomic appearance of periostin expressing glioblastoma in patients and xenograft models. Clin Cancer Res 2018;24:6288–99 doi:10.1158/1078-0432.CCR-17-3420 pmid:30054278
    Abstract/FREE Full Text
  54. 54.↵
    1. Reardon DA,
    2. Wen PY
    . Glioma in 2014: unravelling tumour heterogeneity: implications for therapy. Nat Rev Clin Oncol 2015;12:69–70 doi:10.1038/nrclinonc.2014.223 pmid:25560529
    CrossRefPubMed
  55. 55.↵
    1. Lemee JM,
    2. Clavreul A,
    3. Menei P
    . Intratumoral heterogeneity in glioblastoma: don't forget the peritumoral brain zone. Neuro Oncol 2015;17:1322–32 doi:10.1093/neuonc/nov119 pmid:26203067
    CrossRefPubMed
  56. 56.↵
    1. Li Y,
    2. Liu X,
    3. Qian Z, et al
    . Genotype prediction of ATRX mutation in lower-grade gliomas using an MRI radiomics signature. Eur Radiol 2018;28:2960–68 doi:10.1007/s00330-017-5267-0 pmid:29404769
    CrossRefPubMed
  57. 57.↵
    1. Lin AL,
    2. White M,
    3. Miller-Thomas MM, et al
    . Molecular and histologic characteristics of pseudoprogression in diffuse gliomas. J Neurooncol 2016;130:529–33 doi:10.1007/s11060-016-2247-1 pmid:27704386
    CrossRefPubMed
  58. 58.↵
    1. Xi YB,
    2. Guo F,
    3. Xu ZL, et al
    . Radiomics signature: a potential biomarker for the prediction of MGMT promoter methylation in glioblastoma. J Magn Reson Imaging 2018;47:1380–87 doi:10.1002/jmri.25860 pmid:28926163
    CrossRefPubMed
  59. 59.↵
    1. Korfiatis P,
    2. Kline TL,
    3. Coufalova L, et al
    . MRI texture features as biomarkers to predict MGMT methylation status in glioblastomas. Med Phys 2016;43:2835–44 doi:10.1118/1.4948668 pmid:27277032
    CrossRefPubMed
  60. 60.↵
    1. Akbari H,
    2. Bakas S,
    3. Pisapia JM, et al
    . In vivo evaluation of EGFRvIII mutation in primary glioblastoma patients via complex multiparametric MRI signature. Neuro Oncol 2018;20:1068–79 doi:10.1093/neuonc/noy033 pmid:29617843
    CrossRefPubMed
  61. 61.↵
    1. Chang P,
    2. Grinband J,
    3. Weinberg BD, et al
    . Deep-learning convolutional neural networks accurately classify genetic mutations in gliomas. AJNR Am J Neuroradiol 2018;39:1201–07 doi:10.3174/ajnr.A5667 pmid:29748206
    Abstract/FREE Full Text
  62. 62.↵
    1. Zinn PO,
    2. Singh SK,
    3. Kotrotsou A, et al
    . Distinct radiomic phenotypes define glioblastoma TP53-PTEN-EGFR mutational landscape. Neurosurgery 2017;64:203–10 doi:10.1093/neuros/nyx316 pmid:28899058
    CrossRefPubMed
  63. 63.↵
    1. Kaloshi G,
    2. Benouaich-Amiel A,
    3. Diakite F, et al
    . Temozolomide for low-grade gliomas: predictive impact of 1p/19q loss on response and outcome. Neurology 2007;68:1831–36 doi:10.1212/01.wnl.0000262034.26310.a2 pmid:17515545
    CrossRefPubMed
  64. 64.↵
    1. Cha S
    . Neuroimaging in neuro-oncology. Neurotherapeutics 2009;6:465–77 doi:10.1016/j.nurt.2009.05.002 pmid:19560737
    CrossRefPubMed
  65. 65.↵
    1. Al-Okaili RN,
    2. Krejza J,
    3. Woo JH, et al
    . Intraaxial brain masses: MR imaging-based diagnostic strategy–initial experience. Radiology 2007;243:539–50 doi:10.1148/radiol.2432060493 pmid:17456876
    CrossRefPubMed
  66. 66.↵
    1. Chen C,
    2. Ou X,
    3. Wang J, et al
    . Radiomics-based machine learning in differentiation between glioblastoma and metastatic brain tumors. Front Oncol 2019;9:806 doi:10.3389/fonc.2019.00806 pmid:31508366
    CrossRefPubMed
  67. 67.↵
    1. Kang D,
    2. Park JE,
    3. Kim YH, et al
    . Diffusion radiomics as a diagnostic model for atypical manifestation of primary central nervous system lymphoma: development and multicenter external validation. Neuro Oncol 2018;20:1251–61 doi:10.1093/neuonc/noy021 pmid:29438500
    CrossRefPubMed
  68. 68.↵
    1. Kong Z,
    2. Jiang C,
    3. Zhu R, et al
    . (18)F-FDG-PET-based radiomics features to distinguish primary central nervous system lymphoma from glioblastoma. Neuroimage Clin 2019;23:101912 doi:10.1016/j.nicl.2019.101912 pmid:31491820
    CrossRefPubMed
  69. 69.↵
    1. Ortiz-Ramon R,
    2. Larroza A,
    3. Ruiz-Espana S, et al
    . Classifying brain metastases by their primary site of origin using a radiomics approach based on texture analysis: a feasibility study. Eur Radiology 2018;28:4514–23 doi:10.1007/s00330-018-5463-6 pmid:29761357
    CrossRefPubMed
  70. 70.↵
    1. Sun R,
    2. Limkin EJ,
    3. Vakalopoulou M, et al
    . A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol 2018;19:1180–91 doi:10.1016/S1470-2045(18)30413-3 pmid:30120041
    CrossRefPubMed
  71. 71.↵
    1. Tian Q,
    2. Yan LF,
    3. Zhang X, et al
    . Radiomics strategy for glioma grading using texture features from multiparametric MRI. J Magn Reson Imaging 2018;48:1518–28 doi:10.1002/jmri.26010 pmid:29573085
    CrossRefPubMed
  72. 72.↵
    1. Theresia E,
    2. Malueka RG,
    3. Pranacipta S, et al
    . Association between Ki-67 labeling index and histopathological grading of glioma in Indonesian population. Asian Pac J Cancer Prev 2020;21:1063–68 doi:10.31557/APJCP.2020.21.4.1063 pmid:32334471
    CrossRefPubMed
  73. 73.↵
    1. Grossmann P,
    2. Narayan V,
    3. Chang K, et al
    . Quantitative imaging biomarkers for risk stratification of patients with recurrent glioblastoma treated with bevacizumab. Neuro Oncol 2017;19:1688–97 doi:10.1093/neuonc/nox092 pmid:28499022
    CrossRefPubMed
  74. 74.↵
    1. Szychot E,
    2. Youssef A,
    3. Ganeshan B, et al
    . Predicting outcome in childhood diffuse midline gliomas using magnetic resonance imaging based texture analysis. J Neuroradiol 2021;48:243–47 doi:10.1016/j.neurad.2020.02.005 pmid:32184119
    CrossRefPubMed
  75. 75.↵
    1. McGarry SD,
    2. Hurrell SL,
    3. Kaczmarowski AL, et al
    . Magnetic resonance imaging-based radiomic profiles predict patient prognosis in newly diagnosed glioblastoma before therapy. Tomography 2016;2:223–28 doi:10.18383/j.tom.2016.00250 pmid:27774518
    CrossRefPubMed
  76. 76.↵
    1. Narang S,
    2. Kim D,
    3. Aithala S, et al
    . Tumor image-derived texture features are associated with CD3 T-cell infiltration status in glioblastoma. Oncotarget 2017;8:101244–54 doi:10.18632/oncotarget.20643 pmid:29254160
    CrossRefPubMed
  77. 77.↵
    1. Kotrotsou A,
    2. Zinn PO,
    3. Colen RR
    . Radiomics in brain tumors: an emerging technique for characterization of tumor environment. Magn Reson Imaging Clin N Am 2016;24:719–29 doi:10.1016/j.mric.2016.06.006 pmid:27742112
    CrossRefPubMed
  78. 78.↵
    1. Arevalo OD,
    2. Soto C,
    3. Rabiei P, et al
    . Assessment of glioblastoma response in the era of bevacizumab: longstanding and emergent challenges in the imaging evaluation of pseudoresponse. Front Neurol 2019;10:460 doi:10.3389/fneur.2019.00460 pmid:31133966
    CrossRefPubMed
  79. 79.↵
    1. Okada H,
    2. Weller M,
    3. Huang R, et al
    . Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol 2015;16:e534–42 doi:10.1016/S1470-2045(15)00088-1 pmid:26545842
    CrossRefPubMed
  80. 80.↵
    1. Wan B,
    2. Wang S,
    3. Tu M, et al
    . The diagnostic performance of perfusion MRI for differentiating glioma recurrence from pseudoprogression: a meta-analysis. Medicine (Baltimore) 2017;96:e6333 doi:10.1097/MD.0000000000006333 pmid:28296759
    CrossRefPubMed
  81. 81.↵
    1. Huang RY,
    2. Neagu MR,
    3. Reardon DA, et al
    . Pitfalls in the neuroimaging of glioblastoma in the era of antiangiogenic and immuno/targeted: detecting illusive disease, defining response. Front Neurol 2015;6:33 doi:10.3389/fneur.2015.00033 pmid:25755649
    CrossRefPubMed
  82. 82.↵
    1. Paugh BS,
    2. Qu C,
    3. Jones C, et al
    . Integrated molecular genetic profiling of pediatric high-grade gliomas reveals key differences with the adult disease. J Clin Oncol 2010;28:3061–68 doi:10.1200/JCO.2009.26.7252 pmid:20479398
    Abstract/FREE Full Text
  83. 83.↵
    1. Siegel R,
    2. Naishadham D,
    3. Jemal A
    . Cancer statistics, 2013. CA Cancer J Clin 2013;63:11–30 doi:10.3322/caac.21166 pmid:23335087
    CrossRefPubMed
  84. 84.↵
    1. Dong J,
    2. Li L,
    3. Liang S, et al
    . Differentiation between ependymoma and medulloblastoma in children with radiomics approach. Acad Radiol 2021;28:318–27 doi:10.1016/j.acra.2020.02.012 pmid:32222329
    CrossRefPubMed
  85. 85.↵
    1. Fetit AE,
    2. Novak J,
    3. Rodriguez D, et al
    . Radiomics in paediatric neuro-oncology: a multicentre study on MRI texture analysis. NMR Biomed 2018 Jan 3 [Epub ahead of print] doi:10.1002/nbm.3781 pmid:29073725
    CrossRefPubMed
  86. 86.↵
    1. Khatua S,
    2. Cooper LJN,
    3. Sandberg DI, et al
    . Phase I study of intraventricular infusions of autologous ex-vivo-expanded NK cells in children with recurrent medulloblastoma and ependymoma. Neuro Oncol 2020;22:1214–25 doi:10.1093/neuonc/noaa047 pmid:32152626
    CrossRefPubMed
  87. 87.↵
    1. Zhou H,
    2. Hu R,
    3. Tang O, et al
    . Automatic machine learning to differentiate pediatric posterior fossa tumors on routine MR imaging. AJNR Am J Neuroradiol 2020;41:1279–85 doi:10.3174/ajnr.A6621 pmid:32661052
    Abstract/FREE Full Text
  88. 88.↵
    1. Fetit AE,
    2. Novak J,
    3. Rodriguez D, et al
    . 3D texture analysis of heterogeneous MRI data for diagnostic classification of childhood brain tumours. Stud Health Technol Inform 2015;213:19–22 pmid:26152942
    PubMed
  89. 89.↵
    1. Northcott PA,
    2. Robinson GW,
    3. Kratz CP, et al
    . Medulloblastoma. Nat Rev Dis Primers 2019;5:11 doi:10.1038/s41572-019-0063-6 pmid:30765705
    CrossRefPubMed
  90. 90.↵
    1. Kumar R,
    2. Liu APY,
    3. Orr BA, et al
    . Advances in the classification of pediatric brain tumors through DNA methylation profiling: from research tool to frontline diagnostic. Cancer 2018;124:4168–80 doi:10.1002/cncr.31583 pmid:30255939
    CrossRefPubMed
  91. 91.↵
    1. Wagner MW,
    2. Hainc N,
    3. Khalvati F, et al
    . Radiomics of pediatric low-grade gliomas: toward a pretherapeutic differentiation of BRAF-mutated and BRAF-fused tumors. AJNR Am J Neuroradiol 2021;42:759–65 doi:10.3174/ajnr.A6998 pmid:33574103
    Abstract/FREE Full Text
  92. 92.↵
    1. Foster JB,
    2. Madsen PJ,
    3. Hegde M, et al
    . Immunotherapy for pediatric brain tumors: past and present. Neuro Oncol 2019;21:1226–38 doi:10.1093/neuonc/noz077 pmid:31504801
    CrossRefPubMed
  93. 93.↵
    1. Wang SS,
    2. Bandopadhayay P,
    3. Jenkins MR
    . Towards immunotherapy for pediatric brain tumors. Trends Immunol 2019;40:748–61 doi:10.1016/j.it.2019.05.009 pmid:31229353
    CrossRefPubMed
  94. 94.↵
    1. Barredo Arrieta A,
    2. Díaz-Rodríguez N,
    3. Del Ser J, et al
    . Explainable Artificial Intelligence (XAI): concepts, taxonomies, opportunities and challenges toward responsible AI. Information Fusion 2020;58:82–115 doi:10.1016/j.inffus.2019.12.012
    CrossRef
  • Received February 1, 2021.
  • Accepted after revision July 19, 2021.
  • © 2022 by American Journal of Neuroradiology
PreviousNext
Back to top

In this issue

American Journal of Neuroradiology: 43 (6)
American Journal of Neuroradiology
Vol. 43, Issue 6
1 Jun 2022
  • Table of Contents
  • Index by author
  • Complete Issue (PDF)
Advertisement
Print
Download PDF
Email Article

Thank you for your interest in spreading the word on American Journal of Neuroradiology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Evolving Role and Translation of Radiomics and Radiogenomics in Adult and Pediatric Neuro-Oncology
(Your Name) has sent you a message from American Journal of Neuroradiology
(Your Name) thought you would like to see the American Journal of Neuroradiology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Cite this article
M. Ak, S.A. Toll, K.Z. Hein, R.R. Colen, S. Khatua
Evolving Role and Translation of Radiomics and Radiogenomics in Adult and Pediatric Neuro-Oncology
American Journal of Neuroradiology Jun 2022, 43 (6) 792-801; DOI: 10.3174/ajnr.A7297

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
0 Responses
Respond to this article
Share
Bookmark this article
Radiomics in Neuro-Oncology
M. Ak, S.A. Toll, K.Z. Hein, R.R. Colen, S. Khatua
American Journal of Neuroradiology Jun 2022, 43 (6) 792-801; DOI: 10.3174/ajnr.A7297
del.icio.us logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One
Purchase

Jump to section

  • Article
    • Abstract
    • ABBREVIATIONS:
    • Conclusions
    • Footnotes
    • References
  • Figures & Data
  • Supplemental
  • Info & Metrics
  • Responses
  • References
  • PDF

Related Articles

  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Crossref (21)
  • Google Scholar

This article has been cited by the following articles in journals that are participating in Crossref Cited-by Linking.

  • Role of Artificial Intelligence in Radiogenomics for Cancers in the Era of Precision Medicine
    Sanjay Saxena, Biswajit Jena, Neha Gupta, Suchismita Das, Deepaneeta Sarmah, Pallab Bhattacharya, Tanmay Nath, Sudip Paul, Mostafa M. Fouda, Manudeep Kalra, Luca Saba, Gyan Pareek, Jasjit S. Suri
    Cancers 2022 14 12
  • 18F-FDG PET/CT-based radiomics nomogram could predict bone marrow involvement in pediatric neuroblastoma
    Lijuan Feng, Xu Yang, Xia Lu, Ying Kan, Chao Wang, Dehui Sun, Hui Zhang, Wei Wang, Jigang Yang
    Insights into Imaging 2022 13 1
  • Pre-operative MRI radiomics model non-invasively predicts key genomic markers and survival in glioblastoma patients
    Mathew Pease, Zachary C. Gersey, Murat Ak, Ahmed Elakkad, Aikaterini Kotrotsou, Serafettin Zenkin, Nabil Elshafeey, Priyadarshini Mamindla, Vinodh A. Kumar, Ashok J. Kumar, R. R. Colen, P. O. Zinn
    Journal of Neuro-Oncology 2022 160 1
  • Targeting Isocitrate Dehydrogenase (IDH) in Solid Tumors: Current Evidence and Future Perspectives
    Francesca Carosi, Elisabetta Broseghini, Laura Fabbri, Giacomo Corradi, Riccardo Gili, Valentina Forte, Roberta Roncarati, Daria Maria Filippini, Manuela Ferracin
    Cancers 2024 16 15
  • Radiomics analysis of contrast-enhanced computed tomography in predicting the International Neuroblastoma Pathology Classification in neuroblastoma
    Haoru Wang, Mingye Xie, Xin Chen, Jin Zhu, Li Zhang, Hao Ding, Zhengxia Pan, Ling He
    Insights into Imaging 2023 14 1
  • Prediction of chemotherapy-related complications in pediatric oncology patients: artificial intelligence and machine learning implementations
    Seda Ardahan Sevgili, Selmin Şenol
    Pediatric Research 2023 93 2
  • AI-driven estimation of O6 methylguanine-DNA-methyltransferase (MGMT) promoter methylation in glioblastoma patients: a systematic review with bias analysis
    Mullapudi Venkata Sai Samartha, Navneet Kumar Dubey, Biswajit Jena, Gorantla Maheswar, Wen-Cheng Lo, Sanjay Saxena
    Journal of Cancer Research and Clinical Oncology 2024 150 2
  • Associating T1-Weighted and T2-Weighted Magnetic Resonance Imaging Radiomic Signatures With Preoperative Symptom Severity in Patients With Cervical Spondylotic Myelopathy
    Nima Alan, Serafettin Zenkin, Raj Swaroop Lavadi, Andrew D. Legarreta, Joseph S. Hudson, Daryl P. Fields, Nitin Agarwal, Priyadarshini Mamindla, Murat Ak, Vishal Peddagangireddy, Lauren Puccio, Thomas J. Buell, D. Kojo Hamilton, Adam S. Kanter, David O. Okonkwo, Pascal O. Zinn, Rivka R. Colen
    World Neurosurgery 2024 184
  • Beyond hand-crafted features for pretherapeutic molecular status identification of pediatric low-grade gliomas
    Kareem Kudus, Matthias W. Wagner, Khashayar Namdar, Julie Bennett, Liana Nobre, Uri Tabori, Cynthia Hawkins, Birgit Betina Ertl-Wagner, Farzad Khalvati
    Scientific Reports 2024 14 1
  • Radiomic profiles improve prognostication and reveal targets for therapy in cervical cancer
    Mari Kyllesø Halle, Erlend Hodneland, Kari S. Wagner-Larsen, Njål G. Lura, Kristine E. Fasmer, Hege F. Berg, Tomasz Stokowy, Aashish Srivastava, David Forsse, Erling A. Hoivik, Kathrine Woie, Bjørn I. Bertelsen, Camilla Krakstad, Ingfrid S. Haldorsen
    Scientific Reports 2024 14 1

More in this TOC Section

  • Diagnostic Neuroradiology of Monoclonal Antibodies
  • ML for Glioma Molecular Subtype Prediction
  • Segmentation of Brain Metastases with BLAST
Show more Adult Brain

Similar Articles

Advertisement

Indexed Content

  • Current Issue
  • Accepted Manuscripts
  • Article Preview
  • Past Issues
  • Editorials
  • Editors Choice
  • Fellow Journal Club
  • Letters to the Editor

Cases

  • Case Collection
  • Archive - Case of the Week
  • Archive - Case of the Month
  • Archive - Classic Case

Special Collections

  • Special Collections

Resources

  • News and Updates
  • Turn around Times
  • Submit a Manuscript
  • Author Policies
  • Manuscript Submission Guidelines
  • Evidence-Based Medicine Level Guide
  • Publishing Checklists
  • Graphical Abstract Preparation
  • Imaging Protocol Submission
  • Submit a Case
  • Become a Reviewer/Academy of Reviewers
  • Get Peer Review Credit from Publons

Multimedia

  • AJNR Podcast
  • AJNR SCANtastic
  • Video Articles

About Us

  • About AJNR
  • Editorial Board
  • Not an AJNR Subscriber? Join Now
  • Alerts
  • Feedback
  • Advertise with us
  • Librarian Resources
  • Permissions
  • Terms and Conditions

American Society of Neuroradiology

  • Not an ASNR Member? Join Now

© 2025 by the American Society of Neuroradiology All rights, including for text and data mining, AI training, and similar technologies, are reserved.
Print ISSN: 0195-6108 Online ISSN: 1936-959X

Powered by HighWire