You are on page 1of 15

REVIEW

Artificial Intelligence Applications


in Glioma With 1p/19q Co-Deletion:
A Systematic Review
Simin Zhang, PhD,1 Lijuan Yin, MD, PhD,2 Lu Ma, MD, PhD,3 and Huaiqiang Sun, PhD1*

As an important genomic marker for oligodendrogliomas, early determination of 1p/19q co-deletion status is critical for
guiding therapy and predicting prognosis in patients with glioma. The purpose of this study is to systematically review the
literature concerning the magnetic resonance imaging (MRI) with artificial intelligence (AI) methods for predicting 1p/19q
co-deletion status in glioma. PubMed, Scopus, Embase, and IEEE Xplore were searched in accordance with the Preferred
Reporting Items for systematic reviews and meta-analyses guidelines. Methodological quality of studies was assessed
according to the Quality Assessment of Diagnostic Accuracy Studies-2. Finally, 28 studies were included in the quantitative
analysis. Diagnostic test accuracy reached an area under the ROC curve of 0.71–0.98 were reported in 24 studies. The
remaining four studies with no available AUC provided an accuracy of 0.75–0. 89. The included studies varied widely in
terms of imaging sequences, input features, and modeling methods. The current review highlighted that integrating MRI
with AI technology is a potential tool for determination 1p/19q status pre-operatively and noninvasively, which can possi-
bly help clinical decision-making. However, the reliability and feasibility of this approach still need to be further validated
and improved in a real clinical setting.
Evidence Level: 2.
Technical Efficacy: 2.
J. MAGN. RESON. IMAGING 2023;58:1338–1352.

G liomas are the most common and infiltrative primary


intracranial neoplasms. In 2016, the World Health Orga-
nization (WHO) provided an update of brain tumor classifica-
co-deletion have significantly longer OS/PFS times com-
pared to those with no 1p/19q co-deletion. Aside from its
diagnostic and prognostic values, 1p/19q co-deletion has
tion based on histological and molecular characteristics.1 been revealed as an informative biomarker for surgical plan-
Furtherly, the latest WHO criteria (2021, 5th edition) solid- ning in low-grade glioma (LGG).4–6 A prior study suggested
ifies the centrality of molecular markers in the delineation of 1p/19q intact but not 1p/19q co-deleted LGG would bene-
glioma categories, generating three primary subtypes of glioma: fit from gross total resection.7 More specifically, studies
1) astrocytoma, IDH-mutant; 2) oligodendroglioma, IDH- reported residual tumor has a more negative impact on sur-
mutant, and 1p/19q co-deletion; and 3) glioblastoma, IDH- vival in 1p/19q intact, IDH-mutated astrocytomas than on
wildtype2 (Fig. 1). Therefore, mutations in the IDH and 1p/19q co-deletion, IDH-mutated oligodendrogliomas.5,8
co-deletion of chromosomes 1p and 19q are playing vital role Therefore, determination of 1p/19q co-deletion status at the
in determining glioma subtypes. earliest possible stage, that is, even before surgery is critical
As a favorable prognostic biomarker, 1p/19q co- for tumor subtyping, prognostication, personalized treat-
deletion status is a good indicator for better response to ment, especially an informed surgical planning, which may
adjuvant chemotherapy,3 and patients with 1p/19q avoid nonmaximal resection in 1p/19q intact LGG or

View this article online at wileyonlinelibrary.com. DOI: 10.1002/jmri.28737

Received Dec 21, 2022, Accepted for publication Apr 3, 2023.

*Address reprint requests to: H.S., Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu,
610041, China. E-mail: sunhuaiqiang@scu.edu.cn.
This work was supported by the National Natural Science Foundation of China (Grant No. 81974278).

From the 1Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China; 2Department of
Pathology, West China Hospital of Sichuan University, Chengdu, China; and 3Department of Neurosurgery, West China Hospital of Sichuan University,
Chengdu, China
Additional supporting information may be found in the online version of this article

1338 © 2023 International Society for Magnetic Resonance in Medicine.


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Zhang et al.: A Review of AI Applications in Glioma With 1p/19q Co-Deletion

FIGURE 1: Sample MRI acquisition in three primary subtypes of glioma. Figure displays axial slices of three patients from our own
dataset. In the top row, MRI images from a 24-year-old female with astrocytoma (grade 2, IDH mutant and 1p/19q noncodeleted), in
the middle bottom row, images from a 37-year-old male with oligodendroglioma (grade 2, IDH-mutant and 1p/19q-codeleted), and
in the bottom row, images from a 44-year-old female with glioblastoma (grade 4, IDH-wildtype and 1p/19q noncodeleted).

less-justified surgery-related neurological deficits in 1p/19q as a result, subjective, with interobserver variability in diagno-
co-deleted glioma. sis. Therefore, the accuracy of visual features has not yet been
However, with current technology, identification of sufficiently established to completely avoid histopathological
1p/19q co-deletion in gliomas requires invasive surgical re- evaluation following neurosurgical intervention.
section or biopsy followed by fluorescence in situ hybridiza- Artificial intelligence (AI) is emerging as a solution to
tion.9 Nevertheless, surgical procedures may be associated the limitations of conventional visual assessment, which may
with morbidity, mortality, and high costs.10,11 Additionally, help identify features that hidden to the naked eye and associ-
nonrepresentative tissue samples due to intratumoural hetero- ate them with outcomes. Machine learning (ML), a sub-
geneity of gliomas may bring some bias in determining gene category of AI, focused on developing algorithms that can
mutation status.12 Thus, there is a growing interest in devel- identify patterns within data without explicit specification.
oping noninvasive alternatives that can provide molecular and ML used artificial feature engineering (extraction of hand-
genetic information. crafted radiomics feature, reduction and selection of features)
Magnetic resonance imaging (MRI) has been proposed followed by utilization of ML algorithms for task predic-
as a potential candidate due to its noninvasive nature and tion.16 This pipeline is simplified and, at the same time,
wide clinical applicability. Studies suggested that 1p/19q co- enhanced by using deep learning (DL). Unlike ML, DL fol-
deletion was associated with specific imaging characteristics, lows the neural network architectures approach, which pro-
such as indistinct border, heterogeneous signal intensity, para- vides automatic learning of features and their representation
magnetic susceptibility effect and calcification.13 Recently, in a hierarchical manner at various levels. Additionally, by
studies have also reported the “T2-FLAIR mismatch sign,” employing a hierarchy of nonlinear transforms, DL can model
which represents a highly specific imaging biomarker for the very complex data patterns, making it more suitable for learn-
IDH-mutant, 1p/19q non-co-deletion molecular sub- ing complicated patterns and subtle differences in data, par-
type.14,15 These investigations encourage prospective evalua- ticularly the image data of human brain.17 The introduction
tion of molecular alterations and MRI characteristics of of AI to neuro-oncology imaging has been met with great
glioma. Nonetheless, these image features are qualitative and, enthusiasm from clinicians. Currently, extensive research

November 2023 1339


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Journal of Magnetic Resonance Imaging

articles using AI in neuro-oncology have been published, “oligodendroglioma” OR “LGG” OR “glioblastoma” OR


which mainly focus on the following tasks: 1) brain tumor “GBM”) AND (“1p/19q” OR “phenotyping” OR “molecu-
segmentation18; 2) predictive tasks (tumor type, grade, genet- lar subtypes” OR “genotype”) AND (“magnetic resonance
ics, and molecular marker identification)19,20; 3) survival pre- imaging” OR “MRI”).
diction.21 With regard to the utilization of AI in genetic
status prediction, various studies have reported promising Eligibility Criteria
results on prediction of 1p/19q co-deletion. According to the scope of the review, we considered AI inves-
We believe that it is of utmost importance to evaluate tigations aimed at 1p/19q co-deletion prediction in glioma.
the published research on the prediction 1p/19q co-deletion The imaging modalities we considered were MRI and
of glioma of AI applications due to the growing significance PET/MRI. Studies were excluded under the following cir-
of understanding the pathophysiology of glioma with 1p/19q cumstances: 1) articles that are not written in English; 2) the
co-deletion and the rising dependence on AI for prediction. type of articles are review, meta-analysis, case reports, edito-
In this article, we will review the articles concerning the com- rials or letters, comments, guidelines, and conference proceed-
bination of MRI with AI to predict glioma 1p/19q co- ings; 3) animal studies; 4) research articles focused on
deletion status and discusses the special considerations in methodological aspects; 5) lack of validation in a clinical set-
neuro-oncology, with the hope of yielding more actionable ting; 6) lack of model evaluation metrics (i.e. sensitivity, spec-
change to clinical workflows. ificity, accuracy, and/or AUC) on validation cohorts.
Based on the eligibility criteria, the titles and abstracts
Methods of articles were screened to evaluate their suitability for inclu-
Article Search Strategy sion in this review by two reviewers (Z.S.M and Y. L.J), and
A search was conducted across several databases (PubMed, disagreements were resolved by discussion. The PRISMA
Scopus, Embase, and IEEE Xplore) to identify all relevant flowchart of study is shown in Fig. 2.
publications. Our review included studies published before
August 2022. The search strategy used to find AI studies were Data Extraction
(“radiomics” OR “radiogenomics” OR “machine learning” The extracted data from the included studies were number of
OR “support vector machine” OR “random forest” OR patients, glioma grade, type of dataset, MRI sequences, tumor
“deep learning” OR “deep neural network” OR “deep belief segmentation method, type of feature, software for feature
network” OR “convolutional neural network” OR “CNN” extraction, feature selection method, AI method for classifica-
OR “artificial intelligence” OR “AI” OR “computer aided tion, model performance on validation set (a held-out test set
diagnosis”) AND (“astrocytoma” OR “glioma” OR or external cohort was treated as validation).

FIGURE 2: PRISMA flowchart of systematic literature search.

1340 Volume 58, No. 5


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Zhang et al.: A Review of AI Applications in Glioma With 1p/19q Co-Deletion

Quality Assessment 4 adopted semi-automatic segmentation, 7 studies adopted


The quality assessments were performed based on Quality automated segmentation, 2 studies developed multitask CNN
Assessment of Diagnostic Accuracy Studies-2 (QUADAS- that can predict 1p/19q status simultaneously segmented
2).22,23 The entire processes of evaluation and questionnaire tumor (Fig. 3b).
filling were double-checked independently by Z.S.M and
Y. L. J and disagreement was resolved by consensus. Feature
Totally, 17 of included studies extracted radiomics features
Results such as tumor shape, intensity and texture, among which,
We identified 404 articles from the initial search. After pre- 5 studies combined with Visually Accessible Rembrandt
liminary screening and full text review, most studies were Imaging (VASARI) features and/or clinical characteristics
excluded for a number of reasons outlined (Fig. 2). Finally, (Fig. 3c). Of the remaining 11 studies adopted DL features
28 studies met all eligibility criteria were involved. utilizing 2D/2.5D/3D image patches as input (Fig. 3c).
Quality assessment results are summarized in Table S1, As for radiomics-based ML, the software for feature
five studies were considered as an unclear risk of bias in extraction were Pyradiomics (n = 5), Matlab with extention
patients selection domain as a lack of specification of consecu- (n = 3), 3D Slicer (n = 1), PREDICT toolbox (n = 1),
tive patient selection, four studies were categorized as unclear RadiomiX (n = 1), GLISTRboost (n = 1). Since the high
risk of bias in reference standard domain as lack of descrip- dimensionality and complexity of MRI data obtained through
tion the reference standard and how it was conducted and different sequences, feature dimensionality reduction, and
interpreted. selection were critical steps in the discovery of predictors from
high-dimensional input. The used dimensionality reduction
Study Characteristics and selection techniques of the included studies were as fol-
All studies used retrospectively collected data. Twenty studies lows: least absolute shrinkage and selection operator (LASSO)
drew patients from multiple institutions, commonly using the (n = 2), PCA (n = 2), variance threshold (n = 2), Spearman
TCIA database. The sample size ranged from 42 to 1748. correlation coefficient (n = 2), Bootstrap resampling (n = 1),
Most studies focused on WHO 2–4 glioma or WHO 2–3, ANOVA F-test (n = 1).
with one study including only WHO 2 glioma24 and one
study including only WHO 4 glioma.25 For the target condi- AI Method
tion, 10 studies focused on IDH mutation combined 1p/19q Seventeen studies utilized radiomics-based traditional ML for
co-deletion, while 18 studies on 1p/19q co-deletion only. 1p/19q status perdition, the classification algorithms includ-
ing Random Forest (RF) (n = 6), support vector machine
Imaging Acquisition (SVM) (n = 4), logistic regression (LR) (n = 3), Xtreme gra-
Imaging methods including scanner types and sequence dient boosting (XGBoost) (n = 2), Ensemble Bagged Trees
parameters can affect feature calculations. In total, 24 studies (EBT) (n = 1), neural network (n = 1). Eleven studies uti-
used conventional MRI sequences, including T1-weighted lized DL method, including autoencoder (n = 2), CNN
imaging (T1WI), contrast-enhanced T1-weighted imaging (n = 7), recurrent neural network (RNN) (n = 1), and gen-
(T1CE), followed by a frequent addition of T2-weighted erative adversarial network (GAN) (n = 1). Notably, several
imaging (T2WI) and fluid-attenuated inversion recovery studies used more than one AI method (see Table 1 and
(FLAIR). The use of diffusion-weighted imaging (DWI) was Fig. 3d).
presented in three publications, and the dynamic susceptibil- As for model development and evaluation, the methods
ity contrast perfusion MR imaging (DSC) was investigated in employed are as follows: k-fold cross validation
one publication (see Table 1 and Fig. 3a). (CV) (n = 13), hold out (n = 6), Bootstrap (n = 5), leave
one out CV (n = 1), hold-out and inner K-fold CV (n = 1).
Glioma Segmentation Additionally, 11 studies provided external validation cohorts
Segmentation is a crucial initial step in neuro-oncology imag- to assess model performance (Fig. 3e,f).
ing analysis. Method of segmentation can be manual, semi-
automatic, or fully automated. Manual segmentation involves Model Performance
comprehensively labeling the structure in each slice, which is The proposed models were assessed based on their presented
a time-consuming task with relatively low inter- and intra- area under the curve (AUC). An AUC was available in 24 of
individual reliability. Semi-automatic is a combination of the included studies, with AUC ranging from 0.71 to 0.98.
manual guidance and region growing algorithms. While, the The remaining four studies provided accuracy (ACC) for
automated method does not require any human involvement model performance (0.75–0.89). Table 1 summarized the
and is more suitable for processing large data sets. In the cur- study characteristics, AI method and the performance of the
rent review, 15 studies adopted manual segmentation, AI in terms of AUC or accuracy on the validation set. While

November 2023 1341


TABLE 1. Participant Demographics, Study Characteristics, and AI Performance of the Included Studies

1342
Patients, Glioma MRI Externally Feature Model
Study N Grade Sequences Data AI Methods D&E Validated Segmentation Software Input Selection Performance
Radiomics-based machine learning
Zhou et al 165 2, 3 T1, T2, TCIA LR Bootstrap No Manual Matlab Radiomics; Bootstrap AUC = 0.96
(2017)26 T1C, VASARI resampling
FLAIR
Han et al 277 2, 3 T2 In-house RF K-fold No Manual NA Radiomics; Variance AUC = 0.76
(2018)27 clinical threshold
Journal of Magnetic Resonance Imaging

Lu et al 87 2, 3, 4 T1, T2, TCIA SVM K-fold No Semiautomatic Matlab Radiomics t-test AUC = 0.92
(2018)28 T1C,
FLAIR
Shofty et al 47 2 T2, T1C, In-house EBT K-fold No Manual Matlab Radiomics PCA AUC = 0.87
(2018)29 FLAIR
Kim et al 167 2, 3, 4 T1, T2, TCIA RF Hold-out No Manual Pyradiomics Radiomics PCA AUC = 0.71
(2018)30 T1C,
FLAIR
van der 413 2, 3 T1C, T2 In- SVM K-fold Yes Manual PREDICT Radiomics; Not applied AUC = 0.72
Voort et al house, clinical
(2019)31 TCIA
Zhou et al 281 2, 3, 4 T1C, HUP, RF Bootstrap Yes Manual MATLAB Radiomics; Not applied AUC = 0.72
(2019)32 FLAIR BWH, clinical
MGH,
TCIA
Kong et al 96 2, 3 T1C, T2 In-house RF Hold-out No Manual Pyradiomics Radiomics LASSO AUC = 0.89
(2020)33
Kocak et al 107 2, 3 T1C, T2 TCIA Neural K-fold No Semiautomatic LIFEx Radiomics ReliefF AUC = 0.87
(2020)34 network
Rathore et al 159 2, 3 T1, T2 TCIA SVM Leave- No Semiautomatic NA Radiomics Not applied AUC = 0.86
(2020)35 one-
out

Volume 58, No. 5


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
TABLE 1. Continued

Patients, Glioma MRI Externally Feature Model


Study N Grade Sequences Data AI Methods D&E Validated Segmentation Software Input Selection Performance

November 2023
Casale et al 209 2, 3 T1C, T2 TCIA RF Hold- Yes Manual RadiomiX Radiomics Spearman AUC = 0.87
(2021)36 out; correlation
inner coefficient
K-fold
Fan et al 157 2 T1, T2, CGGA SVM Nested No Manual Matlab Radiomics Elastic Net AUC = 0.81
(2021)24 T1C K-fold
Kha 159 2, 3 T2 TCIA XGBoost K-fold Yes Manual 3D Slicer Radiomics Spearmans AUC = 0.87
et al Correlation
(2021)37 Coefficient
Sun et al 335 2, 3 T1, T2, In-house RF Hold- No Manual Pyradiomics Radiomics; Boruta AUC = 0.76
(2021)38 T1C, out; VASARI;
FLAIR inner clinical
DWI K-fold
Yan et al 357 2, 3, 4 T1, T2, In-house LR Hold- No Manual Pyradiomics Radiomic Variance AUC = 0.82
(2021)39 T1C, out; threshold;
FLAIR inner LASSO
DWI K-fold
Lam et al 106 2, 3 T1C, T2, TCIA XGBoost K-fold Yes Automatic GLISTRboost Radiomics Not applied AUC = 0.78
(2022)40 FLAIR
Li (2022)41 212 2, 3, 4 T1, T2, TCIA LR Hold-out No Automatic Pyradiomics Radiomics ANOVA F- AUC = 0.86
T1C, test
FLAIR
Deep learning
Akkus et al 159 2, 3 T1C, T2 In-house CNN K-fold No Semiautomatic NA Image NA ACC = 0.88
(2017)42 patch
(2D)
Chang et al 259 2, 3, 4 T1, T2, TCIA CNN K-fold No Automatic NA Image NA AUC = 0.88
(2018)43 T1C, patch
FLAIR (2D)
Zhang et al.: A Review of AI Applications in Glioma With 1p/19q Co-Deletion

1343
15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1344
TABLE 1. Continued

Patients, Glioma MRI Externally Feature Model


Study N Grade Sequences Data AI Methods D&E Validated Segmentation Software Input Selection Performance
Choi et al 463 2, 3, 4 DSC In-house RNN Bootstrap No Automatic NA Time series NA AUC = 0.89
(2019)44 perfusion + LSTM
MRI
Ali et al 161 2 T1C, In-house GAN K-fold Yes End to end NA Image NA ACC = 0.75
(2020)45 FLAIR (two patch
Journal of Magnetic Resonance Imaging

sites) (2D)
Tang et al 120 4 T1C, DWI In-house CNN K-fold NO Manual NA Image NA ACC = 0.88
(2020)25 patch
(3D)
Decuyper 628 2, 3, 4 T1, T2, TCIA, CNN Bootstrap Yes Automatic NA Image NA AUC = 0.82
et al T1C, BraTS; patch
(2021)46 FLAIR in- (3D)
house
Tupe- 375 3, 4 T1C, T2, TCIA, Autoencoder NA No Automatic NA Image NA AUC = 0.87
Waghmare FLAIR in- patch
et al house (3D)
(2021)47
Ning et al 431 2, 3, 4 T1C, TCIA, Autoencoder NA Yes Manual NA Image NA AUC = 0.90
(2021)48 FLAIR in- patch
house (2.5D)
van der 1748 2, 3, 4 T1C, T2, TCIA, CNN Bootstrap Yes End to end NA Image NA AUC = 0.85
Voort et al FLAIR in- patch
(2022)49 house (3D)
Cluceru et al 531 2, 3, 4 T1C, T2, TCIA, CNN NA Yes Automatic NA Image NA ACC = 0.89
(2022)50 FLAIR, in- patch
DWI house (3D)

Volume 58, No. 5


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Zhang et al.: A Review of AI Applications in Glioma With 1p/19q Co-Deletion

Fig. 4 provides the associated forest plot for studies assessing

Performance

FLAIR = fluid attenuated inversion recovery; VASARI = visually accessible rembrandt images; LR = logistic regression; RF = random forest; EBT = Ensemble Bagged Trees;
network; DSC = dynamic susceptibility contrast perfusions weighted imaging; T1 = T1-weighted images; T1C = T1-weighted postcontrast images; T2 = T2-weighted images;
DWI = diffusion-weighted images; BraTS = brain tumor segmentation challenge; CNN = convolutional neural network; GAN = generative adversarial network; RNN = recurrent neural
AUC = 0.98
1p/19q co-deletion.
Model
Studies With Code
Seven studies published the analysis code they used, including
six radiomics studies and one DL study. All of the code is
Selection

hosted on Github (https://github.com/). Most of the code is


Feature

NA

written in Python, and most of the ML functions in it are


implemented using methods from the popular machine learn-
ing library scikit-learn. Some of the code also uses the
imbalanced-learn library to deal with data imbalance. It is
patch
(3D)
Image
Input

worth noting that the completeness of the code provided var-


ies from study to study, with some studies publishing code
that contains only the key steps in their analysis process. The
ease of use of these codes also varies, and the easiest one is
Software

PREDICT (https://github.com/Svdvoort/PREDICT), which


NA

puts all the information that needs to be configured into one


file, and one only needs to modify this file when applying the
code to a custom dataset. Given the difficulty of setting up a
Validated Segmentation

DL environment, PrognosAIs_glioma (https://github.com/


Manual

Svdvoort/PrognosAIs_glioma) provides a docker file to deploy


its preprocessing pipeline and the classification model in a vir-
tualized container (see Table 2 for more details).
Externally

CGGA = Chinese Glioma Genome Atlas; D&E = model development and evaluation; ACC = accuracy.

Discussion
Yes

According to the current systematic review, the status of


1p/19q co-deletion of gliomas can be predicted from nonin-
vasive medical imaging data using AI approaches with an
D&E
NA

AUC ranging from 0.71 to 0.98. The implementation of this


approach will help clinicians choose the best treatment plan
for glioma patients, particularly in planning the extent of re-
AI Methods

section prior to surgery. As shown in Fig. 5, there are cur-


CNN

rently two mainstream ways to implement AI in the field of


image-based prediction/classification: conventional ML
methods based on artificial feature engineering, such as radi-
house
TCIA,
Data

in-

omics and DL methods. Although differing in terms of fea-


ture extraction and model development, radiomics and DL
still share some common scientific and technical concerns,
Grade Sequences

FLAIR
T1, T2,

such as data collection and lesion segmentation.


T1C,
MRI

Imaging Modalities
The evaluation of the importance of different imaging modal-
Patients, Glioma

ities for the molecular prediction of glioma patients has


2, 3

always been one of the popular concerns. Given its availabil-


ity in most institutions, conventional MRI modalities
(T1WI, T2WI, T1CE, and FLAIR) are mostly used for gli-
555
N
TABLE 1. Continued

oma diagnosis. Based on our review, AI models based on rou-


tine MRI sequences achieved performance for predicting
1p/19q status with AUC ranging from 0.71 to 0.96. Other
(2022)51

advanced MRI modalities, like diffusion MRI (DWI) and


Yan et al

perfusion MRI (PWI), are also promising to capture more


Study

information that contributed to the prediction of 1p/19q sta-


tus. Compared with conventional MRI, the advantage of

November 2023 1345


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Journal of Magnetic Resonance Imaging

FIGURE 3: Summary of MRI sequences (a), tumor segmentation methods (b), types of imaging features (c), AI methods (d), model
development and evaluation methods (e), external validation (f) used by studies (n = 28) investigating AI models for predicting
1p/19q co-deletion in glioma. VASARI = visually accessible rembrandt imaging; GAN = generative adversarial network;
CNN = convolutional neural network; RNN = recurrent neural network; ML = machine learning.

DWI is the assessment of high cellularity areas, which can and the expertise needed for data postprocessing and interpre-
help identify tissue microstructure and depict neoplastic infil- tation make this approach difficult to be widely implemented
tration in region of brain that appear normal on conventional in clinical practice.
MRI. Sun et al38 reported that the AUC of the radiomics
model without ADC sequence in the testing cohort were Lesion Segmentation
0.65 for IDH mutation combined 1p/19q co-deletion, while Accurate segmentation of lesion regions is crucial for radi-
after incorporating the ADC information into the radiomics omics studies. Although DL methods have the ability to
model, the AUC of prediction model increased to 0.76. extract useful features directly from images, most DL-based
Recent study suggested that the IDH genotype can be predi- studies (9/11 in the current review) still perform lesions seg-
cated using relative cerebral blood volume (rCBV) mapping mentation due to the limitation of sample size. And the pat-
obtained from DSC perfusion MRI,52 which has highlighted ches that contain lesion were used as the input for deep
its potential value used to investigate glioma angiogenesis. network training, allowing the network to focus on the lesion
Further, Choi et al44 assessed feasibility of multidimensional regions without the interference of normal regions and back-
time-series from DSC perfusion MRI in prediction of 1p/19q ground. The irregular morphology, complex tissue compo-
status. The results demonstrate excellent performance with nents (often including tumor parenchyma, necrosis,
both interpretability and reproducibility on the test set peritumour edema), ambiguous boundaries as well as variable
(AUC = 0.95). From the published literature, it is evident location make accurate segmentation of glioma very challeng-
that advanced sequences have the potential to enhance predic- ing. Glioma was segmented manually by clinicians in 15 of
tive ability. However, it remains to be further investigated included studies, however, this process is time-consuming
whether the model should be constructed by combining tra- with high inter-observer variability. Semi-automatic segmen-
ditional and advanced sequences or by choosing one of them. tation is a combination of manual interventions and algo-
More sequences may increase the redundant features in the rithms. It usually requires a manual initialization to guide the
prediction model and reduce its diagnostic specificity. On the algorithm and post correction by experienced physicians. This
other hand, the lack of broad clinical dissemination of method can save part of the labor and was used in 4 of the
advanced MRI sequences in most hospital imaging protocols involved studies. In contrast, fully automated methods do not

1346 Volume 58, No. 5


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Zhang et al.: A Review of AI Applications in Glioma With 1p/19q Co-Deletion

FIGURE 4: 1p/19q status forest plot of included studies with an AUC.

require human involvement and are more suitable for han- Notably, this segmentation architecture demonstrated only a
dling large data sets. Currently, automatic methods are slight degradation in performance when only T1CE and
increasingly employed, and the field has been growing in part FLAIR or T1CE and T2WI scans are available compared to
spurred on by the Brain Tumor Segmentation (BraTS) chal- simultaneous input of all four modalities. This is particularly
lenges, which aiming to automatically segment the gliomas. useful as not all four modalities are available for all patients in
Li et al41 employed a conventional computer vision method, clinical practice. Since U-net architecture showed excellent
named GLISTRboost, to obtain the glioma masks automati- segmentation performance, a series of improved variants
cally. This algorithm achieved 88.5% dice score on BraTS based on original U-net were developed. For example,
2015 dataset.53 However, compared with computer vision Yogananda et al56 developed a 3D-Dense-UNets, which can
methods, recently proposed DL-based methods are more perform 1p/19q co-deletion status classification and simulta-
robust in segmenting targets with large variations. Chang neous single-label tumor segmentation. The whole tumor seg-
et al43 implemented a fully CNN with residual connections mentation mean dice score was 0.80 on TCIA dataset.
to perform whole-tumor segmentation which was the top-
performing algorithm as evaluated in the 2016 international Classification Model Development
Multimodal BraTS Challenge with a dice score of 0.87.54 The classical ML algorithms, like RF, SVM, LR and
Additionally, in our included studies, Choi et al44 used cas- XGBoost are mostly chosen in radiomics style studies. During
cade CNN to segment whole tumors into three sub-regions: 2018–2019, most of studies trained a single ML algorithm in
the enhancing and nonenhancing tumor core and the peri- their workflow. For example, Lu et al28 used and quadratic
tumoral edema. The cascaded CNN architecture proposed by SVM and achieved 80% accuracy in prediction 1p/19q co-
Wang et al,55 achieving a dice score of 0.90 for whole tumor deletion in IDH mutation low-grade glioma on the indepen-
on BraTS 2017 dataset. Recently, deep neural network with dent validation cohort containing only five patients. Han
U-Net architecture showed state-of-the-art performance in et al27 used RF and achieved an AUC of 0.76 on the valida-
brain tumor segmentation. U-Net is an encoder-decoder net- tion cohort, which contains 93 subjects. Recently, there has
work that combines semantic and spatial information through been a clear trend to blend or stack multiple ML algorithms
the use of skip connections from the encoder to the decoder, with different advantages to achieve improved and robust pre-
resulting satisfactory performance in both gross and fine dictive performance. In the work of Li et al41 and Kocak
structure segmentation. Decuyper et al46 applied a 3D U-Net et al,34 they employed various state-of-the-art ML algorithms
similar to the architecture proposed by Isensee et al,18 which to predict the 1p/19q status and compared the performance
achieved a dice score of 90% on BraTS 2019 dataset. of each algorithm. Overall, most existing radiomics studies

November 2023 1347


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Journal of Magnetic Resonance Imaging

TABLE 2. Studies With Code

Study Main How to


Study URL Language Type Input Dependencies Functionalities Re-use
Zhou et al https://github. MATLAB Radiomics Image NA Feature Additional
(2017)26 com/ and extraction; coding is
mvallieres/ mask feature needed to
radiomics processing; call the
model functions
development
van der https://github. Python Radiomics Image scikit-learn Feature Modify the
Voort com/ and extraction; configure
et al Svdvoort/ mask model file
(2019)31 PREDICT development
Kha e al https://github. Python Radiomics Tabular scikit-learn Model Change the
(2021)37 com/ data development path
khanhlee/ specified
LGG- in the
radiomics source
code
Casale https://github. Python Radiomics Tabular scikit-learn Feature selection Change the
et al com/ data imbalanced- model path
(2021)36 roberto- learn development specified
casale/LGG- in source
1p-19q- code
deletion
Yan et al https://github. Python R Radiomics Tabular brnn Feature selection; Change the
(2021)39 com/zhang- data model path
AI-MI/ development specified
glioma- in the
genotype- source
radiomics code
Li et al https://github. Python Radiomics Image pyradiomics Feature Change the
(2022)41 com/ and scikit-learn extraction; path
Yingping- mask imbalanced- feature specified
LI/ learn processing; in the
Radiomics model setting file
development
visualization
van der https://github. Python Deep Image tensorflow Image Build the
Voort com/ learning preprocessing; Docker
et al Svdvoort/ model and
(2022)49 PrognosAIs_ development organize
glioma the data as
requisted

identify ML algorithms mainly based on the researcher’s pref- “automated machine learning (autoML)” has been devel-
erence or the popularity of the algorithm. However, choosing oped to automate key steps of ML and to maximize the
the most appropriate ML pipeline for a given problem tends performance of ML models.58 As a new research direction,
to be complicated, requiring the researcher to have expertise autoML has been successfully applied to H3K27M
in both the ML algorithm and the target problem.57 To over- prediction and IDH combined with MGMT status
come these challenges, a relatively new system called prediction.59,60 However, fewer studies have applied

1348 Volume 58, No. 5


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Zhang et al.: A Review of AI Applications in Glioma With 1p/19q Co-Deletion

FIGURE 5: Characteristic workflow for developing ML glioma 1p/19q status prediction models.

AutoML to 1p/19q status prediction. Thus, further studies learn representations that are related to the task directly from
that focus on the feasibility of this approach for 1p/19q raw images, overcoming the limitations of radiomics in feature
status prediction are warranted. extraction. Akkus et al42 used a multiscale 2D CNN on three
Due to its better feasibility and interpretability on task slices centered at the tumor “equator” and achieved an accu-
datasets with limited sample size, radiomics continues to be the racy of 88% on the test set. Further, Chang et al43 used a com-
preferred tool for predicting clinical target based on imaging bination of ResNet and principal component analysis (PCA) to
features. However, artificially engineered features from radi- predict 1p/19q status, achieving an AUC of 0.88 on their in-
omics method may be a bottle neck limiting the performance house data. It is worth noting that the above models are all
of ML model, with their limited ability to represent complex trained on labelled data. In addition, Tupe-Waghmare et al47
data. In addition, radiomics features are susceptible to varia- built a semi-supervised model that can incorporate unlabeled
tions in imaging devices and imaging parameters, limiting the glioma data and learn to predict 1p/19q status. This model
value of radiomics models for application in real-world clinical combined auto-encoder with multitask CNN, which achieved
settings. The newly proposed DL method uses a neural net- an AUC of 0.82. While still in their pioneer stage, the DL
work with a large number of hidden layers to automatically approaches are promising tools for gaining insight into the

FIGURE 6: Challenges for clinical implementation of ML glioma 1p/19q status prediction models.

November 2023 1349


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Journal of Magnetic Resonance Imaging

relationships between complex data and avoiding the limita- Multicenter study is the current trend in AI research,
tions of artificial feature engineering. which can, on the one hand, attenuate the risk of over-
fitting and, at the same time, due to the inherent variabil-
ity of the data, make the cohort more representative and
Challenges thus lead to more robust inference of the model. TCIA
The small-sample high-dimensional problem is a challenge dataset are mostly used dataset in glioma AI studies, which
faced by almost all medical AI research and it is no exception including patients collected from different institutions with
in 1p/19q status prediction. DL is more data-intensive than different imaging devices or imaging protocols. Thus, the
radiomics as tens of thousands of weights in the deep neural nonbiological batch effects might be introduced.63 Data
network need to be determined during training. In our harmonization may be helpful for standardizing the vari-
included radiomics studies, the sample size ranged from 47 to able images for analysis. This process can mitigate spurious
413. As for DL, the sample size ranged from 120 to 1748. If effects on the scientific conclusions incorporated due to
the feature dimension is high and the training data is insuffi- undesired sources of variation, such as batch effects, intrin-
cient, there will be a great risk of obtaining an overfit model, sic factors within the subjects, and scanning sites. Remark-
which works perfectly on the training set but significantly ably, only Li et al41 applied both Z-score and ComBat for
degrades on the test set.61 The best solution to overfitting is normalization. Thus, for reducing the variability of the
of course collecting more data. However, in field of neuro- image properties, standard imaging protocols, consistent
oncology, large dataset with both radiological and pathologi- image processing together with appropriate harmonization
cal data are often not easy to obtain as data acquisition is method are warranted in the future study. Figure 6 shows
costly and pooling data from multiple institutions is often challenges for clinical implementation of ML glioma
restricted due to privacy protection policies. Alternatively, 1p/19q status prediction models.
data augmentation is proposed to alleviate data insufficiency In summary, the current review corporates two categories
and improve the robustness of predictions by adding trans- of models: traditional ML and DL models. As for traditional
formed copies of already existing data. There are seven studies ML, it cannot work with raw image data directly and must rely
utilized data augmentation, generating equivalent data based on expert design techniques to extract and construct informative
on limited real data according to various transforms (flipping, features (namely “feature engineering”),64 the process is strongly
rotating, cropping and scaling). Transfer learning is another related to the data acquisition. The protocol and scanners
way to mitigate small sample size problem. Two stages of change between different institutions, which may be limiting
model training are involved in transfer learning. The model factors in the development of a robust ML model. As regard to
was pretrained on a large-scale benchmark dataset and then DL models, image features can be “learned” implicitly through
fine-tuned on a small but study-specific dataset. Yogananda the iterative process of optimizing prediction performance, it
et al56 applied transfer learning in 1p/19q status prediction, takes images as input eliminating the steps of feature engineering
using the network “T2-net” previously trained on dataset and selection, hence potentially mitigating human bias.65 How-
regarding IDH classification. The model achieves good per- ever, DL is not without drawbacks, it depends on tremendous
formance (AUC = 0.95) with substantially less training data. volume of high quality, well-annotated data to train a reliable
Generalizability of the AI models is one of the major model,66 such data are difficult to aggregate in large part because
challenges hindering their board clinical applicability. Devel- of regulatory and privacy concerns across institutions. Thus,
oping an AI model on one dataset and evaluating it on data-sharing and federated learning may be future directions for
dataset from other institutions (external validation) is the best overcoming current barriers for supporting clinical implementa-
strategy to assess the performance, robustness, and depend- tion of DL models.
ability of AI models. Unfortunately, this approach is not
prevalent in most of medical AI studies. So far, only about
6% of published medical AI studies used external valida- Conclusions
tion.62 In our review of 1p/19q prediction, only 11 studies Current evidence shows promising results for various AI
validated their models on external datasets. Furthermore, methodologies for 1P/19q status classification, which could
given that studies based on highly curated datasets (including be beneficial for personalized treatment. However, till now,
BraTS or TCIA) have shown consistently high classification there is no agreement about the radiomics pipeline, and the
accuracy, performances of models trained on these datasets prior studies are very heterogeneous regarding the segmenta-
may not be reproducible in real-world scenario. Future works tion methods, the number and category of extracted features,
should validate their model externally using large, less-curated MRI sequences, and AI technique. In addition, only 11 stud-
datasets to evaluate the model’s generalizability on real-world ies have implemented their model on external datasets. All of
data and the potential for broad clinical deployment. the studies were retrospective in design. Before the clinical

1350 Volume 58, No. 5


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Zhang et al.: A Review of AI Applications in Glioma With 1p/19q Co-Deletion

implementation of glioma 1p/19q prediction by AI, more 19. Rathore S, Bakas S, Akbari H, et al. Machine learning radiomic bio-
markers non-invasively assess genetic characteristics of glioma patients.
standardized research is needed. Cancer Res 2019;79(13):1392.

20. Zinn PO, Singh SK, Kotrotsou A, et al. Distinct radiomic phenotypes
Conflicts of Interest define glioblastoma TP53-PTEN-EGFR mutational landscape. Neuro-
surgery 2017;64:203-210.
The authors declare no potential conflicts of interest.
21. Huang H, Zhang W, Fang Y, Hong J, Su S, Lai X. Overall survival pre-
diction for gliomas using a novel compound approach. Front Oncol
References 2021;11:724191.

1. Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Orga- 22. Mongan J, Moy L, Kahn CE. Checklist for artificial intelligence in medi-
nization classification of tumors of the central nervous system: A sum- cal imaging (CLAIM): A guide for authors and reviewers. Radiol Artif
mary. Acta Neuropathol 2016;131(6):803-820. Intell 2020;2(2):e200029.

2. Louis DN, Perry A, Wesseling P, et al. The 2021 WHO classification of 23. Whiting PF, Rutjes AW, Westwood ME, et al. QUADAS-2: A revised
tumors of the central nervous system: A summary. Neuro Oncol 2021; tool for the quality assessment of diagnostic accuracy studies. Ann
23(8):1231-1251. Intern Med 2011;155(8):529-536.

3. Kaloshi G, Benouaich-Amiel A, Diakite F, et al. Temozolomide for low- 24. Fan Z, Sun Z, Fang S, et al. Preoperative radiomics analysis of 1p/19q
grade gliomas: Predictive impact of 1p/19q loss on response and out- status in WHO grade II gliomas. Front Oncol 2021;11:616740.
come. Neurology 2007;68(21):1831-1836.
25. Tang Z, Xu Y, Jin L, et al. Deep learning of imaging phenotype and
4. Cagney DN, Sul J, Huang RY, Ligon KL, Wen PY, Alexander BM. The genotype for predicting overall survival time of glioblastoma patients.
FDA NIH biomarkers, EndpointS, and other tools (BEST) resource in IEEE Trans Med Imaging 2020;39(6):2100-2109.
neuro-oncology. Neuro Oncol 2018;20(9):1162-1172.
26. Zhou H, Vallières M, Bai HX, et al. MRI features predict survival and
5. Wijnenga MMJ, French PJ, Dubbink HJ, et al. The impact of surgery in molecular markers in diffuse lower-grade gliomas. Neuro Oncol 2017;
molecularly defined low-grade glioma: An integrated clinical, radiologi- 19(6):862-870.
cal, and molecular analysis. Neuro Oncol 2018;20(1):103-112.
27. Han Y, Xie Z, Zang Y, et al. Non-invasive genotype prediction of chro-
6. Kawaguchi T, Sonoda Y, Shibahara I, et al. Impact of gross total re- mosome 1p/19q co-deletion by development and validation of an
section in patients with WHO grade III glioma harboring the IDH 1/2 MRI-based radiomics signature in lower-grade gliomas. J Neurooncol
mutation without the 1p/19q co-deletion. J Neurooncol 2016;129(3): 2018;140(2):297-306.
505-514.
28. Lu CF, Hsu FT, Hsieh KL, et al. Machine learning-based radiomics for
7. Ding X, Wang Z, Chen D, et al. The prognostic value of maximal surgi- molecular subtyping of gliomas. Clin Cancer Res 2018;24(18):4429-
cal resection is attenuated in oligodendroglioma subgroups of adult 4436.
diffuse glioma: A multicenter retrospective study. J Neurooncol 2018;
29. Shofty B, Artzi M, Ben Bashat D, et al. MRI radiomics analysis of molec-
140(3):591-603.
ular alterations in low-grade gliomas. Int J Comput Assist Radiol Surg
8. Clark VE, Cahill DP. Extent of resection versus molecular classification: 2018;13(4):563-571.
What matters when? Neurosurg Clin N Am 2019;30(1):95-101. 30. Kim D, Wang N, Ravikumar V, et al. Prediction of 1p/19q codeletion in
9. Scheie D, Andresen PA, Cvancarova M, et al. Fluorescence in situ diffuse glioma patients using pre-operative multiparametric magnetic
hybridization (FISH) on touch preparations: A reliable method for resonance imaging. Front Comput Neurosci 2019;13:52.
detecting loss of heterozygosity at 1p and 19q in oligodendroglial 31. van der Voort SR, Incekara F, Wijnenga MMJ, et al. Predicting the
tumors. Am J Surg Pathol 2006;30(7):828-837. 1p/19q codeletion status of presumed low-grade glioma with an exter-
10. Chen CC, Hsu PW, Erich Wu TW, et al. Stereotactic brain biopsy: Sin- nally validated machine learning algorithm. Clin Cancer Res 2019;
gle center retrospective analysis of complications. Clin Neurol Neuro- 25(24):7455-7462.
surg 2009;111(10):835-839. 32. Zhou H, Chang K, Bai HX, et al. Machine learning reveals multimodal
11. Sanai N, Martino J, Berger MS. Morbidity profile following aggressive MRI patterns predictive of isocitrate dehydrogenase and 1p/19q status
resection of parietal lobe gliomas. J Neurosurg 2012;116(6):1182-1186. in diffuse low- and high-grade gliomas. J Neurooncol 2019;142(2):
299-307.
12. Belden CJ, Valdes PA, Ran C, et al. Genetics of glioblastoma: A win-
dow into its imaging and histopathologic variability. Radiographics 33. Kong Z, Jiang C, Zhang Y, et al. Thin-slice magnetic rsonance imaging-
2011;31(6):1717-1740. based radiomics signature predicts chromosomal 1p/19q co-deletion
status in grade II and III gliomas. Front Neurol 2020;11:551771.
13. Megyesi JF, Kachur E, Lee DH, et al. Imaging correlates of molecular
signatures in oligodendrogliomas. Clin Cancer Res 2004;10(13):4303- 34. Kocak B, Durmaz ES, Ates E, et al. Radiogenomics of lower-grade glio-
4306. mas: Machine learning-based MRI texture analysis for predicting
1p/19q codeletion status. Eur Radiol 2020;30(2):877-886.
14. Patel SH, Poisson LM, Brat DJ, et al. T2–FLAIR mismatch, an imaging
biomarker for IDH and 1p/19q status in lower-grade gliomas: A 35. Rathore S, Chaddad A, Bukhari NH, Niazi T. Imaging signature of
TCGA/TCIA project. Clin Cancer Res 2017;23(20):6078-6085. 1p/19q Co-deletion status derived via machine learning in lower grade
glioma. Radiomics and radiogenomics in neuro-oncology. Cham:
15. Broen MPG, Smits M, Wijnenga MMJ, et al. The T2-FLAIR mismatch Springer International Publishing; 2020. p 61-69.
sign as an imaging marker for non-enhancing IDH-mutant, 1p/19q-
intact lower-grade glioma: A validation study. Neuro Oncol 2018; 36. Casale R, Lavrova E, Sanduleanu S, Woodruff HC, Lambin P. Develop-
20(10):1393-1399. ment and external validation of a non-invasive molecular status predic-
tor of chromosome 1p/19q co-deletion based on MRI radiomics
16. Alzubaidi L, Zhang J, Humaidi AJ, et al. Review of deep learning: Con- analysis of low grade glioma patients. Eur J Radiol 2021;139:109678.
cepts, CNN architectures, challenges, applications, future directions.
J Big Data 2021;8(1):53. 37. Kha QH, Le VH, Hung TNK, Le NQK. Development and validation of
an efficient MRI radiomics signature for improving the predictive per-
17. Sharma N, Sharma R, Jindal N. Machine learning and deep learning formance of 1p/19q Co-deletion in lower-grade gliomas. Cancers
applications-a vision. Glob Transit Proc 2021;2(1):24-28. (Basel) 2021;13(21):5398.

18. Isensee F, Jaeger PF, Kohl SAA, Petersen J, Maier-Hein KH. nnU-net: A 38. Sun C, Fan L, Wang W, et al. Radiomics and qualitative features from
self-configuring method for deep learning-based biomedical image multiparametric MRI predict molecular subtypes in patients with lower-
segmentation. Nat Methods 2021;18(2):203-211. grade glioma. Front Oncol 2021;11:756828.

November 2023 1351


15222586, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jmri.28737 by Manager Information Resources Bond University Library, Wiley Online Library on [14/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Journal of Magnetic Resonance Imaging

39. Yan J, Zhang B, Zhang S, et al. Quantitative MRI-based radiomics for signature which is non-invasively predictable with rCBV imaging in
noninvasively predicting molecular subtypes and survival in glioma human glioma. Sci Rep 2015;5:16238.
patients. NPJ Precis Oncol 2021;5(1):72.
53. Bakas S, Zeng K, Sotiras A, et al. GLISTRboost: Combining multimodal
40. Lam LHT, Do DT, Diep DTN, et al. Molecular subtypes classification of MRI segmentation, registration, and biophysical tumor growth model-
low-grade gliomas patients using MRI-based radiomics and machine ing with gradient boosting Machines for Glioma Segmentation. Brain
learning. NMR Biomed 2022;35(11):e4792. 2016;9556:144-155.

41. Li Y, Ammari S, Lawrance L, et al. Radiomics-based method for 54. Chang PD. Fully convolutional deep residual neural networks for brain
predicting the glioma subtype as defined by tumor grade, IDH muta- tumor segmentation. Brainlesion: Glioma, multiple sclerosis, stroke and
tion, and 1p/19q codeletion. Cancers (Basel) 2022;14(7):1778. traumatic brain injuries. Cham: Springer International Publishing; 2016.
p 108-118.
42. Akkus Z, Ali I, Sedl
ař J, et al. Predicting deletion of chromosomal arms
1p/19q in low-grade gliomas from MR images using machine intelli- 55. Wang G, Li W, Ourselin S, Vercauteren T. Automatic brain tumor seg-
gence. J Digit Imaging 2017;30(4):469-476. mentation based on cascaded convolutional neural networks with
uncertainty estimation. Front Comput Neurosci 2019;13:56.
43. Chang P, Grinband J, Weinberg BD, et al. Deep-learning convolutional
neural networks accurately classify genetic mutations in gliomas. AJNR 56. Yogananda CGB, Shah BR, Yu FF, et al. A novel fully automated MRI-
Am J Neuroradiol 2018;39(7):1201-1207. based deep-learning method for classification of 1p/19q co-deletion
status in brain gliomas. Neurooncol Adv 2020;2(1):vdaa066.
44. Choi KS, Choi SH, Jeong B. Prediction of IDH genotype in gliomas with
dynamic susceptibility contrast perfusion MR imaging using an explain- 57. Wang S, Summers RM. Machine learning and radiology. Med Image
able recurrent neural network. Neuro Oncol 2019;21(9):1197-1209. Anal 2012;16(5):933-951.

45. Ali MB, Gu IY, Berger MS, et al. Domain mapping and deep learning 58. Hutter F, Lücke J, Schmidt-Thieme L. Beyond manual tuning of hyper-
from multiple MRI clinical datasets for prediction of molecular subtypes parameters. Kunstl Intel 2015;29(4):329-337.
in low grade gliomas. Brain Sci 2020;10(7):463. 59. Zhang S, Sun H, Su X, et al. Automated machine learning to predict
46. Decuyper M, Bonte S, Deblaere K, Van Holen R. Automated MRI based the co-occurrence of isocitrate dehydrogenase mutations and O(6)-
pipeline for segmentation and prediction of grade, IDH mutation and methylguanine-DNA methyltransferase promoter methylation in
1p19q co-deletion in glioma. Comput Med Imaging Graph 2021;88: patients with gliomas. J Magn Reson Imaging 2021;54(1):197-205.
101831. 60. Su X, Chen N, Sun H, et al. Automated machine learning based on rad-
47. Tupe-Waghmare P, Malpure P, Kotecha K, et al. Comprehensive geno- iomics features predicts H3 K27M mutation in midline gliomas of the
mic subtyping of glioma using semi-supervised multi-task deep learn- brain. Neuro Oncol 2020;22(3):393-401.
ing on multimodal MRI. IEEE Access 2021;9:167900-167910. 61. Marcus GF. Deep Learning: A critical appraisal. 2018.
48. Ning Z, Tu C, Di X, Feng Q, Zhang Y. Deep cross-view co-regularized 62. Kim DW, Jang HY, Kim KW, Shin Y, Park SH. Design characteristics of
representation learning for glioma subtype identification. Med Image studies reporting the performance of artificial intelligence algorithms
Anal 2021;73:102160. for diagnostic analysis of medical images: Results from recently publi-
shed papers. Korean J Radiol 2019;20(3):405-410.
49. van der Voort SR, Incekara F, Wijnenga MMJ, et al. Combined molecu-
lar subtyping, grading, and segmentation of glioma using multi-task 63. Papadimitroulas P, Brocki L, Christopher Chung N, et al. Artificial intelli-
deep learning. Neuro Oncol 2023;25(2):279-289. gence: Deep learning in oncological radiomics and challenges of inter-
pretability and data harmonization. Phys Med 2021;83:108-121.
50. Cluceru J, Interian Y, Phillips JJ, et al. Improving the noninvasive classi-
fication of glioma genetic subtype with deep learning and diffusion- 64. Chen L, Xia C, Sun H. Recent advances of deep learning in psychiatric
weighted imaging. Neuro Oncol 2022;24(4):639-652. disorders. Prec Clin Med 2020;3(3):202-213.
51. Yan J, Zhang S, Sun Q, et al. Predicting 1p/19q co-deletion status from 65. Krauze AV, Zhuge Y, Zhao R, Tasci E, Camphausen K. AI-driven image
magnetic resonance imaging using deep learning in adult-type diffuse analysis in central nervous system tumors-traditional machine learning,
lower-grade gliomas: A discovery and validation study. Lab Invest deep learning and hybrid models. J Biotechnol Biomed 2022;5(1):1-19.
2022;102(2):154-159.
66. Shaver MM, Kohanteb PA, Chiou C, et al. Optimizing neuro-oncology
52. Kickingereder P, Sahm F, Radbruch A, et al. IDH mutation status is imaging: A review of deep learning approaches for glioma imaging.
associated with a distinct hypoxia/angiogenesis transcriptome Cancers 2019;11(6):829.

1352 Volume 58, No. 5

You might also like