Next Article in Journal
Comparative Analysis of Laparoscopic and Robotic Transperitoneal Adrenalectomy Performed at a Single Institution
Previous Article in Journal
Work-Related Musculoskeletal Disorders among Dentists in the United Arab Emirates: A Cross-Sectional Study
Previous Article in Special Issue
Multi-Modal Brain Tumor Detection Using Deep Neural Network and Multiclass SVM
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Evidence, Limitations and Future Challenges of Survival Prediction for Glioblastoma Based on Advanced Noninvasive Methods: A Narrative Review

by
Sergio García-García
1,*,
Manuel García-Galindo
2,
Ignacio Arrese
1,
Rosario Sarabia
1 and
Santiago Cepeda
1
1
Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
2
Faculty of Medicine, University of Valladolid, Avenida Ramón y Cajal 7, 47003 Valladolid, Spain
*
Author to whom correspondence should be addressed.
Submission received: 17 October 2022 / Revised: 16 November 2022 / Accepted: 28 November 2022 / Published: 29 November 2022

Abstract

:
Background and Objectives: Survival estimation for patients diagnosed with Glioblastoma (GBM) is an important information to consider in patient management and communication. Despite some known risk factors, survival estimation remains a major challenge. Novel non-invasive technologies such as radiomics and artificial intelligence (AI) have been implemented to increase the accuracy of these predictions. In this article, we reviewed and discussed the most significant available research on survival estimation for GBM through advanced non-invasive methods. Materials and Methods: PubMed database was queried for articles reporting on survival prognosis for GBM through advanced image and data management methods. Articles including in their title or abstract the following terms were initially screened: ((glioma) AND (survival)) AND ((artificial intelligence) OR (radiomics)). Exclusively English full-text articles, reporting on humans, published as of 1 September 2022 were considered. Articles not reporting on overall survival, evaluating the effects of new therapies or including other tumors were excluded. Research with a radiomics-based methodology were evaluated using the radiomics quality score (RQS). Results: 382 articles were identified. After applying the inclusion criteria, 46 articles remained for further analysis. These articles were thoroughly assessed, summarized and discussed. The results of the RQS revealed some of the limitations of current radiomics investigation on this field. Limitations of analyzed studies included data availability, patient selection and heterogeneity of methodologies. Future challenges on this field are increasing data availability, improving the general understanding of how AI handles data and establishing solid correlations between image features and tumor’s biology. Conclusions: Radiomics and AI methods of data processing offer a new paradigm of possibilities to tackle the question of survival prognosis in GBM.

1. Introduction

Glioblastoma (GBM), with an incidence of 3.5/100.000 population, is the most common malignant primary neoplasm of the brain in adults [1]. Its fatal prognosis has scarcely improved over the last decades despite intensive research in the field [1]. Some clinical, surgical and radiological features are known independent predictors of survival [2]. However, accurate survival prediction is a key challenge for patients, relatives and physicians in their search for precision medicine strategies to tackle the burden of this devastating tumor [3].
A radiology-based approach to prognosis prediction has gained momentum in recent years fostered by the development of advanced tools to manage an immense quantity of data and thanks to its noninvasiveness, radiomics, a science based on quantitative data mining from medical images, has been extensively used in oncology [4]. Texture analysis, a branch of radiomics, exploits the information concealed in voxels and pixels and provides a quantitative assessment of images that might serve as a virtual biopsy [5]. Thus, different imaging modalities, segmentation algorithms and texture features have successfully contributed to supporting tumor diagnosis, molecular profile estimation, treatment response evaluation and overall survival (OS) prediction in neurooncology [6,7,8].
Handling such an enormous quantity of complex data as derived from radiomic analysis requires specific methods to obtain useful results and interpretable information. Thus, artificial intelligence (AI) methods have been developed and applied to this novel prognostic approach [9]. Machine learning (ML), a discipline within AI, through training datasets on ground truth labels has allowed us to obtain algorithms that can execute complex tasks such as tumor segmentation, tumor grading, molecular classification and survival prognosis [10]. ML may follow a supervised (e.g., logistic regression, support vector machine (SVM), random forest (RF), naïve Bayesian networks, decision trees (DT)) or unsupervised (e.g., K-means cluster) workflow. In a different way, deep learning (DL), a subclass of ML, does not require human intervention or ground truth labels to learn. Instead, DL, mainly present in different forms of neural networks (NN), has been successfully applied to tasks such as survival estimation, tumor segmentation, and estimation of glioma molecular subtypes [6,11,12,13,14,15]. NN learns on its own from previous fails and successful associations and can make more complex correlations. However, DL needs higher volumes of data and requires extensive computational time for training. In all, different AI strategies might be used to perform similar functions, which in the case of survival prognosis often consists of correctly classifying patients into long and short survivors, whether by means of SVM, RF, DT or naïve Bayesian classifiers, for instance, in the case of ML; or NN in the case of DL.
In this review, we present an update of the current evidence on advanced statistics, radiomics and data processing methods for the accurate survival prognosis of patients suffering from high-grade gliomas. The initial attempt to conduct a systematic review or meta-analysis was soon quit given the enormous heterogeneity of methods, the frequent involvement of the same set of patients from public datasets and the lack of consistency in results reporting. Instead, we sought to provide a comprehensive and thorough review of advanced noninvasive methods for survival estimation in GBM, while we discuss present challenges and future perspectives in this field.

2. Materials and Methods

A systematic search of PubMed was performed for articles reporting on the survival prognosis for GBM, involving in their methodology a noninvasive advanced radiological approach including radiomics and/or AI.
The query was built for the following terms to be present on the Title or abstract: ((glioma) AND (survival)) AND ((artificial intelligence) OR (radiomics)). Only English full-text articles, reporting on human subjects, published (even ahead of print) as of 1 September 2022 were considered. Each manuscript was independently reviewed by two authors (S. G-G and S.C.). Articles not reporting on overall survival, including other tumors than GBM, or redundant articles were excluded. Articles assessing the effect of novel therapies on survival were also excluded. In the case of an investigation conducted by the same scientific team with similar methods applied to an updated, previously published dataset, the last manuscript was considered. In case of disagreement on the need to include a given study, a consensus was reached after substantiated discussion held by the signing authors.
Studies involving a methodology based on radiomics were assessed according to the radiomics quality score (RQS) [16]. The RQS is a sixteen-item scale that evaluates the methodology and reporting of an investigation to improve the consistency of radiomics studies, increase its reproducibility and enhance scientific soundness in this field [16].

3. Results

The search resulted in 382 articles with the terms glioma, survival, AI and radiomics, whether in their title or abstract. After applying the inclusion criteria, 311 articles were excluded. Seventy-one full-text articles were thoroughly reviewed, of which 46 articles were ultimately included (Figure 1). The main results of these articles are summarized in Table 1. Similarly, the results of the RQS of articles, whose methodology was based on radiomics, are presented in Table 2. The average and median scores of the RQS for the analysed studies were 10.5 (29%) and 11 (31%), respectively.

4. Discussion

Predicting life expectancy for patients diagnosed with GBM is a major challenge. Many variables might influence the prognosis, and despite numerous efforts and approaches, uncertainty often erodes patients’ and physicians’ will to face and overcome the fateful outlook that this disease entails. New advanced techniques to process images and data have proven their ability to noninvasively mine and display the information shielded in the infinite number of pixels and voxels that compose multimodal neuroimages. Initially, we conducted a meta-analysis or systematic review of the main evidence on survival prognosis through these advanced methods, such as radiomics and AI. However, we soon realized that the tremendous variability of methods, the lack of consistency in reporting the results and the recurrent use of public datasets of the same patients hampered successful fulfillment of this task and rendered it unmanageable. Instead, we present a thorough review of the key available evidence regarding this critical issue while we discuss the main existing limitations, the actual room for improvement and the new horizons and challenges that future research should address.
Conventional morphological MRI features contain valuable prognostic information. As proven by Molina et al., a simple model based on age and morphological features, without texture data, could outperform more complex models in GBM [32] prognosis prediction. Nonetheless, morphologic information might be a loose term in which many features fit. In an effort to unify the common characteristics gliomas display on MRI studies and to harmonize the vocabulary in which we refer to them, Visually Accessible Rembrandt Images (VASARI) were developed [57]. This list of 30 features has been demonstrated to be useful for predicting OS in GBM [58]. In addition, VASARI, which does not consider texture information, has been successfully integrated into ML models based on texture data [56]. However, in a study by Ruan et al., only cortical involvement was strongly associated with poor outcomes. Indeed, VASARI features did not increase the predictive accuracy of the RF model based on radiomic features when combined with them.
As shown in Table 1, there are several examples of predictive algorithms based on texture features extracted from conventional MRI sequences. Studies vary widely in terms of image preprocessing, segmentation methods, number of features and ML classifiers (Figure 2). Different strategies and approaches have been performed with a few outstanding examples, such as those of Ruan et al.; Cepeda et al.; Lu et al.; and Sanghani et al., in whose studies based on conventional MRI sequences and ML algorithms, accurate prognostic classification exceeded 90% [29,52,55,56]. Moreover, advanced MRI sequences can also be employed for prognostic purposes. Thus, some authors have implemented functional resting-state MRI and DTI (Diffusion Tensor Imaging) studies to build structural and connectivity networks, extract features and process them with ML and DL algorithms [19,50,59].
An interesting conceptual approach was implemented by Lee et al., who used relative texture features from perfusion maps to predict the survival status at 12 months [22]. Features extracted from enhancing and nonenhancing regions of the tumor were computed to provide these relative features. In addition, kinetic features calculated from the gadolinium concentration time-series of perfusion data in both regions were also extracted [22]. Nonetheless, relative texture features displayed a higher impact on prognosis than kinetic features [22].
Delta-radiomics, which consists of features extracted at different time points, provides information about how radiomic features change over time. In a small cohort, Chang et al. demonstrated the strong potential of delta-radiomics combined with a DL algorithm [39]. It is still unknown which time lapses might be more informative depending on the studied outcome variable. Regardless, delta features seem to provide higher predictive information than one-time features, which makes it even more logical for survival prognosis.
ML methods may also generate different radiomic profiles of gliomas that could potentially translate their underlying biological features. Itakura et al. and Rathore et al. classified gliomas into three clusters with different survival prognoses using different methodologies [60,61]. Remarkably, both teams reported three different radiomic profiles that actually shared some characteristics [9]. Specifically, rim-enhancing subtypes granted better prognosis in both reports. Indeed, tumor subgroups were associated with molecular subtypes, location and genetic mutations [60,61]. These findings, which require further validation, support the idea of radiomics as a virtual biopsy, turning the corner to more personalized and precise communication with patients and relatives.
Deep learning, as a branch of AI and considered a fully machine learning method able to train on its own without human intervention, has consistently defeated rivals in image recognition competitions such as ImageNet. The increasing amount, complexity, types and availability of data partially explains the swift direction towards DL for medical applications, as DL does not strictly require a human-driven refining of data.
Moradman et al. demonstrated the superior ability of DL to establish complex correlations between multiple clinical, biological and therapeutic variables, and survival in patients diagnosed with glioblastoma [11]. A feed-forward NN offered higher accuracy on survival prognosis than the random survival forest and Cox proportional hazard regression models [11].
Obviating the tasks that texture analysis involves, Ben Ahmed et al. built a convolutional NN based on MRI snapshots that outperformed the accuracy of the best-known predictor by 6% [14]. Using nonlabelled, nonsegmented snapshots offers a fast and low-cost way to feed a DL algorithm [14]. Nonetheless, DL might also be used as a method to mine images. With a hybrid approach, Nie et al. applied a DL method to automatically extract features that would otherwise be difficult to design [34]. These DL features together with key demographic and tumor-related features allowed us to stratify patients into long and short survival groups through an SVM classifier with 90.5% accuracy [34]. DL also supports the inclusion of advanced MRI sequences, as demonstrated by Yan et al., who compared a clinical nomogram with a DL signature based on DTI data [62]. Yan et al. obtained better results with the DL signature, achieving a C-index of 0.9 on an external validation dataset [62]. In addition, the authors suggested the existence of an association of DL features with biological pathways involved in glioma development (synaptic transmission, activation of AMPA receptors, axon guidance, calcium transport, etc.) [62].

4.1. Future Challenges

4.1.1. Data Availability

AI training requires a large, high-quality dataset to build robust algorithms. Creating such datasets is costly and time consuming and demands that professionals shift from care provision to data production. This burden is especially problematic in rare diseases such as GBM. Therefore, a culture of data sharing is needed. Cooperative efforts, such as The Cancer Imaging Archive or the Ivy Glioblastoma Atlas project, have contributed to increasing data availability. Additionally, data could be improved by the harmonization of image acquisition protocols across institutions. Automatic data acquisition, often used by AI in other fields, clashes with the need to preserve the confidentiality of medical data [63].

4.1.2. Opening the Black Box

AI algorithms are built from associations that are not fully disclosed by the algorithm itself. Therefore, drawing conclusions between radiomic features and glioma characteristics might be misleading since their relationships are unknown, and predictive models might be based on variables derived from similar features that might be overrepresented [64]. Indeed, understanding the underlying mechanisms by which biology translates into radiomic features is a classic concern and matter of current investigations. Nonetheless, recent advances, such as principal component analysis and saliency maps, have relieved these concerns by unveiling part of the structure of AI algorithms [65]. Radiomic features might be the fine manifestation of molecular phenotypes in grayscale images [20].

4.1.3. Humanizing AI

When AI is implemented to fulfil a given task, human vs. machine approaches are often used to elucidate who can better perform it. Modern AI applications to the medical field have suggested the benefits of human-in-the-loop strategies to overcome the unique challenges medicine poses to AI. In expert augmented machine learning, researchers combined the knowledge of experts to solve specific problems where AI algorithms might fail the most [66]. Thus, the quality of training data might be notably improved by integrating the information that specialists base their decisions on. Similarly, in active learning, key data are obtained from the expert by the algorithm itself to increase the quality of the training dataset or enhance the ability of the algorithm to extract useful information [67,68].

4.1.4. Integrating AI into Clinical Practice

The ultimate goal of research in medicine is transferring the lessons learned in the laboratory to clinical practice. The topic covered in this review is not an exception. The reports presented herein are commendable efforts to find key features and methods to improve patients’ prognostic estimation. However, the methodology that most of these investigations involve is extremely time-consuming and makes it inefficient for daily implementation in a clinical context. The next paramount advancement in this field, beyond increasing accuracy or simplifying the workflow, will be the production of an open source, easily integrable and precise AI algorithm that requires simple or null intervention of the physician for prognostic estimation from multimodal MRI studies.

4.2. Limitations

The major limitations of previous publications can be summarized in the following sections:
  • Patient selection: In most published articles, patients were included without considering the extent of resection, which is one of the main factors associated with overall survival. Therefore, if the intention is to use the imaging characteristics independently to predict the outcome, it is necessary either to include only patients with gross total resection or perhaps to introduce in the model a variable through which the degree of resectability of the tumor can be quantified [55].
  • Image preprocessing and data extraction: There is significant variability in the methods employed to preprocess MRI images and in the parameters used to extract radiomic features. This pitfall explains the differences in the results obtained on the same patient dataset (such as the TCIA patient cohort) [47,49]. Therefore, the lack of details about the preprocessing pipeline used by the different authors limits the reproducibility of their results [11,35,44,54].
  • Classification task vs. survival regression: There are discrepancies in how different authors approach the challenge of predicting survival in GBM. On the one hand, some studies attempt to carry out a survival analysis, in which the relationship between the radiomic variables and survival in days is expressed by the Harrell index or the hazard ratio [6,18,35,37]. On the other hand, there are works in which a classification task has been carried out to create survival groups. The latter methodology is much easier to interpret and has a clinical orientation [6,19,23]. However, the cut-off point for establishing survival groups is entirely arbitrary in various publications [19]. For example, it does not seem helpful to define a short-term survivor as one who does not exceed ten months of life when the overall median survival is 15 months. Therefore, unifying the criteria for short- and long-term survival definitions in this neoplasm is essential.
  • Lack of validation in multi-institutional data: Although there are studies with promising results, the lack of validation in a multicenter cohort seriously limits the application of predictions in a clinical setting [55]. One of the challenges of models based on radiomic features is to find a set of stable and reproducible features so that they can be used regardless of artifacts produced during image acquisition, MRI acquisition protocols, and scanner manufacturers.

5. Conclusions

Advanced image analysis and data processing methods have gained momentum over the last decade. Methods such as radiomics, texture analysis, ML and DL have been successfully implemented to provide an accurate survival estimation and risk factor identification for patients diagnosed with GBM. The wide variety of available approaches prevents unifying methods and drawing consistent conclusions from reported results. However, despite its limitations, the existing symbiosis between radiomics and AI represents a robust approach to build evidence and address unanswered questions in neuro-oncology. In fact, AI is no longer a matter of future but a living, vibrant and powerful reality.

Author Contributions

Conceptualization, S.G.-G. and S.C.; methodology, S.G.-G. and S.C.; formal analysis, S.G.-G. and M.G.-G.; investigation, S.G.-G.; resources, R.S.; data curation, S.G.-G. and M.G.-G.; writing—original draft preparation, S.G.-G. and S.C.; writing—review and editing, I.A. and R.S.; supervision, S.C. and R.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ostrom, Q.T.; Cioffi, G.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2014–2018. Neuro-Oncology 2021, 23, iii1–iii105. [Google Scholar] [CrossRef]
  2. McKinnon, C.; Nandhabalan, M.; A Murray, S.; Plaha, P. Glioblastoma: Clinical presentation, diagnosis, and management. BMJ 2021, 374, n1560. [Google Scholar] [CrossRef] [PubMed]
  3. Prados, M.D.; Byron, S.; Tran, N.L.; Phillips, J.J.; Molinaro, A.M.; Ligon, K.L.; Wen, P.Y.; Kuhn, J.G.; Mellinghoff, I.K.; De Groot, J.F.; et al. Toward precision medicine in glioblastoma: The promise and the challenges. Neuro-Oncology 2015, 17, 1051–1063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kumar, V.; Gu, Y.; Basu, S.; Berglund, A.; Eschrich, S.A.; Schabath, M.B.; Forster, K.; Aerts, H.J.W.L.; Dekker, A.; Fenstermacher, D.; et al. Radiomics: The process and the challenges. Magn. Reson. Imaging 2012, 30, 1234–1248. [Google Scholar] [CrossRef] [Green Version]
  5. Litvin, A.; Burkin, D.; Kropinov, A.; Paramzin, F. Radiomics and Digital Image Texture Analysis in Oncology (Review). Sovrem. Teh. Med. 2021, 13, 97. [Google Scholar] [CrossRef] [PubMed]
  6. Han, W.; Qin, L.; Bay, C.; Chen, X.; Yu, K.-H.; Miskin, N.; Li, A.; Xu, X.; Young, G. Deep Transfer Learning and Radiomics Feature Prediction of Survival of Patients with High-Grade Gliomas. Am. J. Neuroradiol. 2019, 41, 40–48. [Google Scholar] [CrossRef] [Green Version]
  7. Huang, W.-Y.; Wen, L.-H.; Wu, G.; Hu, M.-Z.; Zhang, C.-C.; Chen, F.; Zhao, J.-N. Comparison of Radiomics Analyses Based on Different Magnetic Resonance Imaging Sequences in Grading and Molecular Genomic Typing of Glioma. J. Comput. Assist. Tomogr. 2021, 45, 110–120. [Google Scholar] [CrossRef]
  8. Yan, J.; Zhang, B.; Zhang, S.; Cheng, J.; Liu, X.; Wang, W.; Dong, Y.; Zhang, L.; Mo, X.; Chen, Q.; et al. Quantitative MRI-based radiomics for noninvasively predicting molecular subtypes and survival in glioma patients. NPJ Precis. Oncol. 2021, 5, 72. [Google Scholar] [CrossRef]
  9. Rudie, J.D.; Rauschecker, A.M.; Bryan, R.N.; Davatzikos, C.; Mohan, S. Emerging Applications of Artificial Intelligence in Neuro-Oncology. Radiology 2019, 290, 607–618. [Google Scholar] [CrossRef] [PubMed]
  10. Xu, J.; Meng, Y.; Qiu, K.; Topatana, W.; Li, S.; Wei, C.; Chen, T.; Chen, M.; Ding, Z.; Niu, G. Applications of Artificial Intelligence Based on Medical Imaging in Glioma: Current State and Future Challenges. Front. Oncol. 2022, 12, 892056. [Google Scholar] [CrossRef]
  11. Moradmand, H.; Aghamiri, S.M.R.; Ghaderi, R.; Emami, H. The role of deep learning-based survival model in improving survival prediction of patients with glioblastoma. Cancer Med. 2021, 10, 7048–7059. [Google Scholar] [CrossRef]
  12. van der Voort, S.R.; Incekara, F.; Wijnenga, M.M.J.; Kapsas, G.; Gahrmann, R.; Schouten, J.W.; Tewarie, R.N.; Lycklama, G.J.; Hamer, P.C.; Eijgelaar, R.S.; et al. Combined molecular subtyping, grading, and segmentation of glioma using multi-task deep learning. Neuro-Oncology 2022, noac166. [Google Scholar] [CrossRef] [PubMed]
  13. Yogananda, C.G.B.; Shah, B.R.; Vejdani-Jahromi, M.; Nalawade, S.S.; Murugesan, G.K.; Yu, F.F.; Pinho, M.C.; Wagner, B.C.; Mickey, B.; Patel, T.R.; et al. A novel fully automated MRI-based deep-learning method for classification of IDH mutation status in brain gliomas. Neuro-Oncology 2020, 22, 402–411. [Google Scholar] [CrossRef]
  14. Ben Ahmed, K.; Hall, L.O.; Goldgof, D.B.; Gatenby, R. Ensembles of Convolutional Neural Networks for Survival Time Estimation of High-Grade Glioma Patients from Multimodal MRI. Diagnostics 2022, 12, 345. [Google Scholar] [CrossRef] [PubMed]
  15. Lao, J.; Chen, Y.; Li, Z.-C.; Li, Q.; Zhang, J.; Liu, J.; Zhai, G. A Deep Learning-Based Radiomics Model for Prediction of Survival in Glioblastoma Multiforme. Sci. Rep. 2017, 7, 10353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Lambin, P.; Leijenaar, R.T.H.; Deist, T.M.; Peerlings, J.; de Jong, E.E.C.; van Timmeren, J.; Sanduleanu, S.; Larue, R.T.H.M.; Even, A.J.G.; Jochems, A.; et al. Radiomics: The bridge between medical imaging and personalized medicine. Nat. Rev. Clin. Oncol. 2017, 14, 749–762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Yang, D.; Rao, G.; Martinez, J.; Veeraraghavan, A.; Rao, A. 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, 6725–6735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Chaddad, A.; Tanougast, C. Extracted magnetic resonance texture features discriminate between phenotypes and are associated with overall survival in glioblastoma multiforme patients. Med. Biol. Eng. Comput. 2016, 54, 1707–1718. [Google Scholar] [CrossRef]
  19. Liu, L.; Zhang, H.; Rekik, I.; Chen, X.; Wang, Q.; Shen, D. Outcome Prediction for Patient with High-Grade Gliomas from Brain Functional and Structural Networks. In Proceedings of the Medical Image Computing and Computer-Assisted Intervention, Athens, Greece, 17–21 October 2016; Volume 9901, pp. 26–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Macyszyn, L.; Akbari, H.; Pisapia, J.M.; Da, X.; Attiah, M.; Pigrish, V.; Bi, Y.; Pal, S.; Davuluri, R.V.; Roccograndi, L.; et al. Imaging patterns predict patient survival and molecular subtype in glioblastoma via machine learning techniques. Neuro-Oncology 2015, 18, 417–425. [Google Scholar] [CrossRef] [PubMed]
  21. Kickingereder, P.; Götz, M.; Muschelli, J.; Wick, A.; Neuberger, U.; Shinohara, R.T.; Sill, M.; Nowosielski, M.; Schlemmer, H.-P.; Radbruch, A.; et al. Large-scale Radiomic Profiling of Recurrent Glioblastoma Identifies an Imaging Predictor for Stratifying Anti-Angiogenic Treatment Response. Clin. Cancer Res. 2016, 22, 5765–5771. [Google Scholar] [CrossRef] [Green Version]
  22. Lee, J.; Jain, R.; Khalil, K.; Griffith, B.; Bosca, R.; Rao, G.; Rao, A. Texture Feature Ratios from Relative CBV Maps of Perfusion MRI Are Associated with Patient Survival in Glioblastoma. Am. J. Neuroradiol. 2015, 37, 37–43. [Google Scholar] [CrossRef]
  23. Ingrisch, M.; Schneider, M.J.; Nörenberg, D.; de Figueiredo, G.N.; Maier-Hein, K.; Suchorska, B.; Schüller, U.; Albert, N.; Brückmann, H.; Reiser, M.; et al. Radiomic Analysis Reveals Prognostic Information in T1-Weighted Baseline Magnetic Resonance Imaging in Patients With Glioblastoma. Investig. Radiol. 2017, 52, 360–366. [Google Scholar] [CrossRef]
  24. Liu, Y.; Xu, X.; Yin, L.; Zhang, X.; Li, L.; Lu, H. Relationship between Glioblastoma Heterogeneity and Survival Time: An MR Imaging Texture Analysis. Am. J. Neuroradiol. 2017, 38, 1695–1701. [Google Scholar] [CrossRef] [Green Version]
  25. Li, Q.; Bai, H.; Chen, Y.; Sun, Q.; Liu, L.; Zhou, S.; Wang, G.; Liang, C.; Li, Z.-C. A Fully-Automatic Multiparametric Radiomics Model: Towards Reproducible and Prognostic Imaging Signature for Prediction of Overall Survival in Glioblastoma Multiforme. Sci. Rep. 2017, 7, 14331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Prasanna, P.; Patel, J.; Partovi, S.; Madabhushi, A.; Tiwari, P. Radiomic features from the peritumoral brain parenchyma on treatment-naïve multi-parametric MR imaging predict long versus short-term survival in glioblastoma multiforme: Preliminary findings. Eur. Radiol. 2016, 27, 4188–4197. [Google Scholar] [CrossRef]
  27. Kickingereder, P.; Neuberger, U.; Bonekamp, D.; Piechotta, P.L.; Goetz, M.; Wick, A.; Sill, M.; Kratz, A.; Shinohara, R.T.; Jones, D.T.W.; et al. Radiomic subtyping improves disease stratification beyond key molecular, clinical, and standard imaging characteristics in patients with glioblastoma. Neuro-Oncology 2017, 20, 848–857. [Google Scholar] [CrossRef]
  28. Bae, S.; Choi, Y.S.; Ahn, S.S.; Chang, J.H.; Kang, S.-G.; Kim, E.H.; Kim, S.H.; Lee, S.-K. Radiomic MRI Phenotyping of Glioblastoma: Improving Survival Prediction. Radiology 2018, 289, 797–806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Sanghani, P.; Ang, B.T.; King, N.K.K.; Ren, H. Overall survival prediction in glioblastoma multiforme patients from volumetric, shape and texture features using machine learning. Surg. Oncol. 2018, 27, 709–714. [Google Scholar] [CrossRef]
  30. Chaddad, A.; Sabri, S.; Niazi, T.; Abdulkarim, B. Prediction of survival with multi-scale radiomic analysis in glioblastoma patients. Med. Biol. Eng. Comput. 2018, 56, 2287–2300. [Google Scholar] [CrossRef]
  31. Liu, Y.; Zhang, X.; Feng, N.; Yin, L.; He, Y.; Xu, X.; Lu, H. The effect of glioblastoma heterogeneity on survival stratification: A multimodal MR imaging texture analysis. Acta Radiol. 2018, 59, 1239–1246. [Google Scholar] [CrossRef]
  32. Molina-García, D.; Vera-Ramírez, L.; Pérez-Beteta, J.; Arana, E.; Pérez-García, V.M. Prognostic models based on imaging findings in glioblastoma: Human versus Machine. Sci. Rep. 2019, 9, 5982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Tan, Y.; Mu, W.; Wang, X.-C.; Yang, G.-Q.; Gillies, R.J.; Zhang, H. Improving survival prediction of high-grade glioma via machine learning techniques based on MRI radiomic, genetic and clinical risk factors. Eur. J. Radiol. 2019, 120, 108609. [Google Scholar] [CrossRef]
  34. Nie, D.; Lu, J.; Zhang, H.; Adeli, E.; Wang, J.; Yu, Z.; Liu, L.; Wang, Q.; Wu, J.; Shen, D. Multi-Channel 3D Deep Feature Learning for Survival Time Prediction of Brain Tumor Patients Using Multi-Modal Neuroimages. Sci. Rep. 2019, 9, 1103. [Google Scholar] [CrossRef] [Green Version]
  35. Choi, Y.; Ahn, K.-J.; Nam, Y.; Jang, J.; Shin, N.-Y.; Choi, H.S.; Jung, S.-L.; Kim, B.-S. Analysis of heterogeneity of peritumoral T2 hyperintensity in patients with pretreatment glioblastoma: Prognostic value of MRI-based radiomics. Eur. J. Radiol. 2019, 120, 108642. [Google Scholar] [CrossRef] [PubMed]
  36. Chen, X.; Fang, M.; Dong, D.; Liu, L.; Xu, X.; Wei, X.; Jiang, X.; Qin, L.; Liu, Z. Development and Validation of a MRI-Based Radiomics Prognostic Classifier in Patients with Primary Glioblastoma Multiforme. Acad. Radiol. 2019, 26, 1292–1300. [Google Scholar] [CrossRef] [PubMed]
  37. Sasaki, T.; Kinoshita, M.; Fujita, K.; Fukai, J.; Hayashi, N.; Uematsu, Y.; Okita, Y.; Nonaka, M.; Moriuchi, S.; Uda, T.; et al. Radiomics and MGMT promoter methylation for prognostication of newly diagnosed glioblastoma. Sci. Rep. 2019, 9, 14435. [Google Scholar] [CrossRef] [Green Version]
  38. Um, H.; Tixier, F.; Bermudez, D.; Deasy, J.O.; Young, R.J.; Veeraraghavan, H. Impact of image preprocessing on the scanner dependence of multi-parametric MRI radiomic features and covariate shift in multi-institutional glioblastoma datasets. Phys. Med. Biol. 2019, 64, 165011. [Google Scholar] [CrossRef] [PubMed]
  39. Chang, Y.; Lafata, K.; Sun, W.; Wang, C.; Chang, Z.; Kirkpatrick, J.P.; Yin, F.-F. An investigation of machine learning methods in delta-radiomics feature analysis. PLoS ONE 2019, 14, e0226348. [Google Scholar] [CrossRef] [Green Version]
  40. Tixier, F.; Um, H.; Bermudez, D.; Iyer, A.; Apte, A.; Graham, M.S.; Nevel, K.S.; Deasy, J.O.; Young, R.J.; Veeraraghavan, H. Preoperative MRI-radiomics features improve prediction of survival in glioblastoma patients over MGMT methylation status alone. Oncotarget 2019, 10, 660–672. [Google Scholar] [CrossRef]
  41. Shboul, Z.; Alam, M.; Vidyaratne, L.; Pei, L.; Elbakary, M.I.; Iftekharuddin, K.M. Feature-Guided Deep Radiomics for Glioblastoma Patient Survival Prediction. Front. Neurosci. 2019, 13, 966. [Google Scholar] [CrossRef]
  42. Chaddad, A.; Daniel, P.; Sabri, S.; Desrosiers, C.; Abdulkarim, B. Integration of Radiomic and Multi-omic Analyses Predicts Survival of Newly Diagnosed IDH1 Wild-Type Glioblastoma. Cancers 2019, 11, 1148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kim, J.Y.; Yoon, M.J.; Park, J.E.; Choi, E.J.; Lee, J.; Kim, H.S. Radiomics in peritumoral non-enhancing regions: Fractional anisotropy and cerebral blood volume improve prediction of local progression and overall survival in patients with glioblastoma. Neuroradiology 2019, 61, 1261–1272. [Google Scholar] [CrossRef]
  44. Liao, X.; Cai, B.; Tian, B.; Luo, Y.; Song, W.; Li, Y. Machine-learning based radiogenomics analysis of MRI features and metagenes in glioblastoma multiforme patients with different survival time. J. Cell. Mol. Med. 2019, 23, 4375–4385. [Google Scholar] [CrossRef]
  45. Osman, A.F.I. A Multi-parametric MRI-Based Radiomics Signature and a Practical ML Model for Stratifying Glioblastoma Patients Based on Survival Toward Precision Oncology. Front. Comput. Neurosci. 2019, 13, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Chaddad, A.; Daniel, P.; Desrosiers, C.; Toews, M.; Abdulkarim, B. Novel Radiomic Features Based on Joint Intensity Matrices for Predicting Glioblastoma Patient Survival Time. IEEE J. Biomed. Health Inform. 2018, 23, 795–804. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, X.; Lu, H.; Tian, Q.; Feng, N.; Yin, L.; Xu, X.; Du, P.; Liu, Y. A radiomics nomogram based on multiparametric MRI might stratify glioblastoma patients according to survival. Eur. Radiol. 2019, 29, 5528–5538. [Google Scholar] [CrossRef] [PubMed]
  48. Zhang, X.; Lu, D.; Gao, P.; Tian, Q.; Lu, H.; Xu, X.; He, X.; Liu, Y. Survival-relevant high-risk subregion identification for glioblastoma patients: The MRI-based multiple instance learning approach. Eur. Radiol. 2020, 30, 5602–5610. [Google Scholar] [CrossRef]
  49. Suter, Y.; Knecht, U.; Alão, M.; Valenzuela, W.; Hewer, E.; Schucht, P.; Wiest, R.; Reyes, M. Radiomics for glioblastoma survival analysis in pre-operative MRI: Exploring feature robustness, class boundaries, and machine learning techniques. Cancer Imaging 2020, 20, 55. [Google Scholar] [CrossRef]
  50. Bakas, S.; Shukla, G.; Akbari, H.; Erus, G.; Sotiras, A.; Rathore, S.; Sako, C.; Ha, S.M.; Rozycki, M.; Singh, A.; et al. Integrative radiomic analysis for pre-surgical prognostic stratification of glioblastoma patients: From advanced to basic MRI protocols. SPIE Med. Imaging 2020, 11315, 113151S. [Google Scholar] [CrossRef] [PubMed]
  51. Park, J.E.; Kim, H.S.; Jo, Y.; Yoo, R.-E.; Choi, S.H.; Nam, S.J.; Kim, J.H. Radiomics prognostication model in glioblastoma using diffusion- and perfusion-weighted MRI. Sci. Rep. 2020, 10, 4250. [Google Scholar] [CrossRef] [Green Version]
  52. Lu, Y.; Patel, M.; Natarajan, K.; Ughratdar, I.; Sanghera, P.; Jena, R.; Watts, C.; Sawlani, V. Machine learning-based radiomic, clinical and semantic feature analysis for predicting overall survival and MGMT promoter methylation status in patients with glioblastoma. Magn. Reson. Imaging 2020, 74, 161–170. [Google Scholar] [CrossRef]
  53. Baid, U.; Rane, S.; Talbar, S.; Gupta, S.; Thakur, M.H.; Moiyadi, A.; Mahajan, A. Overall Survival Prediction in Glioblastoma With Radiomic Features Using Machine Learning. Front. Comput. Neurosci. 2020, 14, 61. [Google Scholar] [CrossRef]
  54. Priya, S.; Agarwal, A.; Ward, C.; Locke, T.; Monga, V.; Bathla, G. Survival prediction in glioblastoma on post-contrast magnetic resonance imaging using filtration based first-order texture analysis: Comparison of multiple machine learning models. Neuroradiol. J. 2021, 34, 355–362. [Google Scholar] [CrossRef]
  55. Cepeda, S.; Pérez-Nuñez, A.; García-García, S.; García-Pérez, D.; Arrese, I.; Jiménez-Roldán, L.; García-Galindo, M.; González, P.; Velasco-Casares, M.; Zamora, T.; et al. Predicting Short-Term Survival after Gross Total or Near Total Resection in Glioblastomas by Machine Learning-Based Radiomic Analysis of Preoperative MRI. Cancers 2021, 13, 5047. [Google Scholar] [CrossRef] [PubMed]
  56. Ruan, Z.; Mei, N.; Lu, Y.; Xiong, J.; Li, X.; Zheng, W.; Liu, L.; Yin, B. A Comparative and Summative Study of Radiomics-based Overall Survival Prediction in Glioblastoma Patients. J. Comput. Assist. Tomogr. 2022, 46, 470–479. [Google Scholar] [CrossRef]
  57. Wiki for the VASARI feature Set the National Cancer Institute Web Site. 2008. Available online: https://wikicancerimagingarchivenet/display/Public/VASARI+Research+Project (accessed on 15 September 2022).
  58. Peeken, J.C.; Hesse, J.; Haller, B.; Kessel, K.A.; Nüsslin, F.; Combs, S.E. Semantic imaging features predict disease progression and survival in glioblastoma multiforme patients. Strahlenther. Onkol. 2018, 194, 580–590. [Google Scholar] [CrossRef]
  59. Nie, D.; Zhang, H.; Adeli, E.; Liu, L.; Shen, D. 3D Deep Learning for Multi-modal Imaging-Guided Survival Time Prediction of Brain Tumor Patients. Med. Image Comput. Comput. Assist. Interv. 2016, 9901, 212–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Itakura, H.; Achrol, A.S.; Mitchell, L.A.; Loya, J.J.; Liu, T.; Westbroek, E.M.; Feroze, A.H.; Rodriguez, S.; Echegaray, S.; Azad, T.D.; et al. Magnetic resonance image features identify glioblastoma phenotypic subtypes with distinct molecular pathway activities. Sci. Transl. Med. 2015, 7, 303ra138. [Google Scholar] [CrossRef] [Green Version]
  61. Rathore, S.; Akbari, H.; Rozycki, M.; Abdullah, K.G.; Nasrallah, M.P.; Binder, Z.A.; Davuluri, R.V.; Lustig, R.A.; Dahmane, N.; Bilello, 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. [Google Scholar] [CrossRef]
  62. Yan, J.; Zhao, Y.; Chen, Y.; Wang, W.; Duan, W.; Wang, L.; Zhang, S.; Ding, T.; Liu, L.; Sun, Q.; et al. Deep learning features from diffusion tensor imaging improve glioma stratification and identify risk groups with distinct molecular pathway activities. EBioMedicine 2021, 72, 103583. [Google Scholar] [CrossRef] [PubMed]
  63. Zlochower, A.; Chow, D.S.; Chang, P.; Khatri, D.; Boockvar, J.A.; Filippi, C.G. Deep Learning AI Applications in the Imaging of Glioma. Top. Magn. Reson. Imaging 2020, 29, 115-00. [Google Scholar] [CrossRef]
  64. Welch, M.L.; McIntosh, C.; Haibe-Kains, B.; Milosevic, M.; Wee, L.; Dekker, A.; Huang, S.H.; Purdie, T.; O’Sullivan, B.; Aerts, H.J.; et al. Vulnerabilities of radiomic signature development: The need for safeguards. Radiother. Oncol. 2019, 130, 2–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Chang, P.; Grinband, J.; Weinberg, B.D.; Bardis, M.; Khy, M.; Cadena, G.; Su, M.-Y.; Cha, S.; Filippi, C.G.; Bota, D.; et al. Deep-Learning Convolutional Neural Networks Accurately Classify Genetic Mutations in Gliomas. Am. J. Neuroradiol. 2018, 39, 1201–1207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Gennatas, E.D.; Friedman, J.H.; Ungar, L.H.; Pirracchio, R.; Eaton, E.; Reichmann, L.G.; Interian, Y.; Luna, J.M.; Ii, C.B.S.; Auerbach, A.; et al. Expert-augmented machine learning. Proc. Natl. Acad. Sci. USA 2020, 117, 4571–4577. [Google Scholar] [CrossRef] [Green Version]
  67. Budd, S.; Robinson, E.C.; Kainz, B. A survey on active learning and human-in-the-loop deep learning for medical image analysis. Med. Image Anal. 2021, 71, 102062. [Google Scholar] [CrossRef]
  68. De Angeli, K.; Gao, S.; Alawad, M.; Yoon, H.; Schaefferkoetter, N.; Wu, X.; Durbin, E.B.; Doherty, J.; Stroup, A.; Coyle, L.; et al. Deep active learning for classifying cancer pathology reports. BMC Bioinform. 2021, 22, 113. [Google Scholar] [CrossRef]
Figure 1. Flowchart depicting the identification, screening and inclusion of studies in the present review.
Figure 1. Flowchart depicting the identification, screening and inclusion of studies in the present review.
Medicina 58 01746 g001
Figure 2. (A) Diagram depicting an example of the conventional process for studies implementing radiomics and machine learning algorithms. (B) Diagram of an example of a survival prediction investigation based on deep learning.
Figure 2. (A) Diagram depicting an example of the conventional process for studies implementing radiomics and machine learning algorithms. (B) Diagram of an example of a survival prediction investigation based on deep learning.
Medicina 58 01746 g002
Table 1. Summary of the studies analyzed in this review.
Table 1. Summary of the studies analyzed in this review.
Author
Year
NCases from Public Database *MRI Sequence
Radiomics Analysis
Segmentation Method
(Labels)
Image
Preprocessing
F. Extraction SoftwareN of F.Feature TypeFeature Selection/
ML Classifier
Validation MethodModel Performance
1Yang [17]
2015
82Yes
TCIA
T1C
FLAIR
Manual
Enhancing tumor
Whole tumor
Intensity Normalization Re-SlicingMATLAB976SFTA, GLRLM, Local Binary Patterns, Histogram of oriented gradients, HaralickRFOut-Bag ValidationSFTA T1C
AUC = 0.69
2Chaddad [18]
2016
40Yes
TCIA
T1
FLAIR
Manual
Enhancing Tumor
Necrosis
Edema
Co-Registration Intensity NormalizationMATLAB22GLCMDA, NB, DT,
SVM
LOOCVAUC = 0.793 Phenotypes with KM significantly different
3Liu [19]
2016
68NoRS-F- MRI
DTI
Automatic Anatomical LabellingRS-F-MRI:
SPM8 and DPARSF
DTI:
FSL and PANDA
GRETNA Toolbox for Connectomics2797Functional and Structural Networks,
Clinical
SVMNoAccuracy = 75%
4Macyszyn [20]
2016
134NoT1C
T1
T2
T2FLAIR
DTI
DS
Automatic
Enhancing Tumor
Non/Enhancing Tumor
Edema
Ventricles
Co-RegistrationN/A216First Order, Tumor Location, GLISTR Outputs,
Intensities
SVM10-FoldCV
VD = 29
Retrospective Accuracy = 77.14%
Prospective Accuracy = 79.17%
5Kickingereder [21]
2016
119NoT1
T1C
FLAIR
DWI
DS-C
Semiautomatic, Enhancing Tumor
Non/Enhancing Tumor
Co-Registration
N4 bias Correction Intensity Normalization
MITK12,190First order, volumetric, Wavelet, Haralick, GLCM, GLRLM,SPCAVD = 40C-index = 0.61
HR = 3.45
KM
6Lee [22]
2016
24Yes
TCIA
DS-CManual
Enhancing tumor
Nonenhancing tumor Normal WM
Co-Registration
Normalization
MATLAB18First order, GLCM, HaralickUnivariate AnalyisisNoAUC = 0.83
HR = 0.019
KM
7Ingrisch [23]
2017
66NoT1CSemiautomatic
Whole Tumor
Resampling
Normalization
Python208First order, Haralick, Parameter-free Threshold Adjacency StatisticsMinimal Depth, RF10-FoldCVC-index = 0.67
HR = 1.04
KM
8Liu [24]
2017
133Yes
TCIA
T1CManual
Enhancing Tumor
ResamplingMATLAB56First order, GLCM, GLRLMRFE-SVM
10-FoldCVAUC = 0.81 Accuracy = 78%
KM
9Li [25]
2017
92Yes
TCIA = 60
T1
T1C
FLAIR
T2
Automatic
Enhancing Tumor
Non/Enhancing Tumor
Necrosis
Edema
N4 bias correction, skull stripping, resampling, co-registration, histogram matchingMATLAB45,792First order, GLCM, GLRLM, GLSZM, NGTDMLASSOVD = 32C-index = 0.71
HR = 3.29
KM
10Liu [24]
2017
133Yes
TCGA
T1CManual
Whole Tumor
ResamplingMATLAB56GLCM, GLRLM, HistogramSVMNoAccuracy = 78.2%
AUC = 0.8104
11Lao [15]
2017
112Yes
TCIA = 75
T1
T1C
FLAIR
T2
Manual
Necrosis
Enhancing tumor
Edema
N4 Bias correction
Resampling
Co-Registration
Histogram matching
MATLAB99,707First order, GLSM, GLRLM, GLSZM, NGTDM, Deep featuresLASSOVD = 37C-index = 0.71
HR = 5.13
KM
Nomogram
12Prasanna [26]
2017
65Yes
TCIA
T1C
FLAIR
T2
Manual
Enhancing Tumor
Necrosis
Edema
Co-Registration
Insensity Normalization Bias Field Correction
MATLAB402Haralick, Laws features, Histogram of oriented gradients, Laplacian pyramidsmRMR, RF3-FoldCVKM
C-index = 0.70
13Kickingereder [27]
2018
181NoT1
T1C
FLAIR
T2
Semiautomatic, Enhancing Tumor
Nonenhancing tumor
Necrosis
Intensity Normalization CoregistrationMITK1043First order, shape, GLCM, GLRLM, GLSZMLASSOVD = 61HR = 2.72
14Bae [28]
2018
217NoT1C
FLAIR
T2
DTI
Manual.
Necrosis
Enhancing Tumor
Non/Enhancing Tumor
Co-Registration
N4-Bias Correction
Normalization
Python796GLCM, GLRLM, GLSZMVHA, RSFVD = 54AUC = 0.652
KM
15Sanghani [29]
2018
163Yes
BRATS
T1
T1C
FLAIR
T2
Manual
Enhancing Tumor
Non/Enhancing Tumor
Edema
Co-Registration ResamplingPython2200Volumetric, Shape, First order, GLCM, Gabor textureRFE-SVM5-FoldCVAccuracy = 98.7%
16Chaddad [30]
2018
40Yes
TCIA
T1
FLAIR
Manual
Enhancing Tumor
Non/Enhancing Tumor
Necrosis
Edema
Co-Registration Resampling
Intensity Normalization
MATLAB9Texture features based on LOG filterRF5-FoldCVAUC = 0.85
17Liu [31]
2018
119YesT1
T1C
FLAIR
T2
Manual
Enhancing tumor
Co-Registration ResamplingMATLAB54First order, GLCM, GLRLMSVM-RFENoT1C
AUC = 0.79, Accuracy = 80.67%
KM
18Molina-Garcia [32]
2019
404Yes
TCIA
T1CManual
Enhancing Tumor
Necrotic Core
NoMATLAB44First Order, GLRLM, GLCMNN
SVM
RT
VD = 93C-Index = 0.817
(Optimal Linear Prognosis Model)
19Tan [33]
2019
147Yes
TCIA = 112
T1C
FLAIR
Manual
Whole tumor
Edema
Contralateral WM
Co-Registration
N4 Bias Correction Resampling
Intensity Normalization
MATLAB1456 LASSOVD = 35Radiomics
C-index = 0.71
HR = 2.18
Nomogram
C-Index = 0.76
20Nie [34]
2019
93NoT1C
DTI
RS-F-MRI
Manual
Whole Tumor
Co-RegistrationN/A2048CNN supervisedCNN
SVM
10-FoldCV
VD =25
Accuracy = 90.46%
(VD = 88%)
21Choi [35]
2019
114Yes
TCIA = 53
T2Manual
Peritumoral
N/APython106First Order, GLCM, GLRLM, GLSZMNo
VD = 34
C-index 0.659
KM
22Chen [36]
2019
127Yes
TCIA
T1CManual
Enhancing tumor
Insensity NormalizationMATLAB3824First order, Shape, GLCM, GLRLMmRMRN/AHR = 3.65
AUC = 0.82
KM
23Sasaki [37]
2019
182NoT1
T1C
T2
Manual
Enhancing tumor
Whole tumor
Co-Registration
Intensity Normalization
MATLAB489First order, GLCM, GLRLM, shapeSPCA,
LASSO
10-FoldCVHR = 1.62
High and Low risk Log Rank Test
p = 0.004
24Um [38]
2019
161Yes
TCIA
T1
T1C
FLAIR
Semiautomatic
Whole tumor
Co-Registration
Rescaling
Bias field Correction Histogram Matching Resampling
CERR420First order, Edge features (LoG, Sober, Gabor, Wavelet), GLCM, GLSZM, HaralickLASSOVD = 47HR = 3.61
KM
25Chang [39]
2019
12NoT1
T2FLAIR
Pretreatment
Posttreatment1
Posttreatment2
Manual
Whole Tumor
Co-RegistrationMATLAB61GLCM, GLDM, GLRLM, GLSZM, Delta RadiomicsRF, Linear- SVM, Kernel-SVM, NN, NB, LRNoAUC = 0.889
Best Result:
RF with SVM
and
NN with Delta Radiomics
26Tixier [40]
2019
159Yes
TCIA = 47
T1
T1C
FLAIR
Semiautomatic
Whole tumor
Co-Registration
Gabor Filtering
Binning
CERR286First order, GLCM, GLSZM, GaborLASSOVD = 61KM
27Shboul [41]
2019
224Yes
BRATS
T1
T1C
FLAIR
T2
Automatic
Whole tumor
Edema
Necrosis
Enhancing Tumor
Co-Registration
Bias Correction Normalization
N/A31,000Texture, Euler, HistogramUnivariate, RFS, RF, XGBoostVD = 61
LOOCV
Accuracy = 73%
VD-Accuracy = 68%
28Chaddad [42]
2019
200Yes
TCIA = 71
T1C, FLAIRManual
Whole tumor
ResamplingMATLAB45First order, GLCM, NGTDM, GLSZMNoVD = 100AUC = 0.752
KM
29Kim [43]
2019
83NoT1
T1C
FLAIR
T2
DTI
DS-C
Semiautomatic
Enhancing Tumor
Non/Enhancing Tumor
Co-Registration
Intensity Normalization Resampling
MATLAB6472First order, Wavelet, GLCM, GLRLMLASSO10-FoldCVDTI Radiomics
AUC = 0.70
C-index 0.63
DS-C
AUC = 0.76
C-index = 0.55
30Liao [44]
2019
137Yes
TCIA
FLAIRManualN/APython72First order, GLCM, GLSZM, GLRLM, NGTDM, GLDMGBDT, SVM, kNNVD = 41Accuracy = 81% Short survival
AUC = 0.79
Long survival
AUC = 0.81
31Osman [45]
2019
163Yes,
BRATS
T1
T1C
FLAIR
T2
Manual
Enhancing Tumor
Non/Enhancing Tumor
Edema
Co-Registration
Smoothing
Interpolation
Intensity Normalization Intensisty Rescaling
MATLAB147First order, GLCM, Histogram of oriented gradients, Local Binary Pattern.LASSO, SVM, kNN, DAVD = 54Accuracy = 57.8% Short survival
AUC = 0.81
Median survival
AUC = 0.47
Long survival
AUC = 0.72
32Chaddad [46]
2019
73Yes
TCIA
T1C
FLAIR
Manual
Enhancing Tumor
Necrosis
Edema
Co-Registration Resampling
Intensity Normalization
MATLAB11JIM, GLCMSpCoR
RF
LOOCVJIM features:
HR = 1.88
AUC = 0.776
33Zhang
2019 [47]
105Yes
TCIA
T1
T1C
FLAIR
T2
Manual
FLAIR Signal
Enhancing Tumor
Necrosis
Edema
Co-Registration Resampling
Collewet Normalization
MATLAB4000First Order, GLCM, GLRLM, GLSZM, WaveletLASSO
LR
VD = 35C-Index = 0.94
Nomogram
34Han [6]
2020
178Yes
TCIA = 128
T1CManual
Whole Tumor
Normalization
Gray-Level Quantization
Resampling
MATLAB (radiomics)
CNN
(Keras-TensorFlow)
Elastic Net/Cox
(R)
8540First order, Nontexture, Histogram,
GLCM, GLRLM, GLSZM, NGTDM
Deep features(CNN)
MAD,
C-Index,
PearsonC
NoLong Rank Test Long/Short Survival p < 0.001 (HR = 3.26)
35Zhang [48]
2020
104Yes
TCIA
T1
T1C
FLAIR
T2
Manual
Whole Tumor
Tumor subregions
Co-Registration Resampling
Normalization
MATLAB180First Order, GLCM, GLRLM, GLSZMMultiple Instance Learning, SVMVD = 33Accuracy = 87.9% Sensitivity = 85.7% Specificity = 89.4%
36Suter [49]
2020
109Yes
TCIA = 76
T1
T1C
FLAIR
T2
Automatic
Enhancing Tumor
Non/Enhancing Tumor
Necrosis
Edema
Co-Registration
Skull Stripping
Resampling
N4 Bias Correction
Python8327First order, GLCM, GLSZM, GLRLM, NGTDM, GLDM, Deep features.13 F selection (RelieF, GINI, CHSQ…) and 12 ML methods
(CNN, SVM, RF, DT…)
VD = 762-Classes:
AUC = 0.66
Accuracy = 64%
3-Classes:
AUC = 0.58
Accuracy = 38%
37Bakas [50]
2020
101NoT1
T1C
FLAIR
T2
DTI
DS-C
Automatic
Enhancing Tumor
Non/Enhancing Tumor
Edema
Co-Registration
Resampling
Noise Filtering
Histogram Matching
CaPTk1612First order, Volumetric, Wavelet, GLCM, GLRLM, GLSZM, NGTDM, Spatial information, diffusion propertiesForward Selection, SVM5-FoldCVAccuracy
Advanced MRI = 73%
Basic MRI = 74.3%
KM
38Park [51]
2020
216NoT1C
FLAIR
DWI
DS-C
Semiautomatic
Enhancing tumor
Co-Registration
Intensity Normalization Resampling
MATLAB1618First order, GLCM, GLRLM, WaveletLASSOVD = 58C-index = 0.64
KM
Nomogram
39Lu [52]
2020
181NoT1CSemiautomatic
Whole tumor
Enhancing tumor
Nonenhancing tumor
Necrosis
Intensity Normalization N4 Bias CorrectionPython333Shape, First order, GLCM, GLDM, GLRLM, GLSZM, NGTDM
VASARI
VHA,
RFS
VD = 78AUC = 0. 96
C-index = 0.90
40Baid [53]
2020
346Yes
BRATS
T1
T1C
FLAIR
T2
Automatic.
Whole Tumor
Enhancing tumor
Tumor core
Co-Registration
N4 Bias Correction Normalization
MATLAB678First order, Wavelet decomposition, GLCMSpCoR,
RF
VD = 53Accuracy = 57.1%
41Moradmand [11]
2021
260Yes
TCGA
IVY
N/AN/AN/APythonN/AClinical, Tumor Data, PostSurgical Treatment, Molecular variablesCoxPH, RF, NNTD = 78C-index = 0.823 Bayesian Hyperparameter Optimization
42Yan [8]
2021
688Yes
TCIA
CGGA
Local
DTI
T2 Flair
Manual
Whole Tumor
Coregistration
Standardization
PythonN/ARadiogenomics
Clinical
CNNVD = 77C-index = 0.825
(VD-C-index = 0.79)
43Priya [54]
2021
85NoT1CManual
Whole Tumor
N/ATexRAD36Texture, AgeSVM
NN
RT
5-FoldCVAUC = 0.811
Accuracy = 67%
AUC CV = 0.71
44Cepeda [55]
2022
203Yes
TCIA = 34
BraTS = 119
T1
T1C
FLAIR
T2
Hybrid (GLISTRboost)
Enhancing tumor
Nonenhancing tumor
Edema
Re-Orientation
Co-Registration
Resampling
Normalization
CaPTk15,720First Order, Histogram, Volumetric, Morphologic, GLCM, GLDM, GLRLM, GLSZM, NGTDMGini Index, FCBF, InfoGain
/
LR, NB, kNN, RF, SVM, NN
TD = 60AUC = 0.98
Accuracy = 94%
(TD-AUC = 0.77
TD-Accuracy = 80%)
Naïve Bayes
45Ben Ahmed [14]
2022
163Yes
BRATS
T1CAutomatic
Enhancing tumor
Tumor core
Whole Tumor
Null-Voxel Reduction
Data Augmentation
2D Transformation
Python35,709SnapshotCNNVD = 46Accuracy = 74%
46Ruan [56]
2022
200Yes
TCGA = 129
T1C
T1
T2FLAIR
Manual
Whole Tumor
StandardizationMATLAB
3D Slicer
665First Order,
VASARI, GLCM, GLDM, GLRLM, GLSZM, NGTDM
LASSOVDRadiomics
C-Index = 0.935
RadiomicsVASARI
C-Index = 0.622
AUC: Area Under the Curve from Receiver Operating Characteristics; BraTS: Brain Tumor Segmentation Challenge Dataset; C: Contrast Enhanced; CGGA: Chinese Glioma Genome Atlas; CNN: Convolutional Neural Networks; CoxPH: Cox proportional hazards; CV: Cross Validation; DA: Discriminated Analysis; DS: Dynamic Susceptibility; DT: Decision Trees; DTI: Diffusion Tensor; DWI: Diffusion Weighted Image; Imaging; F: Radiomic Features; GBDT: Gradient Boosting Decision Tree; GLCM: Gray Level Co-Occurrence Matrix; GLDM: Gray Level Difference Matrix; GLSZM: Gray Level Size Zone Matrix; HR: Hazard Ratio;IVY: Ivy Glioblastoma Atlas Project; JIM: Joint Intensity Matrix; KM: Kaplan-Meier Survival Curves; kNN: K Nearest Neighbor; LASSO: Least Absolute Shrinkage and Selection operator; LOOCV: Leave One Out Cross Validation; LoG: Laplacian of Gaussian; LR: Logistic Regression; MAD: Median Absolute Deviation; mRMR: minimum Redundancy Maximum Relevance; N: Number of patients; NB: Naïve Bayesian; NGTDM: Neighborhood gray tobe difference matrix; NN: Neural Networks; PearsonC: Pearson’s Co-Relation Coeficient; RS-F-MRI: Resting State functional MRI; RF: Random Forest; RFE: Recursive feature elimination; RFS: Recursive Feature Selection; RT: Regression Trees; SPCA: Sparse Principal Component Analysis; SpCoR: Spearman’s Co-Relation; SFTA: Segmentation Fractal Texture Analysis); SVM: Support Vector Machine; TCGA: The Cancer Genome Atlas; TCIA: The Cancer Imaging Archives; TD: Testing Dataset; VD: Validation Dataset; VHA: Variable Hunting Algorithm; WM: White Matter. * (If all cases were from a public dataset, the number is not disclosed again).
Table 2. Itemized score for the Radiomics Quality Score of the included radiomics studies assessing survival prognosis in high grade gliomas.
Table 2. Itemized score for the Radiomics Quality Score of the included radiomics studies assessing survival prognosis in high grade gliomas.
Author and YearYang
2015
Lee
2016
Kickingereder
2016
Macyszyn 2016Chaddad
2016
Lao
2017
Liu
2017
Li
2017
Ingrisch
2017
Prasanna
2017
Bae
2018
Sanghani
2018
Liao
2018
Chaddad
2018
Liu
2018
Choi
2019
Image protocol quality1012111111100011
Multiple segmentations0010011100100111
Phantom study on all scanners0000010100000000
Imaging at multiple time points0000000000000000
Feature reduction or adjustment for multiple testing−3333−333333333−33−3
Multivariate analysis with nonradiomic features0111010111101001
Detect and discuss biologic correlates1111111111111111
Cutoff analysis0111110100100100
Discrimination statistics1111111100201110
Calibration statistics1000010000000000
Prospective study registered in trial data base0001000000000000
Validation2−5222322222−52222
Comparison with criterion standard0220020222200002
Potential clinical utility0000000000000000
Cost-effectiveness analysis0000000000000000
Open science and data1100111101020111
Total points4513124191015101114184106
% RQS11%14%36%33%11%53%28%42%28%31%39%3%22%11%28%17%
Author and YearTixier
2019
Shboul
2019
Chaddad
2019
Chen
2019
Kim
2019
Chang
2019
Osman
2019
Chaddad
2019
Um
2019
Zhang
2019
Han
2020
Zhang
2020
Suter
2020
Bakas
2020
Park
2020
Cepeda
2021
Ruan
2022
Image protocol quality10011010112101111
Multiple segmentations11011111110111101
Phantom study on all scanners00000000100010000
Imaging at multiple time points00000100000000000
Feature reduction or adjustment for multiple testing33−333333333333333
Multivariate analysis with nonradiomic features00111011010100111
Detect and discuss biologic correlates11111011111111101
Cutoff analysis10011111111010111
Discrimination statistics01111111011111111
Calibration statistics00000000010000010
Prospective study registered in trial data base00000000000000000
Validation22222222220222332
Comparison with criterion standard00222122022200221
Potential clinical utility00000000000000000
Cost-effectiveness analysis00000000000000000
Open science and data12110031011112121
Total points101051413101613101511131111151513
% RQS28%28%14%39%36%28%44%36%28%42%31%36%31%31%42%42%36%
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

García-García, S.; García-Galindo, M.; Arrese, I.; Sarabia, R.; Cepeda, S. Current Evidence, Limitations and Future Challenges of Survival Prediction for Glioblastoma Based on Advanced Noninvasive Methods: A Narrative Review. Medicina 2022, 58, 1746. https://0-doi-org.brum.beds.ac.uk/10.3390/medicina58121746

AMA Style

García-García S, García-Galindo M, Arrese I, Sarabia R, Cepeda S. Current Evidence, Limitations and Future Challenges of Survival Prediction for Glioblastoma Based on Advanced Noninvasive Methods: A Narrative Review. Medicina. 2022; 58(12):1746. https://0-doi-org.brum.beds.ac.uk/10.3390/medicina58121746

Chicago/Turabian Style

García-García, Sergio, Manuel García-Galindo, Ignacio Arrese, Rosario Sarabia, and Santiago Cepeda. 2022. "Current Evidence, Limitations and Future Challenges of Survival Prediction for Glioblastoma Based on Advanced Noninvasive Methods: A Narrative Review" Medicina 58, no. 12: 1746. https://0-doi-org.brum.beds.ac.uk/10.3390/medicina58121746

Article Metrics

Back to TopTop