Skip to main content

A review on the applications of artificial intelligence and big data for glioblastoma multiforme management

Abstract

Glioblastoma is known as an aggressive type of brain tumor with a very poor survival rate and resistance to different treatment methods. Considering the difficulties in studying glioblastoma, the development of alternative methods for the identification of prognostic factors in this disease seems necessary. Noteworthy, imaging, pathologic, and molecular data obtained from patients are highly valuable because of their potential for this purpose. Artificial intelligence (AI) has emerged as a powerful tool to perform highly accurate analyses and extract more detailed information from available patient data. AI is usually used for the development of prediction models for prognosis, response/resistance to treatments, and subtype identification in cancers. Today, the number of AI-aided developed algorithms is increasing in the field of glioblastoma. Challenges in the diagnosis of tumors using imaging data, prediction of genetic alterations, and prediction of overall survival are among the most popular studies related to glioblastoma. Hereby, we reviewed peer-reviewed articles in which AI methods were used for various targets in glioblastoma. Reviewing the published articles showed that the use of clinical imaging data is reasonably more popular than other assessments because of its noninvasive nature. However, the use of molecular assessments is becoming extended in this disease. In this regard, we summarized the developed algorithms and their applications for the diagnosis and prognosis of glioblastoma tumors. We also considered the accuracy rates of algorithms to shed light on the advancements of different methodologies in the included studies.

Background

Glioblastoma grade IV, formerly called glioblastoma multiforme (GBM), is known as the most aggressive brain tumor with a survival of less than two years [1]. The prevalence of glioblastoma is approximately 3.2 in 100,000 population, while it has the highest mortality rate of90% in 5 years among all the forms of brain tumors [2, 3]. Glioblastomas are divided into two subgroups based on their origination including primary tumors which originate from brain tissue, and secondary tumors which are developed via metastasis from other tissues and compose 10–45% of glioblastomas [4].The median age of glioblastoma diagnosis is 65 years, and it has been found that the probability of incidence for this disease in males is higher than in females. Based on the classification of WHO in 2021, glioblastoma is pathologically diagnosed with IDH wild type and mutations in TERT promoter, chromosomes 7/10, EGFR gene [5]. In addition, the genomic drivers of glioblastoma include amplification of PDGFRA and MET, deletions in CDK2A/B, and gain mutations of NF1, TET2, DNMT3Am and LTBP4 [6,7,8]. On the other hand, genomic changes in DNA mismatch repair genes such as MSH1, MSH2, etc., and altered gene expression in endothelial marker genes and extracellular-associated genes are among the molecular hallmarks of glioblastoma [9].

Artificial intelligence (AI) is a set of continuous activities in which technology performs smart behaviors without human direct intervention. Amisha et al. described AI as the science of engineering for the smart machines production in 1965 [10]. AI is increasingly used in medical sciences for various targets including improvement of decision-making in diagnosis, prognosis, and treatment of diseases [11]. It is also poised to broadly reshape medicine and improve the experiences of both clinicians and patients. Machine learning (ML) algorithms such as K-nearest neighbor (KNN), random forest (RF), support vector machine (SVM), and logistic regression (LR) are used in various medical fields. For example, a large number of studies have been conducted in the oncology field in order to find prognostic factors, improve diagnostic methods, and predict response to treatments based on the genetic factors. In addition, using the deep learning (DL) approach, it is possible to study cancer signaling systems in a large-scale data-driven fashion [10]. Accordingly, these methods lead to faster and better decision-making processes by physicians [12,13,14,15]. In addition, AI is also exploitable physically in medical sciences by which robots and medical devices are developed to assist nurses, doctors, and surgeons [16].

Despite the advancements of AI in medical sciences and brain tumors, several of concerns remain to be answered. The accuracy of predictions made by ML/DL algorithms, the benefits of designed algorithms for improvement of brain tumor classification, and prediction of response to treatment for this disease can be mentioned as the main concerns. In this review, we searched for the relevant studies using keywords including “glioblastoma,” “GBM,” “machine learning” (ML), “deep learning,” and “artificial intelligence” in databases of PubMed, Embase, and Web of Science. Hereby, we summarize applications of virtual artificial intelligence for glioblastoma in various fields such as the use of radiomics for diagnosis and prognosis, and molecular studies for prediction of response to treatment and its benefits for designing personalized treatment methods.

Radiomics and AI applications for glioblastoma

The pathological and molecular assessments of tumor biopsies are the reference standard for tumor diagnosis and grading. However, a noninvasive tool capable of accurately classifying tumor subtypes and inferring its grade would be highly desirable. There are several noninvasive imaging techniques which help to visualize glioma tumors. Computed tomography (CT), positron emission tomography (PET), and magnetic resonance imaging (MRI) are important diagnostic tools used for decision-making about the standard care of glioblastoma in clinical practice. MRI includes detailed data of the lesion location, size, extent, features, tumor and surrounding environment relationship, and associated mass effect. Beyond structural information, MRI can also assess microstructural features such as lesion cellularity, microvascular architecture, and perfusion. Conventional radiomic programs can generally analyze medical images, but they are not accurate enough for analysis of detailed aspects of images. Advanced imaging techniques may demonstrate more details of brain tumors related to their heterogeneity, aggressiveness, and grade; however, they are limited in assessing the microscopic changes that cause macroscopic ones. Convolutional neural network (CNN) from deep learning (DL) algorithms with several applications in image classification and image segmentations are widely used in brain tumor classification [12, 17]. In turn, deep learning, a subcategory of machine learning, is a network model constructed from features of input data which are connected to output layer via several intermediary layers. DL provides automatic learning of features and their representation in a hierarchical manner at various levels. These characteristics have made DL a robust technique for feature extraction and data modeling processes. The initial layers perform simple processing of input data or learn the easy features, while the upper layers perform complex features learning and lead to the results more precisely. Therefore, DL is suitable for dealing with large and complex data [18, 19].

Prediction of survival for patients with an aggressive tumor such as glioblastoma helps oncologists and surgeons to design treatment protocols appropriately for patients. Punctual preoperative prognosis holds great importance for the personal treatment of glioblastoma patients. DL performs multi-faceted MRI image analysis and accordingly assists specialists in predicting the OS time for these patients. The classification and recognition of glioblastoma tumors using MRI is a crucial process before surgery. In addition, the assessment of tumor location, composition, and extent of tumors are of high importance. On the other hand, manual tumor classification is a time-consuming and error-prone process. In this regard, Eijgelaar et al. designed an algorithm using DL (sparsified algorithm) which can be used to analyze data in a generalized and specific manner. Consequently, glioblastoma tumors are classified automatically, which it significantly assists physicians in accomplishing better surgeries in a shorter time [20] (Fig. 1).

Fig. 1
figure 1

Machine learning and deep learning in brain tumor detection

Lao et al. used a radiomic model that could predict patient OS by searching the intricate details of medical images. Interestingly, this program can play a crucial role in treating individuals with glioblastoma by categorizing them into high-risk and low-risk groups [21]. Biophysical modeling can provide more complex and extensive aspects of glioblastoma tumor growth. In this regard, Pati et al. designed a linear regression (LR) model based on DL that utilizes three components: diffusion coefficient in white matter, mass parameter, and estimated days of tumor growth based on the patient’s conditions. This algorithm notably facilitates tumor growth estimation for physicians [22]. In addition, glioblastoma primary and metastatic tumors often overlap in MRI images, and consequently, their diagnosis becomes challenging for radiologists. Shin et al. developed a DL-based model that effectively distinguishes between these two types of images with high-level accuracy [23]. In addition, glioblastoma and primary central nervous system lymphoma (PCNSL) are recognized as primary brain tumors. These tumors exhibit significant similarities in MRI images. Considering that these two tumors have different treatment methods, the development of an accurate diagnosis method using MRI data is undoubtedly important. Xia et al. introduced a model based on the CNN algorithm that easily elucidates the differences between the MRIs of these two tumor types [24]. Another issue is separation between glioblastoma and brain metastases which remains as an issue in medical imaging. Physicians should be able to accurately differentiate between these two tumor types. Bea et al. presented a CNN model that uses radiomic features to easily distinguish between the mentioned two tumor types [25]. Tumefactive demyelinating lesion (TDL) and PCNSL are common intracranial neuroepithelial lesions in glioblastoma. These regions can be observed as solitary or scattered forms. In solitary form, they are seen as well-defined units with surrounding edema. In scattered form, they appear as irregular scattered masses. Although these tumors have different origins, they show relatively intersections in MRI images. Researchers increased the accuracy of diagnosis of scattered forms by a large number of MRI data and using DL algorithms. This approach also facilitated the process of separating these brain lesions in MRI images. Consequently, it helps physicians to make better decisions in the management of patient treatment [13]. Preoperative multimodal imaging data has been used to predict the OS of patients via extraction of genotype-related features from MRI data [26]. CNN algorithm can accurately classify glioblastoma tumors, which is based on a nifty net framework at the same time. Additionally, CNN algorithms are very helpful in the differentiation between glioblastoma and secondary brain tumors in MRI images [20]. Tumor cells of glioblastoma have a highly invasive phenotype. As a result, there is a 90% chance of tumor recurrence that is very prevalent even after complete resection and performing chemotherapy and other cancer treatments in the tumor area. Also, recurrences usually occur at the site of tumor removal or in areas near the tumor site. Yan et al. recognized the non-enhanced peritumoral region by utilizing the CNN algorithm and MRI data, which helped determining probable regions of glioblastoma cell progression in the future [27] (Table 1).

Table 1 Application of radiomics in GBM

Another research indicated that unsupervised DL such as DBN algorithms with layer models works great for glioblastoma subtype recognition [28]. A designed radiometric model operates based on DL algorithms and shows successful application in finding deep aspects and details of MRI images. In addition, in this model, researchers employ COX regression which leads to precise prediction of OS in glioblastoma patients [21]. Another ML algorithm, called as deep neural network (DNN), supports specialists in discriminating glioblastoma and brain secondary tumors using only imaging data [23]. DL algorithms are also able to diagnose mutated genes that lead to glioblastoma appearance and OS prediction [29]. Researchers claimed that ML algorithms such as LR, SVM, k-nearest neighbor (KNN), and naïve Bayes (NB) are applied for glioblastoma tumor classification; however, the CNN algorithm assists this tumor categorization as well [25, 30] (Fig. 2).

Fig. 2
figure 2

The overview of AI-based GBM management methods

Genomics, precision medicine, and AI applications for glioblastoma

Precision oncology is fed by data extracted from high throughput sequencing (HTS) techniques and is progressing toward flexible treatment methods based on the personal genome diversities. HTS techniques, including next-generation sequencing (NGS), provide a robust resource for the identification of targeted therapy triggers and risk factors for cancers. Integration of NGS data with imaging and clinical data via AI methods leads to various predictions about personal sensitivities, susceptibilities, and responses to treatment precisely. In addition to the mentioned general applications, the use of AI in cancer big data analytics has technical advantages such as scaling up the data processing and accurate predictions where clinicians may have different diagnostics and decisions about treatments [52,53,54]. Thang et al. designed a multi-task CNN that efficiently considered the tumor genotype in predicting OS of patients and it yielded an accurate predictive algorithm [26]. In cancerous cells such as glioblastoma, there are several signaling pathways that are initiated via mutated genes. This signaling pathway may conduct abnormal functionalities in these cells. Analyzing these processes can potentially lead to the development of personalized cancer treatment approaches. In this regard, the DBN algorithm was introduced by Young et al. which examines glioblastoma tumors using genomic data, and these processes considerably help in understanding the underlying mechanisms of tumor subtypes [28]. In this regard, there is a group of mutated genes involved in the development of glioblastoma. DL- and ML-based models can analyze and identify these genes, which result in effective prediction of OS for patients with this type of tumor. This algorithm also helps physicians to make effective decisions for selecting better treatment methods based on the high-risk/low-risk categorization [29]. Molecular features of glioblastoma cells have always been a challenge for medical scientists, but AI has provided valuable assistance in this area. Munquad et al. utilized SVM, LR, and RF algorithms for classification of glioma cells based on molecular variables and gene expression profiles [30] (Fig. 3).

Fig. 3
figure 3

Machine learning and deep learning in genomic data and diagnosis

Challenges and future prospective

Gliomas are a heterogeneous group of common intracranial tumors that cause significant mortality and morbidity. Gliomas are classified into four grades (I to IV) of increasingly aggressive malignancy and worsening prognosis. Indeed, in clinical practice, tumor type and grade influence treatment choice. According to the World Health Organization (WHO) Grade IV tumors, so-called glioblastoma the most aggressive primary brain tumor, with a median survival after diagnosis of just 12–15 months. Applications of AI for this disease in different aspects have also disadvantages. Malfunctioning in designed algorithms or processors along with lack of accurate results or incorrect interpretations of the results can lead to the wrong conclusions and subsequently to wrong decision-making for physicians [55]. On the other hand, overfitting is the main problem for predicting various aspects of glioblastoma diagnosis, prognosis, and treatment which warns physicians to not completely rely on AI-based applications and also leads specialists in medical-AI to develop more accurate and reliable software. For this purpose, a vast amount of data is not only required for training, testing and internal validation, but also for external validation of algorithms. Noteworthy, the use of algorithms for prognosis can be confusing, since the results change based on the slight changes in the included variables [56]. Accordingly, achieving the accurate results and best features for prediction of outcomes in each stage of disease and for every application including prognosis, diagnosis, and treatment and their subgroups seems necessary. In this regard, the determination of the most effective variables for each application will help to not only use a limited number of variables, but also to develop more accurate algorithms. An aggressive type of cancer such as glioblastoma has still a wide area of unclarities around the prognosis of patients and categorizing them to low risk and high risk, while in the diagnosis of glioblastoma and its subtypes based on the pathobiology results and synchronizing these results with imaging results to develop noninvasive AI algorithms, it is important to consider that there are already conflicts between genome profiling of mutations for grading tumors and imaging-based diagnosis. Decision-making about treatment methods for glioblastoma patients is an ongoing concern. Glioblastoma treatment includes standard care including surgical removal, chemotherapy, and radiotherapy. However, according to the genomic variations in patients, different resistance types to standard care and novel treatments such as immunotherapies are observed in glioblastoma patients [57, 58]. Also, the lack of standardized radiomics parameters and data can induce some challenges in the generalization of ML algorithms, which have limited AI model efficiencies in detecting high-grade gliomas and predicting patient survival [59]. Although AI models are beneficial for helping GBM patients, medical data quality and paucity, AI methods transparency, different image imputations, and ethical problems linger as the concerns [60, 61]. As it was mentioned above, there are still lots of questions regarding glioblastoma which can be answered using genomics data such as prediction of response to treatment rate, resistance to treatment, recurrent cases, and survival rate. In this regard, AI algorithms are required in order to predict resistance to each treatment type based on the molecular profiling. These algorithms will help to save time in treating patients especially those who are at high risk and design an effective treatment pathway which can be most effective and lead to longer survival in GBM patients.

Conclusion

Glioblastoma is known for low survival and short lifetime of the patients. Considering lots of uncertainties around the diagnosis and prognosis of the patients and decision-making for the most effective treatment approach, AI can help to resolve these issues. Development of verified and reliable applications with the lowest overfitting, underfitting, false-positive ratio, and false-negative ratios is required to boost progress toward improving the management of glioblastoma patients. In this regard, the identification of the most effective variables for each application is necessary. In total, AI has shown the ability to create a brighter future for physicians in glioblastoma disease management, especially in estimating patient OS and other tumor-related complex information with radiomics and molecular assessment tools, which needs the contribution of physicians and computer scientists.

Abbreviations

AI:

Artificial intelligence

ANN:

Artificial neural network

CNN:

Convolutional neural network

CT:

Computed tomography

DNN:

Deep neural network

DL:

Deep learning

GBM:

Glioblastoma multiforme

HTS:

High throughput sequencing

KNN:

K-nearest neighbor

LR:

Logistic regression

LR:

Linear regression

ML:

Machine learning

MRI:

Magnetic resonance imaging

NB:

Naïve Bayes

NGS:

Next-generation sequencing

OS:

Overall survival

PCNSL:

Primary central nervous system lymphoma

PET:

Positron emission tomography

RF:

Random forest

SVM:

Support vector machine

TDL:

Tumefactive demyelinating lesion

WHO:

World Health Organization

References

  1. Witthayanuwat S, Pesee M, Supaadirek C, Supakalin N, Thamronganantasakul K, Krusun S. Survival analysis of glioblastoma multiforme. Asian Pac J Cancer Prev APJCP. 2018;19(9):2613.

    CAS  PubMed  Google Scholar 

  2. Carlsson SK, Brothers SP, Wahlestedt C. Emerging treatment strategies for glioblastoma multiforme. EMBO Mol Med. 2014;6(11):1359–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Thakkar JP, Dolecek TA, Horbinski C, Ostrom QT, Lightner DD, Barnholtz-Sloan JS, et al. Epidemiologic and molecular prognostic review of glioblastoma. Cancer Epidemiol Biomark Prev. 2014;23(10):1985–96.

    Article  CAS  Google Scholar 

  4. Lah TT, Novak M, Breznik B, editors. Brain malignancies: glioblastoma and brain metastases, Seminars in cancer biology. Amsterdam: Elsevier; 2020.

    Google Scholar 

  5. Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol. 2021;23(8):1231–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Barthel FP, Johnson KC, Varn FS, Moskalik AD, Tanner G, Kocakavuk E, et al. Longitudinal molecular trajectories of diffuse glioma in adults. Nature. 2019;576(7785):112–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Draaisma K, Chatzipli A, Taphoorn M, Kerkhof M, Weyerbrock A, Sanson M, et al. Molecular evolution of IDH wild-type glioblastomas treated with standard of care affects survival and design of precision medicine trials: a report from the EORTC 1542 study. J Clin Oncol. 2020;38(1):81–99.

    Article  CAS  PubMed  Google Scholar 

  8. Wang J, Cazzato E, Ladewig E, Frattini V, Rosenbloom DI, Zairis S, et al. Clonal evolution of glioblastoma under therapy. Nat Genet. 2016;48(7):768–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hoogstrate Y, Draaisma K, Ghisai SA, van Hijfte L, Barin N, de Heer I, et al. Transcriptome analysis reveals tumor microenvironment changes in glioblastoma. Cancer Cell. 2023;41(4):678-92. e7.

    Article  CAS  PubMed  Google Scholar 

  10. Malik P, Pathania M, Rathaur VK. Overview of artificial intelligence in medicine. J Family Med Prim Care. 2019;8(7):2328.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Rajpurkar P, Chen E, Banerjee O, Topol EJ. AI in health and medicine. Nat Med. 2022;28(1):31–8.

    Article  CAS  PubMed  Google Scholar 

  12. Williams S, Layard Horsfall H, Funnell JP, Hanrahan JG, Khan DZ, Muirhead W, et al. Artificial intelligence in brain tumour surgery—an emerging paradigm. Cancers. 2021;13(19):5010.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Zhang Y, Liang K, He J, Ma H, Chen H, Zheng F, et al. Deep learning with data enhancement for the differentiation of solitary and multiple cerebral glioblastoma, lymphoma, and tumefactive demyelinating lesion. Front Oncol. 2021;11: 665891.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Sotoudeh H, Shafaat O, Bernstock JD, Brooks MD, Elsayed GA, Chen JA, et al. Artificial intelligence in the management of glioma: era of personalized medicine. Front Oncol. 2019;9:768.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Luo J, Pan M, Mo K, Mao Y, Zou D, editors. Emerging role of artificial intelligence in diagnosis, classification and clinical management of glioma seminars in cancer biology. Amsterdam: Elsevier; 2023.

    Google Scholar 

  16. Hamet P, Tremblay J. Artificial intelligence in medicine. Metabolism. 2017;69:S36–40.

    Article  CAS  Google Scholar 

  17. Amin J, Sharif M, Haldorai A, Yasmin M, Nayak RS. Brain tumor detection and classification using machine learning: a comprehensive survey. Complex Intell Syst. 2021:1–23.

  18. Sharma N, Sharma R, Jindal N. Machine learning and deep learning applications-a vision. Global Trans Proc. 2021;2(1):24–8.

    Article  Google Scholar 

  19. Xie Y, Zaccagna F, Rundo L, Testa C, Agati R, Lodi R, et al. Convolutional neural network techniques for brain tumor classification (from 2015 to 2022): review, challenges, and future perspectives. Diagnostics. 2022;12(8):1850.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Eijgelaar RS, Visser M, Müller DM, Barkhof F, Vrenken H, van Herk M, et al. Robust deep learning–based segmentation of glioblastoma on routine clinical MRI scans using sparsified training. Radiol Artif Intell. 2020;2(5):190103.

    Article  Google Scholar 

  21. Lao J, Chen Y, Li Z-C, Li Q, Zhang J, Liu J, et al. A deep learning-based radiomics model for prediction of survival in glioblastoma multiforme. Sci Rep. 2017;7(1):10353.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Pati S, Sharma V, Aslam H, Thakur SP, Akbari H, Mang A, et al. (eds). Estimating glioblastoma biophysical growth parameters using deep learning regression. Brainlesion: glioma, multiple sclerosis, stroke and traumatic brain injuries: 6th International Workshop, BrainLes 2020, Held in Conjunction with MICCAI 2020, Lima, Peru, October 4, 2020, Revised Selected Papers, Part I 6; 2021: Springer.

  23. Shin I, Kim H, Ahn S, Sohn B, Bae S, Park J, et al. Development and validation of a deep learning-based model to distinguish glioblastoma from solitary brain metastasis using conventional MR images. Am J Neuroradiol. 2021;42(5):838–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Xia W, Hu B, Li H, Shi W, Tang Y, Yu Y, et al. Deep learning for automatic differential diagnosis of primary central nervous system lymphoma and glioblastoma: multi-parametric magnetic resonance imaging based convolutional neural network model. J Magn Reson Imaging. 2021;54(3):880–7.

    Article  PubMed  Google Scholar 

  25. Bae S, An C, Ahn SS, Kim H, Han K, Kim SW, et al. Robust performance of deep learning for distinguishing glioblastoma from single brain metastasis using radiomic features: model development and validation. Sci Rep. 2020;10(1):12110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Tang Z, Xu Y, Jin L, Aibaidula A, Lu J, Jiao Z, et al. Deep learning of imaging phenotype and genotype for predicting overall survival time of glioblastoma patients. IEEE Trans Med Imaging. 2020;39(6):2100–9.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Yan J-L, Li C, van der Hoorn A, Boonzaier NR, Matys T, Price SJ. A neural network approach to identify the peritumoral invasive areas in glioblastoma patients by using MR radiomics. Sci Rep. 2020;10(1):9748.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Young JD, Cai C, Lu X. Unsupervised deep learning reveals prognostically relevant subtypes of glioblastoma. BMC Bioinf. 2017;18(11):5–17.

    Google Scholar 

  29. Wong KK, Rostomily R, Wong ST. Prognostic gene discovery in glioblastoma patients using deep learning. Cancers. 2019;11(1):53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Munquad S, Si T, Mallik S, Das AB, Zhao Z. A deep learning–based framework for supporting clinical diagnosis of glioblastoma subtypes. Front Genet. 2022;13: 855420.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Faghani S, Khosravi B, Moassefi M, Conte GM, Erickson BJ. A comparison of three different deep learning-based models to predict the MGMT promoter methylation status in glioblastoma using brain MRI. J Digit Imaging. 2023;36(3):837–46.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Zhang H, Zhang H, Zhang Y, Zhou B, Wu L, Lei Y, et al. Deep learning radiomics for the assessment of telomerase reverse transcriptase promoter mutation status in patients with glioblastoma using multiparametric MRI. J Magn Reson Imaging. 2023;58(5):1441–51.

    Article  PubMed  Google Scholar 

  33. Pasquini L, Napolitano A, Tagliente E, Dellepiane F, Lucignani M, Vidiri A, et al. Deep learning can differentiate IDH-mutant from IDH-wild GBM. J Personalized Med. 2021;11(4):290.

    Article  Google Scholar 

  34. Zhang H, Zhou B, Zhang H, Zhang Y, Lei Y, Huang B. Peritumoral radiomics for identification of telomerase reverse transcriptase promoter mutation in patients with glioblastoma based on preoperative MRI. Can Assoc Radiol J. 2023:08465371231183309.

  35. Saxena S, Jena B, Mohapatra B, Gupta N, Kalra M, Scartozzi M, et al. Fused deep learning paradigm for the prediction of o6-methylguanine-DNA methyltransferase genotype in glioblastoma patients: a neuro-oncological investigation. Comput Biol Med. 2023;153: 106492.

    Article  CAS  PubMed  Google Scholar 

  36. Jaruenpunyasak J, Duangsoithong R, Tunthanathip T. Deep learning for image classification between primary central nervous system lymphoma and glioblastoma in corpus callosal tumors. J Neurosci Rural Pract. 2023;14(3):470.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Bathla G, Dhruba DD, Soni N, Liu Y, Larson NB, Kassmeyer BA, et al. AI-based classification of three common malignant tumors in neuro-oncology: a multi-institutional comparison of machine learning and deep learning methods. J Neuroradiol. 2023.

  38. Liu Z, Jiang Z, Meng L, Yang J, Liu Y, Zhang Y, et al. Handcrafted and deep learning-based radiomic models can distinguish GBM from brain metastasis. J Oncol. 2021;2021.

  39. Shoushtari FK, Sina S, Dehkordi AN. Automatic segmentation of glioblastoma multiform brain tumor in MRI images: using Deeplabv3+ with pre-trained Resnet18 weights. Physica Med. 2022;100:51–63.

    Article  Google Scholar 

  40. Rui W, Zhang S, Shi H, Sheng Y, Zhu F, Yao Y, et al. Deep learning-assisted quantitative susceptibility mapping as a tool for grading and molecular subtyping of gliomas. Phenomics. 2023;3(3):243–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Chang K, Beers AL, Bai HX, Brown JM, Ly KI, Li X, et al. Automatic assessment of glioma burden: a deep learning algorithm for fully automated volumetric and bidimensional measurement. Neuro Oncol. 2019;21(11):1412–22.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Ermiş E, Jungo A, Poel R, Blatti-Moreno M, Meier R, Knecht U, et al. Fully automated brain resection cavity delineation for radiation target volume definition in glioblastoma patients using deep learning. Radiat Oncol. 2020;15:1–10.

    Article  Google Scholar 

  43. Ramesh KK, Xu KM, Trivedi AG, Huang V, Sharghi VK, Kleinberg LR, et al. A fully automated post-surgical brain tumor segmentation model for radiation treatment planning and longitudinal tracking. Cancers. 2023;15(15):3956.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Du P, Wu X, Liu X, Chen J, Chen L, Cao A, et al. The application of decision tree model based on clinicopathological risk factors and pre-operative MRI radiomics for predicting short-term recurrence of glioblastoma after total resection: a retrospective cohort study. Am J Cancer Res. 2023;13(8):3449.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Moassefi M, Faghani S, Conte GM, Kowalchuk RO, Vahdati S, Crompton DJ, et al. A deep learning model for discriminating true progression from pseudoprogression in glioblastoma patients. J Neurooncol. 2022;159(2):447–55.

    Article  CAS  PubMed  Google Scholar 

  46. Chiu F-Y, Le NQK, Chen C-Y. A multiparametric MRI-based radiomics analysis to efficiently classify tumor subregions of glioblastoma: a pilot study in machine learning. J Clin Med. 2021;10(9):2030.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Meaney C, Das S, Colak E, Kohandel M. Deep learning characterization of brain tumours with diffusion weighted imaging. J Theor Biol. 2023;557: 111342.

    Article  PubMed  Google Scholar 

  48. Fathi Kazerooni A, Saxena S, Toorens E, Tu D, Bashyam V, Akbari H, et al. Clinical measures, radiomics, and genomics offer synergistic value in AI-based prediction of overall survival in patients with glioblastoma. Sci Rep. 2022;12(1):8784.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Li M, Tang H, Chan MD, Zhou X, Qian X. DC-AL GAN: Pseudoprogression and true tumor progression of glioblastoma multiform image classification based on DCGAN and AlexNet. Med Phys. 2020;47(3):1139–50.

    Article  PubMed  Google Scholar 

  50. Akbari H, Rathore S, Bakas S, Nasrallah MP, Shukla G, Mamourian E, et al. Histopathology-validated machine learning radiographic biomarker for noninvasive discrimination between true progression and pseudo-progression in glioblastoma. Cancer. 2020;126(11):2625–36.

    Article  CAS  PubMed  Google Scholar 

  51. Bacchi S, Zerner T, Dongas J, Asahina AT, Abou-Hamden A, Otto S, et al. Deep learning in the detection of high-grade glioma recurrence using multiple MRI sequences: a pilot study. J Clin Neurosci. 2019;70:11–3.

    Article  PubMed  Google Scholar 

  52. Parimbelli E, Wilk S, Cornet R, Sniatala P, Sniatala K, Glaser S, et al. A review of AI and data science support for cancer management. Artif Intell Med. 2021;117: 102111.

    Article  CAS  PubMed  Google Scholar 

  53. Dlamini Z, Francies FZ, Hull R, Marima R. Artificial intelligence (AI) and big data in cancer and precision oncology. Comput Struct Biotechnol J. 2020;18:2300–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Capobianco E. High-dimensional role of AI and machine learning in cancer research. Br J Cancer. 2022;126(4):523–32.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Gao Y, Sharma T, Cui Y. Addressing the challenge of biomedical data inequality: an artificial intelligence perspective. Annu Rev Biomed Data Sci. 2023;6:153–71.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Pierce R, Sterckx S, Van Biesen W. A riddle, wrapped in a mystery, inside an enigma: How semantic black boxes and opaque artificial intelligence confuse medical decision-making. Bioethics. 2022;36(2):113–20.

    Article  PubMed  Google Scholar 

  57. Dymova MA, Kuligina EV, Richter VA. Molecular mechanisms of drug resistance in glioblastoma. Int J Mol Sci. 2021;22(12):6385.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Jackson CM, Choi J, Lim M. Mechanisms of immunotherapy resistance: lessons from glioblastoma. Nat Immunol. 2019;20(9):1100–9.

    Article  CAS  PubMed  Google Scholar 

  59. Pasquini L, Napolitano A, Lucignani M, Tagliente E, Dellepiane F, Rossi-Espagnet MC, et al. AI and high-grade glioma for diagnosis and outcome prediction: Do all machine learning models perform equally well? Front Oncol. 2021;11: 601425.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ibrahim M, Muhammad Q, Zamarud A, Eiman H, Fazal F. Navigating glioblastoma diagnosis and care: transformative pathway of artificial intelligence in integrative oncology. Cureus. 2023;15(8).

  61. Jin W, Fatehi M, Abhishek K, Mallya M, Toyota B, Hamarneh G. Artificial intelligence in glioma imaging: challenges and advances. J Neural Eng. 2020;17(2): 021002.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable

Funding

Not applicated.

Author information

Authors and Affiliations

Authors

Contributions

MM paraphrased radiomics and AI applications for glioblastoma and genomics, precision medicine, and AI applications for glioblastoma section of this paper and was a major contributor in writing the manuscript, FTK paraphrased background and challenges and future prospective section, and all authors read and approved the final manuscript.

Corresponding author

Correspondence to Elnaz Amanzadeh Jajin.

Ethics declarations

Ethics approval and consent to participate

Not applicated.

Consent for publication

Not applicated.

Availability of data and materials

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mehmandoost, M., Konjin, F.T., Jajin, E.A. et al. A review on the applications of artificial intelligence and big data for glioblastoma multiforme management. Egypt J Neurosurg 39, 51 (2024). https://doi.org/10.1186/s41984-024-00306-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41984-024-00306-4

Keywords