1
|
Yan RE, Greenfield JP. Challenges and Outlooks in Precision Medicine: Expectations Versus Reality. World Neurosurg 2024; 190:573-581. [PMID: 39425299 DOI: 10.1016/j.wneu.2024.06.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 10/21/2024]
Abstract
Recent developments in technology have led to rapid advances in precision medicine, especially due to the rise of next-generation sequencing and molecular profiling. These technological advances have led to rapid advances in research, including increased tumor subtype resolution, new therapeutic agents, and mechanistic insights. Certain therapies have even been approved for molecular biomarkers across histopathological diagnoses; however, translation of research findings to the clinic still faces a number of challenges. In this review, the authors discuss several key challenges to the clinical integration of precision medicine, including the blood-brain barrier, both a lack and excess of molecular targets, and tumor heterogeneity/escape from therapy. They also highlight a few key efforts to address these challenges, including new frontiers in drug delivery, a rapidly expanding treatment repertoire, and improvements in active response monitoring. With continued improvements and developments, the authors anticipate that precision medicine will increasingly become the gold standard for clinical care.
Collapse
Affiliation(s)
- Rachel E Yan
- Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Jeffrey P Greenfield
- Department of Neurological Surgery, NewYork-Presbyterian Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
2
|
Yu D, Zhong Q, Xiao Y, Feng Z, Tang F, Feng S, Cai Y, Gao Y, Lan T, Li M, Yu F, Wang Z, Gao X, Li Z. Combination of MRI-based prediction and CRISPR/Cas12a-based detection for IDH genotyping in glioma. NPJ Precis Oncol 2024; 8:140. [PMID: 38951603 PMCID: PMC11217299 DOI: 10.1038/s41698-024-00632-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/30/2024] [Indexed: 07/03/2024] Open
Abstract
Early identification of IDH mutation status is of great significance in clinical therapeutic decision-making in the treatment of glioma. We demonstrate a technological solution to improve the accuracy and reliability of IDH mutation detection by combining MRI-based prediction and a CRISPR-based automatic integrated gene detection system (AIGS). A model was constructed to predict the IDH mutation status using whole slices in MRI scans with a Transformer neural network, and the predictive model achieved accuracies of 0.93, 0.87, and 0.84 using the internal and two external test sets, respectively. Additionally, CRISPR/Cas12a-based AIGS was constructed, and AIGS achieved 100% diagnostic accuracy in terms of IDH detection using both frozen tissue and FFPE samples in one hour. Moreover, the feature attribution of our predictive model was assessed using GradCAM, and the highest correlations with tumor cell percentages in enhancing and IDH-wildtype gliomas were found to have GradCAM importance (0.65 and 0.5, respectively). This MRI-based predictive model could, therefore, guide biopsy for tumor-enriched, which would ensure the veracity and stability of the rapid detection results. The combination of our predictive model and AIGS improved the early determination of IDH mutation status in glioma patients. This combined system of MRI-based prediction and CRISPR/Cas12a-based detection can be used to guide biopsy, resection, and radiation for glioma patients to improve patient outcomes.
Collapse
Affiliation(s)
- Donghu Yu
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qisheng Zhong
- Department of Neurosurgery, 960 Hospital of PLA, Jinan, Shandong, China
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, China
| | - Zhebin Feng
- Department of Neurosurgery, PLA General Hospital, Beijing, China
| | - Feng Tang
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shiyu Feng
- Department of Neurosurgery, PLA General Hospital, Beijing, China
| | - Yuxiang Cai
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yutong Gao
- Department of Prosthodontics, Wuhan University Hospital of Stomatology, Wuhan, China
| | - Tian Lan
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mingjun Li
- Department of Radiology, Liaocheng People's Hospital, Liaocheng, China
| | - Fuhua Yu
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, China
| | - Zefen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, China.
| | - Xu Gao
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China.
| | - Zhiqiang Li
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
3
|
Lost J, Ashraf N, Jekel L, von Reppert M, Tillmanns N, Willms K, Merkaj S, Petersen GC, Avesta A, Ramakrishnan D, Omuro A, Nabavizadeh A, Bakas S, Bousabarah K, Lin M, Aneja S, Sabel M, Aboian M. Enhancing clinical decision-making: An externally validated machine learning model for predicting isocitrate dehydrogenase mutation in gliomas using radiomics from presurgical magnetic resonance imaging. Neurooncol Adv 2024; 6:vdae157. [PMID: 39659829 PMCID: PMC11630777 DOI: 10.1093/noajnl/vdae157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
Background Glioma, the most prevalent primary brain tumor, poses challenges in prognosis, particularly in the high-grade subclass, despite advanced treatments. The recent shift in tumor classification underscores the crucial role of isocitrate dehydrogenase (IDH) mutation status in the clinical care of glioma patients. However, conventional methods for determining IDH status, including biopsy, have limitations. Exploring the use of machine learning (ML) on magnetic resonance imaging to predict IDH mutation status shows promise but encounters challenges in generalizability and translation into clinical practice because most studies either use single institution or homogeneous datasets for model training and validation. Our study aims to bridge this gap by using multi-institution data for model validation. Methods This retrospective study utilizes data from large, annotated datasets for internal (377 cases from Yale New Haven Hospitals) and external validation (207 cases from facilities outside Yale New Haven Health). The 6-step research process includes image acquisition, semi-automated tumor segmentation, feature extraction, model building with feature selection, internal validation, and external validation. An extreme gradient boosting ML model predicted the IDH mutation status, confirmed by immunohistochemistry. Results The ML model demonstrated high performance, with an Area under the Curve (AUC), Accuracy, Sensitivity, and Specificity in internal validation of 0.862, 0.865, 0.885, and 0.713, and external validation of 0.835, 0.851, 0.850, and 0.847. Conclusions The ML model, built on a heterogeneous dataset, provided robust results in external validation for the prediction task, emphasizing its potential clinical utility. Future research should explore expanding its applicability and validation in diverse global healthcare settings.
Collapse
Affiliation(s)
- Jan Lost
- Department of Neurosurgery, Heinrich-Heine University, Dusseldorf, Germany
| | - Nader Ashraf
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Leon Jekel
- DKFZ Division of Translational Neurooncology at the WTZ, German Cancer Consortium, DKTK Partner Site, University Hospital Essen, Essen, Germany
| | | | - Niklas Tillmanns
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Dusseldorf, Germany
| | | | | | | | - Arman Avesta
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Divya Ramakrishnan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Antonio Omuro
- Department of Neurology and Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ali Nabavizadeh
- Department of Radiology, Perelman School of Medicine, Hospital of University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Spyridon Bakas
- Division of Computational Pathology, Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - MingDe Lin
- Visage Imaging, Inc., San Diego, California, USA
| | - Sanjay Aneja
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Mariam Aboian
- Department of Radiology, Children’s Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Das A, Ding S, Liu R, Huang C. Quantifying the Growth of Glioblastoma Tumors Using Multimodal MRI Brain Images. Cancers (Basel) 2023; 15:3614. [PMID: 37509277 PMCID: PMC10377296 DOI: 10.3390/cancers15143614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Predicting the eventual volume of tumor cells, that might proliferate from a given tumor, can help in cancer early detection and medical procedure planning to prevent their migration to other organs. In this work, a new statistical framework is proposed using Bayesian techniques for detecting the eventual volume of cells expected to proliferate from a glioblastoma (GBM) tumor. Specifically, the tumor region was first extracted using a parallel image segmentation algorithm. Once the tumor region was determined, we were interested in the number of cells that could proliferate from this tumor until its survival time. For this, we constructed the posterior distribution of the tumor cell numbers based on the proposed likelihood function and a certain prior volume. Furthermore, we extended the detection model and conducted a Bayesian regression analysis by incorporating radiomic features to discover those non-tumor cells that remained undetected. The main focus of the study was to develop a time-independent prediction model that could reliably predict the ultimate volume a malignant tumor of the fourth-grade severity could attain and which could also determine if the incorporation of the radiomic properties of the tumor enhanced the chances of no malignant cells remaining undetected.
Collapse
Affiliation(s)
- Anisha Das
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| | - Shengxian Ding
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| | - Rongjie Liu
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| | - Chao Huang
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
5
|
Capobianco E, Dominietto M. Assessment of brain cancer atlas maps with multimodal imaging features. J Transl Med 2023; 21:385. [PMID: 37308956 PMCID: PMC10262565 DOI: 10.1186/s12967-023-04222-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/22/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Glioblastoma Multiforme (GBM) is a fast-growing and highly aggressive brain tumor that invades the nearby brain tissue and presents secondary nodular lesions across the whole brain but generally does not spread to distant organs. Without treatment, GBM can result in death in about 6 months. The challenges are known to depend on multiple factors: brain localization, resistance to conventional therapy, disrupted tumor blood supply inhibiting effective drug delivery, complications from peritumoral edema, intracranial hypertension, seizures, and neurotoxicity. MAIN TEXT Imaging techniques are routinely used to obtain accurate detections of lesions that localize brain tumors. Especially magnetic resonance imaging (MRI) delivers multimodal images both before and after the administration of contrast, which results in displaying enhancement and describing physiological features as hemodynamic processes. This review considers one possible extension of the use of radiomics in GBM studies, one that recalibrates the analysis of targeted segmentations to the whole organ scale. After identifying critical areas of research, the focus is on illustrating the potential utility of an integrated approach with multimodal imaging, radiomic data processing and brain atlases as the main components. The templates associated with the outcome of straightforward analyses represent promising inference tools able to spatio-temporally inform on the GBM evolution while being generalizable also to other cancers. CONCLUSIONS The focus on novel inference strategies applicable to complex cancer systems and based on building radiomic models from multimodal imaging data can be well supported by machine learning and other computational tools potentially able to translate suitably processed information into more accurate patient stratifications and evaluations of treatment efficacy.
Collapse
Affiliation(s)
- Enrico Capobianco
- The Jackson Laboratory, 10 Discovery Drive, Farmington, CT, 06032, USA.
| | - Marco Dominietto
- Paul Scherrer Institute (PSI), Forschungsstrasse 111, 5232, Villigen, Switzerland
- Gate To Brain SA, Via Livio 7, 6830, Chiasso, Switzerland
| |
Collapse
|
6
|
Ruffle JK, Mohinta S, Gray R, Hyare H, Nachev P. Brain tumour segmentation with incomplete imaging data. Brain Commun 2023; 5:fcad118. [PMID: 37124946 PMCID: PMC10144694 DOI: 10.1093/braincomms/fcad118] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/22/2023] [Accepted: 04/08/2023] [Indexed: 05/02/2023] Open
Abstract
Progress in neuro-oncology is increasingly recognized to be obstructed by the marked heterogeneity-genetic, pathological, and clinical-of brain tumours. If the treatment susceptibilities and outcomes of individual patients differ widely, determined by the interactions of many multimodal characteristics, then large-scale, fully-inclusive, richly phenotyped data-including imaging-will be needed to predict them at the individual level. Such data can realistically be acquired only in the routine clinical stream, where its quality is inevitably degraded by the constraints of real-world clinical care. Although contemporary machine learning could theoretically provide a solution to this task, especially in the domain of imaging, its ability to cope with realistic, incomplete, low-quality data is yet to be determined. In the largest and most comprehensive study of its kind, applying state-of-the-art brain tumour segmentation models to large scale, multi-site MRI data of 1251 individuals, here we quantify the comparative fidelity of automated segmentation models drawn from MR data replicating the various levels of completeness observed in real life. We demonstrate that models trained on incomplete data can segment lesions very well, often equivalently to those trained on the full completement of images, exhibiting Dice coefficients of 0.907 (single sequence) to 0.945 (complete set) for whole tumours and 0.701 (single sequence) to 0.891 (complete set) for component tissue types. This finding opens the door both to the application of segmentation models to large-scale historical data, for the purpose of building treatment and outcome predictive models, and their application to real-world clinical care. We further ascertain that segmentation models can accurately detect enhancing tumour in the absence of contrast-enhancing imaging, quantifying the burden of enhancing tumour with an R 2 > 0.97, varying negligibly with lesion morphology. Such models can quantify enhancing tumour without the administration of intravenous contrast, inviting a revision of the notion of tumour enhancement if the same information can be extracted without contrast-enhanced imaging. Our analysis includes validation on a heterogeneous, real-world 50 patient sample of brain tumour imaging acquired over the last 15 years at our tertiary centre, demonstrating maintained accuracy even on non-isotropic MRI acquisitions, or even on complex post-operative imaging with tumour recurrence. This work substantially extends the translational opportunity for quantitative analysis to clinical situations where the full complement of sequences is not available and potentially enables the characterization of contrast-enhanced regions where contrast administration is infeasible or undesirable.
Collapse
Affiliation(s)
- James K Ruffle
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Samia Mohinta
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Robert Gray
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Harpreet Hyare
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Parashkev Nachev
- UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
7
|
Beyond Imaging and Genetic Signature in Glioblastoma: Radiogenomic Holistic Approach in Neuro-Oncology. Biomedicines 2022; 10:biomedicines10123205. [PMID: 36551961 PMCID: PMC9775324 DOI: 10.3390/biomedicines10123205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is a malignant brain tumor exhibiting rapid and infiltrative growth, with less than 10% of patients surviving over 5 years, despite aggressive and multimodal treatments. The poor prognosis and the lack of effective pharmacological treatments are imputable to a remarkable histological and molecular heterogeneity of GBM, which has led, to date, to the failure of precision oncology and targeted therapies. Identification of molecular biomarkers is a paradigm for comprehensive and tailored treatments; nevertheless, biopsy sampling has proved to be invasive and limited. Radiogenomics is an emerging translational field of research aiming to study the correlation between radiographic signature and underlying gene expression. Although a research field still under development, not yet incorporated into routine clinical practice, it promises to be a useful non-invasive tool for future personalized/adaptive neuro-oncology. This review provides an up-to-date summary of the recent advancements in the use of magnetic resonance imaging (MRI) radiogenomics for the assessment of molecular markers of interest in GBM regarding prognosis and response to treatments, for monitoring recurrence, also providing insights into the potential efficacy of such an approach for survival prognostication. Despite a high sensitivity and specificity in almost all studies, accuracy, reproducibility and clinical value of radiomic features are the Achilles heel of this newborn tool. Looking into the future, investigators' efforts should be directed towards standardization and a disciplined approach to data collection, algorithms, and statistical analysis.
Collapse
|
8
|
Zhang H, Huang Y, Yang E, Gao X, Zou P, Sun J, Tian Z, Bao M, Liao D, Ge J, Yang Q, Li X, Zhang Z, Luo P, Jiang X. Identification of a Fibroblast-Related Prognostic Model in Glioma Based on Bioinformatics Methods. Biomolecules 2022; 12:biom12111598. [PMID: 36358948 PMCID: PMC9687522 DOI: 10.3390/biom12111598] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Glioma is the most common primary tumor of the central nervous system with a high lethality rate. This study aims to mine fibroblast-related genes with prognostic value and construct a corresponding prognostic model. Methods: A glioma-related TCGA (The Cancer Genome Atlas) cohort and a CGGA (Chinese Glioma Genome Atlas) cohort were incorporated into this study. Variance expression profiling was executed via the “limma” R package. The “clusterProfiler” R package was applied to perform a GO (Gene Ontology) analysis. The Kaplan–Meier (K–M) curve, LASSO regression analysis, and Cox analyses were implemented to determine the prognostic genes. A fibroblast-related risk model was created and affirmed by independent cohorts. We derived enriched pathways between the fibroblast-related high- and low-risk subgroups using gene set variation analysis (GSEA). The immune infiltration cell and the stromal cell were calculated using the microenvironment cell populations-counter (MCP-counter) method, and the immunotherapy response was assessed with the SubMap algorithm. The chemotherapy sensitivity was estimated using the “pRRophetic” R package. Results: A total of 93 differentially expressed fibroblast-related genes (DEFRGs) were uncovered in glioma. Seven prognostic genes were filtered out to create a fibroblast-related gene signature in the TCGA-glioma cohort training set. We then affirmed the fibroblast-related risk model via TCGA-glioma cohort and CGGA-glioma cohort testing sets. The Cox regression analysis proved that the fibroblast-related risk score was an independent prognostic predictor in prediction of the overall survival of glioma patients. The fibroblast-related gene signature revealed by the GSEA was applicable to the immune-relevant pathways. The MCP-counter algorithm results pointed to significant distinctions in the tumor microenvironment between fibroblast-related high- and low-risk subgroups. The SubMap analysis proved that the fibroblast-related risk score could predict the clinical sensitivity of immunotherapy. The chemotherapy sensitivity analysis indicated that low-risk patients were more sensitive to multiple chemotherapeutic drugs. Conclusion: Our study identified prognostic fibroblast-related genes and generated a novel risk signature that could evaluate the prognosis of glioma and offer a theoretical basis for clinical glioma therapy.
Collapse
Affiliation(s)
- Haofuzi Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Yutao Huang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Erwan Yang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Xiangyu Gao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Peng Zou
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Jidong Sun
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Zhicheng Tian
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Mingdong Bao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Dan Liao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Junmiao Ge
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Qiuzi Yang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Xin Li
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Zhuoyuan Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- Biochemistry and Molecular Biology, College of Life Science, Northwest University, Xi’an 710127, China
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- Correspondence: (P.L.); (X.J.)
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- Correspondence: (P.L.); (X.J.)
| |
Collapse
|
9
|
Kim S, Hoch MJ, Peng L, Somasundaram A, Chen Z, Weinberg BD. A brain tumor reporting and data system to optimize imaging surveillance and prognostication in high-grade gliomas. J Neuroimaging 2022; 32:1185-1192. [PMID: 36045502 DOI: 10.1111/jon.13044] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE High-grade glioma (HGG), including glioblastoma, is the most common primary brain neoplasm and has a dismal prognosis. After initial treatment, follow-up decisions are guided by longitudinal MRI performed at routine intervals. The Brain Tumor Reporting and Data System (BT-RADS) is a proposed structured reporting system for posttreatment brain MRIs. The purpose of this study is to determine the relationship between BT-RADS scores and overall survival in HGG patients. METHODS Chart review of grade 4 glioma patients who had an MRI at a single institution from November 2018 to November 2019 was performed. BT-RADS scores, tumor characteristics, and overall survival were recorded. Likelihood of improvement, stability, or worsening on the subsequent study was calculated for each score. Survival analysis was performed using Kaplan-Meier method, log-rank test, and a time-dependent cox model. Significance level of .05 was used. RESULTS The study identified 91 HGG patients who underwent a total of 538 MRIs. Mean age of patients was 57 years old. Score with the highest likelihood for worsening on the next follow-up was 3b. The risk of death was 53% higher with each incremental increase in BT-RADS scores (hazard ratio, 1.53; 95% confidence interval [CI], 1.07-2.19; p = .019). The risk of death was 167% higher in O-6-methylguanine-DNA-methyltransferase unmethylated tumors (hazard ratio, 2.67; 95% CI, 1.34-5.33; p = .005). CONCLUSIONS BT-RADS scores can be used as a reference guide to anticipate whether patients' subsequent MRI will be improved, stable, or worsened. The scoring system can also be used to predict clinical outcomes and prognosis.
Collapse
Affiliation(s)
- Sera Kim
- Department of Radiology, University of California, San Francisco, San Francisco, California, USA
| | - Michael J Hoch
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lingyi Peng
- Department of Statistics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aravind Somasundaram
- Department of Radiology and Imaging Sciences, Emory University Hospital, Atlanta, Georgia, USA
| | - Zhengjia Chen
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Brent D Weinberg
- Department of Radiology and Imaging Sciences, Emory University Hospital, Atlanta, Georgia, USA
| |
Collapse
|
10
|
Integrated MRI–Immune–Genomic Features Enclose a Risk Stratification Model in Patients Affected by Glioblastoma. Cancers (Basel) 2022; 14:cancers14133249. [PMID: 35805021 PMCID: PMC9265092 DOI: 10.3390/cancers14133249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 11/20/2022] Open
Abstract
Simple Summary Despite crucial scientific advances, Glioblastoma (GB) remains a fatal disease with limited therapeutic options and a lack of suitable biomarkers. The unveiled competence of the brain immune system together with the breakthrough advent of immunotherapy has shifted the present translational research on GB towards an immune-focused perspective. Several clinical trials targeting the immunosuppressive GB background are ongoing. So far, results are inconclusive, underpinning our partial understanding of the complex cancer-immune interplay in brain tumors. High throughput Magnetic Resonance (MR) imaging has shown the potential to decipher GB heterogeneity, including pathologic and genomic clues. However, whether distinct GB immune contextures can be deciphered at an imaging scale is still elusive, leaving unattained the non-invasive achievement of prognostic and predictive biomarkers. Along these lines, we integrated genetic, immunopathologic and imaging features in a series of GB patients. Our results suggest that multiparametric approaches might offer new efficient risk stratification models, opening the possibility to intercept the critical events implicated in the dismal prognosis of GB. Abstract Background: The aim of the present study was to dissect the clinical outcome of GB patients through the integration of molecular, immunophenotypic and MR imaging features. Methods: We enrolled 57 histologically proven and molecularly tested GB patients (5.3% IDH-1 mutant). Two-Dimensional Free ROI on the Biggest Enhancing Tumoral Diameter (TDFRBETD) acquired by MRI sequences were used to perform a manual evaluation of multiple quantitative variables, among which we selected: SD Fluid Attenuated Inversion Recovery (FLAIR), SD and mean Apparent Diffusion Coefficient (ADC). Characterization of the Tumor Immune Microenvironment (TIME) involved the immunohistochemical analysis of PD-L1, and number and distribution of CD3+, CD4+, CD8+ Tumor Infiltrating Lymphocytes (TILs) and CD163+ Tumor Associated Macrophages (TAMs), focusing on immune-vascular localization. Genetic, MR imaging and TIME descriptors were correlated with overall survival (OS). Results: MGMT methylation was associated with a significantly prolonged OS (median OS = 20 months), while no impact of p53 and EGFR status was apparent. GB cases with high mean ADC at MRI, indicative of low cellularity and soft consistency, exhibited increased OS (median OS = 24 months). PD-L1 and the overall number of TILs and CD163+TAMs had a marginal impact on patient outcome. Conversely, the density of vascular-associated (V) CD4+ lymphocytes emerged as the most significant prognostic factor (median OS = 23 months in V-CD4high vs. 13 months in V-CD4low, p = 0.015). High V-CD4+TILs also characterized TIME of MGMTmeth GB, while p53mut appeared to condition a desert immune background. When individual genetic (MGMTunmeth), MR imaging (mean ADClow) and TIME (V-CD4+TILslow) negative predictors were combined, median OS was 21 months (95% CI, 0–47.37) in patients displaying 0–1 risk factor and 13 months (95% CI 7.22–19.22) in the presence of 2–3 risk factors (p = 0.010, HR = 3.39, 95% CI 1.26–9.09). Conclusion: Interlacing MRI–immune–genetic features may provide highly significant risk-stratification models in GB patients.
Collapse
|
11
|
Bernstock JD, Gary SE, Klinger N, Valdes PA, Ibn Essayed W, Olsen HE, Chagoya G, Elsayed G, Yamashita D, Schuss P, Gessler FA, Peruzzi PP, Bag A, Friedman GK. Standard clinical approaches and emerging modalities for glioblastoma imaging. Neurooncol Adv 2022; 4:vdac080. [PMID: 35821676 PMCID: PMC9268747 DOI: 10.1093/noajnl/vdac080] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary adult intracranial malignancy and carries a dismal prognosis despite an aggressive multimodal treatment regimen that consists of surgical resection, radiation, and adjuvant chemotherapy. Radiographic evaluation, largely informed by magnetic resonance imaging (MRI), is a critical component of initial diagnosis, surgical planning, and post-treatment monitoring. However, conventional MRI does not provide information regarding tumor microvasculature, necrosis, or neoangiogenesis. In addition, traditional MRI imaging can be further confounded by treatment-related effects such as pseudoprogression, radiation necrosis, and/or pseudoresponse(s) that preclude clinicians from making fully informed decisions when structuring a therapeutic approach. A myriad of novel imaging modalities have been developed to address these deficits. Herein, we provide a clinically oriented review of standard techniques for imaging GBM and highlight emerging technologies utilized in disease characterization and therapeutic development.
Collapse
Affiliation(s)
- Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Sam E Gary
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Neil Klinger
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Pablo A Valdes
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Walid Ibn Essayed
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Hannah E Olsen
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Gustavo Chagoya
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Galal Elsayed
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Daisuke Yamashita
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Patrick Schuss
- Department of Neurosurgery, Unfallkrankenhaus Berlin , Berlin, Germany
| | | | - Pier Paolo Peruzzi
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Asim Bag
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital , Memphis, TN USA
| | - Gregory K Friedman
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham , Birmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham , AL, USA
| |
Collapse
|
12
|
Merkaj S, Bahar RC, Zeevi T, Lin M, Ikuta I, Bousabarah K, Cassinelli Petersen GI, Staib L, Payabvash S, Mongan JT, Cha S, Aboian MS. Machine Learning Tools for Image-Based Glioma Grading and the Quality of Their Reporting: Challenges and Opportunities. Cancers (Basel) 2022; 14:cancers14112623. [PMID: 35681603 PMCID: PMC9179416 DOI: 10.3390/cancers14112623] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 01/27/2023] Open
Abstract
Technological innovation has enabled the development of machine learning (ML) tools that aim to improve the practice of radiologists. In the last decade, ML applications to neuro-oncology have expanded significantly, with the pre-operative prediction of glioma grade using medical imaging as a specific area of interest. We introduce the subject of ML models for glioma grade prediction by remarking upon the models reported in the literature as well as by describing their characteristic developmental workflow and widely used classifier algorithms. The challenges facing these models-including data sources, external validation, and glioma grade classification methods -are highlighted. We also discuss the quality of how these models are reported, explore the present and future of reporting guidelines and risk of bias tools, and provide suggestions for the reporting of prospective works. Finally, this review offers insights into next steps that the field of ML glioma grade prediction can take to facilitate clinical implementation.
Collapse
Affiliation(s)
- Sara Merkaj
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, P.O. Box 208042, New Haven, CT 06520, USA; (S.M.); (R.C.B.); (T.Z.); (M.L.); (I.I.); (G.I.C.P.); (L.S.); (S.P.)
- Department of Neurosurgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Ryan C. Bahar
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, P.O. Box 208042, New Haven, CT 06520, USA; (S.M.); (R.C.B.); (T.Z.); (M.L.); (I.I.); (G.I.C.P.); (L.S.); (S.P.)
| | - Tal Zeevi
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, P.O. Box 208042, New Haven, CT 06520, USA; (S.M.); (R.C.B.); (T.Z.); (M.L.); (I.I.); (G.I.C.P.); (L.S.); (S.P.)
| | - MingDe Lin
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, P.O. Box 208042, New Haven, CT 06520, USA; (S.M.); (R.C.B.); (T.Z.); (M.L.); (I.I.); (G.I.C.P.); (L.S.); (S.P.)
- Visage Imaging, Inc., 12625 High Bluff Dr, Suite 205, San Diego, CA 92130, USA
| | - Ichiro Ikuta
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, P.O. Box 208042, New Haven, CT 06520, USA; (S.M.); (R.C.B.); (T.Z.); (M.L.); (I.I.); (G.I.C.P.); (L.S.); (S.P.)
| | | | - Gabriel I. Cassinelli Petersen
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, P.O. Box 208042, New Haven, CT 06520, USA; (S.M.); (R.C.B.); (T.Z.); (M.L.); (I.I.); (G.I.C.P.); (L.S.); (S.P.)
| | - Lawrence Staib
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, P.O. Box 208042, New Haven, CT 06520, USA; (S.M.); (R.C.B.); (T.Z.); (M.L.); (I.I.); (G.I.C.P.); (L.S.); (S.P.)
| | - Seyedmehdi Payabvash
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, P.O. Box 208042, New Haven, CT 06520, USA; (S.M.); (R.C.B.); (T.Z.); (M.L.); (I.I.); (G.I.C.P.); (L.S.); (S.P.)
| | - John T. Mongan
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 505 Parnassus Ave., San Francisco, CA 94143, USA; (J.T.M.); (S.C.)
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 505 Parnassus Ave., San Francisco, CA 94143, USA; (J.T.M.); (S.C.)
| | - Mariam S. Aboian
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, P.O. Box 208042, New Haven, CT 06520, USA; (S.M.); (R.C.B.); (T.Z.); (M.L.); (I.I.); (G.I.C.P.); (L.S.); (S.P.)
- Correspondence: ; Tel.: +650-285-7577
| |
Collapse
|
13
|
Bhaduri S, Lesbats C, Sharkey J, Kelly CL, Mukherjee S, Taylor A, Delikatny EJ, Kim SG, Poptani H. Assessing Tumour Haemodynamic Heterogeneity and Response to Choline Kinase Inhibition Using Clustered Dynamic Contrast Enhanced MRI Parameters in Rodent Models of Glioblastoma. Cancers (Basel) 2022; 14:cancers14051223. [PMID: 35267531 PMCID: PMC8909848 DOI: 10.3390/cancers14051223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 12/04/2022] Open
Abstract
To investigate the utility of DCE-MRI derived pharmacokinetic parameters in evaluating tumour haemodynamic heterogeneity and treatment response in rodent models of glioblastoma, imaging was performed on intracranial F98 and GL261 glioblastoma bearing rodents. Clustering of the DCE-MRI-based parametric maps (using Tofts, extended Tofts, shutter speed, two-compartment, and the second generation shutter speed models) was performed using a hierarchical clustering algorithm, resulting in areas with poor fit (reflecting necrosis), low, medium, and high valued pixels representing parameters Ktrans, ve, Kep, vp, τi and Fp. There was a significant increase in the number of necrotic pixels with increasing tumour volume and a significant correlation between ve and tumour volume suggesting increased extracellular volume in larger tumours. In terms of therapeutic response in F98 rat GBMs, a sustained decrease in permeability and perfusion and a reduced cell density was observed during treatment with JAS239 based on Ktrans, Fp and ve as compared to control animals. No significant differences in these parameters were found for the GL261 tumour, indicating that this model may be less sensitive to JAS239 treatment regarding changes in vascular parameters. This study demonstrates that region-based clustered pharmacokinetic parameters derived from DCE-MRI may be useful in assessing tumour haemodynamic heterogeneity with the potential for assessing therapeutic response.
Collapse
Affiliation(s)
- Sourav Bhaduri
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Clémentine Lesbats
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - Jack Sharkey
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Claire Louise Kelly
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Soham Mukherjee
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Arthur Taylor
- Department of Molecular Physiology & Cell Signalling, University of Liverpool, Liverpool L69 3BX, UK;
| | - Edward J. Delikatny
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Sungheon G. Kim
- Department of Radiology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
- Correspondence:
| |
Collapse
|
14
|
Li M, Huang W, Chen H, Jiang H, Yang C, Shen S, Cui Y, Dong G, Ren X, Lin S. T2/FLAIR Abnormity Could be the Sign of Glioblastoma Dissemination. Front Neurol 2022; 13:819216. [PMID: 35185770 PMCID: PMC8849106 DOI: 10.3389/fneur.2022.819216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 01/03/2022] [Indexed: 12/13/2022] Open
Abstract
Purpose Newly emerged or constantly enlarged contrast-enhancing (CE) lesions were the necessary signs for the diagnosis of glioblastoma (GBM) progression. This study aimed to investigate whether the T2-weighted-Fluid-Attenuated Inversion Recovery (T2/FLAIR) abnormal transformation could predict and assess progression for GBMs, especially for tumor dissemination. Methods A consecutive cohort of 246 GBM patients with regular follow-up and sufficient radiological data was included in this study. The series of T2/FLAIR and T1CE images were retrospectively reviewed. The patients were separated into T2/FLAIR and T1CE discordant and accordant subgroups based on the initial progression images. Results A total of 170 qualified patients were finally analyzed. The incidence of discordant T2/FLAIR and T1CE images was 25.9% (44/170). The median time-span of T2/FLAIR indicated tumor progression was 119.5 days (ranging from 57 days-unreached) prior to T1CE. Nearly half of patients (20/44, 45.5%) in the discordant subgroup suffered from tumor dissemination, substantially higher than accordant patients (23/126, 20.6%, p < 0.001). The median time to progression (TTP), post-progression survival (PPS), and overall survival (OS) were not statistically different (all p > 0.05) between discordant and accordant patients. Conclusions T2/FLAIR abnormity could be the sign of GBM progression, especially for newly emerged lesions disseminating from the primary cavity. Physicians should cast more attention on the dynamic change of T2/FLAIR images, which might be of great significance for progression assessment and subsequent clinical decision-making.
Collapse
Affiliation(s)
- Mingxiao Li
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Wei Huang
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Hongyan Chen
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Haihui Jiang
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, China
| | - Chuanwei Yang
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shaoping Shen
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yong Cui
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Gehong Dong
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaohui Ren
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neuroscience, Beijing Key Laboratory of Brain Tumor, Institute for Brain Disorders, Center of Brain Tumor, Beijing, China
- *Correspondence: Xiaohui Ren
| | - Song Lin
- Department of Neurosurgical Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neuroscience, Beijing Key Laboratory of Brain Tumor, Institute for Brain Disorders, Center of Brain Tumor, Beijing, China
- Song Lin
| |
Collapse
|
15
|
Predicting Glioblastoma Cellular Motility from In Vivo MRI with a Radiomics Based Regression Model. Cancers (Basel) 2022; 14:cancers14030578. [PMID: 35158845 PMCID: PMC8833801 DOI: 10.3390/cancers14030578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary A diagnosis of glioblastoma carries a uniformly dismal prognosis. Contributing to this is the near certain chance of aggressive tumor spread and recurrence following treatment. Tumor cell motility may provide one way to characterize the tendencies of glioblastomas to spread and recur. We sought to develop a non-invasive technique for assessing tumor cell motility using quantitative features derived from in vivo preoperative magnetic resonance imaging. Our regression model accurately predicted tumor cell motility in a cohort of participants with preoperative imaging who also had mean cellular motility calculated for their resected tumor cells from time-lapse videos. This work establishes the feasibility of non-invasively characterizing the kinetic properties of tumors and could be used to select patients for motility-targeting precision therapies. Abstract Characterizing the motile properties of glioblastoma tumor cells could provide a useful way to predict the spread of tumors and to tailor the therapeutic approach. Radiomics has emerged as a diagnostic tool in the classification of tumor grade, stage, and prognosis. The purpose of this work is to examine the potential of radiomics to predict the motility of glioblastoma cells. Tissue specimens were obtained from 31 patients undergoing surgical resection of glioblastoma. Mean tumor cell motility was calculated from time-lapse videos of specimen cells. Manual segmentation was used to define the border of the enhancing tumor T1-weighted MR images, and 107 radiomics features were extracted from the normalized image volumes. Model parameter coefficients were estimated using the adaptive lasso technique validated with leave-one-out cross validation (LOOCV) and permutation tests. The R-squared value for the predictive model was 0.60 with p-values for each individual parameter estimate less than 0.0001. Permutation test models trained with scrambled motility failed to produce a model that out-performed the model trained on the true data. The results of this work suggest that it is possible for a quantitative MRI feature-based regression model to non-invasively predict the cellular motility of glioblastomas.
Collapse
|
16
|
Fathi Kazerooni A, Bagley SJ, Akbari H, Saxena S, Bagheri S, Guo J, Chawla S, Nabavizadeh A, Mohan S, Bakas S, Davatzikos C, Nasrallah MP. Applications of Radiomics and Radiogenomics in High-Grade Gliomas in the Era of Precision Medicine. Cancers (Basel) 2021; 13:cancers13235921. [PMID: 34885031 PMCID: PMC8656630 DOI: 10.3390/cancers13235921] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Radiomics and radiogenomics offer new insight into high-grade glioma biology, as well as into glioma behavior in response to standard therapies. In this article, we provide neuro-oncology, neuropathology, and computational perspectives on the role of radiomics in providing more accurate diagnoses, prognostication, and surveillance of patients with high-grade glioma, and on the potential application of radiomics in clinical practice, with the overarching goal of advancing precision medicine for optimal patient care. Abstract Machine learning (ML) integrated with medical imaging has introduced new perspectives in precision diagnostics of high-grade gliomas, through radiomics and radiogenomics. This has raised hopes for characterizing noninvasive and in vivo biomarkers for prediction of patient survival, tumor recurrence, and genomics and therefore encouraging treatments tailored to individualized needs. Characterization of tumor infiltration based on pre-operative multi-parametric magnetic resonance imaging (MP-MRI) scans may allow prediction of the loci of future tumor recurrence and thereby aid in planning the course of treatment for the patients, such as optimizing the extent of resection and the dose and target area of radiation. Imaging signatures of tumor genomics can help in identifying the patients who benefit from certain targeted therapies. Specifying molecular properties of gliomas and prediction of their changes over time and with treatment would allow optimization of treatment. In this article, we provide neuro-oncology, neuropathology, and computational perspectives on the promise of radiomics and radiogenomics for allowing personalized treatments of patients with gliomas and discuss the challenges and limitations of these methods in multi-institutional clinical trials and suggestions to mitigate the issues and the future directions.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Stephen J. Bagley
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hamed Akbari
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Sanjay Saxena
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Sina Bagheri
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Jun Guo
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Ali Nabavizadeh
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Suyash Mohan
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA; (A.F.K.); (H.A.); (S.S.); (J.G.); (A.N.); (S.M.); (S.B.); (C.D.)
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.); (S.C.)
| | - MacLean P. Nasrallah
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence:
| |
Collapse
|
17
|
Pasquini L, Napolitano A, Lucignani M, Tagliente E, Dellepiane F, Rossi-Espagnet MC, Ritrovato M, Vidiri A, Villani V, Ranazzi G, Stoppacciaro A, Romano A, Di Napoli A, Bozzao A. AI and High-Grade Glioma for Diagnosis and Outcome Prediction: Do All Machine Learning Models Perform Equally Well? Front Oncol 2021; 11:601425. [PMID: 34888226 PMCID: PMC8649764 DOI: 10.3389/fonc.2021.601425] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Radiomic models outperform clinical data for outcome prediction in high-grade gliomas (HGG). However, lack of parameter standardization limits clinical applications. Many machine learning (ML) radiomic models employ single classifiers rather than ensemble learning, which is known to boost performance, and comparative analyses are lacking in the literature. We aimed to compare ML classifiers to predict clinically relevant tasks for HGG: overall survival (OS), isocitrate dehydrogenase (IDH) mutation, O-6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation, epidermal growth factor receptor vIII (EGFR) amplification, and Ki-67 expression, based on radiomic features from conventional and advanced magnetic resonance imaging (MRI). Our objective was to identify the best algorithm for each task. One hundred fifty-six adult patients with pathologic diagnosis of HGG were included. Three tumoral regions were manually segmented: contrast-enhancing tumor, necrosis, and non-enhancing tumor. Radiomic features were extracted with a custom version of Pyradiomics and selected through Boruta algorithm. A Grid Search algorithm was applied when computing ten times K-fold cross-validation (K=10) to get the highest mean and lowest spread of accuracy. Model performance was assessed as AUC-ROC curve mean values with 95% confidence intervals (CI). Extreme Gradient Boosting (xGB) obtained highest accuracy for OS (74,5%), Adaboost (AB) for IDH mutation (87.5%), MGMT methylation (70,8%), Ki-67 expression (86%), and EGFR amplification (81%). Ensemble classifiers showed the best performance across tasks. High-scoring radiomic features shed light on possible correlations between MRI and tumor histology.
Collapse
Affiliation(s)
- Luca Pasquini
- Neuroradiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| | - Antonio Napolitano
- Medical Physics Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Martina Lucignani
- Medical Physics Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Emanuela Tagliente
- Medical Physics Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Francesco Dellepiane
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| | - Maria Camilla Rossi-Espagnet
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Matteo Ritrovato
- Unit of Health Technology Assessment (HTA), Biomedical Technology Risk Manager, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Antonello Vidiri
- Radiology and Diagnostic Imaging Department, Regina Elena National Cancer Institute, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Veronica Villani
- Neuro-Oncology Unit, Regina Elena National Cancer Institute, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Giulio Ranazzi
- Department of Clinical and Molecular Medicine, Surgical Pathology Units, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| | - Antonella Stoppacciaro
- Department of Clinical and Molecular Medicine, Surgical Pathology Units, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| | - Andrea Romano
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| | - Alberto Di Napoli
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
- Radiology Department, Castelli Romani Hospital, Rome, Italy
| | - Alessandro Bozzao
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| |
Collapse
|
18
|
Xavier MA, Rezende F, Titze-de-Almeida R, Cornelissen B. BRCAness as a Biomarker of Susceptibility to PARP Inhibitors in Glioblastoma Multiforme. Biomolecules 2021; 11:1188. [PMID: 34439854 PMCID: PMC8394995 DOI: 10.3390/biom11081188] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain cancer. GBMs commonly acquire resistance to standard-of-care therapies. Among the novel means to sensitize GBM to DNA-damaging therapies, a promising strategy is to combine them with inhibitors of the DNA damage repair (DDR) machinery, such as inhibitors for poly(ADP-ribose) polymerase (PARP). PARP inhibitors (PARPis) have already shown efficacy and have received regulatory approval for breast, ovarian, prostate, and pancreatic cancer treatment. In these cancer types, after PARPi administration, patients carrying specific mutations in the breast cancer 1 (BRCA1) and 2 (BRCA2) suppressor genes have shown better response when compared to wild-type carriers. Mutated BRCA genes are infrequent in GBM tumors, but their cells can carry other genetic alterations that lead to the same phenotype collectively referred to as 'BRCAness'. The most promising biomarkers of BRCAness in GBM are related to isocitrate dehydrogenases 1 and 2 (IDH1/2), epidermal growth factor receptor (EGFR), phosphatase and tensin homolog (PTEN), MYC proto-oncogene, and estrogen receptors beta (ERβ). BRCAness status identified by accurate biomarkers can ultimately predict responsiveness to PARPi therapy, thereby allowing patient selection for personalized treatment. This review discusses potential biomarkers of BRCAness for a 'precision medicine' of GBM patients.
Collapse
Affiliation(s)
- Mary-Ann Xavier
- Central Institute of Sciences, Technology for Gene Therapy Laboratory, University of Brasília—UnB/FAV, Brasília 70910-900, Brazil; (F.R.); (R.T.-d.-A.)
| | - Fernando Rezende
- Central Institute of Sciences, Technology for Gene Therapy Laboratory, University of Brasília—UnB/FAV, Brasília 70910-900, Brazil; (F.R.); (R.T.-d.-A.)
| | - Ricardo Titze-de-Almeida
- Central Institute of Sciences, Technology for Gene Therapy Laboratory, University of Brasília—UnB/FAV, Brasília 70910-900, Brazil; (F.R.); (R.T.-d.-A.)
| | - Bart Cornelissen
- Department of Oncology, Radiobiology Research Institute, University of Oxford, Oxford OX3 7LJ, UK;
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, 9700 RB Groningen, The Netherlands
| |
Collapse
|
19
|
Assessing Versatile Machine Learning Models for Glioma Radiogenomic Studies across Hospitals. Cancers (Basel) 2021; 13:cancers13143611. [PMID: 34298824 PMCID: PMC8306149 DOI: 10.3390/cancers13143611] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Radiogenomics enables prediction of the status and prognosis of patients using non-invasively obtained imaging data. Current machine learning (ML) methods used in radiogenomics require huge datasets, which involve the handling of large heterogeneous datasets from multiple cohorts/hospitals. In this study, two different glioma datasets were used to test various ML and image pre-processing methods to confirm whether the models trained on one dataset are universally applicable to other datasets. Our result suggested that the ML method that yielded the highest accuracy in a single dataset was likely to be overfitted. We demonstrated that implementation of standardization and dimension reduction procedures prior to classification, enabled the development of ML methods that are less affected by the multiple cohort difference. We advocate using caution in interpreting the results of radiogenomic studies of the training and testing datasets that are small or mixed, with a view to implementing practical ML methods in radiogenomics. Abstract Radiogenomics use non-invasively obtained imaging data, such as magnetic resonance imaging (MRI), to predict critical biomarkers of patients. Developing an accurate machine learning (ML) technique for MRI requires data from hundreds of patients, which cannot be gathered from any single local hospital. Hence, a model universally applicable to multiple cohorts/hospitals is required. We applied various ML and image pre-processing procedures on a glioma dataset from The Cancer Image Archive (TCIA, n = 159). The models that showed a high level of accuracy in predicting glioblastoma or WHO Grade II and III glioma using the TCIA dataset, were then tested for the data from the National Cancer Center Hospital, Japan (NCC, n = 166) whether they could maintain similar levels of high accuracy. Results: we confirmed that our ML procedure achieved a level of accuracy (AUROC = 0.904) comparable to that shown previously by the deep-learning methods using TCIA. However, when we directly applied the model to the NCC dataset, its AUROC dropped to 0.383. Introduction of standardization and dimension reduction procedures before classification without re-training improved the prediction accuracy obtained using NCC (0.804) without a loss in prediction accuracy for the TCIA dataset. Furthermore, we confirmed the same tendency in a model for IDH1/2 mutation prediction with standardization and application of dimension reduction that was also applicable to multiple hospitals. Our results demonstrated that overfitting may occur when an ML method providing the highest accuracy in a small training dataset is used for different heterogeneous data sets, and suggested a promising process for developing an ML method applicable to multiple cohorts.
Collapse
|
20
|
Awan H, Balasubramaniam S, Odysseos A. A Voxel Model to Decipher the Role of Molecular Communication in the Growth of Glioblastoma Multiforme. IEEE Trans Nanobioscience 2021; 20:296-310. [PMID: 33830926 DOI: 10.1109/tnb.2021.3071922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Glioblastoma Multiforme (GBM), the most malignant human tumour, can be defined by the evolution of growing bio-nanomachine networks within an interplay between self-renewal (Grow) and invasion (Go) potential of mutually exclusive phenotypes of transmitter and receiver cells. Herein, we present a mathematical model for the growth of GBM tumour driven by molecule-mediated inter-cellular communication between two populations of evolutionary bio-nanomachines representing the Glioma Stem Cells (GSCs) and Glioma Cells (GCs). The contribution of each subpopulation to tumour growth is quantified by a voxel model representing the end to end inter-cellular communication models for GSCs and progressively evolving invasiveness levels of glioma cells within a network of diverse cell configurations. Mutual information, information propagation speed and the impact of cell numbers and phenotypes on the communication output and GBM growth are studied by using analysis from information theory. The numerical simulations show that the progression of GBM is directly related to higher mutual information and higher input information flow of molecules between the GSCs and GCs, resulting in an increased tumour growth rate. These fundamental findings contribute to deciphering the mechanisms of tumour growth and are expected to provide new knowledge towards the development of future bio-nanomachine-based therapeutic approaches for GBM.
Collapse
|
21
|
Identification of magnetic resonance imaging features for the prediction of molecular profiles of newly diagnosed glioblastoma. J Neurooncol 2021; 154:83-92. [PMID: 34191225 DOI: 10.1007/s11060-021-03801-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/25/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE We predicted molecular profiles in newly diagnosed glioblastoma patients using magnetic resonance (MR) imaging features and explored the associations between imaging features and major molecular alterations. METHODS This retrospective study included patients with newly diagnosed glioblastoma and available next-generation sequencing results. From preoperative MR imaging, Visually AcceSAble Rembrandt Images (VASARI) features, volumetric parameters, and apparent diffusion coefficient (ADC) values were obtained. First, univariate random forest was performed to identify gene abnormalities that could be predicted by imaging features with high accuracy and stability. Next, multivariate random forest was trained to predict the selected genes in the discovery cohort and was validated in the external cohort. Univariable logistic regression was performed to further explore the associations between imaging features and genes. RESULTS Univariate random forest identified nine genes predicted by imaging features, with high accuracy and stability. The multivariate random forest model showed excellent performance in predicting IDH and PTPN11 mutations in the discovery cohort, which were validated in the external validation cohorts (areas under the receiver operator characteristic curve [AUCs] of 0.855 for IDH and 0.88 for PTPN11). ATRX loss and EGFR mutation were predicted with AUCs of 0.753 and 0.739, respectively, whereas PTEN could not be reliably predicted. Based on univariable logistic regression analyses, IDH, ATRX, and TP53 were clustered according to their shared imaging features, whereas EGFR and CDKN2A/B were clustered in the opposite direction. CONCLUSIONS MR imaging features are related to specific molecular alterations and can be used to predict molecular profiles in patients with newly diagnosed glioblastoma.
Collapse
|
22
|
Min TL, Allen JW, Velazquez Vega JE, Neill SG, Weinberg BD. MRI Imaging Characteristics of Glioblastoma with Concurrent Gain of Chromosomes 19 and 20. ACTA ACUST UNITED AC 2021; 7:228-237. [PMID: 34199376 PMCID: PMC8293438 DOI: 10.3390/tomography7020021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/17/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is the most common and deadly primary brain tumor in adults. Some of the genetic variations identified thus far, such as IDH mutation and MGMT promotor methylation, have implications for survival and response to therapy. A recent analysis of long-term GBM survivors showed that concurrent gain of chromosomes 19 and 20 (19/20 co-gain) is a positive prognostic factor that is independent of IDH mutation status. In this study, we retrospectively identified 18 patients with 19/20 co-gain and compared their imaging features to a control cohort without 19/20 co-gain. Imaging features such as tumor location, size, pial invasion, and ependymal extension were examined manually. When compared without further genetic subclassification, both groups showed similar imaging features except for rates of pial invasion. When each group was subclassified by MGMT promotor methylation status however, the two groups showed different imaging features in a number of additional ways including tumor location, size, and ependymal extension. Our results indicate that different permutations of various genetic mutations that coexist in GBM may interact in unpredictable ways to affect imaging appearance, and that imaging prognostication may be better approached in the context of the global genomic profile rather than individual genetic alterations.
Collapse
Affiliation(s)
- Taejin L. Min
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Emory University Hospital, Suite D112, 1364 Clifton Road NE, Atlanta, GA 30322, USA; (T.L.M.); (J.W.A.)
| | - Jason W. Allen
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Emory University Hospital, Suite D112, 1364 Clifton Road NE, Atlanta, GA 30322, USA; (T.L.M.); (J.W.A.)
| | - Jose E. Velazquez Vega
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Emory University Hospital, Room H184, 1364 Clifton Road NE, Atlanta, GA 30322, USA; (J.E.V.V.); (S.G.N.)
| | - Stewart G. Neill
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Emory University Hospital, Room H184, 1364 Clifton Road NE, Atlanta, GA 30322, USA; (J.E.V.V.); (S.G.N.)
| | - Brent D. Weinberg
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Emory University Hospital, Suite D112, 1364 Clifton Road NE, Atlanta, GA 30322, USA; (T.L.M.); (J.W.A.)
- Correspondence:
| |
Collapse
|
23
|
Pasquini L, Napolitano A, Tagliente E, Dellepiane F, Lucignani M, Vidiri A, Ranazzi G, Stoppacciaro A, Moltoni G, Nicolai M, Romano A, Di Napoli A, Bozzao A. Deep Learning Can Differentiate IDH-Mutant from IDH-Wild GBM. J Pers Med 2021; 11:290. [PMID: 33918828 PMCID: PMC8069494 DOI: 10.3390/jpm11040290] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022] Open
Abstract
Isocitrate dehydrogenase (IDH) mutant and wildtype glioblastoma multiforme (GBM) often show overlapping features on magnetic resonance imaging (MRI), representing a diagnostic challenge. Deep learning showed promising results for IDH identification in mixed low/high grade glioma populations; however, a GBM-specific model is still lacking in the literature. Our aim was to develop a GBM-tailored deep-learning model for IDH prediction by applying convoluted neural networks (CNN) on multiparametric MRI. We selected 100 adult patients with pathologically demonstrated WHO grade IV gliomas and IDH testing. MRI sequences included: MPRAGE, T1, T2, FLAIR, rCBV and ADC. The model consisted of a 4-block 2D CNN, applied to each MRI sequence. Probability of IDH mutation was obtained from the last dense layer of a softmax activation function. Model performance was evaluated in the test cohort considering categorical cross-entropy loss (CCEL) and accuracy. Calculated performance was: rCBV (accuracy 83%, CCEL 0.64), T1 (accuracy 77%, CCEL 1.4), FLAIR (accuracy 77%, CCEL 1.98), T2 (accuracy 67%, CCEL 2.41), MPRAGE (accuracy 66%, CCEL 2.55). Lower performance was achieved on ADC maps. We present a GBM-specific deep-learning model for IDH mutation prediction, with a maximal accuracy of 83% on rCBV maps. Highest predictivity achieved on perfusion images possibly reflects the known link between IDH and neoangiogenesis through the hypoxia inducible factor.
Collapse
Affiliation(s)
- Luca Pasquini
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
- Neuroradiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Antonio Napolitano
- Medical Physics Department, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy; (E.T.); (M.L.)
| | - Emanuela Tagliente
- Medical Physics Department, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy; (E.T.); (M.L.)
| | - Francesco Dellepiane
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| | - Martina Lucignani
- Medical Physics Department, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy; (E.T.); (M.L.)
| | - Antonello Vidiri
- Radiology and Diagnostic Imaging Department, Regina Elena National Cancer Institute, IRCCS, Via Elio Chianesi 53, 00144 Rome, Italy;
| | - Giulio Ranazzi
- Surgical Pathology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (G.R.); (A.S.)
| | - Antonella Stoppacciaro
- Surgical Pathology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (G.R.); (A.S.)
| | - Giulia Moltoni
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| | - Matteo Nicolai
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| | - Andrea Romano
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| | - Alberto Di Napoli
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| | - Alessandro Bozzao
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| |
Collapse
|
24
|
Lin N, Wu L, Xu X, Wu Q, Wang Y, Shen H, Song Y, Wang H, Zhu Z, Kang D, Yang C. Aptamer Generated by Cell-SELEX for Specific Targeting of Human Glioma Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9306-9315. [PMID: 33030015 DOI: 10.1021/acsami.0c11878] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The most prevalent primary brain tumors are gliomas, which start in the glial cells. Although there have been significant technological advances in surgery and radio-chemotherapy, the prognosis and survival of patients with malignant gliomas remain poor. For routine diagnosis of glioma, computed tomography and magnetic resonance imaging primarily depend on anatomical changes and fail to detect the cellular changes that occur early in the development of malignant gliomas. Therefore, it is urgent to find effective molecular diagnostic tools to detect early stages of malignant gliomas. Currently, cell-based Systematic Evolution of Ligands by EXponential enrichment (cell-SELEX) technology is one effective tool to obtain DNA or RNA aptamers capable of differentiating the molecular signatures among different types of cell lines. Using cell-SELEX, we generated and characterized an aptamer, termed S6-1b, that can distinguish the molecular differences between glioma cell line SHG44 and human astrocytes. Under the conditions of 4 and 37 °C, respectively, the dissociation constants of aptamer-cell interaction were both measured in the low nanomolar range. The aptamer S6-1b also exhibited excellent selectivity, making it suitable for use in a complex biological environment. Furthermore, the aptamer can effectively target glioma cells for in vivo fluorescence imaging of tumors. The target type of aptamer S6-1b was identified as a cell membrane protein. Our work indicates that aptamer S6-1b has diagnostic and therapeutic potential to specifically deliver imaging or therapeutic agents to malignant gliomas.
Collapse
Affiliation(s)
- Ningqin Lin
- Department of Neurosurgery, Department of Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Liang Wu
- Department of Neurosurgery, Department of Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Xing Xu
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Qiaoyi Wu
- Department of Neurosurgery, Department of Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Yuzhe Wang
- Department of Neurosurgery, Department of Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Haicong Shen
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yanling Song
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Hongyao Wang
- Department of Neurosurgery, Department of Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Zhi Zhu
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Dezhi Kang
- Department of Neurosurgery, Department of Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Chaoyong Yang
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| |
Collapse
|
25
|
Shi C, Zhou Z, Lin H, Gao J. Imaging Beyond Seeing: Early Prognosis of Cancer Treatment. SMALL METHODS 2021; 5:e2001025. [PMID: 34927817 DOI: 10.1002/smtd.202001025] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/24/2020] [Indexed: 06/14/2023]
Abstract
Assessing cancer response to therapeutic interventions has been realized as an important course to early predict curative efficacy and treatment outcomes due to tumor heterogeneity. Compared to the traditional invasive tissue biopsy method, molecular imaging techniques have fundamentally revolutionized the ability to evaluate cancer response in a spatiotemporal manner. The past few years has witnessed a paradigm shift on the efforts from manufacturing functional molecular imaging probes for seeing a tumor to a vantage stage of interpreting the tumor response during different treatments. This review is to stand by the current development of advanced imaging technologies aiming to predict the treatment response in cancer therapy. Special interest is placed on the systems that are able to provide rapid and noninvasive assessment of pharmacokinetic drug fates (e.g., drug distribution, release, and activation) and tumor microenvironment heterogeneity (e.g., tumor cells, macrophages, dendritic cells (DCs), T cells, and inflammatory cells). The current status, practical significance, and future challenges of the emerging artificial intelligence (AI) technology and machine learning in the applications of medical imaging fields is overviewed. Ultimately, the authors hope that this review is timely to spur research interest in molecular imaging and precision medicine.
Collapse
Affiliation(s)
- Changrong Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Zijian Zhou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Hongyu Lin
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The Key Laboratory for Chemical Biology of Fujian Province and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Jinhao Gao
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The Key Laboratory for Chemical Biology of Fujian Province and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
26
|
Uddin MS, Mamun AA, Alghamdi BS, Tewari D, Jeandet P, Sarwar MS, Ashraf GM. Epigenetics of glioblastoma multiforme: From molecular mechanisms to therapeutic approaches. Semin Cancer Biol 2020; 83:100-120. [PMID: 33370605 DOI: 10.1016/j.semcancer.2020.12.015] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common form of brain cancer and one of the most aggressive cancers found in humans. Most of the signs and symptoms of GBM can be mild and slowly aggravated, although other symptoms might demonstrate it as an acute ailment. However, the precise mechanisms of the development of GBM remain unknown. Due to the improvement of molecular pathology, current researches have reported that glioma progression is strongly connected with different types of epigenetic phenomena, such as histone modifications, DNA methylation, chromatin remodeling, and aberrant microRNA. Furthermore, the genes and the proteins that control these alterations have become novel targets for treating glioma because of the reversibility of epigenetic modifications. In some cases, gene mutations including P16, TP53, and EGFR, have been observed in GBM. In contrast, monosomies, including removals of chromosome 10, particularly q23 and q25-26, are considered the standard markers for determining the development and aggressiveness of GBM. Recently, amid the epigenetic therapies, histone deacetylase inhibitors (HDACIs) and DNA methyltransferase inhibitors have been used for treating tumors, either single or combined. Specifically, HDACIs are served as a good choice and deliver a novel pathway to treat GBM. In this review, we focus on the epigenetics of GBM and the consequence of its mutations. We also highlight various treatment approaches, namely gene editing, epigenetic drugs, and microRNAs to combat GBM.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong Special Administrative Region
| | - Badrah S Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687, Reims Cedex 2, France
| | - Md Shahid Sarwar
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
27
|
Clement P, Booth T, Borovečki F, Emblem KE, Figueiredo P, Hirschler L, Jančálek R, Keil VC, Maumet C, Özsunar Y, Pernet C, Petr J, Pinto J, Smits M, Warnert EAH. GliMR: Cross-Border Collaborations to Promote Advanced MRI Biomarkers for Glioma. J Med Biol Eng 2020; 41:115-125. [PMID: 33293909 PMCID: PMC7712600 DOI: 10.1007/s40846-020-00582-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/04/2020] [Indexed: 01/01/2023]
Abstract
Purpose There is an annual incidence of 50,000 glioma cases in Europe. The optimal treatment strategy is highly personalised, depending on tumour type, grade, spatial localization, and the degree of tissue infiltration. In research settings, advanced magnetic resonance imaging (MRI) has shown great promise as a tool to inform personalised treatment decisions. However, the use of advanced MRI in clinical practice remains scarce due to the downstream effects of siloed glioma imaging research with limited representation of MRI specialists in established consortia; and the associated lack of available tools and expertise in clinical settings. These shortcomings delay the translation of scientific breakthroughs into novel treatment strategy. As a response we have developed the network “Glioma MR Imaging 2.0” (GliMR) which we present in this article. Methods GliMR aims to build a pan-European and multidisciplinary network of experts and accelerate the use of advanced MRI in glioma beyond the current “state-of-the-art” in glioma imaging. The Action Glioma MR Imaging 2.0 (GliMR) was granted funding by the European Cooperation in Science and Technology (COST) in June 2019. Results GliMR’s first grant period ran from September 2019 to April 2020, during which several meetings were held and projects were initiated, such as reviewing the current knowledge on advanced MRI; developing a General Data Protection Regulation (GDPR) compliant consent form; and setting up the website. Conclusion The Action overcomes the pre-existing limitations of glioma research and is funded until September 2023. New members will be accepted during its entire duration.
Collapse
Affiliation(s)
- Patricia Clement
- Ghent Institute for Metabolic and Functional Imaging (GIfMI), Ghent University, Ghent, Belgium
| | - Thomas Booth
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH UK.,Department of Neuroradiology, King's College Hospital NHS Foundation Trust, London, SE5 9RS UK
| | - Fran Borovečki
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Kyrre E Emblem
- Division of Radiology and Nuclear Medicine, Department of Diagnostic Physics, Oslo University Hospital, Oslo, Norway
| | - Patrícia Figueiredo
- Institute for Systems and Robotics - Lisboa and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Lydiane Hirschler
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | - Radim Jančálek
- Department of Neurosurgery, St. Anne's University Hospital and Medical Faculty, Masaryk University, Brno, Czech Republic
| | - Vera C Keil
- Department of Radiology, Amsterdam University Medical Center, VUmc, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Yelda Özsunar
- Department of Radiology, Faculty of Medicine, Adnan Menderes University, Aydın, Turkey
| | - Cyril Pernet
- Centre for Clinical Brain Sciences & Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Jan Petr
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Joana Pinto
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Marion Smits
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Esther A H Warnert
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
28
|
|
29
|
Fu X, Wu C, Han N, Liu N, Han S, Liu X, Li S, Yan C. Depressive and anxiety disorders worsen the prognosis of glioblastoma. Aging (Albany NY) 2020; 12:20095-20110. [PMID: 33113511 PMCID: PMC7655183 DOI: 10.18632/aging.103593] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 05/25/2020] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme (GBM) is one of the most malignant tumors. Depressive and anxiety disorders may co-exist with GBM. We investigated whether depression and anxiety influenced the outcomes of GBM. The Patient Health Questionnaire 9-item (PHQ-9) and Generalized Anxiety Disorder 7-item (GAD-7) scales were used to investigate the mental condition of GBM patients in our department, and the overall survival times of these patients were monitored. The scores on both scales were higher in GBM patients than in healthy controls. For each scale, GBM patients were divided into high- and low-score groups based on the average score. The prognosis was poorer for GBM patients in the high-score groups than for those in the low-score groups. Moreover, magnetic resonance imaging revealed that tumor necrosis was more prevalent among high-scored GBM patients. Cellular experiments were performed on primary GBM cells from patients with either high or low scores on both scales. Sphere formation, EdU and wound healing assays revealed greater proliferation and invasion capacities in GBM cells from patients with high scores on both scales. Western blotting assay revealed significantly different expression of epithelial and mesenchymal markers between the two groups. Thus, our analysis revealed a clinically important correlation between depression/anxiety and GBM prognosis.
Collapse
Affiliation(s)
- Xiaojun Fu
- Sanbo Brain Hospital, Capital Medical University, Beijing, PR China.,Capital Medical University, Beijing, PR China
| | - Chenxing Wu
- Sanbo Brain Hospital, Capital Medical University, Beijing, PR China
| | - Ning Han
- Department of Neurosurgery, Chinese PLA Tianjin Rehabilitation and Recuperation Center of Joint Service Support Force, Tianjin, PR China
| | - Ning Liu
- Sanbo Brain Hospital, Capital Medical University, Beijing, PR China
| | - Song Han
- Sanbo Brain Hospital, Capital Medical University, Beijing, PR China
| | - Xuebin Liu
- Zhong Guang Tianyi Bio Technology Co., Ltd., Beijing, China
| | - Shouwei Li
- Sanbo Brain Hospital, Capital Medical University, Beijing, PR China
| | - Changxiang Yan
- Sanbo Brain Hospital, Capital Medical University, Beijing, PR China
| |
Collapse
|
30
|
Song H, Fu X, Wu C, Li S. Aging-related tumor associated fibroblasts changes could worsen the prognosis of GBM patients. Cancer Cell Int 2020; 20:489. [PMID: 33061843 PMCID: PMC7545944 DOI: 10.1186/s12935-020-01571-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/23/2020] [Indexed: 01/18/2023] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most malignant tumor in human brain, with highly heterogeneity among different patients. Age could function as an incidence and prognosis risk factor for many tumors. Method A series of bioinformatic experiments were conducted to evaluate the differences of incidence, differential expressed genes, enriched pathways with the data from Surveillance, Epidemiology, and End Results (SEER) program, the cancer genome atlas (TCGA) and Chinese glioma genome atlas (CGGA) project. Results We discovered in our present study that distinct difference of incidence and prognosis of different aged GBM patients. By a series of bioinformatic method, we found that the tumor associated fibroblasts (TAFs) was the most crucial tumor microenvironment (TME) component that led to this phenomenon. Epithelial-mesenchymal transition (EMT) could be the mechanism by which TAFs regulate the progression of GBM. Conclusion We have proposed a close correlation between age and GBM incidence and prognosis, and propose the underlying mechanism behind this correlation by mining different databases, which laid the foundation for future research.
Collapse
Affiliation(s)
- Hongwang Song
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaojun Fu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093 China
| | - Chenxing Wu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093 China
| | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093 China
| |
Collapse
|
31
|
Abstract
This manuscript will review emerging applications of artificial intelligence, specifically deep learning, and its application to glioblastoma multiforme (GBM), the most common primary malignant brain tumor. Current deep learning approaches, commonly convolutional neural networks (CNNs), that take input data from MR images to grade gliomas (high grade from low grade) and predict overall survival will be shown. There will be more in-depth review of recent articles that have applied different CNNs to predict the genetics of glioma on pre-operative MR images, specifically 1p19q codeletion, MGMT promoter, and IDH mutations, which are important criteria for the diagnosis, treatment management, and prognostication of patients with GBM. Finally, there will be a brief mention of current challenges with DL techniques and their application to image analysis in GBM.
Collapse
|
32
|
Abstract
Magnetic resonance imaging (MRI) has been the cornerstone of imaging of brain tumors in the past 4 decades. Conventional MRI remains the workhorse for neuro-oncologic imaging, not only for basic information such as location, extent, and navigation but also able to provide information regarding proliferation and infiltration, angiogenesis, hemorrhage, and more. More sophisticated MRI sequences have extended the ability to assess and quantify these features; for example, permeability and perfusion acquisitions can assess blood-brain barrier disruption and angiogenesis, diffusion techniques can assess cellularity and infiltration, and spectroscopy can address metabolism. Techniques such as fMRI and diffusion fiber tracking can be helpful in diagnostic planning for resection and radiation therapy, and more sophisticated iterations of these techniques can extend our understanding of neurocognitive effects of these tumors and associated treatment responses and effects. More recently, MRI has been used to go beyond such morphological, physiological, and functional characteristics to assess the tumor microenvironment. The current review highlights multiple recent and emerging approaches in MRI to characterize the tumor microenvironment.
Collapse
|
33
|
Reuter G, Lommers E, Balteau E, Simon J, Phillips C, Scholtes F, Martin D, Lombard A, Maquet P. Multiparameter quantitative histological MRI values in high-grade gliomas: a potential biomarker of tumor progression. Neurooncol Pract 2020; 7:646-655. [PMID: 33304600 PMCID: PMC7716186 DOI: 10.1093/nop/npaa047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Conventional MRI poorly distinguishes brain parenchyma microscopically invaded by high-grade gliomas (HGGs) from the normal brain. By contrast, quantitative histological MRI (hMRI) measures brain microstructure in terms of physical MR parameters influenced by histochemical tissue composition. We aimed to determine the relationship between hMRI parameters in the area surrounding the surgical cavity and the presence of HGG recurrence. Methods Patients were scanned after surgery with an hMRI multiparameter protocol that allowed for estimations of longitudinal relaxation rate (R1) = 1/T1, effective transverse relaxation rate (R2)*=1/T2*, magnetization transfer saturation (MTsat), and proton density. The initial perioperative zone (IPZ) was segmented on the postoperative MRI. Once recurrence appeared on conventional MRI, the area of relapsing disease was delineated (extension zone, EZ). Conventional MRI showing recurrence and hMRI were coregistered, allowing for the extraction of parameters R1, R2*, MTsat, and PD in 3 areas: the overlap area between the IPZ and EZ (OZ), the peritumoral brain zone, PBZ (PBZ = IPZ - OZ), and the area of recurrence (RZ = EZ - OZ). Results Thirty-one patients with HGG who underwent gross-total resection were enrolled. MTsat and R1 were the most strongly associated with tumor progression. MTsat was significantly lower in the OZ and RZ, compared to PBZ. R1 was significantly lower in RZ compared to PBZ. PD was significantly higher in OZ compared to PBZ, and R2* was higher in OZ compared to PBZ or RZ. These changes were detected 4 to 120 weeks before recurrence recognition on conventional MRI. Conclusions HGG recurrence was associated with hMRI parameters' variation after initial surgery, weeks to months before overt recurrence.
Collapse
Affiliation(s)
- Gilles Reuter
- GIGA Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.,Department of Neurosurgery, University Hospital of Liège, Liège, Belgium
| | - Emilie Lommers
- GIGA Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.,Department of Neurology, University Hospital of Liège, Liège, Belgium
| | - Evelyne Balteau
- GIGA Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium
| | - Jessica Simon
- Psychology and Neuroscience of Cognition-PsyNCogn, University of Liège, Liège, Belgium
| | - Christophe Phillips
- GIGA Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.,GIGA In Silico Medicine, University of Liège, Liège, Belgium
| | - Felix Scholtes
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium.,Laboratory of Developmental Neurobiology, GIGA-Neurosciences Research Center, University of Liège, Liège, Belgium.,Department of Neuroanatomy, University of Liège, Liège, Belgium
| | - Didier Martin
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium
| | - Arnaud Lombard
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium.,Laboratory of Developmental Neurobiology, GIGA-Neurosciences Research Center, University of Liège, Liège, Belgium
| | - Pierre Maquet
- GIGA Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.,Department of Neurology, University Hospital of Liège, Liège, Belgium
| |
Collapse
|
34
|
De Pardieu M, Boucebci S, Herpe G, Fauche C, Velasco S, Ingrand P, Tasu JP. Glioma-grade diagnosis using in-phase and out-of-phase T1-weighted magnetic resonance imaging: A prospective study. Diagn Interv Imaging 2020; 101:451-456. [DOI: 10.1016/j.diii.2020.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 12/15/2022]
|
35
|
Tang Z, Xu Y, Jin L, Aibaidula A, Lu J, Jiao Z, Wu J, Zhang H, Shen D. Deep Learning of Imaging Phenotype and Genotype for Predicting Overall Survival Time of Glioblastoma Patients. IEEE TRANSACTIONS ON MEDICAL IMAGING 2020; 39:2100-2109. [PMID: 31905135 PMCID: PMC7289674 DOI: 10.1109/tmi.2020.2964310] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Glioblastoma (GBM) is the most common and deadly malignant brain tumor. For personalized treatment, an accurate pre-operative prognosis for GBM patients is highly desired. Recently, many machine learning-based methods have been adopted to predict overall survival (OS) time based on the pre-operative mono- or multi-modal imaging phenotype. The genotypic information of GBM has been proven to be strongly indicative of the prognosis; however, this has not been considered in the existing imaging-based OS prediction methods. The main reason is that the tumor genotype is unavailable pre-operatively unless deriving from craniotomy. In this paper, we propose a new deep learning-based OS prediction method for GBM patients, which can derive tumor genotype-related features from pre-operative multimodal magnetic resonance imaging (MRI) brain data and feed them to OS prediction. Specifically, we propose a multi-task convolutional neural network (CNN) to accomplish both tumor genotype and OS prediction tasks jointly. As the network can benefit from learning tumor genotype-related features for genotype prediction, the accuracy of predicting OS time can be prominently improved. In the experiments, multimodal MRI brain dataset of 120 GBM patients, with as many as four different genotypic/molecular biomarkers, are used to evaluate our method. Our method achieves the highest OS prediction accuracy compared to other state-of-the-art methods.
Collapse
|
36
|
Advances in Noninvasive Neurodiagnostics. World Neurosurg 2020; 139:1-3. [PMID: 32194266 DOI: 10.1016/j.wneu.2020.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/02/2020] [Indexed: 11/21/2022]
|
37
|
Vascular habitat analysis based on dynamic susceptibility contrast perfusion MRI predicts IDH mutation status and prognosis in high-grade gliomas. Eur Radiol 2020; 30:3254-3265. [PMID: 32078014 DOI: 10.1007/s00330-020-06702-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/29/2019] [Accepted: 02/03/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The current study aimed to evaluate the clinical practice for hemodynamic tissue signature (HTS) method in IDH genotype prediction in three groups derived from high-grade gliomas. METHODS Preoperative MRI examinations of 44 patients with known grade and IDH genotype were assigned into three study groups: glioblastoma multiforme, grade III, and high-grade gliomas. Perfusion parameters were analyzed and were used to automatically draw the four reproducible habitats (high-angiogenic enhancing tumor habitats, low-angiogenic enhancing tumor habitats, infiltrated peripheral edema habitats, vasogenic peripheral edema habitats) related to vascular heterogeneity. These four habitats were then compared between inter-patient with IDH mutation and their wild-type counterparts at these three groups, respectively. The discriminating potential for HTS in assessing IDH mutation status prediction was assessed by ROC curves. RESULTS Compared with IDH wild type, IDH mutation had significantly decreased relative cerebral blood volume (rCBV) at the high-angiogenic enhancing tumor habitats and low-angiogenic enhancing tumor habitats. ROC analysis revealed that the rCBVs in habitats had great ability to discriminate IDH mutation from their wild type in all groups. In addition, the Kaplan-Meier survival analysis yielded significant differences for the survival times observed from the populations dichotomized by low (< 4.31) and high (> 4.31) rCBV in the low-angiogenic enhancing tumor habitat. CONCLUSIONS The HTS method has been proven to have high prediction capabilities for IDH mutation status in high-grade glioma patients, providing a set of quantifiable habitats associated with tumor vascular heterogeneity. KEY POINTS • The HTS method has a high accuracy for molecular stratification prediction for all subsets of HGG. • The HTS method can give IDH mutation-related hemodynamic information of tumor-infiltrated and vasogenic edema. • IDH-relevant rCBV difference in habitats will be a great prognosis factor in HGG.
Collapse
|
38
|
Extracellular Vesicles Involvement in the Modulation of the Glioblastoma Environment. JOURNAL OF ONCOLOGY 2020; 2020:3961735. [PMID: 32411235 PMCID: PMC7204270 DOI: 10.1155/2020/3961735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/11/2019] [Indexed: 12/24/2022]
Abstract
Glioblastoma (GBM) is the most deadly primary brain tumour and is a paradigmatic example of heterogeneous cancer. Although expanding data propose the phenotypic plasticity exhibited by glioblastoma cells, as a critical feature involved in the tumour development and posttherapy recurrence, the central machinery responsible for their aggressiveness remains elusive. Despite decades of research, the complex biology of the glioblastoma is still unknown. Progress in genetic and epigenetic discoveries has improved diagnostic classification, prognostic information, and therapeutic planning. In the complex model of intercellular signalling, several studies have shown that extracellular vesicles have a key role in the intercellular communication among GBM cells and the tumour microenvironment modulation. The purpose of this review is to summarize the role of the EV-mediated intercellular crosstalk in the glioblastoma physiopathology.
Collapse
|
39
|
Tang Z, Xu Y, Jiao Z, Lu J, Jin L, Aibaidula A, Wu J, Wang Q, Zhang H, Shen D. Pre-operative Overall Survival Time Prediction for Glioblastoma Patients Using Deep Learning on Both Imaging Phenotype and Genotype. MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION : MICCAI ... INTERNATIONAL CONFERENCE ON MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION 2019; 11764:415-422. [PMID: 34085058 PMCID: PMC8171810 DOI: 10.1007/978-3-030-32239-7_46] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Glioblastoma (GBM) is the most common and deadly malignant brain tumor with short yet varied overall survival (OS) time. Per request of personalized treatment, accurate pre-operative prognosis for GBM patients is highly desired. Currently, many machine learning-based studies have been conducted to predict OS time based on pre-operative multimodal MR images of brain tumor patients. However, tumor genotype, such as MGMT and IDH, which has been proven to have strong relationship with OS, is completely not considered in pre-operative prognosis as the genotype information is unavailable until craniotomy. In this paper, we propose a new deep learning based method for OS time prediction. It can derive genotype related features from pre-operative multimodal MR images of brain tumor patients to guide OS time prediction. Particularly, we propose a multi-task convolutional neural network (CNN) to accomplish tumor genotype and OS time prediction tasks. As the network can benefit from learning genotype related features toward genotype prediction, we verify upon a dataset of 120 GBM patients and conclude that the multi-task learning can effectively improve the accuracy of predicting OS time in personalized prognosis.
Collapse
Affiliation(s)
- Zhenyu Tang
- Beijing Advanced Innovation Center for Big Data and Brain Computing, Beihang University, Beijing, China
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yuyun Xu
- Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhicheng Jiao
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Junfeng Lu
- Neurosurgery Department of Huashan Hospital, Shanghai, China
| | - Lei Jin
- Neurosurgery Department of Huashan Hospital, Shanghai, China
| | | | - Jinsong Wu
- Neurosurgery Department of Huashan Hospital, Shanghai, China
| | - Qian Wang
- The Medical Image Computing Lab, Shanghai Jiao Tong University, Shanghai, China
| | - Han Zhang
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dinggang Shen
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
40
|
Wolf KJ, Chen J, Coombes J, Aghi MK, Kumar S. Dissecting and rebuilding the glioblastoma microenvironment with engineered materials. NATURE REVIEWS. MATERIALS 2019; 4:651-668. [PMID: 32647587 PMCID: PMC7347297 DOI: 10.1038/s41578-019-0135-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/24/2019] [Indexed: 05/15/2023]
Abstract
Glioblastoma (GBM) is the most aggressive and common form of primary brain cancer. Several decades of research have provided great insight into GBM progression; however, the prognosis remains poor with a median patient survival time of ~ 15 months. The tumour microenvironment (TME) of GBM plays a crucial role in mediating tumour progression and thus is being explored as a therapeutic target. Progress in the development of treatments targeting the TME is currently limited by a lack of model systems that can accurately recreate the distinct extracellular matrix composition and anatomic features of the brain, such as the blood-brain barrier and axonal tracts. Biomaterials can be applied to develop synthetic models of the GBM TME to mimic physiological and pathophysiological features of the brain, including cellular and ECM composition, mechanical properties, and topography. In this Review, we summarize key features of the GBM microenvironment and discuss different strategies for the engineering of GBM TME models, including 2D and 3D models featuring chemical and mechanical gradients, interfaces and fluid flow. Finally, we highlight the potential of engineered TME models as platforms for mechanistic discovery and drug screening as well as preclinical testing and precision medicine.
Collapse
Affiliation(s)
- Kayla J. Wolf
- University of California, Berkeley – University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Joseph Chen
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Jason Coombes
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA
- Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Manish K. Aghi
- Department of Neurosurgery, University of California San Francisco (UCSF), San Francisco, California, 94158
| | - Sanjay Kumar
- University of California, Berkeley – University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, 94720, USA
| |
Collapse
|
41
|
Soni N, Priya S, Bathla G. Texture Analysis in Cerebral Gliomas: A Review of the Literature. AJNR Am J Neuroradiol 2019; 40:928-934. [PMID: 31122918 DOI: 10.3174/ajnr.a6075] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 04/22/2019] [Indexed: 12/17/2022]
Abstract
Texture analysis is a continuously evolving, noninvasive radiomics technique to quantify macroscopic tissue heterogeneity indirectly linked to microscopic tissue heterogeneity beyond human visual perception. In recent years, systemic oncologic applications of texture analysis have been increasingly explored. Here we discuss the basic concepts and methodologies of texture analysis, along with a review of various MR imaging texture analysis applications in glioma imaging. We also discuss MR imaging texture analysis limitations and the technical challenges that impede its widespread clinical implementation. With continued advancement in computational processing, MR imaging texture analysis could potentially develop into a valuable clinical tool in routine oncologic imaging.
Collapse
Affiliation(s)
- N Soni
- From the Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - S Priya
- From the Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.
| | - G Bathla
- From the Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| |
Collapse
|
42
|
Choi Y, Ahn KJ, Nam Y, Jang J, Shin NY, Choi HS, Jung SL, Kim BS. Analysis of peritumoral hyperintensity on pre-operative T2-weighted MR images in glioblastoma: Additive prognostic value of Minkowski functionals. PLoS One 2019; 14:e0217785. [PMID: 31150499 PMCID: PMC6544273 DOI: 10.1371/journal.pone.0217785] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 05/17/2019] [Indexed: 11/19/2022] Open
Abstract
Objectives The extent of peritumoral tumor cell infiltrations in glioblastoma contributes to poor prognosis. We aimed to assess additive prognostic value of Minkowski functionals in analyzing heterogeneity of peritumoral hyperintensity on T2WI in glioblastoma patients. Methods Clinical data (age, sex, extent of surgical resection), O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status and pre-operative T2WI of 113 pathologically confirmed glioblastoma patients (from our institution, n = 61; from the Cancer Imaging Archive, n = 52) were retrospectively reviewed. The patients were randomly grouped into a training set (n = 80) and a test set (n = 33). Peritumoral T2 hyperintensity was manually segmented and Minkowski functionals—a texture analysis method capturing heterogeneity of MR images—were computed as a function of 11 grayscale thresholds. The Cox proportional hazards models were fitted with clinical variables, Minkowski functionals features as well as both combined. The risk prediction performances of the Minkowski functionals and combined models were validated on a separate test dataset. The sex-specific survival difference of the entire cohort was analyzed according to MGMT methylation status via Kaplan-Meier survival curves. Results Thirty-three Minkowski features (11 area, 11 perimeter and 11 genus) for each patient were acquired giving a total of 3729 features. Cox regression models fitted with clinical data, Minkowski features, and both combined had incremental concordance indices of 0.577 (P = 0.02), 0.706 (P = 0.02) and 0.714 (P = 0.01) respectively. The prediction error rate of the combined model—having clinical and Minkowski features—was lower than that of Minkowski functionals model (0.135 and 0.161, respectively) when validated on a test dataset. No sex-specific survival difference was found according to MGMT methylation status (male, P = 0.2; female, P = 0.22). Conclusions Minkowski functionals features computed from peritumoral hyperintensity can capture heterogeneity of glioblastoma on T2WI and have additive prognostic value in predicting survival, demonstrating their potential in complementing currently available prognostic parameters.
Collapse
Affiliation(s)
- Yangsean Choi
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Kook Jin Ahn
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- * E-mail:
| | - Yoonho Nam
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jinhee Jang
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Na-Young Shin
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyun Seok Choi
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - So-Lyung Jung
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Bum-soo Kim
- Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
43
|
Chaddad A, Kucharczyk MJ, Daniel P, Sabri S, Jean-Claude BJ, Niazi T, Abdulkarim B. Radiomics in Glioblastoma: Current Status and Challenges Facing Clinical Implementation. Front Oncol 2019; 9:374. [PMID: 31165039 PMCID: PMC6536622 DOI: 10.3389/fonc.2019.00374] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
Radiomics analysis has had remarkable progress along with advances in medical imaging, most notability in central nervous system malignancies. Radiomics refers to the extraction of a large number of quantitative features that describe the intensity, texture and geometrical characteristics attributed to the tumor radiographic data. These features have been used to build predictive models for diagnosis, prognosis, and therapeutic response. Such models are being combined with clinical, biological, genetics and proteomic features to enhance reproducibility. Broadly, the four steps necessary for radiomic analysis are: (1) image acquisition, (2) segmentation or labeling, (3) feature extraction, and (4) statistical analysis. Major methodological challenges remain prior to clinical implementation. Essential steps include: adoption of an optimized standard imaging process, establishing a common criterion for performing segmentation, fully automated extraction of radiomic features without redundancy, and robust statistical modeling validated in the prospective setting. This review walks through these steps in detail, as it pertains to high grade gliomas. The impact on precision medicine will be discussed, as well as the challenges facing clinical implementation of radiomic in the current management of glioblastoma.
Collapse
Affiliation(s)
- Ahmad Chaddad
- Division of Radiation Oncology, Department of Oncology, McGill University, Montreal, QC, Canada
| | | | - Paul Daniel
- Division of Radiation Oncology, Department of Oncology, McGill University, Montreal, QC, Canada
| | - Siham Sabri
- Department of Pathology, McGill University, Montreal, QC, Canada.,Research Institute of the McGill University Health Centre, Glen Site, Montreal, QC, Canada
| | - Bertrand J Jean-Claude
- Research Institute of the McGill University Health Centre, Glen Site, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Tamim Niazi
- Division of Radiation Oncology, Department of Oncology, McGill University, Montreal, QC, Canada
| | - Bassam Abdulkarim
- Division of Radiation Oncology, Department of Oncology, McGill University, Montreal, QC, Canada.,Research Institute of the McGill University Health Centre, Glen Site, Montreal, QC, Canada
| |
Collapse
|
44
|
What Neuroradiologists Need to Know About Radiation Treatment for Neural Tumors. Top Magn Reson Imaging 2019; 28:37-47. [PMID: 31022047 DOI: 10.1097/rmr.0000000000000196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Radiation oncologists and radiologists have a unique and mutually dependent relationship. Radiation oncologists rely on diagnostic imaging to locate the tumor and define the treatment target volume, evaluation of response to therapy, and follow-up. Accurate interpretation of post-treatment imaging requires diagnostic radiologists to have a basic understanding of radiation treatment planning and delivery. There are various radiation treatment modalities such as 3D conformal radiation therapy, intensity modulated radiation therapy and stereotactic radiosurgery as well as different radiation modalities such as photons and protons that can be used for treatment. All of these have subtle differences in how the treatment is planned and how the imaging findings might be affected. This paper provides an overview of the basic principles of radiation oncology, different radiation treatment modalities, how radiation therapy is planned and delivered, how knowledge of this process can help interpretation of images, and how the radiologist can contribute to this process.
Collapse
|
45
|
|
46
|
Correlations between metabolic texture features, genetic heterogeneity, and mutation burden in patients with lung cancer. Eur J Nucl Med Mol Imaging 2018; 46:446-454. [DOI: 10.1007/s00259-018-4138-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/16/2018] [Indexed: 12/11/2022]
|
47
|
Abstract
The most aggressive brain malignancy, glioblastoma, accounts for 60-70% of all gliomas and is uniformly fatal. According to the molecular signature, glioblastoma is divided into four subtypes (proneural, neural, classical, and mesenchymal), each with its own genetic background. The Cancer Genome Atlas project provides information about the most common genetic changes in glioblastoma. They involve mutations in TP53, TERT, and PTEN, and amplifications in EFGR, PDGFRA, CDK4, CDK6, MDM2, and MDM4. Recently, epigenetics was used to demonstrate the oncogenic roles of miR-124, miR-137, and miR-128. The most important findings so far are mutations in IDH1/2 and MGMT promoter methylation, which are routinely used as predictive biomarkers in patient care. Current clinical treatment leaves patients with only a 10% chance for 5-year survival. Attempts to define the mutational profile of glioblastoma to identify clinically relevant changes have not yet yielded significant results. This can be attributed to inter- and intra-tumor heterogeneity that is present in most glioblastomas, as well as hypermutation that appears as a consequence of chemotherapy. The evolving field of radiogenomics aims to classify glioblastoma using a combination of magnetic resonance imaging and genomic information. In the era of genomic medicine, next-generation sequencing is extensively used in glioblastoma research because it can detect multiple changes in a single biological sample; its potential in detecting circulating cell-free DNA has been tested in cerebrospinal fluid and plasma, and it shows promise in the examination of the cellular content of extracellular vesicles as a potential source of biomarkers. Next-generation sequencing is making its way into glioblastoma diagnostics. Gene panels like GlioSeq, which includes the most commonly mutated genes, are currently being tested on snap frozen and formalin fixed paraffin embedded tissues. This new methodology is helping to define the "next generation of glioblastomas" - clinically defined and better understood, with greater potential to improve patient care. However, limitations of the necessary infrastructure, space for data storage, technical expertise, and data ownership need to be considered carefully.
Collapse
Affiliation(s)
- Ivana Jovčevska
- a Medical Center for Molecular Biology, Institute of Biochemistry, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| |
Collapse
|