1
|
Yuan J, Siakallis L, Li HB, Brandner S, Zhang J, Li C, Mancini L, Bisdas S. Structural- and DTI- MRI enable automated prediction of IDH Mutation Status in CNS WHO Grade 2-4 glioma patients: a deep Radiomics Approach. BMC Med Imaging 2024; 24:104. [PMID: 38702613 PMCID: PMC11067215 DOI: 10.1186/s12880-024-01274-9] [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: 05/01/2023] [Accepted: 04/15/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND The role of isocitrate dehydrogenase (IDH) mutation status for glioma stratification and prognosis is established. While structural magnetic resonance image (MRI) is a promising biomarker, it may not be sufficient for non-invasive characterisation of IDH mutation status. We investigated the diagnostic value of combined diffusion tensor imaging (DTI) and structural MRI enhanced by a deep radiomics approach based on convolutional neural networks (CNNs) and support vector machine (SVM), to determine the IDH mutation status in Central Nervous System World Health Organization (CNS WHO) grade 2-4 gliomas. METHODS This retrospective study analyzed the DTI-derived fractional anisotropy (FA) and mean diffusivity (MD) images and structural images including fluid attenuated inversion recovery (FLAIR), non-enhanced T1-, and T2-weighted images of 206 treatment-naïve gliomas, including 146 IDH mutant and 60 IDH-wildtype ones. The lesions were manually segmented by experienced neuroradiologists and the masks were applied to the FA and MD maps. Deep radiomics features were extracted from each subject by applying a pre-trained CNN and statistical description. An SVM classifier was applied to predict IDH status using imaging features in combination with demographic data. RESULTS We comparatively assessed the CNN-SVM classifier performance in predicting IDH mutation status using standalone and combined structural and DTI-based imaging features. Combined imaging features surpassed stand-alone modalities for the prediction of IDH mutation status [area under the curve (AUC) = 0.846; sensitivity = 0.925; and specificity = 0.567]. Importantly, optimal model performance was noted following the addition of demographic data (patients' age) to structural and DTI imaging features [area under the curve (AUC) = 0.847; sensitivity = 0.911; and specificity = 0.617]. CONCLUSIONS Imaging features derived from DTI-based FA and MD maps combined with structural MRI, have superior diagnostic value to that provided by standalone structural or DTI sequences. In combination with demographic information, this CNN-SVM model offers a further enhanced non-invasive prediction of IDH mutation status in gliomas.
Collapse
Affiliation(s)
- Jialin Yuan
- Department of Radiology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- Queen Square Institute of Neurology, University College London, London, UK
| | - Loizos Siakallis
- Queen Square Institute of Neurology, University College London, London, UK
| | - Hongwei Bran Li
- Department of Informatics, Technical University of Munich, Munich, Germany
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Sebastian Brandner
- Division of Neuropathology, Queen Square Institute of Neurology, University College London, London, UK
| | - Jianguo Zhang
- Department of Computer Science and Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chenming Li
- Department of Computer Science and Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Laura Mancini
- Queen Square Institute of Neurology, University College London, London, UK
- Lysholm Department of Neuroradiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Sotirios Bisdas
- Queen Square Institute of Neurology, University College London, London, UK.
- Lysholm Department of Neuroradiology, University College London Hospitals NHS Foundation Trust, London, UK.
| |
Collapse
|
2
|
Picca A, Bruno F, Nichelli L, Sanson M, Rudà R. Advances in molecular and imaging biomarkers in lower-grade gliomas. Expert Rev Neurother 2023; 23:1217-1231. [PMID: 37982735 DOI: 10.1080/14737175.2023.2285472] [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: 08/07/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
INTRODUCTION Lower-grade (grade 2-3) gliomas (LGGs) constitutes a group of primary brain tumors with variable clinical behaviors and treatment responses. Recent advancements in molecular biology have redefined their classification, and novel imaging modalities emerged for the noninvasive diagnosis and follow-up. AREAS COVERED This review comprehensively analyses the current knowledge on molecular and imaging biomarkers in LGGs. Key molecular alterations, such as IDH mutations and 1p/19q codeletion, are discussed for their prognostic and predictive implications in guiding treatment decisions. Moreover, the authors explore theranostic biomarkers for the potential of tailored therapies. Additionally, they also describe the utility of advanced imaging modalities, including widely available techniques, as dynamic susceptibility contrast perfusion-weighted imaging and less validated, emerging approaches, for the noninvasive LGGs characterization and follow-up. EXPERT OPINION The integration of molecular markers enhanced the stratification of LGGs, leading to the new concept of integrated histomolecular classification. While the IDH mutation is an established key prognostic and predictive marker, recent results from IDH inhibitors trials showed its potential value as a theranostic marker. In this setting, advanced MRI techniques such as 2-D-hydroxyglutarate spectroscopy are very promising for the noninvasive diagnosis and monitoring of LGGs. This progress offers exciting prospects for personalized medicine and improved treatment outcomes in LGGs.
Collapse
Affiliation(s)
- Alberto Picca
- Service de Neurologie 2 Mazarin, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
- Sorbonne Université, Inserm, CNRS, UMRS1127, Institut du Cerveau-Paris Brain Institute-ICM, AP-HP, Paris, France
| | - Francesco Bruno
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science University Hospital, Turin, Italy
| | - Lucia Nichelli
- Service de Neuroradiologie, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
| | - Marc Sanson
- Service de Neurologie 2 Mazarin, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
- Sorbonne Université, Inserm, CNRS, UMRS1127, Institut du Cerveau-Paris Brain Institute-ICM, AP-HP, Paris, France
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science University Hospital, Turin, Italy
| |
Collapse
|
3
|
MRI features predict tumor grade in isocitrate dehydrogenase (IDH)-mutant astrocytoma and oligodendroglioma. Neuroradiology 2023; 65:121-129. [PMID: 35953567 DOI: 10.1007/s00234-022-03038-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/07/2022] [Indexed: 01/28/2023]
Abstract
PURPOSE Nearly all literature for predicting tumor grade in astrocytoma and oligodendroglioma pre-dates the molecular classification system. We investigated the association between contrast enhancement, ADC, and rCBV with tumor grade separately for IDH-mutant astrocytomas and molecularly-defined oligodendrogliomas. METHODS For this retrospective study, 44 patients with IDH-mutant astrocytomas (WHO grades II, III, or IV) and 39 patients with oligodendrogliomas (IDH-mutant and 1p/19q codeleted) (WHO grade II or III) were enrolled. Two readers independently assessed preoperative MRI for contrast enhancement, ADC, and rCBV. Inter-reader agreement was calculated, and statistical associations between MRI metrics and WHO grade were determined per reader. RESULTS For IDH-mutant astrocytomas, both readers found a stepwise positive association between contrast enhancement and WHO grade (Reader A: OR 7.79 [1.97, 30.80], p = 0.003; Reader B: OR 6.62 [1.70, 25.82], p = 0.006); both readers found that ADC was negatively associated with WHO grade (Reader A: OR 0.74 [0.61, 0.90], p = 0.002); Reader B: OR 0.80 [0.66, 0.96], p = 0.017), and both readers found that rCBV was positively associated with WHO grade (Reader A: OR 2.33 [1.35, 4.00], p = 0.002; Reader B: OR 2.13 [1.30, 3.57], p = 0.003). For oligodendrogliomas, both readers found a positive association between contrast enhancement and WHO grade (Reader A: OR 15.33 [2.56, 91.95], p = 0.003; Reader B: OR 20.00 [2.19, 182.45], p = 0.008), but neither reader found an association between ADC or rCBV and WHO grade. CONCLUSIONS Contrast enhancement predicts WHO grade for IDH-mutant astrocytomas and oligodendrogliomas. ADC and rCBV predict WHO grade for IDH-mutant astrocytomas, but not for oligodendrogliomas.
Collapse
|
4
|
Correlation of Diffusion Tensor Imaging Parameters with the Pathological Grade of Brain Glioma and Expression of Vascular Endothelial Growth Factor and Ki-67. IRANIAN JOURNAL OF RADIOLOGY 2022. [DOI: 10.5812/iranjradiol-118135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background: Most brain gliomas are high-grade and likely to spread locally. Consequently, these patients commonly have a poor prognosis. Accurate identification of the malignancy grade of brain glioma before treatment is of great clinical significance. Objectives: This study aimed to explore the correlation of diffusion tensor imaging (DTI) parameters, fractional anisotropy (FA), and apparent diffusion coefficient (ADC) with the pathological grade of brain glioma and expression of vascular endothelial growth factor (VEGF) and Ki-67. Patients and Methods: A total of 116 patients were selected for this study from January 2018 to December 2019. All the participants underwent magnetic resonance imaging (MRI) and DTI before surgery, and the FA and ADC values were measured for the regions of interest. Surgically resected tumor specimens were collected for immunohistochemical assay. Finally, the FA and ADC values and positive expression rates of VEGF and Ki-67 were compared. Results: A significantly higher FA, besides the positive expression of VEGF and Ki-67, was reported in the high-grade group, whereas a lower ADC was found in this group compared to the low-grade group (P < 0.05). Areas of normal white matter and peritumoral edema had higher FA values, whereas lower ADCs were measured in these areas compared to the cerebrospinal fluid (P < 0.05). The FA of tumor parenchymal area was positively correlated with the World Health Organization (WHO) WHO class of tumors (r = 0.588, P = 0.028), and the expression of VEGF and Ki-67 was positively correlated with the WHO grade (r = 0.843, P = 0.002 and r = 0.743, P = 0.006, respectively). The FA of tumor parenchymal area was positively correlated with the expression of VEGF and Ki-67 (r = 0.654, P = 0.008 and r = 0.567, P = 0.012, respectively). However, the ADC of tumor parenchymal area was not significantly correlated with the WHO grade, VEGF expression, or Ki-67 expression (r = 0.143, P = 0.156, r = 0.232, P = 0.116, and r = 0.054, P = 0.179, respectively). Conclusion: The FA value, as a DTI parameter, is valuable for assessing the malignancy grade of tumor cells and can provide a proper reference for formulating treatment regimens for brain gliomas.
Collapse
|
5
|
Liu S, Zhang Y, Kong Z, Jiang C, Wang Y, Zhao D, You H, Ma W, Feng F. Feasibility of evaluating the histologic and genetic subtypes of WHO grade II-IV gliomas by diffusion-weighted imaging. BMC Neurosci 2022; 23:72. [PMID: 36471242 PMCID: PMC9720933 DOI: 10.1186/s12868-022-00750-8] [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: 08/20/2020] [Accepted: 10/28/2022] [Indexed: 12/07/2022] Open
Abstract
BACKGROUND To explore the feasibility of diffusion-weighted imaging (DWI) metrics to predict the histologic subtypes and genetic status of gliomas (e.g., IDH, MGMT, and TERT) noninvasively. METHODS One hundred and eleven patients with pathologically confirmed WHO grade II-IV gliomas were recruited retrospectively. Apparent diffusion coefficient (ADC) values were measured in solid parts of gliomas on co-registered T2-weighted images and were compared with each other in terms of WHO grading and genotypes using t-tests. Receiver operating characteristic analysis was performed to assess the diagnostic performances of ADC. Subsequently, multiple linear regression was used to find independent variables, which can directly affect ADC values. RESULTS The values of overall mean ADC (omADC) and normalized ADC (nADC) of high grade gliomas and IDH wildtype gliomas were lower than low grade gliomas and IDH mutated gliomas (P < 0.05). nADC values showed better diagnostic performance than omADC in identifying tumor grade (AUC: 0.787 vs. 0.750) and IDH status (AUC: 0.836 vs. 0.777). ADC values had limited abilities in distinguishing TERT status (AUC = 0.607 for nADC and 0.617 for omADC) and MGMT status (AUC = 0.651 for nADC). Only tumor grade and IDH status were tightly associated with ADC values. CONCLUSION DWI metrics can predict glioma grading and IDH mutation noninvasively, but have limited use in detecting TERT mutation and MGMT methylation.
Collapse
Affiliation(s)
- Sirui Liu
- grid.506261.60000 0001 0706 7839Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China ,grid.8547.e0000 0001 0125 2443Department of Radiology, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiwei Zhang
- grid.506261.60000 0001 0706 7839Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China ,grid.411472.50000 0004 1764 1621Department of Radiology, Peking University First Hospital, No.8 Xishiku, Beijing, China
| | - Ziren Kong
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Chendan Jiang
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Yu Wang
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Dachun Zhao
- grid.506261.60000 0001 0706 7839Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui You
- grid.506261.60000 0001 0706 7839Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Wenbin Ma
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Feng Feng
- grid.506261.60000 0001 0706 7839Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| |
Collapse
|
6
|
circRPPH1_025 Overexpression Promotes Migration and Invasion of Glioblastoma Multiforme. DISEASE MARKERS 2022; 2022:4764028. [PMID: 35928926 PMCID: PMC9345728 DOI: 10.1155/2022/4764028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022]
Abstract
Objective To study the effect of circ_0000512 (circRPPH1_025) on the tumorigensis and development of glioblastoma and its molecular mechanism. Methods The expression levels of circ_0000512 in normal astrocytes (NHA) and human glioblastoma cell lines (U87, U251, and A172) and the expression levels of circ_0000512 and linear RNA RPPH1 in U87 cells after RNase R treatment were detected by qRT-PCR. The effects of circ_0000512 knockdown or overexpression on the proliferation, migration, invasion, and epithelial-mesenchymal transition of U87 cells were detected by CCK-8 assay, cell colony formation assay, transwell invasion assay, wound healing assay, and western blot. Results The expression of circ_0000512 was upregulated in glioblastoma cells, and the overexpression of circ_0000512 was beneficial to the proliferation, migration, invasion, and epithelial-mesenchymal transition of U87 cells, while knockdown of circ_0000512 showed the opposite results. Conclusion circ_0000512 can be used as a potential target for early diagnosis and targeted therapy of glioblastoma multiforme.
Collapse
|
7
|
Chen Z, Ye N, Teng C, Li X. Alternations and Applications of the Structural and Functional Connectome in Gliomas: A Mini-Review. Front Neurosci 2022; 16:856808. [PMID: 35478847 PMCID: PMC9035851 DOI: 10.3389/fnins.2022.856808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/28/2022] [Indexed: 12/12/2022] Open
Abstract
In the central nervous system, gliomas are the most common, but complex primary tumors. Genome-based molecular and clinical studies have revealed different classifications and subtypes of gliomas. Neuroradiological approaches have non-invasively provided a macroscopic view for surgical resection and therapeutic effects. The connectome is a structural map of a physical object, the brain, which raises issues of spatial scale and definition, and it is calculated through diffusion magnetic resonance imaging (MRI) and functional MRI. In this study, we reviewed the basic principles and attributes of the structural and functional connectome, followed by the alternations of connectomes and their influences on glioma. To extend the applications of connectome, we demonstrated that a series of multi-center projects still need to be conducted to systemically investigate the connectome and the structural-functional coupling of glioma. Additionally, the brain-computer interface based on accurate connectome could provide more precise structural and functional data, which are significant for surgery and postoperative recovery. Besides, integrating the data from different sources, including connectome and other omics information, and their processing with artificial intelligence, together with validated biological and clinical findings will be significant for the development of a personalized surgical strategy.
Collapse
Affiliation(s)
- Ziyan Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Ningrong Ye
- Department of Neurosurgery, Xiangya Hospital, Central South University, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Chubei Teng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
8
|
RFC2: a prognosis biomarker correlated with the immune signature in diffuse lower-grade gliomas. Sci Rep 2022; 12:3122. [PMID: 35210438 PMCID: PMC8873322 DOI: 10.1038/s41598-022-06197-5] [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] [Received: 08/19/2021] [Accepted: 01/24/2022] [Indexed: 11/08/2022] Open
Abstract
Diffuse lower-grade gliomas (LGG) represent the highly heterogeneous and infiltrative neoplasms in the central nervous system (CNS). Replication factor C 2 (RFC2) is a subunit of the RFC complex that modulates DNA replication and repair. However, the prognosis value of RFC2 and its association with the immune signature of tumor microenvironment (TME) in LGG remains unknown. Based on Oncomine, TCGA, GTEx, TIMER, GEPIA, and HPA databases, we evaluated RFC2 expression levels and its clinical prognostic value in LGG and other cancers. Then we analyzed the correlations between RFC2 expression and tumor mutation burden (TMB), tumor microsatellite instability (MSI), and mismatch repair (MMR) genes across cancers. And CIBERSORT and ESTIMATE algorithms were conducted to estimate the association of RFC2 with immune cell infiltration of LGG. Additionally, we performed the functional enrichment analyses of RFC2 in LGG. Then functional experiments were employed to further validate the oncogenic role of RFC2 in LGG. Our results showed that RFC2 was widely highly expressed in most types of cancer. And its expression was closely related to the clinicopathological features and prognosis in LGG and other cancer types. RFC2 levels were also correlated with TMB and MSI across various cancers. Furthermore, RFC2 was positively associated with the infiltration levels of immune cells and immune checkpoint genes in LGG. Additionally, in vitro experiments revealed that RFC2 played an oncogenic role in LGG progression. In conclusion, our findings revealed that RFC2 could serve as a reliable biomarker to predict the prognosis and immune signature for LGG.
Collapse
|
9
|
Abstract
The central role of MRI in neuro-oncology is undisputed. The technique is used, both in clinical practice and in clinical trials, to diagnose and monitor disease activity, support treatment decision-making, guide the use of focused treatments and determine response to treatment. Despite recent substantial advances in imaging technology and image analysis techniques, clinical MRI is still primarily used for the qualitative subjective interpretation of macrostructural features, as opposed to quantitative analyses that take into consideration multiple pathophysiological features. However, the field of quantitative imaging and imaging biomarker development is maturing. The European Imaging Biomarkers Alliance (EIBALL) and Quantitative Imaging Biomarkers Alliance (QIBA) are setting standards for biomarker development, validation and implementation, as well as promoting the use of quantitative imaging and imaging biomarkers by demonstrating their clinical value. In parallel, advanced imaging techniques are reaching the clinical arena, providing quantitative, commonly physiological imaging parameters that are driving the discovery, validation and implementation of quantitative imaging and imaging biomarkers in the clinical routine. Additionally, computational analysis techniques are increasingly being used in the research setting to convert medical images into objective high-dimensional data and define radiomic signatures of disease states. Here, I review the definition and current state of MRI biomarkers in neuro-oncology, and discuss the clinical potential of quantitative image analysis techniques.
Collapse
|
10
|
Wang Y, Wahafu A, Wu W, Xiang J, Huo L, Ma X, Wang N, Liu H, Bai X, Xu D, Xie W, Wang M, Wang J. FABP5 enhances malignancies of lower-grade gliomas via canonical activation of NF-κB signaling. J Cell Mol Med 2021; 25:4487-4500. [PMID: 33837625 PMCID: PMC8093984 DOI: 10.1111/jcmm.16536] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 01/06/2023] Open
Abstract
Low‐grade gliomas (LGGs) are grade III gliomas based on the WHO classification with significant genetic heterogeneity and clinical properties. Traditional histological classification of gliomas has been challenged by the improvement of molecular stratification; however, the reproducibility and diagnostic accuracy of LGGs classification still remain poor. Herein, we identified fatty acid binding protein 5 (FABP5) as one of the most enriched genes in malignant LGGs and elevated FABP5 revealed severe outcomes in LGGs. Functionally, lentiviral suppression of FABP5 reduced malignant characters including proliferation, cloning formation, immigration, invasion and TMZ resistance, contrarily, the malignancies of LGGs were enhanced by exogenous overexpression of FABP5. Mechanistically, epithelial‐mesenchymal transition (EMT) was correlated to FABP5 expression in LGGs and tumour necrosis factor α (TNFα)‐dependent NF‐κB signalling was involved in this process. Furthermore, FABP5 induced phosphorylation of inhibitor of nuclear factor kappa‐B kinase α (IKKα) thus activated nuclear factor kappa‐B (NF‐κB) signalling. Taken together, our study indicated that FABP5 enhances malignancies of LGGs through canonical activation of NF‐κB signalling, which could be used as individualized prognostic biomarker and potential therapeutic target of LGGs.
Collapse
Affiliation(s)
- Yichang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Alafate Wahafu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianyang Xiang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Longwei Huo
- Department of Neurosurgery, The First Hospital of Yulin, Yulin, China
| | - Xudong Ma
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ning Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hao Liu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaobin Bai
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dongze Xu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wanfu Xie
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Maode Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jia Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
11
|
Huang Z, Lu C, Li G, Li Z, Sun S, Zhang Y, Hou Z, Xie J. Prediction of Lower Grade Insular Glioma Molecular Pathology Using Diffusion Tensor Imaging Metric-Based Histogram Parameters. Front Oncol 2021; 11:627202. [PMID: 33777772 PMCID: PMC7988075 DOI: 10.3389/fonc.2021.627202] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 01/18/2021] [Indexed: 12/20/2022] Open
Abstract
Objectives To explore whether a simplified lesion delineation method and a set of diffusion tensor imaging (DTI) metric-based histogram parameters (mean, 25th percentile, 75th percentile, skewness, and kurtosis) are efficient at predicting the molecular pathology status (MGMT methylation, IDH mutation, TERT promoter mutation, and 1p19q codeletion) of lower grade insular gliomas (grades II and III). Methods 40 lower grade insular glioma patients in two medical centers underwent preoperative DTI scanning. For each patient, the entire abnormal area in their b-non (b0) image was defined as region of interest (ROI), and a set of histogram parameters were calculated for two DTI metrics, fractional anisotropy (FA) and mean diffusivity (MD). Then, we compared how these DTI metrics varied according to molecular pathology and glioma grade, with their predictive performance individually and jointly assessed using receiver operating characteristic curves. The reliability of the combined prediction was evaluated by the calibration curve and Hosmer and Lemeshow test. Results The mean, 25th percentile, and 75th percentile of FA were associated with glioma grade, while the mean, 25th percentile, 75th percentile, and skewness of both FA and MD predicted IDH mutation. The mean, 25th percentile, and 75th percentile of FA, and all MD histogram parameters significantly distinguished TERT promoter status. Similarly, all MD histogram parameters were associated with 1p19q status. However, none of the parameters analyzed for either metric successfully predicted MGMT methylation. The 25th percentile of FA yielded the highest prediction efficiency for glioma grade, IDH mutation, and TERT promoter mutation, while the 75th percentile of MD gave the best prediction of 1p19q codeletion. The combined prediction could enhance the discrimination of grading, IDH and TERT mutation, and also with a good fitness. Conclusions Overall, more invasive gliomas showed higher FA and lower MD values. The simplified ROI delineation method presented here based on the combination of appropriate histogram parameters yielded a more practical and efficient approach to predicting molecular pathology in lower grade insular gliomas. This approach could help clinicians to determine the extent of tumor resection required and reduce complications, enabling more precise treatment of insular gliomas in combination with radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Zhenxing Huang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Neurological Diseases (China), Beijing, China
| | - Changyu Lu
- Department of Neurosurgery, Peking University International Hospital, Beijing, China
| | - Gen Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Neurological Diseases (China), Beijing, China
| | - Zhenye Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Neurological Diseases (China), Beijing, China
| | - Shengjun Sun
- National Clinical Research Center for Neurological Diseases (China), Beijing, China.,Neuroimaging Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yazhuo Zhang
- National Clinical Research Center for Neurological Diseases (China), Beijing, China.,Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zonggang Hou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Neurological Diseases (China), Beijing, China
| | - Jian Xie
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Neurological Diseases (China), Beijing, China
| |
Collapse
|
12
|
Sanvito F, Castellano A, Falini A. Advancements in Neuroimaging to Unravel Biological and Molecular Features of Brain Tumors. Cancers (Basel) 2021; 13:cancers13030424. [PMID: 33498680 PMCID: PMC7865835 DOI: 10.3390/cancers13030424] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Advanced neuroimaging is gaining increasing relevance for the characterization and the molecular profiling of brain tumor tissue. On one hand, for some tumor types, the most widespread advanced techniques, investigating diffusion and perfusion features, have been proven clinically feasible and rather robust for diagnosis and prognosis stratification. In addition, 2-hydroxyglutarate spectroscopy, for the first time, offers the possibility to directly measure a crucial molecular marker. On the other hand, numerous innovative approaches have been explored for a refined evaluation of tumor microenvironments, particularly assessing microstructural and microvascular properties, and the potential applications of these techniques are vast and still to be fully explored. Abstract In recent years, the clinical assessment of primary brain tumors has been increasingly dependent on advanced magnetic resonance imaging (MRI) techniques in order to infer tumor pathophysiological characteristics, such as hemodynamics, metabolism, and microstructure. Quantitative radiomic data extracted from advanced MRI have risen as potential in vivo noninvasive biomarkers for predicting tumor grades and molecular subtypes, opening the era of “molecular imaging” and radiogenomics. This review presents the most relevant advancements in quantitative neuroimaging of advanced MRI techniques, by means of radiomics analysis, applied to primary brain tumors, including lower-grade glioma and glioblastoma, with a special focus on peculiar oncologic entities of current interest. Novel findings from diffusion MRI (dMRI), perfusion-weighted imaging (PWI), and MR spectroscopy (MRS) are hereby sifted in order to evaluate the role of quantitative imaging in neuro-oncology as a tool for predicting molecular profiles, stratifying prognosis, and characterizing tumor tissue microenvironments. Furthermore, innovative technological approaches are briefly addressed, including artificial intelligence contributions and ultra-high-field imaging new techniques. Lastly, after providing an overview of the advancements, we illustrate current clinical applications and future perspectives.
Collapse
Affiliation(s)
- Francesco Sanvito
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.S.); (A.F.)
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Unit of Radiology, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Antonella Castellano
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.S.); (A.F.)
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Correspondence: ; Tel.: +39-02-2643-3015
| | - Andrea Falini
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.S.); (A.F.)
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
13
|
Automated apparent diffusion coefficient analysis for genotype prediction in lower grade glioma: association with the T2-FLAIR mismatch sign. J Neurooncol 2020; 149:325-335. [PMID: 32909115 DOI: 10.1007/s11060-020-03611-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/31/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE The prognosis of lower grade glioma (LGG) patients depends (in large part) on both isocitrate dehydrogenase (IDH) gene mutation and chromosome 1p/19q codeletion status. IDH-mutant LGG without 1p/19q codeletion (IDHmut-Noncodel) often exhibit a unique imaging appearance that includes high apparent diffusion coefficient (ADC) values not observed in other subtypes. The purpose of this study was to develop an ADC analysis-based approach that can automatically identify IDHmut-Noncodel LGG. METHODS Whole-tumor ADC metrics, including fractional tumor volume with ADC > 1.5 × 10-3mm2/s (VADC>1.5), were used to identify IDHmut-Noncodel LGG in a cohort of N = 134 patients. Optimal threshold values determined in this dataset were then validated using an external dataset containing N = 93 cases collected from The Cancer Imaging Archive. Classifications were also compared with radiologist-identified T2-FLAIR mismatch sign and evaluated concurrently to identify added value from a combined approach. RESULTS VADC>1.5 classified IDHmut-Noncodel LGG in the internal cohort with an area under the curve (AUC) of 0.80. An optimal threshold value of 0.35 led to sensitivity/specificity = 0.57/0.93. Classification performance was similar in the validation cohort, with VADC>1.5 ≥ 0.35 achieving sensitivity/specificity = 0.57/0.91 (AUC = 0.81). Across both groups, 37 cases exhibited positive T2-FLAIR mismatch sign-all of which were IDHmut-Noncodel. Of these, 32/37 (86%) also exhibited VADC>1.5 ≥ 0.35, as did 23 additional IDHmut-Noncodel cases which were negative for T2-FLAIR mismatch sign. CONCLUSION Tumor subregions with high ADC were a robust indicator of IDHmut-Noncodel LGG, with VADC>1.5 achieving > 90% classification specificity in both internal and validation cohorts. VADC>1.5 exhibited strong concordance with the T2-FLAIR mismatch sign and the combination of both parameters improved sensitivity in detecting IDHmut-Noncodel LGG.
Collapse
|
14
|
Darvishi P, Batchala PP, Patrie JT, Poisson LM, Lopes MB, Jain R, Fadul CE, Schiff D, Patel SH. Prognostic Value of Preoperative MRI Metrics for Diffuse Lower-Grade Glioma Molecular Subtypes. AJNR Am J Neuroradiol 2020; 41:815-821. [PMID: 32327434 DOI: 10.3174/ajnr.a6511] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/29/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND PURPOSE Despite the improved prognostic relevance of the 2016 WHO molecular-based classification of lower-grade gliomas, variability in clinical outcome persists within existing molecular subtypes. Our aim was to determine prognostically significant metrics on preoperative MR imaging for lower-grade gliomas within currently defined molecular categories. MATERIALS AND METHODS We undertook a retrospective analysis of 306 patients with lower-grade gliomas accrued from an institutional data base and The Cancer Genome Atlas. Two neuroradiologists in consensus analyzed preoperative MRIs of each lower-grade glioma to determine the following: tumor size, tumor location, number of involved lobes, corpus callosum involvement, hydrocephalus, midline shift, eloquent cortex involvement, ependymal extension, margins, contrast enhancement, and necrosis. Adjusted hazard ratios determined the association between MR imaging metrics and overall survival per molecular subtype, after adjustment for patient age, patient sex, World Health Organization grade, and surgical resection status. RESULTS For isocitrate dehydrogenase (IDH) wild-type lower-grade gliomas, tumor size (hazard ratio, 3.82; 95% CI, 1.94-7.75; P < .001), number of involved lobes (hazard ratio, 1.70; 95% CI, 1.28-2.27; P < .001), hydrocephalus (hazard ratio, 4.43; 95% CI, 1.12-17.54; P = .034), midline shift (hazard ratio, 1.16; 95% CI, 1.03-1.30; P = .013), margins (P = .031), and contrast enhancement (hazard ratio, 0.34; 95% CI, 0.13-0.90; P = .030) were associated with overall survival. For IDH-mutant 1p/19q-codeleted lower-grade gliomas, tumor size (hazard ratio, 2.85; 95% CI, 1.06-7.70; P = .039) and ependymal extension (hazard ratio, 6.34; 95% CI, 1.07-37.59; P = .042) were associated with overall survival. CONCLUSIONS MR imaging metrics offers prognostic information for patients with lower-grade gliomas within molecularly defined classes, with the greatest prognostic value for IDH wild-type lower-grade gliomas.
Collapse
Affiliation(s)
- P Darvishi
- From the Departments of Radiology and Medical Imaging (P.D., P.P.B., S.H.P.)
| | - P P Batchala
- From the Departments of Radiology and Medical Imaging (P.D., P.P.B., S.H.P.)
| | | | - L M Poisson
- Department of Public Health (L.M.P.), Henry Ford Health System, Detroit, Michigan
| | - M-B Lopes
- Pathology, Divisions of Neuropathology and Molecular Diagnostics (M.-B.L.)
| | - R Jain
- Departments of Radiology (R.J.) and Neurosurgery (R.J.), New York University School of Medicine, New York, New York
| | - C E Fadul
- Division of Neuro-Oncology (C.E.F., D.S.), University of Virginia Health System, Charlottesville, Virginia
| | - D Schiff
- Division of Neuro-Oncology (C.E.F., D.S.), University of Virginia Health System, Charlottesville, Virginia
| | - S H Patel
- From the Departments of Radiology and Medical Imaging (P.D., P.P.B., S.H.P.)
| |
Collapse
|
15
|
Matsui Y, Maruyama T, Nitta M, Saito T, Tsuzuki S, Tamura M, Kusuda K, Fukuya Y, Asano H, Kawamata T, Masamune K, Muragaki Y. Prediction of lower-grade glioma molecular subtypes using deep learning. J Neurooncol 2019; 146:321-327. [PMID: 31865510 DOI: 10.1007/s11060-019-03376-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/18/2019] [Indexed: 12/16/2022]
Abstract
INTRODUCTION It is useful to know the molecular subtype of lower-grade gliomas (LGG) when deciding on a treatment strategy. This study aims to diagnose this preoperatively. METHODS A deep learning model was developed to predict the 3-group molecular subtype using multimodal data including magnetic resonance imaging (MRI), positron emission tomography (PET), and computed tomography (CT). The performance was evaluated using leave-one-out cross validation with a dataset containing information from 217 LGG patients. RESULTS The model performed best when the dataset contained MRI, PET, and CT data. The model could predict the molecular subtype with an accuracy of 96.6% for the training dataset and 68.7% for the test dataset. The model achieved test accuracies of 58.5%, 60.4%, and 59.4% when the dataset contained only MRI, MRI and PET, and MRI and CT data, respectively. The conventional method used to predict mutations in the isocitrate dehydrogenase (IDH) gene and the codeletion of chromosome arms 1p and 19q (1p/19q) sequentially had an overall accuracy of 65.9%. This is 2.8 percent point lower than the proposed method, which predicts the 3-group molecular subtype directly. CONCLUSIONS A deep learning model was developed to diagnose the molecular subtype preoperatively based on multi-modality data in order to predict the 3-group classification directly. Cross-validation showed that the proposed model had an overall accuracy of 68.7% for the test dataset. This is the first model to double the expected value for a 3-group classification problem, when predicting the LGG molecular subtype.
Collapse
Affiliation(s)
- Yutaka Matsui
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
- Technical Department, Atom Medical Corporation, Tokyo, Japan
| | - Takashi Maruyama
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
- Department of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Masayuki Nitta
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
- Department of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Taiichi Saito
- Department of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Shunsuke Tsuzuki
- Department of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Manabu Tamura
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
- Department of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Kaori Kusuda
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Yasukazu Fukuya
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Hidetsugu Asano
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
- Technical Department, Atom Medical Corporation, Tokyo, Japan
| | - Takakazu Kawamata
- Department of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Ken Masamune
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Yoshihiro Muragaki
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
- Department of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|