1
|
Badve C, Nirappel A, Lo S, Orringer DA, Olson JJ. Congress of neurological surgeons systematic review and evidence-based guidelines for the role of imaging in newly diagnosed WHO grade II diffuse glioma in adults: update. J Neurooncol 2025:10.1007/s11060-025-05043-8. [PMID: 40338482 DOI: 10.1007/s11060-025-05043-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/09/2025] [Indexed: 05/09/2025]
Abstract
TARGET POPULATION Adult patients with suspected or histologically proven WHO Grade II diffuse glioma. QUESTION 1: In adult patients with suspected or histologically proven WHO Grade II diffuse glioma, do advanced MRI techniques using magnetic resonance spectroscopy, perfusion weighted imaging or diffusion weighted imaging provide superior assessment of tumor grade, margins, progression, treatment-related effects, and prognosis compared to standard neuroimaging? RECOMMENDATION Level II: The use of diffusion imaging and dynamic susceptibility contrast (DSC), dynamic contrast enhancement (DCE) and arterial spin labeling (ASL) sequences are suggested to differentiate WHO Grade II diffuse glioma from higher grade gliomas when this is not accomplished by T2 weighted and pre- and post-gadolinium contrast enhanced T1 weighted imaging. LEVEL III The use of diffusion and perfusion is suggested for obtaining information in genomics, prognosis, and post treatment monitoring when this information would be of value to the clinician and is not obtained through other methods. LEVEL III The use of MR Spectroscopy is suggested to differentiate WHO Grade II diffuse glioma from higher grade gliomas when this is not accomplished by standard MRI, perfusion and diffusion techniques and when such information would be of value to the clinician. QUESTION 2: In adult patients with suspected or histologically proven WHO Grade II diffuse glioma, does molecular imaging using amino acid PET tracers provide superior assessment of tumor grade, margins, progression, treatment-related effects, and prognosis compared to standard neuroimaging? RECOMMENDATION Level III: If not already evident by MRI studies, the addition of amino acid PET with FET and FDOPA as a tracer is suggested to help determine if a brain lesion is a low grade glioma or high grade glioma. LEVEL III If the standard clinical prognostic parameters are unclear and novel PET tracers are available, the clinician may consider FET to assist in determination of prognosis in an individual with grade II diffuse glioma. LEVEL III Clinicians may use FDOPA PET in addition to MRI if additional information is required for detection of tumor progression.
Collapse
Affiliation(s)
- Chaitra Badve
- University Hospitals Cleveland Medical Center, Cleveland, USA.
- Department of Radiology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - Abraham Nirappel
- Department of Radiology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Simon Lo
- Department of Radiation Oncology, University of Washington, Seattle, WA, USA
| | - Daniel A Orringer
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Júdice de Mattos Farina EM, Kuriki PEDA. Seeing the Unseen: How Unsupervised Learning Can Predict Genetic Mutations from Radiologic Images. Radiol Artif Intell 2025; 7:e250243. [PMID: 40366245 DOI: 10.1148/ryai.250243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
|
3
|
Awais M, Rehman A, Bukhari SS. Advances in liquid biopsy and virtual biopsy for care of patients with glioma: a narrative review. Expert Rev Anticancer Ther 2025; 25:529-550. [PMID: 40183671 DOI: 10.1080/14737140.2025.2489629] [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: 01/19/2025] [Accepted: 04/02/2025] [Indexed: 04/05/2025]
Abstract
INTRODUCTION The World Health Organization's 2021 classification of central nervous system neoplasms incorporated molecular and genetic features for classifying gliomas. Classification of gliomas located in deep-seated structures became a clinical conundrum given the absence of crucial pathological and molecular data. Advances in noninvasive imaging modalities offered virtual biopsy as a novel solution to this problem by identifying surrogate radiomic signatures. Liquid biopsies of blood or cerebrospinal fluid provided another enormous opportunity for identifying genomic, metabolomic and proteomic signatures. AREAS COVERED We summarize and appraise the current state of evidence with regards to virtual biopsy and liquid biopsy in the care of patients with gliomas. PubMed, Embase and Google Scholar were searched on 7/30/2024 for relevant articles published after the year 2013 in the English language. EXPERT OPINION A large body of preclinical and preliminary clinical evidence suggests that virtual biopsy is possible with the combined use of multiple novel imaging modalities in conjunction with machine learning and radiomics. Likewise, liquid biopsy in conjunction with focused ultrasound may be a valuable tool to obtain proteomic and genomic data regarding glioma in a minimally invasive manner. These modalities will likely become an integral part of care for patients with glioma in the future.
Collapse
Affiliation(s)
- Muhammad Awais
- Department of Radiology, The Aga Khan University, Karachi, Pakistan
| | - Abdul Rehman
- Department of Medicine, Tidal Health Peninsula Regional, Salisbury, MD, USA
| | - Syed Sarmad Bukhari
- Department of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
4
|
Roh YH, Cheong EN, Park JE, Choi Y, Jung SC, Song SW, Kim YH, Hong CK, Kim JH, Kim HS. Imaging-Based Molecular Characterization of Adult-Type Diffuse Glioma Using Diffusion and Perfusion MRI in Pre- and Post-Treatment Stage Considering Spatial and Temporal Heterogeneity. J Magn Reson Imaging 2025. [PMID: 40197845 DOI: 10.1002/jmri.29781] [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: 11/05/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Imaging-based molecular characterization is important for identifying treatment targets in adult-type diffuse gliomas. PURPOSE To assess isocitrate dehydrogenase (IDH) mutation and epidermal growth factor receptor (EGFR) amplification status in primary and recurrent gliomas using diffusion and perfusion MRI, addressing spatial and temporal heterogeneity. STUDY TYPE Retrospective. SUBJECTS Three-hundred and twelve newly diagnosed (cross-sectional set, 57.9 ± 13.2 years, 52.2% male, 235 IDH-wildtype, 71 EGFR-amplified) and 38 recurrent (longitudinal set, 53.1 ± 13.4 years, 44.7% male, 30 IDH-wildtype, 13 EGFR-amplified) adult-type diffuse glioma patients. FIELD STRENGTH/SEQUENCE 3.0T; diffusion weighted and dynamic susceptibility contrast-perfusion weighted imaging. ASSESSMENT Radiomics features from contrast-enhancing tumors (CET) and non-enhancing lesions (NEL) were extracted from apparent diffusion coefficient and perfusion maps. Spatial heterogeneity was assessed using intersection and Bhattacharyya distance between CET and NEL. Stable imaging features were identified in patients with unchanged genetic profiles in the longitudinal set. The "best model," using features from the cross-sectional set (n = 312), and the "concordant model," using stable features identified in the longitudinal set (n = 38), were constructed using the LASSO for IDH and EGFR status. STATISTICAL TESTS The area under the receiver-operating-characteristic curve (AUC). RESULTS For IDH mutations, both best and concordant models demonstrated high AUCs in the cross-sectional set (0.936; 95% confidence interval [CI]: 0.903-0.969 and 0.964 [0.943-0.986], respectively). Only the concordant model maintained strong performance in recurrent tumors (AUC, 0.919 vs. 0.656). For EGFR amplification in IDH-wildtype, the best and concordant models showed AUCs of 0.821 (95% CI: 0.761-0.881) and 0.746 (95% CI: 0.675-0.817) in newly diagnosed gliomas, but poor performance in recurrent tumors with AUCs of 0.503 (95% CI: 0.34-0.665) and 0.518 (95% CI: 0.357-0.678). DATA CONCLUSION Diffusion and perfusion MRI characterized IDH status in both newly diagnosed and recurrent gliomas, but showed limited diagnostic performance for EGFR, especially for recurrent tumors. EVIDENCE LEVEL 3 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Yun Hwa Roh
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - E-Nae Cheong
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Yangsean Choi
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Seung Chai Jung
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Sang Woo Song
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Young-Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Chang-Ki Hong
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jeong Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
5
|
Ganesan G, Rangasami R, Chandrasekharan A, Marreddy S, Ramachandran R. Role of Advanced Magnetic Resonance Imaging in Differentiating among Glioma Subtypes and Predicting Tumor-Proliferative Behavior. Asian J Neurosurg 2025; 20:34-42. [PMID: 40041591 PMCID: PMC11875706 DOI: 10.1055/s-0044-1790508] [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] [Indexed: 03/06/2025] Open
Abstract
Objective Gliomas are a devastating and heterogeneous group of primary brain tumors. Previously, the source of glioma was undetermined. Recent literature indicates that neural stem cells, or progenitors, are proposed to be the source of glioma. The prognosis of different types of gliomas differs due to their various biological tissue types. Besides the histological grade, the two useful immunohistochemistry markers that show the tumor's biological behavior are isocitrate dehydrogenase (IDH) labeling and the K i -67 labeling index. We sought to determine the magnetic resonance imaging (MRI) characteristics associated with IDH mutational status and ascertain whether MRI combined with IDH mutational status, can better predict the clinical outcomes of gliomas. Materials and Methods This period study was conducted in the Department of Radiology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India for 5 years (May 2016-May 2021). The study cohort included 30 patients diagnosed with gliomas who underwent preoperative MRI followed by surgical resection and histopathological examination. Preoperative MRI images were done to assess qualitative tumor characteristics such as location, margin of tumor, extent, cortical involvement, cystic component, mineralization or hemorrhage, and contrast enhancement. Discussion Differences in MRI features between IDH-mutant (MT) and IDH-wild-type (WT) groups were analyzed using the chi-square test for categorical variables and the Mann-Whitney U test for continuous variables. Statistical analysis was conducted using SPSS software. Results Among the 30 patients evaluated, 18 had IDH-WT and 12 had IDH-MT type gliomas. Male predominance (73.33%) was noted in our study. Brainstem location, indistinct borders (83.33%), less cortical involvement (72.22%), less cystic changes (88.89%), more area of necrotic component (44.44%), significantly increased choline/creatine (Cho/Cr) ratio, and choline/N-acetyl aspartate (Cho/NAA) ratio favors IDH-WT tumors. Positive T2-fluid-attenuated inversion recovery mismatch sign is more frequently seen in IDH-MT (7/12; 58.33%) tumors than in IDH-WT (4/18; 22.22%) tumors. Whereas well-defined contours (66.67%), more cortical involvement (83.33%), more cystic changes (58.33%), and less area of necrotic component favor IDH-MT type tumors. Conclusion MRI is a very promising and valuable tool for differentiating among glioma subtypes and predicting tumor-proliferative behavior in glioma cases. The combination of MRI characteristics with IDH mutation status enhances the predictive accuracy for clinical outcomes in glioma patients. This approach could potentially guide treatment planning and improve prognostic assessments.
Collapse
Affiliation(s)
- Gunalan Ganesan
- Department of Radiology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Rajeswaran Rangasami
- Department of Radiology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Anupama Chandrasekharan
- Department of Radiology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Sahithi Marreddy
- Department of Radiology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Rajoo Ramachandran
- Department of Radiology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| |
Collapse
|
6
|
Yan J, Guo C, Zheng H, Li Y, Duan M, Zhang C, Cui L, Lv X, Fu G, Cheng J. Noninvasive prediction of BRAF V600E mutation status of pleomorphic xanthoastrocytomas with MRI morphologic features and diffusion-weighted imaging. BMC Cancer 2024; 24:1022. [PMID: 39160463 PMCID: PMC11331820 DOI: 10.1186/s12885-024-12713-9] [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: 10/07/2023] [Accepted: 07/26/2024] [Indexed: 08/21/2024] Open
Abstract
OBJECTIVES Seeking a noninvasive predictor for BRAF V600E mutation status of pleomorphic xanthoastrocytomas (PXAs) is essential for their prognoses and therapeutic use of BRAF inhibitors. We aimed to noninvasively diagnose BRAF V600E-mutated PXAs using MRI morphologic, DWI and clinical parameters. METHODS The clinical findings, anatomical MRI characteristics, and diffusion parameters of 36 pathologically confirmed PXAs were retrospectively analyzed, and BRAF V600E-mutated (n = 16) and wild-type (n = 20) groups were compared. A binary logistic-regression analysis was performed, and a ROC curve was calculated to determine the independent predictors of BRAF V600E mutation status, diagnostic accuracy, and optimal cut-off value. RESULTS A comparison of findings between groups showed that BRAF V600E-mutated PXAs were more frequent in children and young adults (≤ 35 years; P = 0.042) who often had histories of seizures (P = 0.004). Furthermore, BRAF V600E-mutated PXAs generally presented as solitary masses (P = 0.024), superficial locations with meningeal attachment (P < 0.001), predominantly cystic with mural nodules (P = 0.005), and had greater minimal ADC ratio (ADCratio) values of the tumor and peritumoral edema (P < 0.001). Binary logistic regression showed that age ≤ 35 years, solitary mass, superficial locations with meningeal attachment, and a greater minimal ADCratio of the tumor were independent predictors of BRAF V600E-mutated PXAs. The combination of all four independent predictors resulted in the highest sensitivity (100%) and specificity (90%), with AUC = 0.984. CONCLUSION The BRAF V600E mutation status of PXAs could be noninvasively predicted using clinical and MRI characteristics. CRITICAL RELEVANCE STATEMENT The noninvasive diagnostic criteria for BRAF V600E-mutated PXAs could offer guidance for the administration of BRAF V600E mutation inhibitors in the future.
Collapse
Affiliation(s)
- Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Road 1, Zhengzhou, Henan Province, 450052, China
| | - Cuiping Guo
- Department of Radiology, Children's Hospital of Hangzhou, 195 Wenhui Road, Hangzhou, Zhejiang Province, 310014, China
| | - Hongwei Zheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Road 1, Zhengzhou, Henan Province, 450052, China
| | - Yinhua Li
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Road 1, Zhengzhou, Henan Province, 450052, China
| | - Mengjiao Duan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Road 1, Zhengzhou, Henan Province, 450052, China
| | - Chaoli Zhang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Road 1, Zhengzhou, Henan Province, 450052, China
| | - Li Cui
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Road 1, Zhengzhou, Henan Province, 450052, China
| | - Xiaofei Lv
- Department of Medical Imaging, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong Province, 510060, China
| | - Gui Fu
- Department of Medical Imaging, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong Province, 510060, China.
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Road 1, Zhengzhou, Henan Province, 450052, China.
| |
Collapse
|
7
|
Han W, Zhou H, Zhang X, Li H, Han X, Su L, Tian L, Xue X. HMGB2 is a biomarker associated with poor prognosis promoting radioresistance in glioma by targeting base excision repair pathway. Transl Oncol 2024; 45:101977. [PMID: 38728871 PMCID: PMC11107350 DOI: 10.1016/j.tranon.2024.101977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/27/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND High mobility group box 2 (HMGB2) is considered as a biomarker of poor prognosis in various cancers.This study aims to investigate the effect and mechanism of HMGB2 in gliomas. METHODS With the glioma related on-line and our local hospital databases, the expression differences of HMGB2,Kaplan-Meier survival analysis and COX regression analysis were performed.The correlation analysis between the clinicopathological features and imaging parameters with the HMGB2 expression had been done. Then GSEA and PPI networks were carried out to find out the most significant pathway. The pathway inhibitor was applied to verify HMGB2's participation. CCK8,EDU assays,γ-H2AX immunofluorescence staining and colony formation assay were conducted to observe effects on glioma cells. RESULTS Available datasets showed that HMGB2 was highly expressed in glioma and patients with high expression of HMGB2 had poorer prognosis and molecular characteristics. Protein level evidence of western blot and immunohistochemistry from our center supported the conclusions above. Analysis on imaging features suggested that HMGB2 expression level had an inverse association with ADCmean but positively with the thickness of enhancing margin. Results from GSEA and PPI network analysis exhibited that HMGB2 was involved in base excision repair (BER) signaling pathway. Experimental evidence demonstrated that the overexpression of HMGB2 promoted the proliferation of glioma cells and enhanced the radio-resistance. CONCLUSIONS HMGB2 could promote glioma development and enhance the radioresistance of glioma cells, potentially related to the BER pathway, suggesting it may serve as an underlying biomarker for patients with glioma.
Collapse
Affiliation(s)
- Wei Han
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Huandi Zhou
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xinyuan Zhang
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Haonan Li
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xuetao Han
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Linlin Su
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Tian
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoying Xue
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
8
|
Truong NCD, Bangalore Yogananda CG, Wagner BC, Holcomb JM, Reddy D, Saadat N, Hatanpaa KJ, Patel TR, Fei B, Lee MD, Jain R, Bruce RJ, Pinho MC, Madhuranthakam AJ, Maldjian JA. Two-Stage Training Framework Using Multicontrast MRI Radiomics for IDH Mutation Status Prediction in Glioma. Radiol Artif Intell 2024; 6:e230218. [PMID: 38775670 PMCID: PMC11294953 DOI: 10.1148/ryai.230218] [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: 06/26/2023] [Revised: 03/18/2024] [Accepted: 04/25/2024] [Indexed: 06/21/2024]
Abstract
Purpose To develop a radiomics framework for preoperative MRI-based prediction of isocitrate dehydrogenase (IDH) mutation status, a crucial glioma prognostic indicator. Materials and Methods Radiomics features (shape, first-order statistics, and texture) were extracted from the whole tumor or the combination of nonenhancing, necrosis, and edema regions. Segmentation masks were obtained via the federated tumor segmentation tool or the original data source. Boruta, a wrapper-based feature selection algorithm, identified relevant features. Addressing the imbalance between mutated and wild-type cases, multiple prediction models were trained on balanced data subsets using random forest or XGBoost and assembled to build the final classifier. The framework was evaluated using retrospective MRI scans from three public datasets (The Cancer Imaging Archive [TCIA, 227 patients], the University of California San Francisco Preoperative Diffuse Glioma MRI dataset [UCSF, 495 patients], and the Erasmus Glioma Database [EGD, 456 patients]) and internal datasets collected from the University of Texas Southwestern Medical Center (UTSW, 356 patients), New York University (NYU, 136 patients), and University of Wisconsin-Madison (UWM, 174 patients). TCIA and UTSW served as separate training sets, while the remaining data constituted the test set (1617 or 1488 testing cases, respectively). Results The best performing models trained on the TCIA dataset achieved area under the receiver operating characteristic curve (AUC) values of 0.89 for UTSW, 0.86 for NYU, 0.93 for UWM, 0.94 for UCSF, and 0.88 for EGD test sets. The best performing models trained on the UTSW dataset achieved slightly higher AUCs: 0.92 for TCIA, 0.88 for NYU, 0.96 for UWM, 0.93 for UCSF, and 0.90 for EGD. Conclusion This MRI radiomics-based framework shows promise for accurate preoperative prediction of IDH mutation status in patients with glioma. Keywords: Glioma, Isocitrate Dehydrogenase Mutation, IDH Mutation, Radiomics, MRI Supplemental material is available for this article. Published under a CC BY 4.0 license. See also commentary by Moassefi and Erickson in this issue.
Collapse
Affiliation(s)
- Nghi C. D. Truong
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Chandan Ganesh Bangalore Yogananda
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Benjamin C. Wagner
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - James M. Holcomb
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Divya Reddy
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Niloufar Saadat
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Kimmo J. Hatanpaa
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Toral R. Patel
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Baowei Fei
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Matthew D. Lee
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Rajan Jain
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Richard J. Bruce
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Marco C. Pinho
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Ananth J. Madhuranthakam
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| | - Joseph A. Maldjian
- From the Departments of Radiology (N.C.D.T., C.G.B.Y., B.C.W.,
J.M.H., D.R., N.S., B.F., M.C.P., A.J.M., J.A.M.), Pathology (K.J.H.), and
Neurologic Surgery (T.R.P.), The University of Texas Southwestern Medical
Center, 5323 Harry Hines Blvd, Dallas, TX 75390; Department of Bioengineering,
The University of Texas at Dallas, Richardson, Tex (B.F.); Departments of
Radiology (M.D.L., R.J.) and Neurosurgery (R.J.), New York University Grossman
School of Medicine, New York, NY; and Department of Radiology, University of
Wisconsin–Madison, Madison, Wis (R.J.B.)
| |
Collapse
|
9
|
Park CJ, Kim S, Han K, Ahn SS, Kim D, Park YW, Chang JH, Kim SH, Lee SK. Diffusion- and Perfusion-Weighted MRI Radiomics for Survival Prediction in Patients with Lower-Grade Gliomas. Yonsei Med J 2024; 65:283-292. [PMID: 38653567 PMCID: PMC11045349 DOI: 10.3349/ymj.2023.0323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/27/2023] [Accepted: 12/13/2023] [Indexed: 04/25/2024] Open
Abstract
PURPOSE Lower-grade gliomas of histologic grades 2 and 3 follow heterogenous clinical outcomes, which necessitates risk stratification. This study aimed to evaluate whether diffusion-weighted and perfusion-weighted MRI radiomics allow overall survival (OS) prediction in patients with lower-grade gliomas and investigate its prognostic value. MATERIALS AND METHODS In this retrospective study, radiomic features were extracted from apparent diffusion coefficient, relative cerebral blood volume map, and Ktrans map in patients with pathologically confirmed lower-grade gliomas (January 2012-February 2019). The radiomics risk score (RRS) calculated from selected features constituted a radiomics model. Multivariable Cox regression analysis, including clinical features and RRS, was performed. The models' integrated area under the receiver operating characteristic curves (iAUCs) were compared. The radiomics model combined with clinical features was presented as a nomogram. RESULTS The study included 129 patients (median age, 44 years; interquartile range, 37-57 years; 63 female): 90 patients for training set and 39 patients for test set. The RRS was an independent risk factor for OS with a hazard ratio of 6.01. The combined clinical and radiomics model achieved superior performance for OS prediction compared to the clinical model in both training (iAUC, 0.82 vs. 0.72, p=0.002) and test sets (0.88 vs. 0.76, p=0.04). The radiomics nomogram combined with clinical features exhibited good agreement between the actual and predicted OS with C-index of 0.83 and 0.87 in the training and test sets, respectively. CONCLUSION Adding diffusion- and perfusion-weighted MRI radiomics to clinical features improved survival prediction in lower-grade glioma.
Collapse
Affiliation(s)
- Chae Jung Park
- Department of Radiology, Research Institute of Radiological Science, Yongin Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sooyon Kim
- Department of Applied Statistics, Yonsei University, Seoul, Korea
| | - Kyunghwa Han
- Department of Radiology, Center for Clinical Imaging Data Science, Research Institute of Radiological Sciences, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Soo Ahn
- Department of Radiology, Center for Clinical Imaging Data Science, Research Institute of Radiological Sciences, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| | - Dain Kim
- Graduate School of Artificial Intelligence, Pohang University of Science and Technology, Pohang, Korea
| | - Yae Won Park
- Department of Radiology, Center for Clinical Imaging Data Science, Research Institute of Radiological Sciences, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Koo Lee
- Department of Radiology, Center for Clinical Imaging Data Science, Research Institute of Radiological Sciences, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
10
|
Pons-Escoda A, Majos C, Smits M, Oleaga L. Presurgical diagnosis of diffuse gliomas in adults: Post-WHO 2021 practical perspectives from radiologists in neuro-oncology units. RADIOLOGIA 2024; 66:260-277. [PMID: 38908887 DOI: 10.1016/j.rxeng.2024.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/31/2023] [Indexed: 06/24/2024]
Abstract
The 2021 World Health Organization classification of CNS tumours was greeted with enthusiasm as well as an initial potential overwhelm. However, with time and experience, our understanding of its key aspects has notably improved. Using our collective expertise gained in neuro-oncology units in hospitals in different countries, we have compiled a practical guide for radiologists that clarifies the classification criteria for diffuse gliomas in adults. Its format is clear and concise to facilitate its incorporation into everyday clinical practice. The document includes a historical overview of the classifications and highlights the most important recent additions. It describes the main types in detail with an emphasis on their appearance on imaging. The authors also address the most debated issues in recent years. It will better prepare radiologists to conduct accurate presurgical diagnoses and collaborate effectively in clinical decision making, thus impacting decisions on treatment, prognosis, and overall patient care.
Collapse
Affiliation(s)
- A Pons-Escoda
- Radiology Department, Hospital Universitari de Bellvitge, Barcelona, Spain; Facultat de Medicina i Ciencies de La Salut, Universitat de Barcelona (UB), Barcelona, Spain.
| | - C Majos
- Radiology Department, Hospital Universitari de Bellvitge, Barcelona, Spain; Neuro-Oncology Unit, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, Barcelona, Spain; Diagnostic Imaging and Nuclear Medicine Research Group, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, Barcelona, Spain
| | - M Smits
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands; Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, The Netherlands; Medical Delta, Delft, The Netherlands
| | - L Oleaga
- Radiology Department, Hospital Clínic Barcelona, Barcelona, Spain
| |
Collapse
|
11
|
Zhou W, Wen J, Huang Q, Zeng Y, Zhou Z, Zhu Y, Chen L, Guan Y, Xie F, Zhuang D, Hua T. Development and validation of clinical-radiomics analysis for preoperative prediction of IDH mutation status and WHO grade in diffuse gliomas: a consecutive L-[methyl-11C] methionine cohort study with two PET scanners. Eur J Nucl Med Mol Imaging 2024; 51:1423-1435. [PMID: 38110710 DOI: 10.1007/s00259-023-06562-0] [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: 09/04/2023] [Accepted: 12/04/2023] [Indexed: 12/20/2023]
Abstract
PURPOSE Determination of isocitrate dehydrogenase (IDH) genotype is crucial in the stratification of diagnosis and prognostication in diffuse gliomas. We sought to build and validate radiomics models and clinical features incorporated nomogram for preoperative prediction of IDH mutation status and WHO grade of diffuse gliomas with L-[methyl-11C] methionine ([11C]MET) PET/CT imaging according to the 2016 WHO classification of tumors of the central nervous system. METHODS Consecutive 178 preoperative [11C]MET PET/CT images were retrospectively studied for radiomics analysis. One hundred six patients from PET scanner 1 were used as training dataset, and 72 patients from PET scanner 2 were used for validation dataset. [11C]MET PET and integrated CT radiomics features were extracted, respectively; three independent predictive models were built based on PET features, CT features, and combined PET/CT features, respectively. The SelectKBest method, Spearman correlation analysis, Least Absolute Shrinkage and Selection Operator (LASSO) regression, and machine learning algorithms were applied for feature selection and model building. After filtering the satisfactory predictive model, key clinical features were incorporated for the nomogram establishment. RESULTS The combined [11C]MET PET/CT radiomics model, which consisted of four PET features and eight integrated CT features, was significantly associated with IDH genotype (p < 0.0001 for both training and validation datasets). Nomogram based on the [11C]MET PET/CT radiomics score, patients' age, and dichotomous tumor location status showed satisfactory discrimination capacity, and the AUC was 0.880 (95% CI, 0.726-0.998) in the training dataset and 0.866 (95% CI, 0.777-0.956) in the validation dataset. In IDH stratified WHO grade prediction, the final radiomics model consists of four PET features and two CT features had reasonable and stable differential efficacy of WHO grade II and III patients from grade IV patients in IDH-wildtype patients, and the AUC was 0.820 (95% CI, 0.541-1.000) in the training dataset and 0.766 (95% CI, 0.612-0.921) in the validation dataset. CONCLUSION [11C]MET PET radiomics features could benefit non-invasive IDH genotype prediction, and integrated CT radiomics features could enhance the efficacy. Radiomics and clinical features incorporation could establish satisfactory nomogram for clinical application. This non-invasive predictive investigation based on our consecutive cohort from two PET scanners could provide the perspective to observe the differential efficacy and the stability of radiomics-based investigation in untreated diffuse gliomas.
Collapse
Affiliation(s)
- Weiyan Zhou
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianbo Wen
- Department of Radiology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Huang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yan Zeng
- Department of Research Center, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Zhirui Zhou
- Radiation Oncology Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuhua Zhu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Lei Chen
- Department of Research Center, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Dongxiao Zhuang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| | - Tao Hua
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Mendes Serrão E, Klug M, Moloney BM, Jhaveri A, Lo Gullo R, Pinker K, Luker G, Haider MA, Shinagare AB, Liu X. Current Status of Cancer Genomics and Imaging Phenotypes: What Radiologists Need to Know. Radiol Imaging Cancer 2023; 5:e220153. [PMID: 37921555 DOI: 10.1148/rycan.220153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Ongoing discoveries in cancer genomics and epigenomics have revolutionized clinical oncology and precision health care. This knowledge provides unprecedented insights into tumor biology and heterogeneity within a single tumor, among primary and metastatic lesions, and among patients with the same histologic type of cancer. Large-scale genomic sequencing studies also sparked the development of new tumor classifications, biomarkers, and targeted therapies. Because of the central role of imaging in cancer diagnosis and therapy, radiologists need to be familiar with the basic concepts of genomics, which are now becoming the new norm in oncologic clinical practice. By incorporating these concepts into clinical practice, radiologists can make their imaging interpretations more meaningful and specific, facilitate multidisciplinary clinical dialogue and interventions, and provide better patient-centric care. This review article highlights basic concepts of genomics and epigenomics, reviews the most common genetic alterations in cancer, and discusses the implications of these concepts on imaging by organ system in a case-based manner. This information will help stimulate new innovations in imaging research, accelerate the development and validation of new imaging biomarkers, and motivate efforts to bring new molecular and functional imaging methods to clinical radiology. Keywords: Oncology, Cancer Genomics, Epignomics, Radiogenomics, Imaging Markers Supplemental material is available for this article. © RSNA, 2023.
Collapse
Affiliation(s)
- Eva Mendes Serrão
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Maximiliano Klug
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Brian M Moloney
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Aaditeya Jhaveri
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Roberto Lo Gullo
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Katja Pinker
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Gary Luker
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Masoom A Haider
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Atul B Shinagare
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Xiaoyang Liu
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| |
Collapse
|
13
|
Pan T, Su CQ, Tang WT, Lin J, Lu SS, Hong XN. Combined texture analysis of dynamic contrast-enhanced MRI with histogram analysis of diffusion kurtosis imaging for predicting IDH mutational status in gliomas. Acta Radiol 2023; 64:2552-2560. [PMID: 37331987 DOI: 10.1177/02841851231180291] [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] [Indexed: 06/20/2023]
Abstract
BACKGROUND Non-invasive detection of isocitrate dehydrogenase (IDH) mutational status in gliomas is clinically meaningful for molecular stratification of glioma; however, it remains challenging. PURPOSE To investigate the usefulness of texture analysis (TA) of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and histogram analysis of diffusion kurtosis imaging (DKI) maps for evaluating IDH mutational status in gliomas. MATERIAL AND METHODS This retrospective study enrolled 84 patients with histologically confirmed gliomas, comprising IDH-mutant (n = 34) and IDH-wildtype (n = 50). TA was performed for the quantitative parameters derived by DCE-MRI. Histogram analysis was performed for the quantitative parameters derived by DKI. Unpaired Student's t-test was used to identify IDH-mutant and IDH-wildtype gliomas. Logistic regression and receiver operating characteristic (ROC) curve analyses were used to compare the diagnostic performance of each parameter and their combination for predicting the IDH mutational status in gliomas. RESULTS Significant statistical differences in the TA of DCE-MRI and histogram analysis of DKI were observed between IDH-mutant and IDH-wildtype gliomas (all P < 0.05). Using multivariable logistic regression, the entropy of Ktrans, skewness of Ve, and Kapp-90th had higher prediction potential for IDH mutations with areas under the ROC curve (AUC) of 0.915, 0.735, and 0.830, respectively. A combination of these analyses for the identification of IDH mutation improved the AUC to 0.978, with a sensitivity and specificity of 94.1% and 96.0%, respectively, which was higher than the single analysis (P < 0.05). CONCLUSION Integrating the TA of DCE-MRI and histogram analysis of DKI may help to predict the IDH mutational status.
Collapse
Affiliation(s)
- Ting Pan
- Department of Radiology, Northern Jiangsu People's Hospital, Clinical Medical School of Yangzhou University, Yangzhou, PR China
| | - Chun-Qiu Su
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Wen-Tian Tang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Jie Lin
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Shan-Shan Lu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Xun-Ning Hong
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| |
Collapse
|
14
|
Yuzkan S, Mutlu S, Han M, Akkurt TS, Sencan F, Kusku Cabuk F, Gunaldi O, Tugcu B, Kocak B. Predicting Isocitrate Dehydrogenase Mutation Status of Grade 2-4 Gliomas with Diffusion Tensor Imaging (DTI) Parameters Derived from Model-Based DTI and Model-Free Q-Sampling Imaging Reconstructions. World Neurosurg 2023; 177:e580-e592. [PMID: 37390902 DOI: 10.1016/j.wneu.2023.06.099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/02/2023]
Abstract
OBJECTIVE To determine whether diffusion tensor imaging (DTI) parameters acquired with model-based DTI and model-free generalized Q-sampling imaging (GQI) reconstructions may noninvasively predict isocitrate dehydrogenase (IDH) mutational status in patients with grade 2-4 gliomas. METHODS Forty patients with known IDH genotype (28 IDH wild-type; 12 IDH mutant) who underwent preoperative DTI evaluation on a 3-Tesla magnetic resonance imaging scanner were analyzed retrospectively. Absolute values obtained from model-based and model-free reconstructions were compared. Using the intraclass correlation coefficient, interobserver agreement was assessed for various sampling techniques. Variables having statistically significant distributions between IDH groups were subjected to a receiver operating characteristic (ROC) analysis. Using multivariable logistic regression analysis, independent predictors, if present, were identified and a model was developed. RESULTS Six imaging parameters (3 from model-based DTI and 3 from model-free GQI reconstructions) showed statistically significant differences between groups (P < 0.001, power >0.97), with very high correlation to each other (P < 0.001). Age difference between the groups was statistically significant (P < 0.001). The optimal logistic regression model comprised a GQI-based parameter and age, which were independent predictors as well, producing an area under the ROC curve, accuracy, sensitivity, and specificity of 0.926, 85%, 75%, and 89.3%, respectively. Using the GQI reconstruction feature alone with a cut-off of 1.60, an 85% of accuracy was also achieved with ROC analysis. CONCLUSIONS The imaging parameters acquired from model-based DTI and model-free GQI reconstructions, combined with the clinical variable age, may have the ability to noninvasively predict the IDH genotype in gliomas, either alone or in particular combinations.
Collapse
Affiliation(s)
- Sabahattin Yuzkan
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey.
| | - Samet Mutlu
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Mehmet Han
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Tuce Soylemez Akkurt
- Department of Pathology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Fahir Sencan
- Department of Neurosurgery, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Fatmagul Kusku Cabuk
- Department of Pathology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Omur Gunaldi
- Department of Neurosurgery, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Bekir Tugcu
- Department of Neurosurgery, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Burak Kocak
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| |
Collapse
|
15
|
Hervey-Jumper SL, Zhang Y, Phillips JJ, Morshed RA, Young JS, McCoy L, Lafontaine M, Luks T, Ammanuel S, Kakaizada S, Egladyous A, Gogos A, Villanueva-Meyer J, Shai A, Warrier G, Rice T, Crane J, Wrensch M, Wiencke JK, Daras M, Oberheim Bush NA, Taylor JW, Butowski N, Clarke J, Chang S, Chang E, Aghi M, Theodosopoulos P, McDermott M, Jakola AS, Kavouridis VK, Nawabi N, Solheim O, Smith T, Berger MS, Molinaro AM. Interactive Effects of Molecular, Therapeutic, and Patient Factors on Outcome of Diffuse Low-Grade Glioma. J Clin Oncol 2023; 41:2029-2042. [PMID: 36599113 PMCID: PMC10082290 DOI: 10.1200/jco.21.02929] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 08/18/2022] [Accepted: 11/14/2022] [Indexed: 01/06/2023] Open
Abstract
PURPOSE In patients with diffuse low-grade glioma (LGG), the extent of surgical tumor resection (EOR) has a controversial role, in part because a randomized clinical trial with different levels of EOR is not feasible. METHODS In a 20-year retrospective cohort of 392 patients with IDH-mutant grade 2 glioma, we analyzed the combined effects of volumetric EOR and molecular and clinical factors on overall survival (OS) and progression-free survival by recursive partitioning analysis. The OS results were validated in two external cohorts (n = 365). Propensity score analysis of the combined cohorts (n = 757) was used to mimic a randomized clinical trial with varying levels of EOR. RESULTS Recursive partitioning analysis identified three survival risk groups. Median OS was shortest in two subsets of patients with astrocytoma: those with postoperative tumor volume (TV) > 4.6 mL and those with preoperative TV > 43.1 mL and postoperative TV ≤ 4.6 mL. Intermediate OS was seen in patients with astrocytoma who had chemotherapy with preoperative TV ≤ 43.1 mL and postoperative TV ≤ 4.6 mL in addition to oligodendroglioma patients with either preoperative TV > 43.1 mL and residual TV ≤ 4.6 mL or postoperative residual volume > 4.6 mL. Longest OS was seen in astrocytoma patients with preoperative TV ≤ 43.1 mL and postoperative TV ≤ 4.6 mL who received no chemotherapy and oligodendroglioma patients with preoperative TV ≤ 43.1 mL and postoperative TV ≤ 4.6 mL. EOR ≥ 75% improved survival outcomes, as shown by propensity score analysis. CONCLUSION Across both subtypes of LGG, EOR beginning at 75% improves OS while beginning at 80% improves progression-free survival. Nonetheless, maximal resection with preservation of neurological function remains the treatment goal. Our findings have implications for surgical strategies for LGGs, particularly oligodendroglioma.
Collapse
Affiliation(s)
- Shawn L. Hervey-Jumper
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Yalan Zhang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Joanna J. Phillips
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Ramin A. Morshed
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Jacob S. Young
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Lucie McCoy
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Marisa Lafontaine
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Tracy Luks
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Simon Ammanuel
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Sofia Kakaizada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Andrew Egladyous
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Andrew Gogos
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Javier Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Anny Shai
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Gayathri Warrier
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Terri Rice
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Jason Crane
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - John K. Wiencke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Mariza Daras
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
- Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Nancy Ann Oberheim Bush
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
- Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Jennie W. Taylor
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
- Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Jennifer Clarke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
- Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Susan Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
- Department of Neurology, University of California, San Francisco, San Francisco, CA
| | - Edward Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Manish Aghi
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Philip Theodosopoulos
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Michael McDermott
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Asgeir S. Jakola
- Department of Neurological Surgery, St Olavs University Hospital, Trondheim, Norway
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience, University of Gothenburg, Sahlgrenska Academy, Gothenburg, Sweden
| | | | - Noah Nawabi
- Department of Neurological Surgery, Brigham and Women's Hospital, Boston, MA
| | - Ole Solheim
- Department of Neurological Surgery, St Olavs University Hospital, Trondheim, Norway
- Norwegian University of Science and Technology, Trondheim, Norway
| | - Timothy Smith
- Department of Neurological Surgery, Brigham and Women's Hospital, Boston, MA
| | - Mitchel S. Berger
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Annette M. Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
16
|
Laudicella R, Mantarro C, Catalfamo B, Alongi P, Gaeta M, Minutoli F, Baldari S, Bisdas S. PET Imaging in Gliomas. RADIOLOGY‐NUCLEAR MEDICINE DIAGNOSTIC IMAGING 2023:194-218. [DOI: 10.1002/9781119603627.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
17
|
Glioma radiogenomics and artificial intelligence: road to precision cancer medicine. Clin Radiol 2023; 78:137-149. [PMID: 36241568 DOI: 10.1016/j.crad.2022.08.138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/19/2022] [Indexed: 01/18/2023]
Abstract
Radiogenomics refers to the study of the relationship between imaging phenotypes and gene expression patterns/molecular characteristics, which might allow improved diagnosis, decision-making, and predicting patient outcomes in the context of multiple diseases. Central nervous system (CNS) tumours contribute to significant cancer-related mortality in the present age. Although historically CNS neoplasms were classified and graded based on microscopic appearance, there was discordance between two histologically similar tumours that showed varying prognosis and behaviour, attributable to their molecular signatures. These led to the incorporation of molecular markers in the classification of CNS neoplasms. Meanwhile, advancements in imaging technology such as diffusion-based imaging (including tractography), perfusion, and spectroscopy in addition to the conventional imaging of glial neoplasms, have opened an avenue for radiogenomics. This review touches upon the schema of the current classification of gliomas, concepts behind molecular markers, and parameters that are used in radiogenomics to characterise gliomas and the role of artificial intelligence for the same. Further, the role of radiomics in the grading of brain tumours, prediction of treatment response and prognosis has been discussed. Use of automated and semi-automated tumour segmentation for radiotherapy planning and follow-up has also been discussed briefly.
Collapse
|
18
|
Kibe Y, Motomura K, Ohka F, Aoki K, Shimizu H, Yamaguchi J, Nishikawa T, Saito R. Imaging features of localized IDH wild-type histologically diffuse astrocytomas: a single-institution case series. Sci Rep 2023; 13:23. [PMID: 36646712 PMCID: PMC9842655 DOI: 10.1038/s41598-022-25928-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 12/07/2022] [Indexed: 01/18/2023] Open
Abstract
Isocitrate dehydrogenase wild-type (IDHwt) diffuse astrocytomas feature highly infiltrative patterns, such as a gliomatosis cerebri growth pattern with widespread involvement. Among these tumors, localized IDHwt histologically diffuse astrocytomas are rarer than the infiltrative type. The aim of this study was to assess and describe the clinical, radiographic, histopathological, and molecular characteristics of this rare type of IDHwt histologically diffuse astrocytomas and thereby provide more information on how its features affect clinical prognoses and outcomes. We retrospectively analyzed the records of five patients with localized IDHwt histologically diffuse astrocytomas between July 2017 and January 2020. All patients were female, and their mean age at the time of the initial treatment was 55.0 years. All patients had focal disease that did not include gliomatosis cerebri or multifocal disease. All patients received a histopathological diagnosis of diffuse astrocytomas at the time of the initial treatment. For recurrent tumors, second surgeries were performed at a mean of 12.4 months after the initial surgery. A histopathological diagnosis of glioblastoma was made in four patients and one of gliosarcoma in one patient. The initial status of IDH1, IDH2, H3F3A, HIST1H3B, and BRAF was "wild-type" in all patients. TERT promoter mutations (C250T or C228T) were detected in four patients. No tumors harbored a 1p/19q codeletion, EGFR amplification, or chromosome 7 gain/10 loss (+ 7/ - 10). We assessed clinical cases of localized IDHwt histologically diffuse astrocytomas that resulted in malignant recurrence and a poor clinical prognosis similar to that of glioblastomas. Our case series suggests that even in patients with histologically diffuse astrocytomas and those who present with radiographic imaging findings suggestive of a localized tumor mass, physicians should consider the possibility of IDHwt histologically diffuse astrocytomas.
Collapse
Affiliation(s)
- Yuji Kibe
- grid.27476.300000 0001 0943 978XDepartment of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan
| | - Kazuya Motomura
- grid.27476.300000 0001 0943 978XDepartment of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan
| | - Fumiharu Ohka
- grid.27476.300000 0001 0943 978XDepartment of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan
| | - Kosuke Aoki
- grid.27476.300000 0001 0943 978XDepartment of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan
| | - Hiroyuki Shimizu
- grid.27476.300000 0001 0943 978XDepartment of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan
| | - Junya Yamaguchi
- grid.27476.300000 0001 0943 978XDepartment of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan
| | - Tomohide Nishikawa
- grid.27476.300000 0001 0943 978XDepartment of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan
| | - Ryuta Saito
- grid.27476.300000 0001 0943 978XDepartment of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550 Japan
| |
Collapse
|
19
|
Kamble AN, Agrawal NK, Koundal S, Bhargava S, Kamble AN, Joyner DA, Kalelioglu T, Patel SH, Jain R. Imaging-based stratification of adult gliomas prognosticates survival and correlates with the 2021 WHO classification. Neuroradiology 2023; 65:41-54. [PMID: 35876874 DOI: 10.1007/s00234-022-03015-7] [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: 03/31/2022] [Accepted: 07/08/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND Because of the lack of global accessibility, delay, and cost-effectiveness of genetic testing, there is a clinical need for an imaging-based stratification of gliomas that can prognosticate survival and correlate with the 2021-WHO classification. METHODS In this retrospective study, adult primary glioma patients with pre-surgery/pre-treatment MRI brain images having T2, FLAIR, T1, T1 post-contrast, DWI sequences, and survival information were included in TCIA training-dataset (n = 275) and independent validation-dataset (n = 200). A flowchart for imaging-based stratification of adult gliomas(IBGS) was created in consensus by three authors to encompass all adult glioma types. Diagnostic features used were T2-FLAIR mismatch sign, central necrosis with peripheral enhancement, diffusion restriction, and continuous cortex sign. Roman numerals (I, II, and III) denote IBGS types. Two independent teams of three and two radiologists, blinded to genetic, histology, and survival information, manually read MRI into three types based on the flowchart. Overall survival-analysis was done using age-adjusted Cox-regression analysis, which provided both hazard-ratio (HR) and area-under-curve (AUC) for each stratification system(IBGS and 2021-WHO). The sensitivity and specificity of each IBSG type were analyzed with cross-table to identify the corresponding 2021-WHO genotype. RESULTS Imaging-based stratification was statistically significant in predicting survival in both datasets with good inter-observer agreement (age-adjusted Cox-regression, AUC > 0.5, k > 0.6, p < 0.001). IBGS type-I, type-II, and type-III gliomas had good specificity in identifying IDHmut 1p19q-codel oligodendroglioma (training - 97%, validation - 85%); IDHmut 1p19q non-codel astrocytoma (training - 80%, validation - 85.9%); and IDHwt glioblastoma (training - 76.5%, validation- 87.3%) respectively (p-value < 0.01). CONCLUSIONS Imaging-based stratification of adult diffuse gliomas predicted patient survival and correlated well with 2021-WHO glioma classification.
Collapse
Affiliation(s)
- Akshaykumar N Kamble
- University Hospitals Coventry & Warwickshire, Coventry, UK.
- Deep Learning Institute of Radiological Sciences (DeLoRIS), Mumbai, India.
| | - Nidhi K Agrawal
- Deep Learning Institute of Radiological Sciences (DeLoRIS), Mumbai, India
- Max Super-Specialty Hospital, Mohali, India
| | - Surabhi Koundal
- Department of Radiology, Institute of Nuclear Medicine & Allied Sciences (INMAS), New Delhi, India
| | | | | | - David A Joyner
- Department of Radiology, University of Virginia Health System, Charlottesville, VA, USA
| | - Tuba Kalelioglu
- Department of Radiology, University of Virginia Health System, Charlottesville, VA, USA
| | - Sohil H Patel
- Department of Radiology, University of Virginia Health System, Charlottesville, VA, USA
| | - Rajan Jain
- Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
20
|
Sahu A, Patnam NG, Goda JS, Epari S, Sahay A, Mathew R, Choudhari AK, Desai SM, Dasgupta A, Chatterjee A, Pratishad P, Shetty P, Moiyadi AA, Gupta T. Multiparametric Magnetic Resonance Imaging Correlates of Isocitrate Dehydrogenase Mutation in WHO high-Grade Astrocytomas. J Pers Med 2022; 13:jpm13010072. [PMID: 36675733 PMCID: PMC9865247 DOI: 10.3390/jpm13010072] [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: 10/09/2022] [Revised: 12/18/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022] Open
Abstract
Purpose and background: Isocitrate dehydrogenase (IDH) mutation and O-6 methyl guanine methyl transferase (MGMT) methylation are surrogate biomarkers of improved survival in gliomas. This study aims at studying the ability of semantic magnetic resonance imaging (MRI) features to predict the IDH mutation status confirmed by the gold standard molecular tests. Methods: The MRI of 148 patients were reviewed for various imaging parameters based on the Visually AcceSAble Rembrandt Images (VASARI) study. Their IDH status was determined using immunohistochemistry (IHC). Fisher’s exact or chi-square tests for univariate and logistic regression for multivariate analysis were used. Results: Parameters such as mild and patchy enhancement, minimal edema, necrosis < 25%, presence of cysts, and less rCBV (relative cerebral blood volume) correlated with IDH mutation. The median age of IDH-mutant and IDH-wild patients were 34 years (IQR: 29−43) and 52 years (IQR: 45−59), respectively. Mild to moderate enhancement was observed in 15/19 IDH-mutant patients (79%), while 99/129 IDH-wildtype (77%) had severe enhancement (p-value <0.001). The volume of edema with respect to tumor volume distinguished IDH-mutants from wild phenotypes (peritumoral edema volume < tumor volume was associated with higher IDH-mutant phenotypes; p-value < 0.025). IDH-mutant patients had a median rCBV value of 1.8 (IQR: 1.4−2.0), while for IDH-wild phenotypes, it was 2.6 (IQR: 1.9−3.5) {p-value = 0.001}. On multivariate analysis, a cut-off of 25% necrosis was able to differentiate IDH-mutant from IDH-wildtype (p-value < 0.001), and a cut-off rCBV of 2.0 could differentiate IDH-mutant from IDH-wild phenotypes (p-value < 0.007). Conclusion: Semantic imaging features could reliably predict the IDH mutation status in high-grade gliomas. Presurgical prediction of IDH mutation status could help the treating oncologist to tailor the adjuvant therapy or use novel IDH inhibitors.
Collapse
Affiliation(s)
- Arpita Sahu
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
- Correspondence: (A.S.); (J.S.G.); Tel.: +91-7049000101 (A.S.); +91-22-24177000 (ext. 7027) (J.S.G.); Fax: +91-22-24146937 (A.S.); +91-22-24146937 (J.S.G.)
| | - Nandakumar G. Patnam
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
| | - Jayant Sastri Goda
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai 400012, India
- Correspondence: (A.S.); (J.S.G.); Tel.: +91-7049000101 (A.S.); +91-22-24177000 (ext. 7027) (J.S.G.); Fax: +91-22-24146937 (A.S.); +91-22-24146937 (J.S.G.)
| | - Sridhar Epari
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
- Department of Pathology, Tata Memorial Centre, Mumbai 400012, India
| | - Ayushi Sahay
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
- Department of Pathology, Tata Memorial Centre, Mumbai 400012, India
| | - Ronny Mathew
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
| | - Amit Kumar Choudhari
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
| | - Subhash M. Desai
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
| | - Archya Dasgupta
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai 400012, India
| | - Abhishek Chatterjee
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai 400012, India
| | - Pallavi Pratishad
- Homi Bhabha National Institute, Mumbai 400012, India
- Department of Biostatistics, Tata Memorial Centre, Mumbai 400012, India
| | - Prakash Shetty
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
- Department of Neurosurgery, Tata Memorial Centre, Mumbai 400012, India
| | - Ali Asgar Moiyadi
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
- Department of Neurosurgery, Tata Memorial Centre, Mumbai 400012, India
| | - Tejpal Gupta
- Neuro-Oncology Disease Management Group, Tata Memorial Centre, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400012, India
- Department of Neurosurgery, Tata Memorial Centre, Mumbai 400012, India
| |
Collapse
|
21
|
Giantini-Larsen AM, Pannullo S, Juthani RG. Challenges in the Diagnosis and Management of Low-Grade Gliomas. World Neurosurg 2022; 166:313-320. [PMID: 36192863 DOI: 10.1016/j.wneu.2022.06.074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 12/15/2022]
Abstract
Low-grade gliomas are clinically challenging entities. Patients with these tumors tend to be relatively young at presentation, and lesions are often incidental findings or are identified because the patient presents with a seizure. Rapidly emerging and evolving molecular classifications of gliomas have influenced treatment paradigms. Importantly, low-grade gliomas can be classified on the basis of IDH mutation status, whereby low-grade astrocytomas harbor the IDH mutation, while oligodendrogliomas are defined by both IDH mutant status and 1p/19q co-deletion. Given the importance of molecular classification for diagnosis, treatment planning, and prognostication, tissue samples are necessary for proper management. Literature supports improved overall survival and outcomes with increased extent of resection for low-grade glioma. Awake craniotomies and resection of insular low-grade gliomas both have been demonstrated as safe and improve outcomes for patients with lesions located in eloquent areas. Given the younger age at diagnosis of these lesions compared with higher-grade gliomas, fertility, fertility preservation, and potential malignant transformation should be discussed with patients of childbearing age.
Collapse
Affiliation(s)
- Alexandra M Giantini-Larsen
- Department of Neurological Surgery, Weill Cornell Medical College, New York-Presbyterian Hospital, New York, New York, USA
| | - Susan Pannullo
- Department of Neurological Surgery, Weill Cornell Medical College, New York-Presbyterian Hospital, New York, New York, USA
| | - Rupa Gopalan Juthani
- Department of Neurological Surgery, Weill Cornell Medical College, New York-Presbyterian Hospital, New York, New York, USA.
| |
Collapse
|
22
|
Du N, Zhou X, Mao R, Shu W, Xiao L, Ye Y, Xu X, Shen Y, Lin G, Fang X, Li S. Preoperative and Noninvasive Prediction of Gliomas Histopathological Grades and IDH Molecular Types Using Multiple MRI Characteristics. Front Oncol 2022; 12:873839. [PMID: 35712483 PMCID: PMC9196247 DOI: 10.3389/fonc.2022.873839] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/05/2022] [Indexed: 01/30/2023] Open
Abstract
Background and Purpose Gliomas are one of the most common tumors in the central nervous system. This study aimed to explore the correlation between MRI morphological characteristics, apparent diffusion coefficient (ADC) parameters and pathological grades, as well as IDH gene phenotypes of gliomas. Methods Preoperative MRI data from 166 glioma patients with pathological confirmation were retrospectively analyzed to compare the differences of MRI characteristics and ADC parameters between the low-grade and high-grade gliomas (LGGs vs. HGGs), IDH mutant and wild-type gliomas (IDHmut vs. IDHwt). Multivariate models were constructed to predict the pathological grades and IDH gene phenotypes of gliomas and the performance was assessed by the receiver operating characteristic (ROC) analysis. Results Two multivariable logistic regression models were developed by incorporating age, ADC parameters, and MRI morphological characteristics to predict pathological grades, and IDH gene phenotypes of gliomas, respectively. The Noninvasive Grading Model classified tumor grades with areas under the ROC curve (AUROC) of 0.934 (95% CI=0.895-0.973), sensitivity of 91.2%, and specificity of 78.6%. The Noninvasive IDH Genotyping Model differentiated IDH types with an AUROC of 0.857 (95% CI=0.787-0.926), sensitivity of 88.2%, and specificity of 63.8%. Conclusion MRI features were correlated with glioma grades and IDH mutation status. Multivariable logistic regression models combined with MRI morphological characteristics and ADC parameters may provide a noninvasive and preoperative approach to predict glioma grades and IDH mutation status.
Collapse
Affiliation(s)
- Ningfang Du
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China
| | - Xiaotao Zhou
- Department of Emergency, Changhai Hospital, Naval Medical University, Second Military Medical University, Shanghai, China
| | - Renling Mao
- Department of Neurosurgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Weiquan Shu
- Department of Neurosurgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Li Xiao
- Department of Pathology, Huadong Hospital, Fudan University, Shanghai, China
| | - Yao Ye
- Department of Pathology, Huadong Hospital, Fudan University, Shanghai, China
| | - Xinxin Xu
- Clinical Research Center for Gerontology, Huadong Hospital, Fudan University, Shanghai, China
| | - Yilang Shen
- Institute of Business Analytics, Adelphi University, Garden City, NY, United States
| | - Guangwu Lin
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China
| | - Xuhao Fang
- Department of Neurosurgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Shihong Li
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Cindil E, Sendur HN, Cerit MN, Erdogan N, Celebi F, Dag N, Celtikci E, Inan A, Oner Y, Tali T. Prediction of IDH Mutation Status in High-grade Gliomas Using DWI and High T1-weight DSC-MRI. Acad Radiol 2022; 29 Suppl 3:S52-S62. [PMID: 33685792 DOI: 10.1016/j.acra.2021.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/24/2021] [Accepted: 02/03/2021] [Indexed: 01/09/2023]
Abstract
RATIONALE AND OBJECTIVES We aimed to evaluate the diagnostic performance of diffusion-weighted imaging (DWI) and dynamic susceptibility contrast-enhanced (DSC) magnetic resonance imaging (MRI) parameters in the noninvasive prediction of the isocitrate dehydrogenase (IDH) mutation status in high-grade gliomas (HGGs). MATERIALS AND METHODS A total of 58 patients with histopathologically proved HGGs were included in this retrospective study. All patients underwent multiparametric MRI on 3-T, including DSC-MRI and DWI before surgery. The mean apparent diffusion coefficient (ADC), relative maximum cerebral blood volume (rCBV), and percentage signal recovery (PSR) of the tumor core were measured and compared depending on the IDH mutation status and tumor grade. The Mann-Whitney U test was used to detect statistically significant differences in parameters between IDH-mutant-type (IDH-m-type) and IDH-wild-type (IDH-w-type) HGGs. Receiver operating characteristic curve (ROC) analysis was performed to evaluate the diagnostic performance. RESULTS The rCBV was significantly higher, and the PSR value was significantly lower in IDH-w-type tumors than in the IDH-m group (p = 0.002 and <0.001, respectively).The ADC value in IDH-w-type tumors was significantly lower compared with the one in IDH-m types (p = 0.023), but remarkable overlaps were found between the groups. The PSR showed the best diagnostic performance with an AUC of 0.938 and with an accuracy rate of 0.87 at the optimal cutoff value of 86.85. The combination of the PSR and the rCBV for the identification of the IDH mutation status increased the discrimination ability at the AUC level of 0.955. In terms of each tumor grade, the PSR and rCBV showed significant differences between the IDH-m and IDH-w groups (p ≤0.001). CONCLUSION The rCBV and PSR from DSC-MRI may be feasible noninvasive imaging parameters for predicting the IDH mutation status in HGGs. The standardization of the imaging protocol is indispensable to the utility of DSC perfusion MRI in wider clinical usage.
Collapse
|
24
|
T2-Fluid-Attenuated Inversion Recovery Mismatch Sign in Grade II and III Gliomas: Is There a Coexisting T2-Diffusion-Weighted Imaging Mismatch? J Comput Assist Tomogr 2022; 46:251-256. [PMID: 35297581 DOI: 10.1097/rct.0000000000001267] [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
OBJECTIVE To determine whether the T2 fluid-attenuated inversion recovery (T2-FLAIR) mismatch sign in diffuse gliomas is associated with an equivalent pattern of disparity in signal intensities when comparing T2- and diffusion-weighted imaging (DWI). METHODS The level of correspondence between T2-FLAIR and T2-DWI evaluations in 34 World Health Organization grade II/III gliomas and interreader agreement among 3 neuroradiologists were assessed by calculating intraclass correlation coefficient and κ statistics, respectively. Tumoral apparent diffusion coefficient values were compared using t test. RESULTS There was an almost perfect correspondence between the 2 mismatch signs (intraclass correlation coefficient = 0.824 [95% confidence interval, 0.68-0.91]) that were associated with higher mean tumoral apparent diffusion coefficient (P < 0.01). Interreader agreement was substantial for T2-FLAIR (Fleiss κ = 0.724) and moderate for T2-DWI comparisons (Fleiss κ = 0.589) (P < 0.001). CONCLUSIONS The T2-FLAIR mismatch sign is usually reflected by a distinct microstructural pattern on DWI. The management of this tumor subtype may benefit from specifically tailored imaging assessments.
Collapse
|
25
|
Hagiwara A, Tatekawa H, Yao J, Raymond C, Everson R, Patel K, Mareninov S, Yong WH, Salamon N, Pope WB, Nghiemphu PL, Liau LM, Cloughesy TF, Ellingson BM. Visualization of tumor heterogeneity and prediction of isocitrate dehydrogenase mutation status for human gliomas using multiparametric physiologic and metabolic MRI. Sci Rep 2022; 12:1078. [PMID: 35058510 PMCID: PMC8776874 DOI: 10.1038/s41598-022-05077-2] [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: 08/13/2021] [Accepted: 12/10/2021] [Indexed: 01/19/2023] Open
Abstract
This study aimed to differentiate isocitrate dehydrogenase (IDH) mutation status with the voxel-wise clustering method of multiparametric magnetic resonance imaging (MRI) and to discover biological underpinnings of the clusters. A total of 69 patients with treatment-naïve diffuse glioma were scanned with pH-sensitive amine chemical exchange saturation transfer MRI, diffusion-weighted imaging, fluid-attenuated inversion recovery, and contrast-enhanced T1-weighted imaging at 3 T. An unsupervised two-level clustering approach was used for feature extraction from acquired images. The logarithmic ratio of the labels in each class within tumor regions was applied to a support vector machine to differentiate IDH status. The highest performance to predict IDH mutation status was found for 10-class clustering, with a mean area under the curve, accuracy, sensitivity, and specificity of 0.94, 0.91, 0.90, and 0.91, respectively. Targeted biopsies revealed that the tissues with labels 7-10 showed high expression levels of hypoxia-inducible factor 1-alpha, glucose transporter 3, and hexokinase 2, which are typical of IDH wild-type glioma, whereas those with labels 1 showed low expression of these proteins. In conclusion, A machine learning model successfully predicted the IDH mutation status of gliomas, and the resulting clusters properly reflected the metabolic status of the tumors.
Collapse
Affiliation(s)
- Akifumi Hagiwara
- grid.19006.3e0000 0000 9632 6718UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA 90024 USA ,grid.19006.3e0000 0000 9632 6718Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA ,grid.258269.20000 0004 1762 2738Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroyuki Tatekawa
- grid.19006.3e0000 0000 9632 6718UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA 90024 USA ,grid.19006.3e0000 0000 9632 6718Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA ,grid.261445.00000 0001 1009 6411Department of Diagnostic and Interventional Radiology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Jingwen Yao
- grid.19006.3e0000 0000 9632 6718UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA 90024 USA ,grid.19006.3e0000 0000 9632 6718Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA USA
| | - Catalina Raymond
- grid.19006.3e0000 0000 9632 6718UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA 90024 USA ,grid.19006.3e0000 0000 9632 6718Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Richard Everson
- grid.19006.3e0000 0000 9632 6718Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Kunal Patel
- grid.19006.3e0000 0000 9632 6718Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Sergey Mareninov
- grid.19006.3e0000 0000 9632 6718Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
| | - William H. Yong
- grid.19006.3e0000 0000 9632 6718Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
| | - Noriko Salamon
- grid.19006.3e0000 0000 9632 6718Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Whitney B. Pope
- grid.19006.3e0000 0000 9632 6718Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Phioanh L. Nghiemphu
- grid.19006.3e0000 0000 9632 6718UCLA Neuro-Oncology Program, University of California, Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
| | - Linda M. Liau
- grid.19006.3e0000 0000 9632 6718Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Timothy F. Cloughesy
- grid.19006.3e0000 0000 9632 6718UCLA Neuro-Oncology Program, University of California, Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
| | - Benjamin M. Ellingson
- grid.19006.3e0000 0000 9632 6718UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA 90024 USA ,grid.19006.3e0000 0000 9632 6718Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718UCLA Neuro-Oncology Program, University of California, Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
| |
Collapse
|
26
|
Nalawade SS, Yu FF, Bangalore Yogananda CG, Murugesan GK, Shah BR, Pinho MC, Wagner BC, Xi Y, Mickey B, Patel TR, Fei B, Madhuranthakam AJ, Maldjian JA. Brain tumor IDH, 1p/19q, and MGMT molecular classification using MRI-based deep learning: an initial study on the effect of motion and motion correction. J Med Imaging (Bellingham) 2022; 9:016001. [PMID: 35118164 PMCID: PMC8794036 DOI: 10.1117/1.jmi.9.1.016001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 01/03/2022] [Indexed: 01/29/2023] Open
Abstract
Purpose: Deep learning has shown promise for predicting the molecular profiles of gliomas using MR images. Prior to clinical implementation, ensuring robustness to real-world problems, such as patient motion, is crucial. The purpose of this study is to perform a preliminary evaluation on the effects of simulated motion artifact on glioma marker classifier performance and determine if motion correction can restore classification accuracies. Approach: T2w images and molecular information were retrieved from the TCIA and TCGA databases. Simulated motion was added in the k-space domain along the phase encoding direction. Classifier performance for IDH mutation, 1p/19q co-deletion, and MGMT methylation was assessed over the range of 0% to 100% corrupted k-space lines. Rudimentary motion correction networks were trained on the motion-corrupted images. The performance of the three glioma marker classifiers was then evaluated on the motion-corrected images. Results: Glioma marker classifier performance decreased markedly with increasing motion corruption. Applying motion correction effectively restored classification accuracy for even the most motion-corrupted images. For isocitrate dehydrogenase (IDH) classification, 99% accuracy was achieved, exceeding the original performance of the network and representing a new benchmark in non-invasive MRI-based IDH classification. Conclusions: Robust motion correction can facilitate highly accurate deep learning MRI-based molecular marker classification, rivaling invasive tissue-based characterization methods. Motion correction may be able to increase classification accuracy even in the absence of a visible artifact, representing a new strategy for boosting classifier performance.
Collapse
Affiliation(s)
- Sahil S. Nalawade
- University of Texas Southwestern Medical Center, Advanced Neuroscience Imaging Research Lab, Department of Radiology, Dallas, Texas, United States
| | - Fang F. Yu
- University of Texas Southwestern Medical Center, Advanced Neuroscience Imaging Research Lab, Department of Radiology, Dallas, Texas, United States
| | - Chandan Ganesh Bangalore Yogananda
- University of Texas Southwestern Medical Center, Advanced Neuroscience Imaging Research Lab, Department of Radiology, Dallas, Texas, United States
| | - Gowtham K. Murugesan
- University of Texas Southwestern Medical Center, Advanced Neuroscience Imaging Research Lab, Department of Radiology, Dallas, Texas, United States
| | - Bhavya R. Shah
- University of Texas Southwestern Medical Center, Advanced Neuroscience Imaging Research Lab, Department of Radiology, Dallas, Texas, United States
| | - Marco C. Pinho
- University of Texas Southwestern Medical Center, Advanced Neuroscience Imaging Research Lab, Department of Radiology, Dallas, Texas, United States
| | - Benjamin C. Wagner
- University of Texas Southwestern Medical Center, Advanced Neuroscience Imaging Research Lab, Department of Radiology, Dallas, Texas, United States
| | - Yin Xi
- University of Texas Southwestern Medical Center, Advanced Neuroscience Imaging Research Lab, Department of Radiology, Dallas, Texas, United States
| | - Bruce Mickey
- University of Texas Southwestern Medical Center, Department of Neurological Surgery, Dallas, Texas, United States
| | - Toral R. Patel
- University of Texas Southwestern Medical Center, Department of Neurological Surgery, Dallas, Texas, United States
| | - Baowei Fei
- University of Texas at Dallas, Department of Bioengineering, Richardson, Texas, United States
| | - Ananth J. Madhuranthakam
- University of Texas Southwestern Medical Center, Advanced Neuroscience Imaging Research Lab, Department of Radiology, Dallas, Texas, United States
| | - Joseph A. Maldjian
- University of Texas Southwestern Medical Center, Advanced Neuroscience Imaging Research Lab, Department of Radiology, Dallas, Texas, United States,Address all correspondence to Joseph A. Maldjian,
| |
Collapse
|
27
|
Gore S, Chougule T, Jagtap J, Saini J, Ingalhalikar M. A Review of Radiomics and Deep Predictive Modeling in Glioma Characterization. Acad Radiol 2021; 28:1599-1621. [PMID: 32660755 DOI: 10.1016/j.acra.2020.06.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/22/2022]
Abstract
Recent developments in glioma categorization based on biological genotypes and application of computational machine learning or deep learning based predictive models using multi-modal MRI biomarkers to assess these genotypes provides potential assurance for optimal and personalized treatment plans and efficacy. Artificial intelligence based quantified assessment of glioma using MRI derived hand-crafted or auto-extracted features have become crucial as genomic alterations can be associated with MRI based phenotypes. This survey integrates all the recent work carried out in state-of-the-art radiomics, and Artificial Intelligence based learning solutions related to molecular diagnosis, prognosis, and treatment monitoring with the aim to create a structured resource on radiogenomic analysis of glioma. Challenges such as inter-scanner variability, requirement of benchmark datasets, prospective validations for clinical applicability are discussed with further scope for designing optimal solutions for glioma stratification with immediate recommendations for further diagnostic decisions and personalized treatment plans for glioma patients.
Collapse
|
28
|
Cluceru J, Interian Y, Phillips JJ, Molinaro AM, Luks TL, Alcaide-Leon P, Olson MP, Nair D, LaFontaine M, Shai A, Chunduru P, Pedoia V, Villanueva-Meyer JE, Chang SM, Lupo JM. Improving the noninvasive classification of glioma genetic subtype with deep learning and diffusion-weighted imaging. Neuro Oncol 2021; 24:639-652. [PMID: 34653254 DOI: 10.1093/neuonc/noab238] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Diagnostic classification of diffuse gliomas now requires an assessment of molecular features, often including IDH-mutation and 1p19q-codeletion status. Because genetic testing requires an invasive process, an alternative noninvasive approach is attractive, particularly if resection is not recommended. The goal of this study was to evaluate the effects of training strategy and incorporation of biologically relevant images on predicting genetic subtypes with deep learning. METHODS Our dataset consisted of 384 patients with newly-diagnosed gliomas who underwent preoperative MR imaging with standard anatomical and diffusion-weighted imaging, and 147 patients from an external cohort with anatomical imaging. Using tissue samples acquired during surgery, each glioma was classified into IDH-wildtype (IDHwt), IDH-mutant/1p19q-noncodeleted (IDHmut-intact), and IDH-mutant/1p19q-codeleted (IDHmut-codel) subgroups. After optimizing training parameters, top performing convolutional neural network (CNN) classifiers were trained, validated, and tested using combinations of anatomical and diffusion MRI with either a 3-class or tiered structure. Generalization to an external cohort was assessed using anatomical imaging models. RESULTS The best model used a 3-class CNN containing diffusion-weighted imaging as an input, achieving 85.7% (95% CI:[77.1,100]) overall test accuracy and correctly classifying 95.2%, 88.9%, 60.0% of the IDHwt, IDHmut-intact, and IDHmut-codel tumors. In general, 3-class models outperformed tiered approaches by 13.5-17.5%, and models that included diffusion-weighted imaging were 5-8.8% more accurate than those that used only anatomical imaging. CONCLUSION Training a classifier to predict both IDH-mutation and 1p19q-codeletion status outperformed a tiered structure that first predicted IDH-mutation, then1p19q-codeletion. Including ADC, a surrogate marker of cellularity, more accurately captured differences between subgroups.
Collapse
Affiliation(s)
- Julia Cluceru
- Department of Radiology & Biomedical Imaging, University of California San Francisco
| | | | - Joanna J Phillips
- Department of Neurological Surgery, University of California San Francisco.,Department of Pathology, University of California San Francisco
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco
| | - Tracy L Luks
- Department of Radiology & Biomedical Imaging, University of California San Francisco
| | - Paula Alcaide-Leon
- Department of Radiology & Biomedical Imaging, University of California San Francisco.,Department of Medical Imaging, University of Toronto
| | - Marram P Olson
- Department of Radiology & Biomedical Imaging, University of California San Francisco
| | - Devika Nair
- Department of Radiology & Biomedical Imaging, University of California San Francisco
| | - Marisa LaFontaine
- Department of Radiology & Biomedical Imaging, University of California San Francisco
| | - Anny Shai
- Department of Neurological Surgery, University of California San Francisco
| | - Pranathi Chunduru
- Department of Neurological Surgery, University of California San Francisco
| | - Valentina Pedoia
- Department of Radiology & Biomedical Imaging, University of California San Francisco
| | | | - Susan M Chang
- Department of Neurological Surgery, University of California San Francisco
| | - Janine M Lupo
- Department of Radiology & Biomedical Imaging, University of California San Francisco
| |
Collapse
|
29
|
Morrison MA, Lupo JM. 7-T Magnetic Resonance Imaging in the Management of Brain Tumors. Magn Reson Imaging Clin N Am 2021; 29:83-102. [PMID: 33237018 DOI: 10.1016/j.mric.2020.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This article provides an overview of the current status of ultrahigh-field 7-T magnetic resonance (MR) imaging in neuro-oncology, specifically for the management of patients with brain tumors. It includes a discussion of areas across the pretherapeutic, peritherapeutic, and posttherapeutic stages of patient care where 7-T MR imaging is currently being exploited and holds promise. This discussion includes existing technical challenges, barriers to clinical integration, as well as our impression of the future role of 7-T MR imaging as a clinical tool in neuro-oncology.
Collapse
Affiliation(s)
- Melanie A Morrison
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143, USA.
| |
Collapse
|
30
|
Çelik S, Öven BB, Demir MK, Yılmaz EÇ, Kanan D, Özdamarlar U, Emirzeoglu L, Yapıcıer Ö, Kılıç T. Magnetic resonance imaging criteria for prediction of isocitrate dehydrogenase (IDH) mutation status in patients with grade II-III astrocytoma and oligodendroglioma. Clin Neurol Neurosurg 2021; 207:106745. [PMID: 34146841 DOI: 10.1016/j.clineuro.2021.106745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND IDH mutation status is an important prognostic marker for glial tumors, which is detected immunohistochemically after surgery. Since this method is invasive, easy and noninvasive magnetic resonance imaging (MRI) methods have recently been used in predicting the IDH mutation status. However, there is currently no standard MRI technique to predict IDH mutation. We analyzed the value of conventional MRI to predict IDH mutation and its effect on survival among grade II-III astrocytoma and oligodendroglioma patients. MATERIAL AND METHODS We included WHO grade II-III astrocytoma and oligodendroglioma patients who underwent surgery at Bahcesehir University Goztepe Medical Park Hospital. All patients were analyzed according to their immunohistochemical IDH mutation status. Preoperative conventional MRI studies with respect to their location, diffusion restriction, contrast enhancement, calcification and hemorrhage on susceptibility-weighted image (SWI) or T2*- weighted imaging (T2*WI), and T2 -FLAIR mismatch properties were retrospectively assessed by a neuroradiologist. The relation between MRI characteristics and IDH mutation was analyzed using a chi-square test. The sensitivity and specificity of radiological IDH mutation were determined by ROC analysis. The impact of IDH mutation on survival was also analyzed by Kaplan-Meier tests. RESULTS IDH mutation was found to be positive in 82.5% of tumors histopathologically and 54.4% radiologically. The sensitivity and specificity were 63.8% and 90%, respectively (Area under the curve/AUC = 0.369, p = 0.08). IDH wild gliomas were predominantly diffusion-restricted tumors. IDH mutant tumors were less likely to have contrast enhancement and had lower grades compared to the IDH wild tumors. The median survival time could not be reached and the overall survival was not related to any tumor characteristics or IDH mutation. CONCLUSIONS Conventional MRI predicts IDH-mutation status in Grade II-III astrocytoma and oligodendroglioma. Contrast-enhancement and restricted diffusion were strongly associated with grade III astrocytoma and oligodendroglioma, IDH-wild type. Location, T2-FLAIR mismatch, and SWI did not contribute to making a decision on the IDH mutation status. There was no significant difference between the survival times of patients and their IDH status.
Collapse
Affiliation(s)
- Serkan Çelik
- Department of Medical Oncology, Bahcesehir University School of Medicine, Istanbul, Turkey.
| | - Bala Başak Öven
- Department of Medical Oncology, Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Mustafa Kemal Demir
- Department of Radiology, Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Enis Çağatay Yılmaz
- Department of Radiology, Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Duaa Kanan
- Department of Medical Oncology, Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Umut Özdamarlar
- Department of Radiology, Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Levent Emirzeoglu
- Department of Medical Oncology, Sultan Abdulhamid Han Training Hospital, Istanbul, Turkey
| | - Özlem Yapıcıer
- Department of Pathology, Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Türker Kılıç
- Department of Neurosurgery, Bahcesehir University School of Medicine, Istanbul, Turkey
| |
Collapse
|
31
|
Steidl E, Langen KJ, Hmeidan SA, Polomac N, Filss CP, Galldiks N, Lohmann P, Keil F, Filipski K, Mottaghy FM, Shah NJ, Steinbach JP, Hattingen E, Maurer GD. Sequential implementation of DSC-MR perfusion and dynamic [ 18F]FET PET allows efficient differentiation of glioma progression from treatment-related changes. Eur J Nucl Med Mol Imaging 2021; 48:1956-1965. [PMID: 33241456 PMCID: PMC8113145 DOI: 10.1007/s00259-020-05114-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/08/2020] [Indexed: 11/21/2022]
Abstract
PURPOSE Perfusion-weighted MRI (PWI) and O-(2-[18F]fluoroethyl-)-l-tyrosine ([18F]FET) PET are both applied to discriminate tumor progression (TP) from treatment-related changes (TRC) in patients with suspected recurrent glioma. While the combination of both methods has been reported to improve the diagnostic accuracy, the performance of a sequential implementation has not been further investigated. Therefore, we retrospectively analyzed the diagnostic value of consecutive PWI and [18F]FET PET. METHODS We evaluated 104 patients with WHO grade II-IV glioma and suspected TP on conventional MRI using PWI and dynamic [18F]FET PET. Leakage corrected maximum relative cerebral blood volumes (rCBVmax) were obtained from dynamic susceptibility contrast PWI. Furthermore, we calculated static (i.e., maximum tumor to brain ratios; TBRmax) and dynamic [18F]FET PET parameters (i.e., Slope). Definitive diagnoses were based on histopathology (n = 42) or clinico-radiological follow-up (n = 62). The diagnostic performance of PWI and [18F]FET PET parameters to differentiate TP from TRC was evaluated by analyzing receiver operating characteristic and area under the curve (AUC). RESULTS Across all patients, the differentiation of TP from TRC using rCBVmax or [18F]FET PET parameters was moderate (AUC = 0.69-0.75; p < 0.01). A rCBVmax cutoff > 2.85 had a positive predictive value for TP of 100%, enabling a correct TP diagnosis in 44 patients. In the remaining 60 patients, combined static and dynamic [18F]FET PET parameters (TBRmax, Slope) correctly discriminated TP and TRC in a significant 78% of patients, increasing the overall accuracy to 87%. A subgroup analysis of isocitrate dehydrogenase (IDH) mutant tumors indicated a superior performance of PWI to [18F]FET PET (AUC = 0.8/< 0.62, p < 0.01/≥ 0.3). CONCLUSION While marked hyperperfusion on PWI indicated TP, [18F]FET PET proved beneficial to discriminate TP from TRC when PWI remained inconclusive. Thus, our results highlight the clinical value of sequential use of PWI and [18F]FET PET, allowing an economical use of diagnostic methods. The impact of an IDH mutation needs further investigation.
Collapse
Affiliation(s)
- Eike Steidl
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt am Main, Schleusenweg 2-16, Frankfurt am Main, 60528, Germany.
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Karl-Josef Langen
- Inst. of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Research Center Juelich, Juelich, Germany
- Dept. of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Aachen, Germany
| | - Sarah Abu Hmeidan
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt am Main, Schleusenweg 2-16, Frankfurt am Main, 60528, Germany
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Nenad Polomac
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt am Main, Schleusenweg 2-16, Frankfurt am Main, 60528, Germany
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Christian P Filss
- Inst. of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Research Center Juelich, Juelich, Germany
- Dept. of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Norbert Galldiks
- Inst. of Neuroscience and Medicine, Cognitive Neuroscience (INM-3), Research Center Juelich, Juelich, Germany
- Dept. of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Cologne, Germany
| | - Philipp Lohmann
- Inst. of Neuroscience and Medicine, Cognitive Neuroscience (INM-3), Research Center Juelich, Juelich, Germany
- Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Fee Keil
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt am Main, Schleusenweg 2-16, Frankfurt am Main, 60528, Germany
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Katharina Filipski
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Felix M Mottaghy
- Dept. of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Aachen, Germany
- Dept. of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Nadim Jon Shah
- Inst. of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Research Center Juelich, Juelich, Germany
- Inst. of Neuroscience and Medicine, Molecular Neuroscience and Neuroimaging (INM-11), JARA, Research Center Juelich, Juelich, Germany
- Dept. of Neurology, University Hospital RWTH Aachen, Aachen, Germany
- JARA - BRAIN - Translational Medicine, Aachen, Germany
| | - Joachim P Steinbach
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Dr. Senckenberg Institute of Neurooncology, University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Elke Hattingen
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt am Main, Schleusenweg 2-16, Frankfurt am Main, 60528, Germany
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gabriele D Maurer
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Dr. Senckenberg Institute of Neurooncology, University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
32
|
Kihira S, Tsankova NM, Bauer A, Sakai Y, Mahmoudi K, Zubizarreta N, Houldsworth J, Khan F, Salamon N, Hormigo A, Nael K. Multiparametric MRI texture analysis in prediction of glioma biomarker status: added value of MR diffusion. Neurooncol Adv 2021; 3:vdab051. [PMID: 34056604 PMCID: PMC8156980 DOI: 10.1093/noajnl/vdab051] [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] [Indexed: 12/16/2022] Open
Abstract
Background Early identification of glioma molecular phenotypes can lead to understanding of patient prognosis and treatment guidance. We aimed to develop a multiparametric MRI texture analysis model using a combination of conventional and diffusion MRI to predict a wide range of biomarkers in patients with glioma. Methods In this retrospective study, patients were included if they (1) had diagnosis of gliomas with known IDH1, EGFR, MGMT, ATRX, TP53, and PTEN status from surgical pathology and (2) had preoperative MRI including FLAIR, T1c+ and diffusion for radiomic texture analysis. Statistical analysis included logistic regression and receiver-operating characteristic (ROC) curve analysis to determine the optimal model for predicting glioma biomarkers. A comparative analysis between ROCs (conventional only vs conventional + diffusion) was performed. Results From a total of 111 patients included, 91 (82%) were categorized to training and 20 (18%) to test datasets. Constructed cross-validated model using a combination of texture features from conventional and diffusion MRI resulted in overall AUC/accuracy of 1/79% for IDH1, 0.99/80% for ATRX, 0.79/67% for MGMT, and 0.77/66% for EGFR. The addition of diffusion data to conventional MRI features significantly (P < .05) increased predictive performance for IDH1, MGMT, and ATRX. The overall accuracy of the final model in predicting biomarkers in the test group was 80% (IDH1), 70% (ATRX), 70% (MGMT), and 75% (EGFR). Conclusion Addition of MR diffusion to conventional MRI features provides added diagnostic value in preoperative determination of IDH1, MGMT, and ATRX in patients with glioma.
Collapse
Affiliation(s)
- Shingo Kihira
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nadejda M Tsankova
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adam Bauer
- Department of Radiology, Kaiser Permanente Fontana Medical Center, Fontana, California, USA
| | - Yu Sakai
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Keon Mahmoudi
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nicole Zubizarreta
- Institute for Health Care Delivery Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jane Houldsworth
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fahad Khan
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Noriko Salamon
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Adilia Hormigo
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kambiz Nael
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
33
|
Zhu L, Wu J, Zhang H, Niu H, Wang L. The value of intravoxel incoherent motion imaging in predicting the survival of patients with astrocytoma. Acta Radiol 2021; 62:423-429. [PMID: 32551800 DOI: 10.1177/0284185120926907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND The evaluation of the prognosis of gliomas may have great value in individualized treatment. PURPOSE To evaluate the value of intravoxel incoherent motion (IVIM) in predicting the survival of patients with astrocytoma and comparing it to apparent diffusion coefficients (ADC). MATERIAL AND METHODS Sixty patients with pathologically confirmed cerebral astrocytomas underwent IVIM scans before any treatment was performed. Patients were divided into death group and survival group according to a two-year follow-up. ADC and quantitative parameters of IVIM including D, D*, and f were measured. Independent sample t test was used to compare the two groups of parameters. The accuracy of each parameter for two-year survival rate was analyzed by receiver operating characteristic (ROC) curve and Kaplan-Meier survival curves. The correlation between quantitative parameters and survival days was analyzed by Pearson correlation analysis. RESULTS The ADC, D*, and f values were statistically significant different between the death and the survival groups (P < 0.05). The AUC of the ADC, D*, and f were 0.811, 0.858, and 0.892, respectively. The ADC cut-off value of 0.668 × 10-3 mm2/s corresponded to 82.6% sensitivity and 73% specificity. The D* cut-off value of 3.913 × 10-3 mm2/s corresponded to 78.4% sensitivity and 87% specificity. The f cut-off value of 0.487 corresponded to 83.8% sensitivity and 87% specificity. Significant log rank test was performed for each parameter to predict overall survival (P < 0.05). There was a correlation between ADC (r = 0.625, P = 0.023), D* (r = -0.655, P = 0.012), f (r = -0.725, P = 0.000) and survival days. CONCLUSION The D* and f values demonstrated great potential in predicting the two-year survival rate for patients with astrocytoma.
Collapse
Affiliation(s)
- Lina Zhu
- Department of Magnetic Resonance, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi, PR China
| | - Jiang Wu
- Department of Magnetic Resonance, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi, PR China
| | - Hui Zhang
- Department of Magnetic Resonance, the First Hospital of Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Heng Niu
- Department of Magnetic Resonance, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi, PR China
| | - Le Wang
- Department of Magnetic Resonance, the First Hospital of Shanxi Medical University, Taiyuan, Shanxi, PR China
| |
Collapse
|
34
|
Thust SC, Maynard JA, Benenati M, Wastling SJ, Mancini L, Jaunmuktane Z, Brandner S, Jäger HR. Regional and Volumetric Parameters for Diffusion-Weighted WHO Grade II and III Glioma Genotyping: A Method Comparison. AJNR Am J Neuroradiol 2021; 42:441-447. [PMID: 33414227 DOI: 10.3174/ajnr.a6965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE Studies consistently report lower ADC values in isocitrate dehydrogenase (IDH) wild-type gliomas than in IDH mutant tumors, but their methods and thresholds vary. This research aimed to compare volumetric and regional ADC measurement techniques for glioma genotyping, with a focus on IDH status prediction. MATERIALS AND METHODS Treatment-naïve World Health Organization grade II and III gliomas were analyzed by 3 neuroradiologist readers blinded to tissue results. ADC minimum and mean ROIs were defined in tumor and in normal-appearing white matter to calculate normalized values. T2-weighted tumor VOIs were registered to ADC maps with histogram parameters (mean, 2nd and 5th percentiles) extracted. Nonparametric testing (eta2 and ANOVA) was performed to identify associations between ADC metrics and glioma genotypes. Logistic regression was used to probe the ability of VOI and ROI metrics to predict IDH status. RESULTS The study included 283 patients with 79 IDH wild-type and 204 IDH mutant gliomas. Across the study population, IDH status was most accurately predicted by ROI mean normalized ADC and VOI mean normalized ADC, with areas under the curve of 0.83 and 0.82, respectively. The results for ROI-based genotyping of nonenhancing and solid-patchy enhancing gliomas were comparable with volumetric parameters (area under the curve = 0.81-0.84). In rim-enhancing, centrally necrotic tumors (n = 23), only volumetric measurements were predictive (0.90). CONCLUSIONS Regional normalized mean ADC measurements are noninferior to volumetric segmentation for defining solid glioma IDH status. Partially necrotic, rim-enhancing tumors are unsuitable for ROI assessment and may benefit from volumetric ADC quantification.
Collapse
Affiliation(s)
- S C Thust
- From the Neuroradiological Academic Unit (S.C.T., J.A.M, S.J.W., L.M., H.R.J.), Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, UK
- Lysholm Department of Neuroradiology (S.C.T., J.A.M., M.B., S.J.W., L.M., H.R.J.), National Hospital for Neurology and Neurosurgery, London, UK
- Imaging Department (S.C.T., H.R.J.), University College London Foundation Hospital, London, UK
| | - J A Maynard
- From the Neuroradiological Academic Unit (S.C.T., J.A.M, S.J.W., L.M., H.R.J.), Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, UK
- Lysholm Department of Neuroradiology (S.C.T., J.A.M., M.B., S.J.W., L.M., H.R.J.), National Hospital for Neurology and Neurosurgery, London, UK
| | - M Benenati
- Lysholm Department of Neuroradiology (S.C.T., J.A.M., M.B., S.J.W., L.M., H.R.J.), National Hospital for Neurology and Neurosurgery, London, UK
- Dipartimento di Diagnostica per Immagini (M.B.), Radioterapia, Oncologia ed Ematologia, Fondazione Policlinico Universitario A. Gemelli Institute for Research, Hospitalization and Health Care, Rome, Italy
| | - S J Wastling
- From the Neuroradiological Academic Unit (S.C.T., J.A.M, S.J.W., L.M., H.R.J.), Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, UK
- Lysholm Department of Neuroradiology (S.C.T., J.A.M., M.B., S.J.W., L.M., H.R.J.), National Hospital for Neurology and Neurosurgery, London, UK
| | - L Mancini
- From the Neuroradiological Academic Unit (S.C.T., J.A.M, S.J.W., L.M., H.R.J.), Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, UK
- Lysholm Department of Neuroradiology (S.C.T., J.A.M., M.B., S.J.W., L.M., H.R.J.), National Hospital for Neurology and Neurosurgery, London, UK
| | - Z Jaunmuktane
- Department of Clinical and Movement Neurosciences (Z.J.)
| | - S Brandner
- Neurodegenerative Disease (S.B.), UCL Queen Square Institute of Neurology, and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - H R Jäger
- From the Neuroradiological Academic Unit (S.C.T., J.A.M, S.J.W., L.M., H.R.J.), Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, UK
- Lysholm Department of Neuroradiology (S.C.T., J.A.M., M.B., S.J.W., L.M., H.R.J.), National Hospital for Neurology and Neurosurgery, London, UK
- Imaging Department (S.C.T., H.R.J.), University College London Foundation Hospital, London, UK
| |
Collapse
|
35
|
Nagaraja TN, Lee IY. Cerebral microcirculation in glioblastoma: A major determinant of diagnosis, resection, and drug delivery. Microcirculation 2021; 28:e12679. [PMID: 33474805 DOI: 10.1111/micc.12679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/12/2021] [Indexed: 12/25/2022]
Abstract
Glioblastoma (GBM) is the most common primary brain tumor with a dismal prognosis. Current standard of treatment is safe maximal tumor resection followed by chemotherapy and radiation. Altered cerebral microcirculation and elevated blood-tumor barrier (BTB) permeability in tumor periphery due to glioma-induced vascular dysregulation allow T1 contrast-enhanced visualization of resectable tumor boundaries. Newer tracers that label the tumor and its vasculature are being increasingly used for intraoperative delineation of glioma boundaries for even more precise resection. Fluorescent 5-aminolevulinic acid (5-ALA) and indocyanine green (ICG) are examples of such intraoperative tracers. Recently, magnetic resonance imaging (MRI)-based MR thermometry is being employed for laser interstitial thermal therapy (LITT) for glioma debulking. However, aggressive, fatal recurrence always occurs. Postsurgical chemotherapy is hampered by the inability of most drugs to cross the blood-brain barrier (BBB). Understanding postsurgical changes in brain microcirculation and permeability is crucial to improve chemotherapy delivery. It is important to understand whether any microcirculatory indices can differentiate between true recurrence and radiation necrosis. LITT leads to peri-ablation BBB opening that persists for several weeks. Whether it can be a conduit for chemotherapy delivery is yet to be explored. This review will address the role of cerebral microcirculation in such emerging ideas in GBM diagnosis and therapy.
Collapse
Affiliation(s)
| | - Ian Y Lee
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
36
|
Zhang HW, Lyu GW, He WJ, Lei Y, Lin F, Wang MZ, Zhang H, Liang LH, Feng YN, Yang JH. DSC and DCE Histogram Analyses of Glioma Biomarkers, Including IDH, MGMT, and TERT, on Differentiation and Survival. Acad Radiol 2020; 27:e263-e271. [PMID: 31983532 DOI: 10.1016/j.acra.2019.12.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/06/2019] [Accepted: 12/07/2019] [Indexed: 12/25/2022]
Abstract
RATIONALE AND OBJECTIVES The World Health Organization 2016 classification of central nervous system tumors added the molecular classification of gliomas and has guiding significance for the operation and prognosis of glioma patients. At present, the perfusion technique plays an important role in judging the malignant degree of glioma. To evaluate the performance of dynamic susceptibility contrast (DSC)- and dynamic contrast-enhanced (DCE)-magnetic resonance imaging (MRI) histogram analyses in discriminating the states of molecular biomarkers and survival in glioma patients. MATERIALS AND METHODS Forty-three glioma patients who underwent DCE- and DSC-MRI were enrolled. Relevant molecular test results, including those on isocitrate dehydrogenase (IDH), O6-methylguanine-DNA methyltransferase (MGMT) and telomere reverse transcriptase (TERT), were collected. The mean relative cerebral blood volume of DSC-MRI and histogram parameters derived from DCE-MRI (volume transfer coefficient (Ktrans), fractional volume of the extravascular extracellular space (Ve), fractional blood plasma volume (Vp), rate constant between the extravascular extracellular space and blood plasma (Kep) and area under the curve (AUC)) were calculated. Differences in each parameter between gliomas with different expression states (IDH, MGMT, and TERT) were evaluated. The diagnostic efficiency of each parameter was analyzed. The overall survival of all patients was assessed. RESULTS The 10th percentile AUC (AUC = 0.830, sensitivity = 0.78, specificity = 0.80), the 90th percentile Ve (AUC = 0.816, sensitivity = 0.84, specificity = 0.79), and the mean Kep (AUC = 0.818, sensitivity = 0.76, specificity = 0.78) provided the highest differential efficiency for IDH, MGMT, and TERT, respectively. Kaplan-Meier curves showed a significant difference between subjects with a 10th percentile AUC higher or lower than 0.028 (log-rank = 7.535; p = 0.006) for IDH and between subjects with different 90th percentile Ve values (log-rank = 6.532; p = 0.011) for MGMT. CONCLUSION Histogram DCE-MRI demonstrates good diagnostic performance in identifying different molecular types and for the prognostic assessment of glioma.
Collapse
Affiliation(s)
- Han-Wen Zhang
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 SunGangXi Road, Shenzhen 518035, China
| | - Gui-Wen Lyu
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 SunGangXi Road, Shenzhen 518035, China
| | - Wen-Jie He
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 SunGangXi Road, Shenzhen 518035, China
| | - Yi Lei
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 SunGangXi Road, Shenzhen 518035, China.
| | - Fan Lin
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 SunGangXi Road, Shenzhen 518035, China
| | - Meng-Zhu Wang
- Department of MR Scientific Marketing, Siemens Healthineers, Guangzhou, Guangdong Province, China
| | - Hong Zhang
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 SunGangXi Road, Shenzhen 518035, China
| | - Li-Hong Liang
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 SunGangXi Road, Shenzhen 518035, China
| | - Yu-Ning Feng
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, 3002 SunGangXi Road, Shenzhen 518035, China
| | - Ji-Hu Yang
- Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| |
Collapse
|
37
|
Choi KS, Choi SH, Jeong B. Prediction of IDH genotype in gliomas with dynamic susceptibility contrast perfusion MR imaging using an explainable recurrent neural network. Neuro Oncol 2020; 21:1197-1209. [PMID: 31127834 DOI: 10.1093/neuonc/noz095] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The aim of this study was to predict isocitrate dehydrogenase (IDH) genotypes of gliomas using an interpretable deep learning application for dynamic susceptibility contrast (DSC) perfusion MRI. METHODS Four hundred sixty-three patients with gliomas who underwent preoperative MRI were enrolled in the study. All the patients had immunohistopathologic diagnoses of either IDH-wildtype or IDH-mutant gliomas. Tumor subregions were segmented using a convolutional neural network followed by manual correction. DSC perfusion MRI was performed to obtain T2* susceptibility signal intensity-time curves from each subregion of the tumors: enhancing tumor, non-enhancing tumor, peritumoral edema, and whole tumor. These, with arterial input functions, were fed into a neural network as multidimensional inputs. A convolutional long short-term memory model with an attention mechanism was developed to predict IDH genotypes. Receiver operating characteristics analysis was performed to evaluate the model. RESULTS The IDH genotype predictions had an accuracy, sensitivity, and specificity of 92.8%, 92.6%, and 93.1%, respectively, in the validation set (area under the curve [AUC], 0.98; 95% confidence interval [CI], 0.969-0.991) and 91.7%, 92.1%, and 91.5%, respectively, in the test set (AUC, 0.95; 95% CI, 0.898-0.982). In temporal feature analysis, T2* susceptibility signal intensity-time curves obtained from DSC perfusion MRI with attention weights demonstrated high attention on the combination of the end of the pre-contrast baseline, up/downslopes of signal drops, and/or post-bolus plateaus for the curves used to predict IDH genotype. CONCLUSIONS We developed an explainable recurrent neural network model based on DSC perfusion MRI to predict IDH genotypes in gliomas.
Collapse
Affiliation(s)
- Kyu Sung Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, Republic of Korea.,KAIST Institute for Health Science and Technology, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, Republic of Korea.,KAIST Institute for Artificial Intelligence, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, Republic of Korea
| | - Bumseok Jeong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, Republic of Korea.,KAIST Institute for Health Science and Technology, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, Republic of Korea.,KAIST Institute for Artificial Intelligence, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, Republic of Korea
| |
Collapse
|
38
|
A Comparative Study of 2 Different Segmentation Methods of ADC Histogram for Differentiation Genetic Subtypes in Lower-Grade Diffuse Gliomas. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9549361. [PMID: 33062706 PMCID: PMC7539099 DOI: 10.1155/2020/9549361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/03/2020] [Accepted: 09/15/2020] [Indexed: 01/04/2023]
Abstract
Background To evaluate the diagnostic performance of apparent diffusion coefficient (ADC) histogram parameters for differentiating the genetic subtypes in lower-grade diffuse gliomas and explore which segmentation method (ROI-1, the entire tumor ROI; ROI2, the tumor ROI excluding cystic and necrotic portions) performs better. Materials and Methods We retrospectively evaluated 56 lower-grade diffuse gliomas and divided them into three categories: IDH-wild group (IDHwt, 16cases); IDH mutant with the intact 1p or 19q group (IDHmut/1p19q+, 18cases); and IDH mutant with the 1p/19q codeleted group (IDHmut/1p19q-, 22cases). Histogram parameters of ADC maps calculated with the two different ROI methods: ADCmean, min, max, mode, P5, P10, P25, P75, P90, P95, kurtosis, skewness, entropy, StDev, and inhomogenity were compared between these categories using the independent t test or Mann-Whitney U test. For statistically significant results, a receiver operating characteristic (ROC) curves were constructed, and the optimal cutoff value was determined by maximizing Youden's index. Area under the curve (AUC) results were compared using the method of Delong et al. Results The inhomogenity from the two different ROI methods for distinguishing IDHwt gliomas from IDHmut gliomas both showed the biggest AUC (0.788, 0.930), the optimal cutoff value was 0.229 (sensitivity, 81.3%; specificity, 75.0%) for the ROI-1 and 0.186 (sensitivity, 93.8%; specificity, 82.5%) for the ROI-2, and the AUC of the inhomogenity from the ROI-2 was significantly larger than that from another segmentation, but no significant differences were identified between the AUCs of other same parameters from the two different ROI methods. For the differentiaiton of IDHmut/1p19q- tumors and IDHmut/1p19q+ tumors, with the ROI-1, the ADCmode showed the biggest AUC (AUC: 0.784; sensitivity, 61.1%; specificity, 90.9%), with the ROI-2, and the skewness performed best (AUC, 0.821; sensitivity, 81.8%; specificity, 77.8%), but no significant differences were identified between the AUCs of the same parameters from the two different ROI methods. Conclusion ADC values analyzed by the histogram method could help to classify the genetic subtypes in lower-grade diffuse gliomas, no matter which ROI method was used. Extracting cystic and necrotic portions from the entire tumor lesions is preferable for evaluating the difference of the intratumoral heterogeneity and classifying IDH-wild tumors, but not significantly beneficial to predicting the 1p19q genotype in the lower-grade gliomas.
Collapse
|
39
|
Kang H, Chen P, Guo H, Zhang L, Tan Y, Xiao H, Yang A, Fang J, Zhang W. Vessel Size Imaging is Associated with IDH Mutation and Patient Survival in Diffuse Lower-Grade Glioma. Cancer Manag Res 2020; 12:9801-9811. [PMID: 33116839 PMCID: PMC7550213 DOI: 10.2147/cmar.s266533] [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: 06/07/2020] [Accepted: 09/07/2020] [Indexed: 11/23/2022] Open
Abstract
Background Patients with isocitrate dehydrogenase (IDH) mutant gliomas have better survival and appear to be more sensitive to chemotherapy than their IDH wild-type counterparts. We attempted to assess the correlations of vessel size imaging (VSI) values with IDH mutation status and patient survival in diffuse lower-grade glioma (LGG). Methods We enrolled 60 patients with diffuse LGGs, among which 43 had IDH-mutant tumors. All patients underwent VSI examination and VSI values for active tumors were calculated. Receiver operating characteristic (ROC) curves were established to evaluate the detection efficiency. Logistic regression was employed to determine the ability of variables to discriminate IDH mutational status. Kaplan–Meier survival analysis and Cox proportional hazards models were utilized to estimate the correlations of VSI values and other risk factors with patient survival. Results We observed that VSI values were lower in IDH-mutant LGGs than IDH wild-type LGGs. The VSImax and VSImean values had AUC values of 0.7305 and 0.7401, respectively, in distinguishing IDH-mutant LGGs from IDH wild-type LGGs. Logistic regression showed that VSImean values, age and tumor location were associated with IDH-mutant status, and the formula integrating the three factors had an AUC value of 0.7798 when distinguishing IDH-mutant LGGs from IDH wild-type LGGs. Moreover, LGG patients with high VSI values exhibited worse survival rates than those with low VSI values for both progression-free survival (PFS) and overall survival (OS). Multivariate Cox proportional hazards regression analysis suggested that IDH mutation status, VSImean values and multiple lesions or lobes were risk factors for PFS of LGG patients. Conclusion VSI value is associated with IDH genotype and maybe an independent predictor of the survival of patients with LGGs.
Collapse
Affiliation(s)
- Houyi Kang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Peng Chen
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Hong Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Letian Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Yong Tan
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Hualiang Xiao
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Ao Yang
- Department of Traffic Injury Research Office, Daping Hospital, Army Medical Center of PLA, Chongqing, People's Republic of China
| | - Jingqin Fang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| | - Weiguo Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, People's Republic of China
| |
Collapse
|
40
|
Li J, Tan T, Zhao L, Liu M, You Y, Zeng Y, Chen D, Xie T, Zhang L, Fu C, Zeng Z. Recent Advancements in Liposome-Targeting Strategies for the Treatment of Gliomas: A Systematic Review. ACS APPLIED BIO MATERIALS 2020; 3:5500-5528. [PMID: 35021787 DOI: 10.1021/acsabm.0c00705] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Malignant tumors represent some of the most intractable diseases that endanger human health. A glioma is a tumor of the central nervous system that is characterized by severe invasiveness, blurred boundaries between the tumor and surrounding normal tissue, difficult surgical removal, and high recurrence. Moreover, the blood-brain barrier (BBB) and multidrug resistance (MDR) are important factors that contribute to the lack of efficacy of chemotherapy in treating gliomas. A liposome is a biofilm-like drug delivery system with a unique phospholipid bilayer that exhibits high affinities with human tissues/organs (e.g., BBB). After more than five decades of development, classical and engineered liposomes consist of four distinct generations, each with different characteristics: (i) traditional liposomes, (ii) stealth liposomes, (iii) targeting liposomes, and (iv) biomimetic liposomes, which offer a promising approach to promote drugs across the BBB and to reverse MDR. Here, we review the history, preparatory methods, and physicochemical properties of liposomes. Furthermore, we discuss the mechanisms by which liposomes have assisted in the diagnosis and treatment of gliomas, including drug transport across the BBB, inhibition of efflux transporters, reversal of MDR, and induction of immune responses. Finally, we highlight ongoing and future clinical trials and applications toward further developing and testing the efficacies of liposomes in treating gliomas.
Collapse
Affiliation(s)
- Jie Li
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Tiantian Tan
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Liping Zhao
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Mengmeng Liu
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Yu You
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Yiying Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Dajing Chen
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Tian Xie
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Lele Zhang
- School of Medicine, Chengdu University, Chengdu 610106, Sichuan, China
| | - Chaomei Fu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Zhaowu Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| |
Collapse
|
41
|
Pandey U, Saini J, Kumar M, Gupta R, Ingalhalikar M. Normative Baseline for Radiomics in Brain MRI: Evaluating the Robustness, Regional Variations, and Reproducibility on FLAIR Images. J Magn Reson Imaging 2020; 53:394-407. [PMID: 32864820 DOI: 10.1002/jmri.27349] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/14/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Radiomics in neuroimaging has gained momentum as a noninvasive prediction tool not only to differentiate between types of brain tumors, but also to create phenotypic signatures in neurological and neuropsychiatric disorders. However, there is currently little understating about the robustness and reproducibility of radiomic features in a baseline normative population. PURPOSE To investigate the intra- and interscanner reproducibility, spatial robustness, and sensitivity of radiomics on fluid attenuation inversion recovery (FLAIR) images, which are widely used in neuro-oncology investigations. STUDY TYPE Retrospective. POPULATION Three separate datasets of healthy controls: 1) 87 subjects (age range 12-64 years), 2) intrascanner three timepoints, four subjects, and 3) interscanner, eight subjects at three different sites. FIELD STRENGTH/SEQUENCE T2 -weighted FLAIR at 1.5T and 3.0T. ASSESSMENT Spatial variance across lobes, and their relation with age/gender, intra- and inter-scanner reproducibility (with and without site harmonization) of radiomics. STATISTICAL TESTS Analysis of variance (ANOVA), interclass correlation (ICC), coefficient of variation (CoV), Bland-Altman analysis. RESULTS Analysis of data revealed no differences between genders; however, multiple radiomic features were highly associated with age (P < 0.05). Spatial variability was also evaluated where only 29.04% gray matter and 38.7% white matter features demonstrated an ICC >0.5. Furthermore, the results demonstrated intra-scanner reliability (ICC >0.5); however, inter-scanner reproducibility was poor, with ICC < 0.5 for 82% gray matter and 78.5% white matter features. The inter-scanner reliability improved (ICC < 0.5 for 39.67% gray matter and 38% white matter features) using site-harmonization techniques. DATA CONCLUSION These findings suggest that, accounting for age, spatial locations in radiomics-based analysis and use of intersite radiomics harmonization is crucial before interpreting these features for pathological inference. Level of Evidence 3. Technical Efficacy Stage 1. J. MAGN. RESON. IMAGING 2021;53:394-407.
Collapse
Affiliation(s)
- Umang Pandey
- Symbiosis Center for Medical Image Analysis, Symbiosis International University, Pune, India
| | - Jitender Saini
- Department of Radiology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Manoj Kumar
- Department of Radiology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Rakesh Gupta
- Department of Radiology, Fortis Hospital, Gurgaon, India
| | - Madhura Ingalhalikar
- Symbiosis Center for Medical Image Analysis, Symbiosis International University, Pune, India
| |
Collapse
|
42
|
Conventional MRI features of adult diffuse glioma molecular subtypes: a systematic review. Neuroradiology 2020; 63:353-362. [PMID: 32840682 DOI: 10.1007/s00234-020-02532-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/17/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE Molecular parameters have become integral to glioma diagnosis. Much of radiogenomics research has focused on the use of advanced MRI techniques, but conventional MRI sequences remain the mainstay of clinical assessments. The aim of this research was to synthesize the current published data on the accuracy of standard clinical MRI for diffuse glioma genotyping, specifically targeting IDH and 1p19q status. METHODS A systematic search was performed in September 2019 using PubMed and the Cochrane Library, identifying studies on the diagnostic value of T1 pre-/post-contrast, T2, FLAIR, T2*/SWI and/or 3-directional diffusion-weighted imaging sequences for the prediction of IDH and/or 1p19q status in WHO grade II-IV diffuse astrocytic and oligodendroglial tumours as defined in the WHO 2016 Classification of CNS Tumours. RESULTS Forty-four studies including a total of 5286 patients fulfilled the inclusion criteria. Correlations between key glioma molecular markers, namely IDH and 1p19q, and distinctive MRI findings have been established, including tumour location, signal composition (including the T2-FLAIR mismatch sign) and apparent diffusion coefficient values. CONCLUSION Consistent trends have emerged indicating that conventional MRI is valuable for glioma genotyping, particularly in presumed lower grade glioma. However, due to limited interobserver testing, the reproducibility of qualitatively assessed visual features remains an area of uncertainty.
Collapse
|
43
|
Chiang GC, Pisapia DJ, Liechty B, Magge R, Ramakrishna R, Knisely J, Schwartz TH, Fine HA, Kovanlikaya I. The Prognostic Value of MRI Subventricular Zone Involvement and Tumor Genetics in Lower Grade Gliomas. J Neuroimaging 2020; 30:901-909. [PMID: 32721076 DOI: 10.1111/jon.12763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/20/2020] [Accepted: 07/07/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Glioblastomas (GBMs) that involve the subventricular zone (SVZ) have a poor prognosis, possibly due to recruitment of neural stem cells. The purpose of this study was to evaluate whether SVZ involvement by lower grade gliomas (LGG), WHO grade II and III, similarly predicts poorer outcomes. We further assessed whether tumor genetics and cellularity are associated with SVZ involvement and outcomes. METHODS Forty-five consecutive LGG patients with preoperative imaging and next generation sequencing were included in this study. Regional SVZ involvement and whole tumor apparent diffusion coefficient (ADC) values, as a measure of cellularity, were assessed on magnetic resonance imaging. Progression was determined by RANO criteria. Kaplan-Meier curves and Cox regression analyses were used to determine the hazard ratios (HR) for progression and survival. RESULTS Frontal, parietal, temporal, and overall SVZ involvement and ADC values were not associated with progression or survival (P ≥ .05). However, occipital SVZ involvement, seen in two patients, was associated with a higher risk of tumor progression (HR = 6.6, P = .016) and death (HR = 31.5, P = .015), CDKN2A/B mutations (P = .03), and lower ADC histogram values at the 5th (P = .026) and 10th percentiles (P = .046). Isocitrate dehydrogenase, phosphatase and tensin homolog, epidermal growth factor receptor, and cyclin-dependent kinase 4 mutations were also prognostic (P ≤ .05). CONCLUSIONS Unlike in GBM, overall SVZ involvement was not found to strongly predict poor prognosis in LGGs. However, occipital SVZ involvement, though uncommon, was prognostic and found to be associated with CDKN2A/B mutations and tumor hypercellularity. Further investigation into these molecular mechanisms underlying occipital SVZ involvement in larger cohorts is warranted.
Collapse
Affiliation(s)
- Gloria C Chiang
- Department of Radiology, Division of Neuroradiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - David J Pisapia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Benjamin Liechty
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Rajiv Magge
- Department of Neurology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Rohan Ramakrishna
- Department of Neurosurgery, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Jonathan Knisely
- Department of Radiation Oncology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Theodore H Schwartz
- Department of Neurosurgery, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Howard A Fine
- Department of Neurology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Ilhami Kovanlikaya
- Department of Radiology, Division of Neuroradiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| |
Collapse
|
44
|
Simplified perfusion fraction from diffusion-weighted imaging in preoperative prediction of IDH1 mutation in WHO grade II-III gliomas: comparison with dynamic contrast-enhanced and intravoxel incoherent motion MRI. Radiol Oncol 2020; 54:301-310. [PMID: 32559177 PMCID: PMC7409598 DOI: 10.2478/raon-2020-0037] [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: 03/23/2020] [Accepted: 05/13/2020] [Indexed: 11/20/2022] Open
Abstract
Background Effect of isocitr ate dehydrogenase 1 (IDH1) mutation in neovascularization might be linked with tissue perfusion in gliomas. At present, the need of injection of contrast agent and the increasing scanning time limit the application of perfusion techniques. We used a simplified intravoxel incoherent motion (IVIM)-derived perfusion fraction (SPF) calculated from diffusion-weighted imaging (DWI) using only three b-values to quantitatively assess IDH1-linked tissue perfusion changes in WHO grade II-III gliomas (LGGs). Additionally, by comparing accuracy with dynamic contrast-enhanced (DCE) and full IVIM MRI, we tried to find the optimal imaging markers to predict IDH1 mutation status. Patients and methods Thirty patients were prospectively examined using DCE and multi-b-value DWI. All parameters were compared between the IDH1 mutant and wild-type LGGs using the Mann-Whitney U test, including the DCE MRI-derived Ktrans, ve and vp, the conventional apparen t diffusion coefficient (ADC0,1000), IVIM-de rived perfusion fraction (f), diffusion coefficient (D) and pseudo-diffusion coefficient (D*), SPF. We evaluated the diagnostic performance by receive r operating characteristic (ROC) analysis. Results Significant differences were detected between WHO grade II-III gliomas for all perfusion and diffusion parameters (P < 0.05). When compared to IDH1 mutant LGGs, IDH1 wild-type LGGs exhibited significantly higher perfusion metrics (P < 0.05) and lower diffusion metrics (P < 0.05). Among all parameters, SPF showed a higher diagnostic performance (area under the curve 0.861), with 94.4% sensitivity and 75% specificity. Conclusions DWI, DCE and IVIM MRI may noninvasively help discriminate IDH1 mutation statuses in LGGs. Specifically, simplified DWI-derived SPF showed a superior diagnostic performance.
Collapse
|
45
|
Bulakbaşı N, Paksoy Y. Correction to: Advanced imaging in adult diffusely infiltrating low-grade gliomas. Insights Imaging 2020; 11:57. [PMID: 32323033 PMCID: PMC7176752 DOI: 10.1186/s13244-020-00862-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The original article [1] contains errors in Table 1 in rows ktrans and Ve; the correct version of Table 1 can be viewed in this Correction article.
Collapse
Affiliation(s)
- Nail Bulakbaşı
- Medical Faculty, University of Kyrenia, Sehit Yahya Bakır Street, Karakum, Mersin-10, Kyrenia, Turkish Republic of Northern Cyprus, Turkey.
| | | |
Collapse
|
46
|
Maynard J, Okuchi S, Wastling S, Busaidi AA, Almossawi O, Mbatha W, Brandner S, Jaunmuktane Z, Koc AM, Mancini L, Jäger R, Thust S. World Health Organization Grade II/III Glioma Molecular Status: Prediction by MRI Morphologic Features and Apparent Diffusion Coefficient. Radiology 2020; 296:111-121. [PMID: 32315266 DOI: 10.1148/radiol.2020191832] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background A readily implemented MRI biomarker for glioma genotyping is currently lacking. Purpose To evaluate clinically available MRI parameters for predicting isocitrate dehydrogenase (IDH) status in patients with glioma. Materials and Methods In this retrospective study of patients studied from July 2008 to February 2019, untreated World Health Organization (WHO) grade II/III gliomas were analyzed by three neuroradiologists blinded to tissue results. Apparent diffusion coefficient (ADC) minimum (ADCmin) and mean (ADCmean) regions of interest were defined in tumor and normal appearing white matter (ADCNAWM). A visual rating of anatomic features (T1 weighted, T1 weighted with contrast enhancement, T2 weighted, and fluid-attenuated inversion recovery) was performed. Interobserver comparison (intraclass correlation coefficient and Cohen κ) was followed by nonparametric (Kruskal-Wallis analysis of variance) testing of associations between ADC metrics and glioma genotypes, including Bonferroni correction for multiple testing. Descriptors with sufficient concordance (intraclass correlation coefficient, >0.8; κ > 0.6) underwent univariable analysis. Predictive variables (P < .05) were entered into a multivariable logistic regression and tested in an additional test sample of patients with glioma. Results The study included 290 patients (median age, 40 years; interquartile range, 33-52 years; 169 male patients) with 82 IDH wild-type, 107 IDH mutant/1p19q intact, and 101 IDH mutant/1p19q codeleted gliomas. Two predictive models incorporating ADCmean-to-ADCNAWM ratio, age, and morphologic characteristics, with model A mandating calcification result and model B recording cyst formation, classified tumor type with areas under the receiver operating characteristic curve of 0.94 (95% confidence interval [CI]: 0.91, 0.97) and 0.96 (95% CI: 0.93, 0.98), respectively. In the test sample of 49 gliomas (nine IDH wild type, 21 IDH mutant/1p19q intact, and 19 IDH mutant/1p19q codeleted), the classification accuracy was 40 of 49 gliomas (82%; 95% CI: 71%, 92%) for model A and 42 of 49 gliomas (86%; 95% CI: 76%, 96%) for model B. Conclusion Two algorithms that incorporated apparent diffusion coefficient values, age, and tumor morphologic characteristics predicted isocitrate dehydrogenase status in World Health Organization grade II/III gliomas on the basis of standard clinical MRI sequences alone. © RSNA, 2020 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- John Maynard
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| | - Sachi Okuchi
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| | - Stephen Wastling
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| | - Ayisha Al Busaidi
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| | - Ofran Almossawi
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| | - Wonderboy Mbatha
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| | - Sebastian Brandner
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| | - Zane Jaunmuktane
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| | - Ali Murat Koc
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| | - Laura Mancini
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| | - Rolf Jäger
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| | - Stefanie Thust
- From the Neuroradiological Academic Unit, Department of Brain, Repair and Rehabilitation, UCL Institute of Neurology, London, England (J.M., S.O., S.W., L.M., R.J., S.T.); Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, England (S.W., A.A.B., W.M., A.M.K., L.M., R.J., S.T.); Population, Policy and Practice Research Unit, UCL Great Ormond Street Institute of Child Health, London, England (O.A.); Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, England (S.B., Z.J.); Department of Radiology, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey (A.M.K.); and Department of Imaging, University College London Foundation Hospital, London, England (R.J., S.T.)
| |
Collapse
|
47
|
Bai J, Varghese J, Jain R. Adult Glioma WHO Classification Update, Genomics, and Imaging: What the Radiologists Need to Know. Top Magn Reson Imaging 2020; 29:71-82. [PMID: 32271284 DOI: 10.1097/rmr.0000000000000234] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Recent advances in the understanding of the genetic makeup of gliomas have led to a paradigm shift in the diagnosis and classification of these tumors. Driven by these changes, the World Health Organization (WHO) introduced an update to its classification system of central nervous system (CNS) tumors in 2016. The updated glioma classification system incorporates molecular markers into tumor subgrouping, which has been shown to better correlate with tumor biology and behavior as well as patient prognosis than the previous purely histology-based classification system. Familiarity with this new classification scheme, the individual molecular markers, and corresponding imaging findings is critical for the radiologists who play an important role in diagnostic and surveillance imaging of patients with CNS tumors. The goals of this article are to review these updates to the WHO classification of CNS tumors with a focus on adult gliomas, provide an overview of key genomic markers of gliomas, and review imaging features pertaining to various genomic subgroups of adult gliomas.
Collapse
Affiliation(s)
- James Bai
- Department of Radiology, New York University Langone Health, New York, NY
| | - Jerrin Varghese
- Department of Radiology, New York University Langone Health, New York, NY
| | - Rajan Jain
- Department of Radiology, New York University Langone Health, New York, NY
- Department of Neurosurgery, New York University Langone Health, New York, NY
| |
Collapse
|
48
|
Ding H, Huang Y, Li Z, Li S, Chen Q, Xie C, Zhong Y. Prediction of IDH Status Through MRI Features and Enlightened Reflection on the Delineation of Target Volume in Low-Grade Gliomas. Technol Cancer Res Treat 2020; 18:1533033819877167. [PMID: 31564237 PMCID: PMC6767744 DOI: 10.1177/1533033819877167] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Isocitrate dehydrogenase mutational status defines distinct biologic behavior and
clinical outcomes in low-grade gliomas. We sought to determine magnetic resonance imaging
characteristics associated with isocitrate dehydrogenase mutational status to evaluate the
predictive roles of magnetic resonance imaging features in isocitrate dehydrogenase
mutational status and therefore their potential impact on the determination of clinical
target volume in radiotherapy. Forty-eight isocitrate dehydrogenase-mutant and 28
isocitrate dehydrogenase–wild-type low-grade gliomas were studied. Isocitrate
dehydrogenase mutation was related to more frequency of cortical involvement compared to
isocitrate dehydrogenase–wild-type group (34/46 vs 6/24, P = .0001).
Peritumoral edema was less frequent in isocitrate dehydrogenase–mutant tumors (32.6% vs
58.3% for isocitrate dehydrogenase–wild-type tumors, P = .0381).
Isocitrate dehydrogenase–wild-type tumors were more likely to have a nondefinable border,
while isocitrate dehydrogenase–mutant tumors had well-defined borders (66.7% vs 39.1%,
P = .0287). Only 8 (17.4%) of 46 of the isocitrate dehydrogenase–mutant
tumors demonstrated marked enhancement, while this was 66.7% in isocitrate–wild-type
tumors (P < .0001). Choline–creatinine ratio for isocitrate
dehydrogenase–wild-type tumors was significantly higher than that for isocitrate
dehydrogenase–mutant tumors. In conclusion, frontal location, well-defined border,
cortical involvement, less peritumoral edema, lack of enhancement, and low
choline–creatinine ratio were predictive for the definition of isocitrate
dehydrogenase–mutant low-grade gliomas. Magnetic resonance imaging can provide an
advantage in the detection of isocitrate dehydrogenase status indirectly and indicate the
need to explore new design for treatment planning in gliomas. Choline–creatinine ratio in
magnetic resonance spectroscopy could be a potential more reasonable reference for the new
design of delineation of target volume in low-grade gliomas.
Collapse
Affiliation(s)
- Haixia Ding
- Department of Chemotherapy and Radiation Therapy, Zhongnan Hospital, Wuhan University, Wuchang District, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuchang District, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yong Huang
- Department of Chemotherapy and Radiation Therapy, Zhongnan Hospital, Wuhan University, Wuchang District, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuchang District, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Zhiqiang Li
- Department of Neurologic Surgery, Zhongnan Hospital, Wuhan University, Wuchang District, Wuhan, China
| | - Sirui Li
- Department of Radiology, Zhongnan Hospital, Wuhan University, Wuchang District, Wuhan, China
| | - Qiongrong Chen
- Department of Pathology, Zhongnan Hospital, Wuhan University, Wuchang District, Wuhan, China
| | - Conghua Xie
- Department of Chemotherapy and Radiation Therapy, Zhongnan Hospital, Wuhan University, Wuchang District, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuchang District, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yahua Zhong
- Department of Chemotherapy and Radiation Therapy, Zhongnan Hospital, Wuhan University, Wuchang District, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuchang District, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| |
Collapse
|
49
|
MR image phenotypes may add prognostic value to clinical features in IDH wild-type lower-grade gliomas. Eur Radiol 2020; 30:3035-3045. [PMID: 32060714 DOI: 10.1007/s00330-020-06683-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/06/2020] [Accepted: 01/28/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE To identify significant prognostic magnetic resonance imaging (MRI) features and their prognostic value when added to clinical features in patients with isocitrate dehydrogenase wild-type (IDHwt) lower-grade gliomas. MATERIALS AND METHODS Preoperative MR images of 158 patients (discovery set = 112, external validation set = 46) with IDHwt lower-grade gliomas (WHO grade II or III) were retrospectively analyzed using the Visually Accessible Rembrandt Images feature set. Radiologic risk scores (RRSs) for overall survival were derived from the least absolute shrinkage and selection operator and elastic net. Multivariable Cox regression analysis, including age, Karnofsky Performance score, extent of resection, WHO grade, and RRS, was performed. The added prognostic value of RRS was calculated by comparing the integrated area under the receiver operating characteristic curve (iAUC) between models with and without RRS. RESULTS The presence of cysts, pial invasion, and cortical involvement were favorable prognostic factors, while ependymal extension, multifocal or multicentric distribution, nonlobar location, proportion of necrosis > 33%, satellites, and eloquent cortex involvement were significantly associated with worse prognosis. RRS independently predicted survival and significantly enhanced model performance for survival prediction when integrated to clinical features (iAUC increased to 0.773-0.777 from 0.737), which was successfully validated on the validation set (iAUC increased to 0.805-0.830 from 0.735). CONCLUSION MRI features associated with prognosis in patients with IDHwt lower-grade gliomas were identified. RRSs derived from MRI features independently predicted survival and significantly improved performance of survival prediction models when integrated into clinical features. KEY POINTS • Comprehensive analysis of MRI features conveys prognostic information in patients with isocitrate dehydrogenase wild-type lower-grade gliomas. • Presence of cysts, pial invasion, and cortical involvement of the tumor were favorable prognostic factors. • Radiological phenotypes derived from MRI independently predict survival and have the potential to improve survival prediction when added to clinical features.
Collapse
|
50
|
Imaging of Central Nervous System Tumors Based on the 2016 World Health Organization Classification. Neurol Clin 2020; 38:95-113. [DOI: 10.1016/j.ncl.2019.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|