1
|
Onishi S, Yamasaki F, Akiyama Y, Kawahara D, Amatya VJ, Yonezawa U, Taguchi A, Ozono I, Khairunnisa NI, Takeshima Y, Horie N. Usefulness of synthetic MRI for differentiation of IDH-mutant diffuse gliomas and its comparison with the T2-FLAIR mismatch sign. J Neurooncol 2024; 170:429-436. [PMID: 39133381 PMCID: PMC11538156 DOI: 10.1007/s11060-024-04794-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION The T2-FLAIR mismatch sign is a characteristic imaging biomarker for astrocytoma, isocitrate dehydrogenase (IDH)-mutant. However, investigators have provided varying interpretations of the positivity/negativity of this sign given for individual cases the nature of qualitative visual assessment. Moreover, MR sequence parameters also influence the appearance of the T2-FLAIR mismatch sign. To resolve these issues, we used synthetic MR technique to quantitatively evaluate and differentiate astrocytoma from oligodendroglioma. METHODS This study included 20 patients with newly diagnosed non-enhanced IDH-mutant diffuse glioma who underwent preoperative synthetic MRI using the Quantification of Relaxation Times and Proton Density by Multiecho acquisition of a saturation-recovery using Turbo spin-Echo Readout (QRAPMASTER) sequence at our institution. Two independent reviewers evaluated preoperative conventional MR images to determine the presence or absence of the T2-FLAIR mismatch sign. Synthetic MRI was used to measure T1, T2 and proton density (PD) values in the tumor lesion. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic performance. RESULTS The pathological diagnoses included astrocytoma, IDH-mutant (n = 12) and oligodendroglioma, IDH-mutant and 1p/19q-codeleted (n = 8). The sensitivity and specificity of T2-FLAIR mismatch sign for astrocytoma were 66.7% and 100% [area under the ROC curve (AUC) = 0.833], respectively. Astrocytoma had significantly higher T1, T2, and PD values than did oligodendroglioma (p < 0.0001, < 0.0001, and 0.0154, respectively). A cutoff lesion T1 value of 1580 ms completely differentiated astrocytoma from oligodendroglioma (AUC = 1.00). CONCLUSION Quantitative evaluation of non-enhanced IDH-mutant diffuse glioma using synthetic MRI allowed for better differentiation between astrocytoma and oligodendroglioma than did conventional T2-FLAIR mismatch sign. Measurement of T1 and T2 value by synthetic MRI could improve the differentiation of IDH-mutant diffuse gliomas.
Collapse
Affiliation(s)
- Shumpei Onishi
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8551, Japan
| | - Fumiyuki Yamasaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8551, Japan.
| | - Yuji Akiyama
- Department of Clinical Radiology, Hiroshima University Hospital, Hiroshima, Japan
| | - Daisuke Kawahara
- Department of Radiation Oncology, Graduate School of Biomedical Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Vishwa Jeet Amatya
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ushio Yonezawa
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8551, Japan
| | - Akira Taguchi
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8551, Japan
| | - Iori Ozono
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8551, Japan
| | - Novita Ikbar Khairunnisa
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8551, Japan
| | - Yukio Takeshima
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobutaka Horie
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8551, Japan
| |
Collapse
|
2
|
Hu C, Fang K, Du Q, Chen J, Wang L, Zhang J, Bai R, Wang Y. Diffusion-weighted MRI precisely predicts telomerase reverse transcriptase promoter mutation status in World Health Organization grade IV gliomas using a residual convolutional neural network. Br J Radiol 2024; 97:1806-1815. [PMID: 39152999 DOI: 10.1093/bjr/tqae146] [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: 12/19/2023] [Revised: 06/18/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024] Open
Abstract
OBJECTIVES Telomerase reverse transcriptase promoter (pTERT) mutation status plays a key role in making decisions and predicting prognoses for patients with World Health Organization (WHO) grade IV glioma. This study was conducted to assess the value of diffusion-weighted imaging (DWI) for predicting pTERT mutation status in WHO grade IV glioma. METHODS MRI data and molecular information were obtained for 266 patients with WHO grade IV glioma at the hospital and divided into training and validation sets. The ratio of training to validation set was approximately 10:3. We trained the same residual convolutional neural network (ResNet) for each MR modality, including structural MRIs (T1-weighted, T2-weighted, and contrast-enhanced T1-weighted) and DWI*, to compare the predictive capacities between DWI and conventional structural MRI. We also explored the effects of different regions of interest on pTERT mutation status prediction outcomes. RESULTS Structural MRI modalities poorly predicted the pTERT mutation status (accuracy = 51%-54%; area under the curve [AUC]=0.545-0.571), whereas DWI combined with its apparent diffusive coefficient maps yielded the best predictive performance (accuracy = 85.2%, AUC = 0.934). Including the radiological and clinical characteristics did not further improve the performance for predicting pTERT mutation status. The entire tumour volume yielded the best prediction performance. CONCLUSIONS DWI technology shows promising potential for predicting pTERT mutations in WHO grade IV glioma and should be included in the MRI protocol for WHO grade IV glioma in clinical practice. ADVANCES IN KNOWLEDGE This is the first large-scale model study to validate the predictive value of DWI for pTERT in WHO grade IV glioma.
Collapse
Affiliation(s)
- Congman Hu
- Department of Neurosurgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China
- Department of Neurosurgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Ke Fang
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou 310020, China
| | - Quan Du
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Jiarui Chen
- Department of Neurosurgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China
| | - Lin Wang
- Department of Neurosurgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China
| | - Jianmin Zhang
- Department of Neurosurgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China
| | - Ruiliang Bai
- Department of Physical Medicine and Rehabilitation of the Affiliated Sir Run Run Shaw Hospital and Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou 310020, China
- Department of Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310058, China
| | - Yongjie Wang
- Department of Neurosurgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China
| |
Collapse
|
3
|
Ozono I, Onishi S, Yonezawa U, Taguchi A, Khairunnisa NI, Amatya VJ, Yamasaki F, Takeshima Y, Horie N. Super T2-FLAIR mismatch sign: a prognostic imaging biomarker for non-enhancing astrocytoma, IDH-mutant. J Neurooncol 2024; 169:571-579. [PMID: 38995493 PMCID: PMC11341624 DOI: 10.1007/s11060-024-04758-4] [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: 12/19/2023] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
PURPOSE The T2-FLAIR mismatch sign is a highly specific diagnostic imaging biomarker for astrocytoma, IDH-mutant. However, a definitive prognostic imaging biomarker has yet to be identified. This study investigated imaging prognostic markers, specifically analyzing T2-weighted and FLAIR images of this tumor. METHODS We retrospectively analyzed 31 cases of non-enhancing astrocytoma, IDH-mutant treated at our institution, and 30 cases from The Cancer Genome Atlas (TCGA)/The Cancer Imaging Archive (TCIA). We defined "super T2-FLAIR mismatch sign" as having a significantly strong low signal comparable to cerebrospinal fluid at non-cystic lesions rather than just a pale FLAIR low-signal tumor lesion as in conventional T2-FLAIR mismatch sign. Cysts were defined as having a round or oval shape and were excluded from the criteria for the super T2-FLAIR mismatch sign. We evaluated the presence or absence of the T2-FLAIR mismatch sign and super T2-FLAIR mismatch sign using preoperative MRI and analyzed the progression-free survival (PFS) and overall survival (OS) by log-rank test. RESULTS The T2-FLAIR mismatch sign was present in 17 cases (55%) in our institution and 9 cases (30%) within the TCGA-LGG dataset without any correlation with PFS or OS. However, the super T2-FLAIR mismatch sign was detected in 8 cases (26%) at our institution and 13 cases (43%) in the TCGA-LGG dataset. At our institution, patients displaying the super T2-FLAIR mismatch sign showed significantly extended PFS (122.7 vs. 35.9 months, p = 0.0491) and OS (not reached vs. 116.7 months, p = 0.0232). Similarly, in the TCGA-LGG dataset, those with the super T2-FLAIR mismatch sign exhibited notably longer OS (not reached vs. 44.0 months, p = 0.0177). CONCLUSION The super T2-FLAIR mismatch is a promising prognostic imaging biomarker for non-enhancing astrocytoma, IDH-mutant.
Collapse
Affiliation(s)
- Iori Ozono
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shumpei Onishi
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Ushio Yonezawa
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Akira Taguchi
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Novita Ikbar Khairunnisa
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Vishwa Jeet Amatya
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Fumiyuki Yamasaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Yukio Takeshima
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobutaka Horie
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
4
|
Onishi S, Kojima M, Yamasaki F, Amatya VJ, Yonezawa U, Taguchi A, Ozono I, Go Y, Takeshima Y, Hiyama E, Horie N. T2-FLAIR mismatch sign, an imaging biomarker for CDKN2A-intact in non-enhancing astrocytoma, IDH-mutant. Neurosurg Rev 2024; 47:412. [PMID: 39117984 PMCID: PMC11310237 DOI: 10.1007/s10143-024-02632-5] [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: 12/23/2023] [Revised: 07/09/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
INTRODUCTION The WHO classification of central nervous system tumors (5th edition) classified astrocytoma, IDH-mutant accompanied with CDKN2A/B homozygous deletion as WHO grade 4. Loss of immunohistochemical (IHC) staining for methylthioadenosine phosphorylase (MTAP) was developed as a surrogate marker for CDKN2A-HD. Identification of imaging biomarkers for CDKN2A status is of immense clinical relevance. In this study, we explored the association between radiological characteristics of non-enhancing astrocytoma, IDH-mutant to the CDKN2A/B status. METHODS Thirty-one cases of astrocytoma, IDH-mutant with MTAP results by IHC were included in this study. The status of CDKN2A was diagnosed by IHC staining for MTAP in all cases, which was further confirmed by comprehensive genomic analysis in 12 cases. The T2-FLAIR mismatch sign, cystic component, calcification, and intratumoral microbleeding were evaluated. The relationship between the radiological features and molecular pathological diagnosis was analyzed. RESULTS Twenty-six cases were identified as CDKN2A-intact while 5 cases were CDKN2A-HD. The presence of > 33% and > 50% T2-FLAIR mismatch was observed in 23 cases (74.2%) and 14 cases (45.2%), respectively, and was associated with CDKN2A-intact astrocytoma (p = 0.0001, 0.0482). None of the astrocytoma, IDH-mutant with CDKN2A-HD showed T2-FLAIR mismatch sign. Cystic component, calcification, and intratumoral microbleeding were not associated with CDKN2A status. CONCLUSION In patients with non-enhancing astrocytoma, IDH-mutant, the T2-FLAIR mismatch sign is a potential imaging biomarker for the CDKN2A-intact subtype. This imaging biomarker may enable preoperative prediction of CDKN2A status among astrocytoma, IDH-mutant.
Collapse
Affiliation(s)
- Shumpei Onishi
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, 734-8551, Hiroshima, Japan
| | - Masato Kojima
- Department of Pediatric Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima, Japan
| | - Fumiyuki Yamasaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, 734-8551, Hiroshima, Japan.
| | - Vishwa Jeet Amatya
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ushio Yonezawa
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, 734-8551, Hiroshima, Japan
| | - Akira Taguchi
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, 734-8551, Hiroshima, Japan
| | - Iori Ozono
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, 734-8551, Hiroshima, Japan
| | - Yukari Go
- Medical Division Technical Center, Hiroshima University, Hiroshima, Japan
| | - Yukio Takeshima
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Eiso Hiyama
- Department of Pediatric Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima, Japan
| | - Nobutaka Horie
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, 734-8551, Hiroshima, Japan
| |
Collapse
|
5
|
Hwa JC, Wong AMC, Jung SM, Wu CT. Pediatric-type diffuse low-grade glioma with T2-FLAIR mismatch sign: a case report and literature review. Childs Nerv Syst 2024; 40:2271-2278. [PMID: 38884778 DOI: 10.1007/s00381-024-06487-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 06/01/2024] [Indexed: 06/18/2024]
Abstract
INTRODUCTION Pediatric-type diffuse low-grade gliomas are a new entity that was introduced in the fifth edition of the World Health Organization Classification of Tumors of the Central Nervous System, which was published in 2021. Notably, the information regarding the radiophenotypes of this new entity is limited. OBJECTIVE T2-FLAIR mismatch sign has been mostly studied in adult-type diffuse gliomas so far. We aimed to present more pediatric cases for future research about T2-FLAIR mismatch signs in pediatric-type diffuse low-grade gliomas. CASE PRESENTATION The current study presents a case of a 2-year-old boy who has a subcortical tumor at the right precentral frontal region. This tumor exhibited a T2-fluid-attenuated inversion recovery (FLAIR) mismatch sign that was identified as specific for isocitrate dehydrogenase (IDH)-mutant 1p/19q non-co-deleted astrocytomas. The tumor was pathologically identified as pediatric-type diffuse low-grade gliomas, and it tested negative for IDH-1 immunohistochemistry. The whole-exome sequencing of tumor tissue revealed negative results for IDH mutation, 1p/19q co-deletion, MYB rearrangement, and all other potential pathogenic mutations. CONCLUSION The T2-FLAIR mismatch sign may not be 100% specific for IDH-mutant gliomas, especially in children, and researchers must further investigate the pathophysiology of the T2-FLAIR mismatch sign in brain tumors and the radiophenotypes of entities of pediatric brain tumors.
Collapse
Affiliation(s)
- Jia-Ching Hwa
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Alex Mun-Ching Wong
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Keelung and Linkou, Chang Gung University College of Medicine, Keelung and Linkou, Taiwan
| | - Shih-Ming Jung
- Department of Pathology, Chang Gung Memorial Hospital, Taoyuan, Linkou, Taiwan
| | - Chieh-Tsai Wu
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Linkou, Taiwan.
| |
Collapse
|
6
|
Dagher SA, Lochner RH, Ozkara BB, Schomer DF, Wintermark M, Fuller GN, Ucisik FE. The T2-FLAIR mismatch sign in oncologic neuroradiology: History, current use, emerging data, and future directions. Neuroradiol J 2024; 37:441-453. [PMID: 37924213 PMCID: PMC11366202 DOI: 10.1177/19714009231212375] [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: 11/06/2023] Open
Abstract
The T2-Fluid-Attenuated Inversion Recovery (T2-FLAIR) mismatch sign is a radiogenomic marker that is easily discernible on preoperative conventional MR imaging. Application of strict criteria (adult population, cerebral hemisphere location, and classic imaging morphology) permits the noninvasive preoperative diagnosis of isocitrate dehydrogenase (IDH)-mutant 1p/19q-non-codeleted diffuse astrocytoma with near-perfect specificity, albeit with variably low sensitivity. This leads to improved preoperative planning and patient counseling. More recent research has shown that the application of less strict criteria compromises the near-perfect specificity of the sign but remains adequate for ruling out IDH-wildtype (glioblastoma) phenotype, which bears a far grimmer prognosis compared to IDH-mutant diffuse astrocytic disease. In this review, we elaborate on the various definitions of the T2-FLAIR mismatch sign present in the literature, illustrate these with images obtained at a comprehensive cancer center, discuss the potential of the mismatch sign for application to certain pediatric-type brain tumors, namely dysembryoplastic neuroepithelial tumor and diffuse midline glioma, and elaborate upon the clinical, histologic, and molecular associations of the T2-FLAIR mismatch sign as recognized to date. Finally, the sign's correlates in diffusion- and perfusion-weighted imaging are presented, and opportunities to further maximize the diagnostic and prognostic applications of the sign in the context of the 2021 revision of the WHO Classification of Central Nervous System Tumors are discussed.
Collapse
Affiliation(s)
- Samir A Dagher
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Riley Hideo Lochner
- Section of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Burak Berksu Ozkara
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Donald F Schomer
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Max Wintermark
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory N Fuller
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Section of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - F Eymen Ucisik
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
7
|
Yu X, Li S, Mai W, Hua X, Sun M, Lai M, Zhang D, Xiao Z, Wang L, Shi C, Luo L, Cai L. Pediatric diffuse intrinsic pontine glioma radiotherapy response prediction: MRI morphology and T2 intensity-based quantitative analyses. Eur Radiol 2024:10.1007/s00330-024-10855-9. [PMID: 38907098 DOI: 10.1007/s00330-024-10855-9] [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: 12/27/2023] [Revised: 03/31/2024] [Accepted: 04/25/2024] [Indexed: 06/23/2024]
Abstract
OBJECTIVES An easy-to-implement MRI model for predicting partial response (PR) postradiotherapy for diffuse intrinsic pontine glioma (DIPG) is lacking. Utilizing quantitative T2 signal intensity and introducing a visual evaluation method based on T2 signal intensity heterogeneity, and compared MRI radiomic models for predicting radiotherapy response in pediatric patients with DIPG. METHODS We retrospectively included patients with brainstem gliomas aged ≤ 18 years admitted between July 2011 and March 2023. Applying Response Assessment in Pediatric Neuro-Oncology criteria, we categorized patients into PR and non-PR groups. For qualitative analysis, tumor heterogeneity vision was classified into four grades based on T2-weighted images. Quantitative analysis included the relative T2 signal intensity ratio (rT2SR), extra pons volume ratio, and tumor ring-enhancement volume. Radiomic features were extracted from T2-weighted and T1-enhanced images of volumes of interest. Univariate analysis was used to identify independent variables related to PR. Multivariate logistic regression was performed using significant variables (p < 0.05) from univariate analysis. RESULTS Of 140 patients (training n = 109, and test n = 31), 64 (45.7%) achieved PR. The AUC of the predictive model with extrapontine volume ratio, rT2SRmax-min (rT2SRdif), and grade was 0.89. The AUCs of the T2-weighted and T1WI-enhanced models with radiomic signatures were 0.84 and 0.81, respectively. For the 31 DIPG test sets, the AUCs were 0.91, 0.83, and 0.81, for the models incorporating the quantitative features, radiomic model (T2-weighted images, and T1W1-enhanced images), respectively. CONCLUSION Combining T2-weighted quantification with qualitative and extrapontine volume ratios reliably predicted pediatric DIPG radiotherapy response. CLINICAL RELEVANCE STATEMENT Combining T2-weighted quantification with qualitative and extrapontine volume ratios can accurately predict diffuse intrinsic pontine glioma (DIPG) radiotherapy response, which may facilitate personalized treatment and prognostic assessment for patients with DIPG. KEY POINTS Early identification is crucial for radiotherapy response and risk stratification in diffuse intrinsic pontine glioma. The model using tumor heterogeneity and quantitative T2 signal metrics achieved an AUC of 0.91. Using a combination of parameters can effectively predict radiotherapy response in this population.
Collapse
Affiliation(s)
- Xiaojun Yu
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Shaoqun Li
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Wenfeng Mai
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Xiaoyu Hua
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Mengnan Sun
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Mingyao Lai
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Dong Zhang
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Zeyu Xiao
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Lichao Wang
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Changzheng Shi
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China.
| | - Liangping Luo
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China.
| | - Linbo Cai
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China.
| |
Collapse
|
8
|
Malik P, Soliman R, Chen YA, Munoz DG, Das S, Bharatha A, Mathur S. Patterns of T2-FLAIR discordance across a cohort of adult-type diffuse gliomas and deviations from the classic T2-FLAIR mismatch sign. Neuroradiology 2024; 66:521-530. [PMID: 38347151 DOI: 10.1007/s00234-024-03297-z] [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: 10/25/2023] [Accepted: 01/25/2024] [Indexed: 03/14/2024]
Abstract
PURPOSE T2-FLAIR mismatch serves as a highly specific but insensitive marker for IDH-mutant (IDHm) astrocytoma with potential limitations in real-world application. We aimed to assess the utility of a broader definition of T2-FLAIR discordance across a cohort of adult-type diffuse lower-grade gliomas (LrGG) to see if specific patterns emerge and additionally examine factors determining deviation from the classic T2-FLAIR mismatch sign. METHODS Preoperative MRIs of non-enhancing adult-type diffuse LrGGs were reviewed. Relevant demographic, molecular, and MRI data were compared across tumor subgroups. RESULTS Eighty cases satisfied the inclusion criteria. Highest discordance prevalence and > 50% T2-FLAIR discordance volume were noted with IDHm astrocytomas (P < 0.001), while < 25% discordance volume was associated with oligodendrogliomas (P = 0.03) and IDH-wildtype (IDHw) LrGG (P = 0.004). "T2-FLAIR matched pattern" was associated with IDHw LrGG (P < 0.001) and small or minimal areas of discordance with oligodendrogliomas (P = 0.03). Sensitivity and specificity of classic mismatch sign for IDHm astrocytoma were 25.7% and 100%, respectively (P = 0.06). Retained ATRX expression and/or non-canonical IDH mutation (n = 10) emerged as a significant factor associated with absence of classic T2-FLAIR mismatch sign in IDHm astrocytomas (100%, P = 0.02) and instead had minimal discordance or matched pattern (40%, P = 0.04). CONCLUSION T2-FLAIR discordance patterns in adult-type diffuse LrGGs exist on a diverging but distinct spectrum of classic mismatch to T2-FLAIR matched patterns. Specific molecular markers may play a role in deviations from classic mismatch sign.
Collapse
Affiliation(s)
- Prateek Malik
- Division of Neuroradiology, Department of Medical Imaging, St. Michael's Hospital, University of Toronto, 30 Bond St., Toronto, ON, M5B 1W8, Canada
| | - Radwa Soliman
- Diagnostic and Interventional Radiology Department, Assiut University, Asyut, Egypt
| | - Yingming Amy Chen
- Division of Neuroradiology, Department of Medical Imaging, St. Michael's Hospital, University of Toronto, 30 Bond St., Toronto, ON, M5B 1W8, Canada
| | - David G Munoz
- Department of Pathology, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Sunit Das
- Division of Neurosurgery, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Aditya Bharatha
- Division of Neuroradiology, Department of Medical Imaging, St. Michael's Hospital, University of Toronto, 30 Bond St., Toronto, ON, M5B 1W8, Canada
| | - Shobhit Mathur
- Division of Neuroradiology, Department of Medical Imaging, St. Michael's Hospital, University of Toronto, 30 Bond St., Toronto, ON, M5B 1W8, Canada.
| |
Collapse
|
9
|
He L, Zhang H, Li T, Yang J, Zhou Y, Wang J, Saidaer T, Bai X, Liu X, Wang Y, Wang L. Identifying IDH-mutant and 1p/19q noncodeleted astrocytomas from nonenhancing gliomas: Manual recognition followed by artificial intelligence recognition. Neurooncol Adv 2024; 6:vdae013. [PMID: 38405203 PMCID: PMC10894653 DOI: 10.1093/noajnl/vdae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024] Open
Abstract
Background The T2-FLAIR mismatch sign (T2FM) has nearly 100% specificity for predicting IDH-mutant and 1p/19q noncodeleted astrocytomas (astrocytomas). However, only 18.2%-56.0% of astrocytomas demonstrate a positive T2FM. Methods must be considered for distinguishing astrocytomas from negative T2FM gliomas. In this study, positive T2FM gliomas were manually distinguished from nonenhancing gliomas, and then a support vector machine (SVM) classification model was used to distinguish astrocytomas from negative T2FM gliomas. Methods Nonenhancing gliomas (regardless of pathological type or grade) diagnosed between January 2022 and October 2022 (N = 300) and November 2022 and March 2023 (N = 196) will comprise the training and validation sets, respectively. Our method for distinguishing astrocytomas from nonenhancing gliomas was examined and validated using the training set and validation set. Results The specificity of T2FM for predicting astrocytomas was 100% in both the training and validation sets, while the sensitivity was 42.75% and 67.22%, respectively. Using a classification model of SVM based on radiomics features, among negative T2FM gliomas, the accuracy was above 85% when the prediction score was greater than 0.70 in identifying astrocytomas and above 95% when the prediction score was less than 0.30 in identifying nonastrocytomas. Conclusions Manual screening of positive T2FM gliomas, followed by the SVM classification model to differentiate astrocytomas from negative T2FM gliomas, may be a more effective method for identifying astrocytomas in nonenhancing gliomas.
Collapse
Affiliation(s)
- Lei He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Hong Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Tianshi Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jianing Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yanpeng Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jiaxiang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Tuerhong Saidaer
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoyan Bai
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xing Liu
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, People’s Republic of China
- Chinese Institute for Brain Research, Beijing, People’s Republic of China
| | - Lei Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
10
|
Jen JP, Li X, Patel M, Haq H, Pohl U, Nagaraju S, Wykes V, Sanghera P, Watts C, Sawlani V. Beyond T2-FLAIR mismatch sign in isocitrate dehydrogenase mutant 1p19q non-codeleted astrocytoma: Analysis of tumor core and evolution with multiparametric magnetic resonance imaging. Neurooncol Adv 2024; 6:vdae065. [PMID: 39071736 PMCID: PMC11275453 DOI: 10.1093/noajnl/vdae065] [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] [Indexed: 07/30/2024] Open
Abstract
Background The T2-FLAIR mismatch sign is an imaging correlate for isocitrate dehydrogenase (IDH)-mutant 1p19q non-codeleted astrocytomas. However, it is only seen in a part of the cases at certain stages. Many of the tumors likely lose T2 homogeneity as they grow in size, and become heterogenous. The aim of this study was to investigate the timecourse of T2-FLAIR mismatch sign, and assess intratumoral heterogeneity using multiparametric magnetic resonance imaging techniques. Methods A total of 128 IDH-mutant gliomas were retrospectively analyzed. Observers blinded to molecular status used strict criteria to select T2-FLAIR mismatch astrocytomas. Pre-biopsy and follow-up standard structural sequences of T2, FLAIR and apparent diffusion coefficient, MR spectroscopy (both single- and multi-voxel techniques), and DSC perfusion were observed. Results Nine T2-FLAIR mismatch astrocytomas were identified. 7 had MR spectroscopy and perfusion data. The smallest astrocytomas began as rounded T2 homogeneous lesions without FLAIR suppression, and developed T2-FLAIR mismatch during follow-up with falls in NAA and raised Cho/Cr ratio. Larger tumors at baseline with T2-FLAIR mismatch signs developed intratumoral heterogeneity, and showed elevated Cho/Cr ratio and raised relative cerebral blood volume (rCBV). The highest levels of intratumoral Cho/Cr and rCBV changes were located within the tumor core, and this area signifies the progression of the tumors toward high grade. Conclusions T2-FLAIR mismatch sign is seen at a specific stage in the development of astrocytoma. By assessing the subsequent heterogeneity, MR spectroscopy and perfusion imaging are able to predict the progression of the tumor towards high grade, thereby can assist targeting for biopsy and selective debulking.
Collapse
Affiliation(s)
- Jian Ping Jen
- Department of Neuroradiology, University Hospitals Birmingham, Birmingham, UK
| | - Xuanxuan Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Markand Patel
- Department of Neuroradiology, University Hospitals Birmingham, Birmingham, UK
| | - Huzaifah Haq
- Department of Neuroradiology, University Hospitals Birmingham, Birmingham, UK
| | - Ute Pohl
- Department of Cellular Pathology, University Hospitals Birmingham, Birmingham, UK
| | - Santhosh Nagaraju
- Department of Cellular Pathology, University Hospitals Birmingham, Birmingham, UK
| | - Victoria Wykes
- Neuroimaging, University of Birmingham, Birmingham, UK
- Department of Neurosurgery, University Hospitals Birmingham, Birmingham, UK
| | - Paul Sanghera
- Neuroimaging, University of Birmingham, Birmingham, UK
| | - Colin Watts
- Neuroimaging, University of Birmingham, Birmingham, UK
- Department of Neurosurgery, University Hospitals Birmingham, Birmingham, UK
| | - Vijay Sawlani
- Department of Neuroradiology, University Hospitals Birmingham, Birmingham, UK
- Neuroimaging, University of Birmingham, Birmingham, UK
| |
Collapse
|
11
|
Naeem A, Aziz N, Nasir M, Rangwala HS, Fatima H, Mubarak F. Accuracy of MRI in Detecting 1p/19q Co-deletion Status of Gliomas: A Single-Center Retrospective Study. Cureus 2024; 16:e51863. [PMID: 38327950 PMCID: PMC10848880 DOI: 10.7759/cureus.51863] [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] [Accepted: 01/08/2024] [Indexed: 02/09/2024] Open
Abstract
Background Oligodendrogliomas, rare brain tumors in the frontal lobe's white matter, are reshaped by molecular markers like isocitrate dehydrogenase mutations and 1p/19q co-deletion, influencing treatment outcomes. Despite the initial indolence, these tumors pose a significant risk, with a median survival of 10-12 years. Non-invasive alternatives, such as magnetic resonance imaging (MRI) for assessing T2-fluid-attenuated inversion recovery (FLAIR) mismatch and calcifications, provide insights into molecular subtypes and aid prognosis. Our study explored these features to predict the oligodendroglioma status and refine patient management to improve outcomes. Methods In this retrospective study, patient data identified patients with suspected central nervous system tumors undergoing MRI, revealing low-grade gliomas. Surgical biopsy and 1p/19q fluorescence in situ hybridization confirmed the co-deletion status. MRI was used to assess various morphological features. Statistical analyses included x2 tests, Fisher's exact tests, Kruskal-Wallis tests, and binary logistic regression models, with significance set at p < 0.05. Results Seventy-three patients (median age, 37 years) were stratified according to 1p/19q co-deletion. Most (61.6%) were 18-40 years old and mostly male (67.1%). Co-deletion cases, primarily frontal lobe lesions (67.6%), were unilateral (88.2%), with 55.9% non-circumscribed margins and 58.8% ill-defined contours. Smooth contrast enhancement and no necrosis were observed in 48.1% of 1p/19q co-deletion cases. Logistic regression analysis showed a significant association between ill-defined/irregular contours and 1p/19q co-deletion. Fisher's exact test confirmed this but raised concerns about the small sample size influencing the conclusions. Conclusions This study established a significant link between glioma tumor contour characteristics, particularly irregular and ill-defined contours, and the likelihood of 1p/19q co-deletion. Our findings underscore the clinical relevance of using tumor contours in treatment decisions and prognosis assessments.
Collapse
Affiliation(s)
- Adnan Naeem
- Department of Radiology, Aga Khan University Hospital, Karachi, PAK
| | - Namrah Aziz
- Department of Radiology, Aga Khan Health Service, Karachi, PAK
| | - Manal Nasir
- Department of Radiology, Aga Khan University Hospital, Karachi, PAK
| | | | - Hareer Fatima
- Department of Medicine, Jinnah Sindh Medical University, Karachi, PAK
| | - Fatima Mubarak
- Department of Radiology, Aga Khan University Hospital, Karachi, PAK
| |
Collapse
|
12
|
Godoy LFDS, Paes VR, Ayres AS, Bandeira GA, Moreno RA, Hirata FDCC, Silva FAB, Nascimento F, Campos Neto GDC, Gentil AF, Lucato LT, Amaro Junior E, Young RJ, Malheiros SMF. Advances in diffuse glial tumors diagnosis. ARQUIVOS DE NEURO-PSIQUIATRIA 2023; 81:1134-1145. [PMID: 38157879 PMCID: PMC10756793 DOI: 10.1055/s-0043-1777729] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/27/2023] [Indexed: 01/03/2024]
Abstract
In recent decades, there have been significant advances in the diagnosis of diffuse gliomas, driven by the integration of novel technologies. These advancements have deepened our understanding of tumor oncogenesis, enabling a more refined stratification of the biological behavior of these neoplasms. This progress culminated in the fifth edition of the WHO classification of central nervous system (CNS) tumors in 2021. This comprehensive review article aims to elucidate these advances within a multidisciplinary framework, contextualized within the backdrop of the new classification. This article will explore morphologic pathology and molecular/genetics techniques (immunohistochemistry, genetic sequencing, and methylation profiling), which are pivotal in diagnosis, besides the correlation of structural neuroimaging radiophenotypes to pathology and genetics. It briefly reviews the usefulness of tractography and functional neuroimaging in surgical planning. Additionally, the article addresses the value of other functional imaging techniques such as perfusion MRI, spectroscopy, and nuclear medicine in distinguishing tumor progression from treatment-related changes. Furthermore, it discusses the advantages of evolving diagnostic techniques in classifying these tumors, as well as their limitations in terms of availability and utilization. Moreover, the expanding domains of data processing, artificial intelligence, radiomics, and radiogenomics hold great promise and may soon exert a substantial influence on glioma diagnosis. These innovative technologies have the potential to revolutionize our approach to these tumors. Ultimately, this review underscores the fundamental importance of multidisciplinary collaboration in employing recent diagnostic advancements, thereby hoping to translate them into improved quality of life and extended survival for glioma patients.
Collapse
Affiliation(s)
- Luis Filipe de Souza Godoy
- Hospital Israelita Albert Einstein, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | - Vitor Ribeiro Paes
- Hospital Israelita Albert Einstein, Laboratório de Patologia Cirúrgica, São Paulo SP, Brazil.
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Patologia, São Paulo SP, Brazil.
| | - Aline Sgnolf Ayres
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | - Gabriela Alencar Bandeira
- Instituto do Câncer do Estado de São Paulo, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | - Raquel Andrade Moreno
- Instituto do Câncer do Estado de São Paulo, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
- Rede D'Or São Luiz, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | | | | | - Felipe Nascimento
- Hospital Israelita Albert Einstein, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | | | - Andre Felix Gentil
- Hospital Israelita Albert Einstein, Departamento de Neurocirurgia, São Paulo SP, Brazil.
| | - Leandro Tavares Lucato
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Seção de Neuroradiologia, São Paulo SP, Brazil.
- Grupo Fleury, São Paulo SP, Brazil.
| | - Edson Amaro Junior
- Hospital Israelita Albert Einstein, Departamento de Radiologia, Seção de Neuroradiologia, São Paulo SP, Brazil.
| | - Robert J. Young
- Memorial Sloan-Kettering Cancer Center, Neuroradiology Service, New York, New York, United States.
| | | |
Collapse
|
13
|
Lasocki A, Roberts-Thomson SJ, Gaillard F. Radiogenomics of adult intracranial gliomas after the 2021 World Health Organisation classification: a review of changes, challenges and opportunities. Quant Imaging Med Surg 2023; 13:7572-7581. [PMID: 37969636 PMCID: PMC10644132 DOI: 10.21037/qims-22-1365] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/29/2023] [Indexed: 11/17/2023]
Abstract
The classification of diffuse gliomas has undergone substantial changes over the last decade, starting with the 2016 World Health Organisation (WHO) classification, which introduced the importance of molecular markers for glioma diagnosis, in particular, isocitrate dehydrogenase (IDH) status and 1p/19-codeletion. This has spurred research into the correlation of imaging features with the key molecular markers, known as "radiogenomics" or "imaging genomics". Radiogenomics has a variety of possible benefits, including supplementing immunohistochemistry to refine the histological diagnosis and overcoming some of the limitations of the histological assessment. The recent 2021 WHO classification has introduced a variety of changes and continues the trend of increasing the importance of molecular markers in the diagnosis. Key changes include a formal distinction between adult- and paediatric-type diffuse gliomas, the addition of new diagnostic entities, refinements to the nomenclature for IDH-mutant (IDHmut) and IDH-wildtype (IDHwt) gliomas, a shift to grading within tumour types, and the addition of molecular markers as a determinant of tumour grade in addition to phenotype. These changes provide both challenges and opportunities for the field of radiogenomics, which are discussed in this review. This includes implications for the interpretation of research performed prior to the 2021 classification, based on the shift to first classifying gliomas based on genotype ahead of grade, as well as opportunities for future research and priorities for clinical integration.
Collapse
Affiliation(s)
- Arian Lasocki
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Radiology, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Frank Gaillard
- Department of Radiology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Radiology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
14
|
Lasocki A, Buckland ME, Molinaro T, Xie J, Gaillard F. Radiogenomics Provides Insights into Gliomas Demonstrating Single-Arm 1p or 19q Deletion. AJNR Am J Neuroradiol 2023; 44:1270-1274. [PMID: 37884300 PMCID: PMC10631530 DOI: 10.3174/ajnr.a8034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND AND PURPOSE IDH-mutant gliomas are further divided on the basis of 1p/19q status: oligodendroglioma, IDH-mutant and 1p/19q-codeleted, and astrocytoma, IDH-mutant (without codeletion). Occasionally, testing may reveal single-arm 1p or 19q deletion (unideletion), which remains within the diagnosis of astrocytoma. Molecular assessment has some limitations, however, raising the possibility that some unideleted tumors could actually be codeleted. This study assessed whether unideleted tumors had MR imaging features and survival more consistent with astrocytomas or oligodendrogliomas. MATERIALS AND METHODS One hundred twenty-one IDH-mutant grade 2-3 gliomas with 1p/19q results were identified. Two neuroradiologists assessed the T2-FLAIR mismatch sign and calcifications, as differentiators of astrocytomas and oligodendrogliomas. MR imaging features and survival were compared among the unideleted tumors, codeleted tumors, and those without 1p or 19q deletion. RESULTS The cohort comprised 65 tumors without 1p or 19q deletion, 12 unideleted tumors, and 44 codeleted. The proportion of unideleted tumors demonstrating the T2-FLAIR mismatch sign (33%) was similar to that in tumors without deletion (49%; P = .39), but significantly higher than codeleted tumors (0%; P = .001). Calcifications were less frequent in unideleted tumors (0%) than in codeleted tumors (25%), but this difference did not reach statistical significance (P = .097). The median survival of patients with unideleted tumors was 7.8 years, which was similar to that in tumors without deletion (8.5 years; P = .72) but significantly shorter than that in codeleted tumors (not reaching median survival after 12 years; P = .013). CONCLUSIONS IDH-mutant gliomas with single-arm 1p or 19q deletion have MR imaging appearance and survival that are similar to those of astrocytomas without 1p or 19q deletion and significantly different from those of 1p/19q-codeleted oligodendrogliomas.
Collapse
Affiliation(s)
- Arian Lasocki
- From the Department of Cancer Imaging (A.L.), Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology (A.L.), The University of Melbourne, Parkville, Victoria, Australia
- Department of Radiology (A.L., F.G.), The University of Melbourne, Parkville, Victoria, Australia
| | - Michael E Buckland
- Department of Neuropathology (M.E.B.), Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- School of Medical Sciences (M.E.B.), University of Sydney, Camperdown, New South Wales, Australia
| | - Tahlia Molinaro
- Department of Medical Oncology (T.M.), Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jing Xie
- Centre for Biostatistics and Clinical Trials (J.X.), Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Frank Gaillard
- Department of Radiology (A.L., F.G.), The University of Melbourne, Parkville, Victoria, Australia
- Department of Radiology (F.G.), The Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
15
|
Branzoli F, Liserre R, Deelchand DK, Poliani PL, Bielle F, Nichelli L, Sanson M, Lehéricy S, Marjańska M. Neurochemical Differences between 1p/19q Codeleted and Noncodeleted IDH-mutant Gliomas by in Vivo MR Spectroscopy. Radiology 2023; 308:e223255. [PMID: 37668523 PMCID: PMC10546286 DOI: 10.1148/radiol.223255] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 06/19/2023] [Accepted: 06/29/2023] [Indexed: 09/06/2023]
Abstract
Background Noninvasive identification of glioma subtypes is important for optimizing treatment strategies. Purpose To compare the in vivo neurochemical profiles between isocitrate dehydrogenase (IDH) 1-mutant 1p/19q codeleted gliomas and their noncodeleted counterparts measured by MR spectroscopy at 3.0 T with a point-resolved spectroscopy (PRESS) sequence optimized for D-2-hydroxyglutarate (2HG) detection. Materials and Methods Adults with IDH1-mutant gliomas were retrospectively included for this study from two university hospitals (inclusion period: January 2015 to July 2016 and September 2019 to June 2021, respectively) based on availability of 1p/19q codeletion status and a PRESS acquisition optimized for 2HG detection (echo time, 97 msec) at 3.0 T before any treatment. Spectral analysis was performed using LCModel and a simulated basis set. Metabolite quantification was performed using the water signal as a reference and correcting for water and metabolite longitudinal and transverse relaxation time constants. Concentration ratios were computed using total creatine (tCr) and total choline. A two-tailed unpaired t test was used to compare metabolite concentrations obtained in codeleted versus noncodeleted gliomas, accounting for multiple comparisons. Results Thirty-one adults (mean age, 39 years ± 8 [SD]; 19 male) were included, and 19 metabolites were quantified. Cystathionine concentration was higher in codeleted (n = 13) than noncodeleted (n = 18) gliomas when quantification was performed using the water signal or tCr as references (2.33 mM ± 0.98 vs 0.93 mM ± 0.94, and 0.34 mM ± 0.14 vs 0.14 mM ± 0.14, respectively; both P < .001). The sensitivity and specificity of PRESS to detect codeletion by means of cystathionine quantification were 92% and 61%, respectively. Other metabolites did not show evidence of a difference between groups (P > .05). Conclusion Higher cystathionine levels were detected in IDH1-mutant 1p/19q codeleted gliomas than in their noncodeleted counterparts with use of a PRESS sequence optimized for 2HG detection. Of 19 metabolites quantified, only cystathionine showed evidence of a difference in concentration between groups. Clinical trial registry no. NCT01703962 © RSNA, 2023 See also the editorial by Lin in this issue.
Collapse
Affiliation(s)
- Francesca Branzoli
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Roberto Liserre
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Dinesh K. Deelchand
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Pietro Luigi Poliani
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Franck Bielle
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Lucia Nichelli
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Marc Sanson
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Stéphane Lehéricy
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| | - Małgorzata Marjańska
- From the Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Paris Brain Institute–L’Institut du Cerveau et de la Moelle Épinière (ICM), 47 boulevard de l’Hôpital, 75013 Paris, France (F. Branzoli, L.N., M.S., S.L.); Center for Neuroimaging Research (CENIR), L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (F. Branzoli, S.L.); Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy (R.L.); Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minn (D.K.D., M.M.); Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (P.L.P.); Laboratory R Escourolle (F. Bielle), Department of Neuroradiology (L.N., S.L.), and Department of Neurology 2 (M.S.), University Hospital La Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France; and Onconeurotek Tumor Bank, L’Institut du Cerveau et de la Moelle Épinière (ICM), Paris, France (M.S.)
| |
Collapse
|
16
|
Park YW, Vollmuth P, Foltyn-Dumitru M, Sahm F, Ahn SS, Chang JH, Kim SH. The 2021 WHO Classification for Gliomas and Implications on Imaging Diagnosis: Part 1-Key Points of the Fifth Edition and Summary of Imaging Findings on Adult-Type Diffuse Gliomas. J Magn Reson Imaging 2023; 58:677-689. [PMID: 37069792 DOI: 10.1002/jmri.28743] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/19/2023] Open
Abstract
The fifth edition of the World Health Organization (WHO) classification of central nervous system tumors published in 2021 advances the role of molecular diagnostics in the classification of gliomas by emphasizing integrated diagnoses based on histopathology and molecular information and grouping tumors based on genetic alterations. Importantly, molecular biomarkers that provide important prognostic information are now a parameter for establishing tumor grades in gliomas. Understanding the 2021 WHO classification is crucial for radiologists for daily imaging interpretation as well as communication with clinicians. Although imaging features are not included in the 2021 WHO classification, imaging can serve as a powerful tool to impact the clinical practice not only prior to tissue confirmation but beyond. This review represents the first of a three-installment review series on the 2021 WHO classification for gliomas, glioneuronal tumors, and neuronal tumors and implications on imaging diagnosis. This Part 1 Review focuses on the major changes to the classification of gliomas and imaging findings on adult-type diffuse gliomas. EVIDENCE LEVEL: 3. TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Yae Won Park
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Philipp Vollmuth
- Section for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University College of Medicine, Heidelberg, Germany
| | - Martha Foltyn-Dumitru
- Section for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University College of Medicine, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Heidelberg University College of Medicine, Heidelberg, Germany
| | - Sung Soo Ahn
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, 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
| |
Collapse
|
17
|
Kalaroopan D, Lasocki A. MRI-based deep learning techniques for the prediction of isocitrate dehydrogenase and 1p/19q status in grade 2-4 adult gliomas. J Med Imaging Radiat Oncol 2023; 67:492-498. [PMID: 36919468 DOI: 10.1111/1754-9485.13522] [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: 11/24/2021] [Accepted: 02/16/2023] [Indexed: 03/16/2023]
Abstract
Molecular biomarkers are becoming increasingly important in the classification of intracranial gliomas. While tissue sampling remains the gold standard, there is growing interest in the use of deep learning (DL) techniques to predict these markers. This narrative review with a systematic approach identifies and synthesises the current published data on DL techniques using conventional MRI sequences for predicting isocitrate dehydrogenase (IDH) and 1p/19q-codeletion status in World Health Organisation grade 2-4 gliomas. Three databases were searched for relevant studies. In all, 13 studies met the inclusion criteria after exclusions. Key results, limitations and discrepancies between studies were synthesised. High accuracy has been reported in some studies, but the existing literature has several limitations, including generally small cohort sizes, a paucity of studies with independent testing cohorts and a lack of studies assessing IDH and 1p/19q together. While DL shows promise as a non-invasive means of predicting glioma genotype, addressing these limitations in future research will be important for facilitating clinical translation.
Collapse
Affiliation(s)
- Dinusha Kalaroopan
- Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Arian Lasocki
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Radiology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Ebiko Y, Tamura K, Hara S, Inaji M, Tanaka Y, Nariai T, Ishii K, Maehara T. T2-FLAIR mismatch sign correlates with 11C-methionine uptake in lower-grade diffuse gliomas. J Neurooncol 2023; 164:257-265. [PMID: 37589920 DOI: 10.1007/s11060-023-04417-0] [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: 07/04/2023] [Accepted: 08/07/2023] [Indexed: 08/18/2023]
Abstract
PURPOSE The T2-FLAIR mismatch sign is recognized as an imaging finding highly suggestive of IDH-mutant astrocytomas. This study was designed to determine whether the T2-FLAIR mismatch sign correlates with uptake of 11C-methionine in lower-grade gliomas. METHODS We included 78 histopathologically verified lower-grade gliomas (grade 2: 31 cases, grade 3: 47 cases) in this study. 78 patients underwent 11C-methionine positron emission tomography (MET-PET) scans and magnetic resonance (MR) imaging scans prior to histological diagnosis. The tumor-to-normal ratio (T/N) of 11C-methionine uptake was calculated by dividing the maximum standardized uptake value (SUV) for the tumor by the mean SUV of the normal brain. MR imaging scans were evaluated for the presence of the T2-FLAIR mismatch sign by three independent reviewers. We compared molecular status, the T2-FLAIR mismatch sign and 11C-methionine uptake among patients with different lower-grade glioma molecular types. RESULTS The 78 lower-grade gliomas were assigned to one of three molecular groups: Group A (IDH-mutant and 1p/19q non-codeleted, n = 22), Group O (IDH-mutant and 1p/19q codeleted, n = 20), and Group W (IDH wildtype, n = 36). T2-FLAIR mismatch was found in 16 cases (20.5%) that were comprised of 8 (36.4%), 0 (0%), 8 (22.2%) cases in the molecular group A, O and W, respectively. The median T/N ratio of MET-PET in tumors with T2-FLAIR mismatch was 1.50, which was significantly lower than that of tumors without T2-FLAIR mismatch (1.83, p < 0.001, Mann-Whitney U test). In the Groups A and W (excluding Group O), the median T/N ratio on MET-PET in groups A and W (but not group O) with T2-FLAIR mismatch was 1.50, which was significantly lower than that of tumors without T2-FLAIR mismatch (1.81, p = 0.002, Mann-Whitney U test). CONCLUSION The T2-FLAIR mismatch sign correlated with lower 11C-methionine uptake in lower grade gliomas.
Collapse
Affiliation(s)
- Yusuke Ebiko
- Department of Neurosurgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo- ku, Tokyo, 113-8519, Japan
| | - Kaoru Tamura
- Department of Neurosurgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo- ku, Tokyo, 113-8519, Japan.
| | - Shoko Hara
- Department of Neurosurgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo- ku, Tokyo, 113-8519, Japan
- Research Team of Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Motoki Inaji
- Department of Neurosurgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo- ku, Tokyo, 113-8519, Japan
- Research Team of Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yoji Tanaka
- Department of Neurosurgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo- ku, Tokyo, 113-8519, Japan
| | - Tadashi Nariai
- Department of Neurosurgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo- ku, Tokyo, 113-8519, Japan
- Research Team of Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Kenji Ishii
- Research Team of Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Taketoshi Maehara
- Department of Neurosurgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo- ku, Tokyo, 113-8519, Japan
| |
Collapse
|
19
|
Lasocki A, Buckland ME, Molinaro T, Xie J, Whittle JR, Wei H, Gaillard F. Correlating MRI features with additional genetic markers and patient survival in histological grade 2-3 IDH-mutant astrocytomas. Neuroradiology 2023; 65:1215-1223. [PMID: 37316586 PMCID: PMC10338396 DOI: 10.1007/s00234-023-03175-0] [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: 03/31/2023] [Accepted: 06/04/2023] [Indexed: 06/16/2023]
Abstract
PURPOSE The increasing importance of molecular markers for classification and prognostication of diffuse gliomas has prompted the use of imaging features to predict genotype ("radiogenomics"). CDKN2A/B homozygous deletion has only recently been added to the diagnostic paradigm for IDH[isocitrate dehydrogenase]-mutant astrocytomas; thus, associated radiogenomic literature is sparse. There is also little data on whether different IDH mutations are associated with different imaging appearances. Furthermore, given that molecular status is now generally obtained routinely, the additional prognostic value of radiogenomic features is less clear. This study correlated MRI features with CDKN2A/B status, IDH mutation type and survival in histological grade 2-3 IDH-mutant brain astrocytomas. METHODS Fifty-eight grade 2-3 IDH-mutant astrocytomas were identified, 50 with CDKN2A/B results. IDH mutations were stratified into IDH1-R132H and non-canonical mutations. Background and survival data were obtained. Two neuroradiologists independently assessed the following MRI features: T2-FLAIR mismatch (<25%, 25-50%, >50%), well-defined tumour margins, contrast-enhancement (absent, wispy, solid) and central necrosis. RESULTS 8/50 tumours with CDKN2A/B results demonstrated homozygous deletion; slightly shorter survival was not significant (p=0.571). IDH1-R132H mutations were present in 50/58 (86%). No MRI features correlated with CDKN2A/B status or IDH mutation type. T2-FLAIR mismatch did not predict survival (p=0.977), but well-defined margins predicted longer survival (HR 0.36, p=0.008), while solid enhancement predicted shorter survival (HR 3.86, p=0.004). Both correlations remained significant on multivariate analysis. CONCLUSION MRI features did not predict CDKN2A/B homozygous deletion, but provided additional positive and negative prognostic information which correlated more strongly with prognosis than CDKN2A/B status in our cohort.
Collapse
Affiliation(s)
- Arian Lasocki
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Grattan St, Melbourne, Melbourne, Victoria, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
- Department of Radiology, The University of Melbourne, Parkville, Victoria, Australia.
| | - Michael E Buckland
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Tahlia Molinaro
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jing Xie
- Centre for Biostatistics and Clinical Trials, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - James R Whittle
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Personalised Oncology Division, Walter and Eliza Hall Institute, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Heng Wei
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Frank Gaillard
- Department of Radiology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Radiology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
20
|
Picca A, Bruno F, Nichelli L, Sanson M, Rudà R. Advances in molecular and imaging biomarkers in lower-grade gliomas. Expert Rev Neurother 2023; 23:1217-1231. [PMID: 37982735 DOI: 10.1080/14737175.2023.2285472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
INTRODUCTION Lower-grade (grade 2-3) gliomas (LGGs) constitutes a group of primary brain tumors with variable clinical behaviors and treatment responses. Recent advancements in molecular biology have redefined their classification, and novel imaging modalities emerged for the noninvasive diagnosis and follow-up. AREAS COVERED This review comprehensively analyses the current knowledge on molecular and imaging biomarkers in LGGs. Key molecular alterations, such as IDH mutations and 1p/19q codeletion, are discussed for their prognostic and predictive implications in guiding treatment decisions. Moreover, the authors explore theranostic biomarkers for the potential of tailored therapies. Additionally, they also describe the utility of advanced imaging modalities, including widely available techniques, as dynamic susceptibility contrast perfusion-weighted imaging and less validated, emerging approaches, for the noninvasive LGGs characterization and follow-up. EXPERT OPINION The integration of molecular markers enhanced the stratification of LGGs, leading to the new concept of integrated histomolecular classification. While the IDH mutation is an established key prognostic and predictive marker, recent results from IDH inhibitors trials showed its potential value as a theranostic marker. In this setting, advanced MRI techniques such as 2-D-hydroxyglutarate spectroscopy are very promising for the noninvasive diagnosis and monitoring of LGGs. This progress offers exciting prospects for personalized medicine and improved treatment outcomes in LGGs.
Collapse
Affiliation(s)
- Alberto Picca
- Service de Neurologie 2 Mazarin, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
- Sorbonne Université, Inserm, CNRS, UMRS1127, Institut du Cerveau-Paris Brain Institute-ICM, AP-HP, Paris, France
| | - Francesco Bruno
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science University Hospital, Turin, Italy
| | - Lucia Nichelli
- Service de Neuroradiologie, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
| | - Marc Sanson
- Service de Neurologie 2 Mazarin, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
- Sorbonne Université, Inserm, CNRS, UMRS1127, Institut du Cerveau-Paris Brain Institute-ICM, AP-HP, Paris, France
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science University Hospital, Turin, Italy
| |
Collapse
|
21
|
Kihira S, Derakhshani A, Leung M, Mahmoudi K, Bauer A, Zhang H, Polson J, Arnold C, Tsankova NM, Hormigo A, Salehi B, Pham N, Ellingson BM, Cloughesy TF, Nael K. Multi-Parametric Radiomic Model to Predict 1p/19q Co-Deletion in Patients with IDH-1 Mutant Glioma: Added Value to the T2-FLAIR Mismatch Sign. Cancers (Basel) 2023; 15:cancers15041037. [PMID: 36831380 PMCID: PMC9954034 DOI: 10.3390/cancers15041037] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/28/2023] [Accepted: 01/28/2023] [Indexed: 02/10/2023] Open
Abstract
PURPOSE The T2-FLAIR mismatch sign has shown promise in determining IDH mutant 1p/19q non-co-deleted gliomas with a high specificity and modest sensitivity. To develop a multi-parametric radiomic model using MRI to predict 1p/19q co-deletion status in patients with newly diagnosed IDH1 mutant glioma and to perform a comparative analysis to T2-FLAIR mismatch sign+. METHODS In this retrospective study, patients with diagnosis of IDH1 mutant gliomas with known 1p/19q status who had preoperative MRI were included. T2-FLAIR mismatch was evaluated independently by two board-certified neuroradiologists. Texture features were extracted from glioma segmentation of FLAIR images. eXtremeGradient Boosting (XGboost) classifiers were used for model development. Leave-one-out-cross-validation (LOOCV) and external validation performances were reported for both the training and external validation sets. RESULTS A total of 103 patients were included for model development and 18 patients for external testing validation. The diagnostic performance (sensitivity/specificity/accuracy) in the determination of the 1p/19q co-deletion status was 59%/83%/67% (training) and 62.5%/70.0%/66.3% (testing) for the T2-FLAIR mismatch sign. This was significantly improved (p = 0.04) using the radiomics model to 77.9%/82.8%/80.3% (training) and 87.5%/89.9%/88.8% (testing), respectively. The addition of radiomics as a computer-assisted tool resulted in significant (p = 0.02) improvement in the performance of the neuroradiologist with 13 additional corrected cases in comparison to just using the T2-FLAIR mismatch sign. CONCLUSION The proposed radiomic model provides much needed sensitivity to the highly specific T2-FLAIR mismatch sign in the determination of the 1p/19q non-co-deletion status and improves the overall diagnostic performance of neuroradiologists when used as an assistive tool.
Collapse
Affiliation(s)
- Shingo Kihira
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Ahrya Derakhshani
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Michael Leung
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Keon Mahmoudi
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Adam Bauer
- Department of Radiology, Kaiser Permanente Fontana Medical Center, Fontana, CA 92335, USA
| | - Haoyue Zhang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jennifer Polson
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Corey Arnold
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Nadejda M. Tsankova
- Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adilia Hormigo
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Banafsheh Salehi
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Nancy Pham
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Benjamin M. Ellingson
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
- UCLA Brain Tumor Imaging Laboratory, Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Timothy F. Cloughesy
- UCLA Brain Tumor Imaging Laboratory, Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Kambiz Nael
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
- Correspondence: ; Tel.: +1-310-267-5932
| |
Collapse
|
22
|
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: 3.0] [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
|
23
|
Fukuya Y, Tamura M, Nitta M, Saito T, Tsuzuki S, Koriyama S, Kuwano A, Kawamata T, Muragaki Y. Tumor volume and calcifications as indicators for preoperative differentiation of grade II/III diffuse gliomas. J Neurooncol 2023; 161:555-562. [PMID: 36749444 DOI: 10.1007/s11060-023-04244-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/18/2023] [Indexed: 02/08/2023]
Abstract
PURPOSE To retrospectively evaluate preoperative clinical factors for their ability to preoperatively differentiate malignancy grades in patients with incipient supratentorial grade II/III diffuse gliomas. METHODS This retrospective study included 206 adult patients with incipient supratentorial grade II/III diffuse gliomas according to the 2016 World Health Organization classification of tumors of the central nervous system. The cohort included 136 men and 70 women, with a median age of 41 years. Preoperative factors included age, sex, presence of calcifications on computed tomography scans, and preoperative tumor volume measured using preoperative magnetic resonance imaging. RESULTS In patients with oligodendrogliomas (IDH-mutant and 1p/19q-codeleted), calcifications were significantly more frequent (p = 0.0034) and tumor volume was significantly larger (p < 0.001) in patients with grade III tumors than in those with grade II tumors. Moreover, in patients with IDH-mutant astrocytomas, preoperative tumor volume was significantly larger (p = 0.0042) in patients with grade III tumors than in those with grade II tumors. In contrast, none of the evaluated preoperative clinical factors were significantly different between the patients with grade II and III IDH-wildtype astrocytomas. CONCLUSION In adult patients with suspicison incipient supratentorial grade II/III diffuse gliomas, presence of calcifications and larger preoperative tumor volume might be used as preoperative indices to differentiate between malignancy grades II and III in oligodendrogliomas (IDH-mutant and 1p/19q-codeleted) and larger preoperative tumor volume might have similar utility in IDH-mutant astrocytomas.
Collapse
Affiliation(s)
- Yasukazu Fukuya
- Department of Radiology, Kobe Comprehensive Medical College, 7-1-21 Tomugaoka, Suma-ku, Kobe-shi, Hyogo 654-0142, Japan
| | - Manabu Tamura
- Faculty of Advanced Techno‑Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan. .,Department of Neurosurgery, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan.
| | - Masayuki Nitta
- Faculty of Advanced Techno‑Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan.,Department of Neurosurgery, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan
| | - Taiichi Saito
- Faculty of Advanced Techno‑Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan.,Department of Neurosurgery, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan
| | - Shunsuke Tsuzuki
- Faculty of Advanced Techno‑Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan.,Department of Neurosurgery, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan
| | - Shunichi Koriyama
- Department of Neurosurgery, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan
| | - Atsushi Kuwano
- Department of Neurosurgery, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan
| | - Takakazu Kawamata
- Department of Neurosurgery, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan
| | - Yoshihiro Muragaki
- Faculty of Advanced Techno‑Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan.,Department of Neurosurgery, Tokyo Women's Medical University, 8‑1 Kawada‑cho, Shinjuku‑ku, Tokyo 162‑8666, Japan
| |
Collapse
|
24
|
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
|
25
|
Lasocki A, Abdalla G, Chow G, Thust SC. Imaging features associated with H3 K27-altered and H3 G34-mutant gliomas: a narrative systematic review. Cancer Imaging 2022; 22:63. [DOI: 10.1186/s40644-022-00500-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/23/2022] [Indexed: 11/18/2022] Open
Abstract
Abstract
Background
Advances in molecular diagnostics accomplished the discovery of two malignant glioma entities harboring alterations in the H3 histone: diffuse midline glioma, H3 K27-altered and diffuse hemispheric glioma, H3 G34-mutant. Radiogenomics research, which aims to correlate tumor imaging features with genotypes, has not comprehensively examined histone-altered gliomas (HAG). The aim of this research was to synthesize the current published data on imaging features associated with HAG.
Methods
A systematic search was performed in March 2022 using PubMed and the Cochrane Library, identifying studies on the imaging features associated with H3 K27-altered and/or H3 G34-mutant gliomas.
Results
Forty-seven studies fulfilled the inclusion criteria, the majority on H3 K27-altered gliomas. Just under half (21/47) were case reports or short series, the remainder being diagnostic accuracy studies. Despite heterogeneous methodology, some themes emerged. In particular, enhancement of H3 K27M-altered gliomas is variable and can be less than expected given their highly malignant behavior. Low apparent diffusion coefficient values have been suggested as a biomarker of H3 K27-alteration, but high values do not exclude this genotype. Promising correlations between high relative cerebral blood volume values and H3 K27-alteration require further validation. Limited data on H3 G34-mutant gliomas suggest some morphologic overlap with 1p/19q-codeleted oligodendrogliomas.
Conclusions
The existing data are limited, especially for H3 G34-mutant gliomas and artificial intelligence techniques. Current evidence indicates that imaging-based predictions of HAG are insufficient to replace histological assessment. In particular, H3 K27-altered gliomas should be considered when occurring in typical midline locations irrespective of enhancement characteristics.
Collapse
|
26
|
Radiomics features based on MRI predict BRAF V600E mutation in pediatric low-grade gliomas: A non-invasive method for molecular diagnosis. Clin Neurol Neurosurg 2022; 222:107478. [DOI: 10.1016/j.clineuro.2022.107478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/31/2022] [Accepted: 09/27/2022] [Indexed: 11/23/2022]
|
27
|
Valentino WL, Okada D, Bhanu S. A curious case of T2-FLAIR mismatch in H3K27M mutant glioma. Radiol Case Rep 2022; 17:2930-2935. [PMID: 35755103 PMCID: PMC9218295 DOI: 10.1016/j.radcr.2022.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/04/2022] [Accepted: 05/11/2022] [Indexed: 11/19/2022] Open
Abstract
Diffuse midline gliomas are a rare relatively new classification of primary central nervous system tumors which include astrocytomas, oligodendrogliomas, and glioblastomas. The T2-FLAIR mismatch sign is regarded as a highly specific imaging feature of IDH-mutant, 1p/19q non-codeleted astrocytomas. The case presented herein demonstrates this sign, however, in a non-IDH mutated diffuse midline glioma with a H3K27M mutation, a World Health Organization Grade IV neoplasm. Although preoperative diagnosis can provide important treatment and prognostic information, it is often quite difficult particularly in primary central nervous system tumors.
Collapse
Affiliation(s)
- William L. Valentino
- Riverside Community Hospital, 4445 Magnolia Avenue, Riverside, CA, 92501 USA
- HCA Healthcare, Nashville, TN, USA
- Corresponding author.
| | - Darren Okada
- Riverside Community Hospital, 4445 Magnolia Avenue, Riverside, CA, 92501 USA
- HCA Healthcare, Nashville, TN, USA
| | - Shiv Bhanu
- Riverside Community Hospital, 4445 Magnolia Avenue, Riverside, CA, 92501 USA
- HCA Healthcare, Nashville, TN, USA
| |
Collapse
|
28
|
Johnson DR, Giannini C, Vaubel RA, Morris JM, Eckel LJ, Kaufmann TJ, Guerin JB. A Radiologist's Guide to the 2021 WHO Central Nervous System Tumor Classification: Part I-Key Concepts and the Spectrum of Diffuse Gliomas. Radiology 2022; 304:494-508. [PMID: 35880978 DOI: 10.1148/radiol.213063] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The fifth edition of the World Health Organization (WHO) classification of tumors of the central nervous system, published in 2021, contains substantial updates in the classification of tumor types. Many of these changes are relevant to radiologists, including "big picture" changes to tumor diagnosis methods, nomenclature, and grading, which apply broadly to many or all central nervous system tumor types, as well as the addition, elimination, and renaming of multiple specific tumor types. Radiologists are integral in interpreting brain tumor imaging studies and have a considerable impact on patient care. Thus, radiologists must be aware of pertinent changes in the field. Staying updated with the most current guidelines allows radiologists to be informed and effective at multidisciplinary tumor boards and in interactions with colleagues in neuro-oncology, neurosurgery, radiation oncology, and neuropathology. This review represents the first of a two-installment review series on the most recent changes to the WHO brain tumor classification system. This first installment focuses on the changes to the classification of adult and pediatric gliomas of greatest relevance for radiologists.
Collapse
Affiliation(s)
- Derek R Johnson
- From the Departments of Radiology (D.R.J., J.M.M., L.J.E., T.J.K., J.B.G.), Neurology (D.R.J.), and Laboratory Medicine and Pathology (C.G., R.A.V.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy (C.G.)
| | - Caterina Giannini
- From the Departments of Radiology (D.R.J., J.M.M., L.J.E., T.J.K., J.B.G.), Neurology (D.R.J.), and Laboratory Medicine and Pathology (C.G., R.A.V.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy (C.G.)
| | - Rachael A Vaubel
- From the Departments of Radiology (D.R.J., J.M.M., L.J.E., T.J.K., J.B.G.), Neurology (D.R.J.), and Laboratory Medicine and Pathology (C.G., R.A.V.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy (C.G.)
| | - Jonathan M Morris
- From the Departments of Radiology (D.R.J., J.M.M., L.J.E., T.J.K., J.B.G.), Neurology (D.R.J.), and Laboratory Medicine and Pathology (C.G., R.A.V.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy (C.G.)
| | - Laurence J Eckel
- From the Departments of Radiology (D.R.J., J.M.M., L.J.E., T.J.K., J.B.G.), Neurology (D.R.J.), and Laboratory Medicine and Pathology (C.G., R.A.V.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy (C.G.)
| | - Timothy J Kaufmann
- From the Departments of Radiology (D.R.J., J.M.M., L.J.E., T.J.K., J.B.G.), Neurology (D.R.J.), and Laboratory Medicine and Pathology (C.G., R.A.V.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy (C.G.)
| | - Julie B Guerin
- From the Departments of Radiology (D.R.J., J.M.M., L.J.E., T.J.K., J.B.G.), Neurology (D.R.J.), and Laboratory Medicine and Pathology (C.G., R.A.V.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy (C.G.)
| |
Collapse
|
29
|
Xu J, Meng Y, Qiu K, Topatana W, Li S, Wei C, Chen T, Chen M, Ding Z, Niu G. Applications of Artificial Intelligence Based on Medical Imaging in Glioma: Current State and Future Challenges. Front Oncol 2022; 12:892056. [PMID: 35965542 PMCID: PMC9363668 DOI: 10.3389/fonc.2022.892056] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/22/2022] [Indexed: 12/24/2022] Open
Abstract
Glioma is one of the most fatal primary brain tumors, and it is well-known for its difficulty in diagnosis and management. Medical imaging techniques such as magnetic resonance imaging (MRI), positron emission tomography (PET), and spectral imaging can efficiently aid physicians in diagnosing, treating, and evaluating patients with gliomas. With the increasing clinical records and digital images, the application of artificial intelligence (AI) based on medical imaging has reduced the burden on physicians treating gliomas even further. This review will classify AI technologies and procedures used in medical imaging analysis. Additionally, we will discuss the applications of AI in glioma, including tumor segmentation and classification, prediction of genetic markers, and prediction of treatment response and prognosis, using MRI, PET, and spectral imaging. Despite the benefits of AI in clinical applications, several issues such as data management, incomprehension, safety, clinical efficacy evaluation, and ethical or legal considerations, remain to be solved. In the future, doctors and researchers should collaborate to solve these issues, with a particular emphasis on interdisciplinary teamwork.
Collapse
Affiliation(s)
- Jiaona Xu
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuting Meng
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kefan Qiu
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Win Topatana
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shijie Li
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Wei
- Department of Neurology, Affiliated Ningbo First Hospital, Ningbo, China
| | - Tianwen Chen
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingyu Chen
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| | - Guozhong Niu
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| |
Collapse
|
30
|
Vagvala S, Guenette JP, Jaimes C, Huang RY. Imaging diagnosis and treatment selection for brain tumors in the era of molecular therapeutics. Cancer Imaging 2022; 22:19. [PMID: 35436952 PMCID: PMC9014574 DOI: 10.1186/s40644-022-00455-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/29/2022] [Indexed: 01/12/2023] Open
Abstract
Currently, most CNS tumors require tissue sampling to discern their molecular/genomic landscape. However, growing research has shown the powerful role imaging can play in non-invasively and accurately detecting the molecular signature of these tumors. The overarching theme of this review article is to provide neuroradiologists and neurooncologists with a framework of several important molecular markers, their associated imaging features and the accuracy of those features. A particular emphasis is placed on those tumors and mutations that have specific or promising imaging correlates as well as their respective therapeutic potentials.
Collapse
Affiliation(s)
- Saivenkat Vagvala
- Division of Neuroradiology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, 75 Francis St, Boston, MA, 02115, USA
| | - Jeffrey P Guenette
- Division of Neuroradiology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, 75 Francis St, Boston, MA, 02115, USA
| | - Camilo Jaimes
- Division of Neuroradiology, Boston Children's, 300 Longwood Ave., 2nd floor, Main Building, Boston, MA, 02115, USA
| | - Raymond Y Huang
- Division of Neuroradiology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, 75 Francis St, Boston, MA, 02115, USA.
| |
Collapse
|
31
|
Radiogenomic association between the T2-FLAIR mismatch sign and IDH mutation status in adult patients with lower-grade gliomas: an updated systematic review and meta-analysis. Eur Radiol 2022; 32:5339-5352. [PMID: 35169897 DOI: 10.1007/s00330-022-08607-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/24/2021] [Accepted: 01/22/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES To reveal a radiogenomic correlation between the presence of the T2-fluid-attenuated inversion recovery resection (T2-FLAIR) mismatch sign on MR images and isocitrate dehydrogenase (IDH) mutation status in adult patients with lower-grade gliomas (LGGs). METHODS A web-based systemic search for eligible literature up to April 13, 2021, was conducted on PubMed, Embase, and the Cochrane Library databases by two independent reviewers. This study was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. We included studies evaluating the accuracy of the T2-FLAIR mismatch sign in diagnosing the IDH mutation in adult patients with LGGs. The T2-FLAIR mismatch sign was defined as a T2-hyperintense lesion that is hypointense on FLAIR except for a hyperintense rim. RESULTS Fourteen studies (n = 1986) were finally identified. The mean age of patients in the included studies ranged from 38.5 to 56 years. The pooled area under the curve (AUC), sensitivity, and specificity were obtained for each molecular profile: IDHmut-Codel: 0.46 (95% confidence interval [CI]: 0.42-0.50), 1% (95%CI: 0-7%), and 69% (95%CI: 62-75%), respectively; IDHmut-Noncodel: 0.75 (95%CI: 0.71-0.79), 42% (95%CI: 34-50%), and 99% (95%CI: 96-100%), respectively; IDH-Mutation regardless of 1p/19q codeletion status: 0.77 (95%CI: 0.73-0.80), 29% (95%CI: 21-40%), and 99% (95%CI: 92-100%), respectively. CONCLUSIONS The T2-FLAIR mismatch sign was an insensitive but highly specific marker for IDHmut-Noncodel and IDH-Mutation LGGs, whereas it was not a useful marker for IDHmut-Codel LGGs. The findings might identify the T2-FLAIR mismatch sign as a non-invasive imaging biomarker for the selection of patients with IDH-mutant LGGs. KEY POINTS • The T2-FLAIR mismatch sign was not a sensitive sign for IDH mutation in LGGs. • The T2-FLAIR mismatch sign was related to IDHmut-Noncodel with a specificity of 99%. • The pooled specificity (69%) of the T2-FLAIR mismatch sign for IDHmut-Codel was low.
Collapse
|
32
|
Aboud O, Shah R, Vera E, Burton E, Theeler B, Wu J, Boris L, Quezado M, Reyes J, Wall K, R Gilbert M, S Armstrong T, Penas-Prado M. Challenges of imaging interpretation to predict oligodendroglioma grade: a report from the Neuro-Oncology Branch. CNS Oncol 2022; 11:CNS83. [PMID: 35142534 PMCID: PMC8988255 DOI: 10.2217/cns-2021-0005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background: To illustrate challenges of imaging interpretation in patients with oligodendroglioma seen at a referral center and evaluate interrater reliability. Methods: Two neuro-oncologists reviewed diagnostic preradiation MRIs of oligodendroglioma patients; interrater reliability was calculated with the kappa coefficient (k). A neuroradiologist measured presurgical apparent diffusion coefficient (ADC), if available. Results: Extensive enhancement was noted in four of 58 patients, k = 0.7; necrosis in seven of 58, k = 0.61; calcification in seven of 17, k = 1.0; diffusion restriction in two of 39 patients, k = 1.0 (all only in grade 3). ADC values with receiver operator characteristic analysis for area under the curve were 0.473, not significantly different from the null hypothesis (p = 0.14). Conclusions: Extensive enhancement, necrosis and calcification correlated with grade 3 oligodendroglioma in our sample. However, interrater variability is an important limitation when assessing radiographic features, supporting the need for standardization of imaging protocols and their interpretation.
Collapse
Affiliation(s)
- Orwa Aboud
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Ritu Shah
- Department of Neuro radiology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Elizabeth Vera
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Eric Burton
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Brett Theeler
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,Walter Reed National Military Medical Center, Bethesda, MD 20814, USA
| | - Jing Wu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Lisa Boris
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Martha Quezado
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814 USA
| | - Jennifer Reyes
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Kathleen Wall
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Terri S Armstrong
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Marta Penas-Prado
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
33
|
Biswas A, Amirabadi A, Wagner M, Ertl-Wagner B. Features of Visually AcceSAble Rembrandt Images: Interrater Reliability in Pediatric Brain Tumors. AJNR Am J Neuroradiol 2022; 43:304-308. [PMID: 35058297 PMCID: PMC8985665 DOI: 10.3174/ajnr.a7399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/20/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND PURPOSE At present, no evidence-based lexicon exists for pediatric intracranial tumors. The Visually AcceSAble Rembrandt Images terminology describes reproducible MR imaging features of adult gliomas for prediction of tumor grade, molecular markers, and survival. Our aim was to assess the interrater reliability of the pre-resection features of Visually AcceSAble Rembrandt Images in pediatric brain tumors. MATERIALS AND METHODS Fifty consecutive pre-resection brain MR imaging examinations of pediatric intracranial neoplasms were independently reviewed by 3 neuroradiologists. The intraclass correlation coefficient for continuous variables and the Krippendorf alpha were used to evaluate the interrater agreement. Subgroup analysis was performed for 30 gliomas. RESULTS Parameters with almost perfect agreement (α > .8) included tumor location (F1) and proportion of enhancing tumor (F5). Parameters with substantial agreement (α = .61-.80) were side of tumor epicenter (F2), involvement of eloquent brain (F3), enhancement quality (F4), proportion of non-contrast-enhancing tumor (F6), and deep white matter invasion (F21). The other parameters showed either moderate (α = .41-.60; n = 11), fair (α = .21-.40; n = 5), or slight agreement (α = 0-.20; n = 1). Subgroup analysis of 30 gliomas showed almost perfect agreement for tumor location (F1), involvement of eloquent brain (F3), and proportion of enhancing tumor (F5); and substantial agreement for side of tumor epicenter (F2), enhancement quality (F4), proportion of noncontrast enhancing tumor (F6), cysts (F8), thickness of enhancing margin (F11), and deep white matter invasion (F21). The intraclass correlation coefficient for measurements in the axial plane was excellent in both the main group (0.984 [F29] and 0.982 [F30]) and the glioma subgroup (0.973 [F29] and 0.973 [F30]). CONCLUSIONS Nine features of Visually AcceSAble Rembrandt Images have an acceptable interrater agreement in pediatric brain tumors. For the subgroup of pediatric gliomas, 11 features of Visually AcceSAble Rembrandt Images have an acceptable interrater agreement. The low degree of reproducibility of the remainder of the features necessitates the use of features tailored to the pediatric age group and is likely related to the more heterogeneous imaging morphology of pediatric brain tumors.
Collapse
Affiliation(s)
- A. Biswas
- From the Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada,Department of Medical Imaging, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - A. Amirabadi
- From the Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada,Department of Medical Imaging, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - M.W. Wagner
- From the Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada,Department of Medical Imaging, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - B.B. Ertl-Wagner
- From the Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada,Department of Medical Imaging, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
34
|
Combining hyperintense FLAIR rim and radiological features in identifying IDH mutant 1p/19q non-codeleted lower-grade glioma. Eur Radiol 2022; 32:3869-3879. [DOI: 10.1007/s00330-021-08500-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023]
|
35
|
Mancini L, Casagranda S, Gautier G, Peter P, Lopez B, Thorne L, McEvoy A, Miserocchi A, Samandouras G, Kitchen N, Brandner S, De Vita E, Torrealdea F, Rega M, Schmitt B, Liebig P, Sanverdi E, Golay X, Bisdas S. CEST MRI provides amide/amine surrogate biomarkers for treatment-naïve glioma sub-typing. Eur J Nucl Med Mol Imaging 2022; 49:2377-2391. [PMID: 35029738 PMCID: PMC9165287 DOI: 10.1007/s00259-022-05676-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/31/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Accurate glioma classification affects patient management and is challenging on non- or low-enhancing gliomas. This study investigated the clinical value of different chemical exchange saturation transfer (CEST) metrics for glioma classification and assessed the diagnostic effect of the presence of abundant fluid in glioma subpopulations. METHODS Forty-five treatment-naïve glioma patients with known isocitrate dehydrogenase (IDH) mutation and 1p/19q codeletion status received CEST MRI (B1rms = 2μT, Tsat = 3.5 s) at 3 T. Magnetization transfer ratio asymmetry and CEST metrics (amides: offset range 3-4 ppm, amines: 1.5-2.5 ppm, amide/amine ratio) were calculated with two models: 'asymmetry-based' (AB) and 'fluid-suppressed' (FS). The presence of T2/FLAIR mismatch was noted. RESULTS IDH-wild type had higher amide/amine ratio than IDH-mutant_1p/19qcodel (p < 0.022). Amide/amine ratio and amine levels differentiated IDH-wild type from IDH-mutant (p < 0.0045) and from IDH-mutant_1p/19qret (p < 0.021). IDH-mutant_1p/19qret had higher amides and amines than IDH-mutant_1p/19qcodel (p < 0.035). IDH-mutant_1p/19qret with AB/FS mismatch had higher amines than IDH-mutant_1p/19qret without AB/FS mismatch ( < 0.016). In IDH-mutant_1p/19qret, the presence of AB/FS mismatch was closely related to the presence of T2/FLAIR mismatch (p = 0.014). CONCLUSIONS CEST-derived biomarkers for amides, amines, and their ratio can help with histomolecular staging in gliomas without intense contrast enhancement. T2/FLAIR mismatch is reflected in the presence of AB/FS CEST mismatch. The AB/FS CEST mismatch identifies glioma subgroups that may have prognostic and clinical relevance.
Collapse
Affiliation(s)
- Laura Mancini
- Box65, Lysholm Department of Neuroradiology, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, 8-11 Queen Square, London, WC1N 3BG, UK.
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK.
| | | | | | | | | | - Lewis Thorne
- Department of Neurosurgery, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Andrew McEvoy
- Department of Neurosurgery, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Anna Miserocchi
- Department of Neurosurgery, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - George Samandouras
- Department of Neurosurgery, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Neil Kitchen
- Department of Neurosurgery, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Sebastian Brandner
- Division of Neuropathology, UCL Queen Square Institute of Neurology, London, UK
- The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Enrico De Vita
- Box65, Lysholm Department of Neuroradiology, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, 8-11 Queen Square, London, WC1N 3BG, UK
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| | - Francisco Torrealdea
- University College Hospital, University College of London Hospitals NHS Foundation Trust, London, UK
| | - Marilena Rega
- University College Hospital, University College of London Hospitals NHS Foundation Trust, London, UK
| | | | | | - Eser Sanverdi
- Box65, Lysholm Department of Neuroradiology, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, 8-11 Queen Square, London, WC1N 3BG, UK
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| | - Xavier Golay
- Box65, Lysholm Department of Neuroradiology, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, 8-11 Queen Square, London, WC1N 3BG, UK
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| | - Sotirios Bisdas
- Box65, Lysholm Department of Neuroradiology, The National Hospital for Neurology & Neurosurgery, University College London Hospitals NHS Foundation Trust, 8-11 Queen Square, London, WC1N 3BG, UK
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
36
|
Mohammed S, Ravikumar V, Warner E, Patel S, Bakas S, Rao A, Jain R. Quantifying T2-FLAIR Mismatch Using Geographically Weighted Regression and Predicting Molecular Status in Lower-Grade Gliomas. AJNR Am J Neuroradiol 2022; 43:33-39. [PMID: 34764084 PMCID: PMC8757555 DOI: 10.3174/ajnr.a7341] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/03/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND PURPOSE The T2-FLAIR mismatch sign is a validated imaging sign of isocitrate dehydrogenase-mutant 1p/19q noncodeleted gliomas. It is identified by radiologists through visual inspection of preoperative MR imaging scans and has been shown to identify isocitrate dehydrogenase-mutant 1p/19q noncodeleted gliomas with a high positive predictive value. We have developed an approach to quantify the T2-FLAIR mismatch signature and use it to predict the molecular status of lower-grade gliomas. MATERIALS AND METHODS We used multiparametric MR imaging scans and segmentation labels of 108 preoperative lower-grade glioma tumors from The Cancer Imaging Archive. Clinical information and T2-FLAIR mismatch sign labels were obtained from supplementary material of relevant publications. We adopted an objective analytic approach to estimate this sign through a geographically weighted regression and used the residuals for each case to construct a probability density function (serving as a residual signature). These functions were then analyzed using an appropriate statistical framework. RESULTS We observed statistically significant (P value = .05) differences between the averages of residual signatures for an isocitrate dehydrogenase-mutant 1p/19q noncodeleted class of tumors versus other categories. Our classifier predicts these cases with area under the curve of 0.98 and high specificity and sensitivity. It also predicts the T2-FLAIR mismatch sign within these cases with an under the curve of 0.93. CONCLUSIONS On the basis of this retrospective study, we show that geographically weighted regression-based residual signatures are highly informative of the T2-FLAIR mismatch sign and can identify isocitrate dehydrogenase-mutation and 1p/19q codeletion status with high predictive power. The utility of the proposed quantification of the T2-FLAIR mismatch sign can be potentially validated through a prospective multi-institutional study.
Collapse
Affiliation(s)
- S. Mohammed
- From the Departments of Biostatistics (S.M., A.R.),Computational Medicine & Bioinformatics (S.M., V.R., E.W., A.R.)
| | - V. Ravikumar
- Computational Medicine & Bioinformatics (S.M., V.R., E.W., A.R.)
| | - E. Warner
- Computational Medicine & Bioinformatics (S.M., V.R., E.W., A.R.)
| | - S.H. Patel
- Department of Radiology & Medical Imaging (S.H.P.), University of Virginia School of Medicine, Charlottesville, Virginia
| | - S. Bakas
- Departments of Radiology (S.B.),Pathology & Laboratory Medicine (S.B.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - A. Rao
- From the Departments of Biostatistics (S.M., A.R.),Computational Medicine & Bioinformatics (S.M., V.R., E.W., A.R.),Radiation Oncology (A.R.),Michigan Institute for Data Sciences (A.R.),Department of Biomedical Engineering (A.R.), University of Michigan, Ann Arbor, Michigan
| | - R. Jain
- Departments of Radiology (R.J.),Neurosurgery (R.J.), New York University Grossman School of Medicine, New York, New York
| |
Collapse
|
37
|
Tyurina AN, Vikhrova NB, Batalov AI, Kalaeva DB, Shults EI, Postnov AA, Pronin IN. [Radiological biomarkers of brain gliomas]. ZHURNAL VOPROSY NEIROKHIRURGII IMENI N. N. BURDENKO 2022; 86:121-126. [PMID: 36534633 DOI: 10.17116/neiro202286061121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The most important objective of modern neuroimaging is comparison of data on genotype and phenotype of brain gliomas. Radiogenomics as a new branch of science is devoted to searching for such relationships based on MRI and PET/CT parameters. The 2021 WHO classification of tumors of the central nervous system poses the most important tasks for physicians in assessment of biological behavior of tumors and their response to combined treatment. The review demonstrates the possibilities and prospects of preoperative MRI and PET/CT with amino acids in assessing the genetic profile of brain gliomas.
Collapse
Affiliation(s)
- A N Tyurina
- Burdenko Neurosurgery Center, Moscow, Russia
| | | | - A I Batalov
- Burdenko Neurosurgery Center, Moscow, Russia
| | - D B Kalaeva
- Burdenko Neurosurgery Center, Moscow, Russia
- Moscow Engineering Physics Institute, Moscow, Russia
| | - E I Shults
- Research Practical Clinical Center of Diagnosis and Telemedicine Technologies, Moscow, Russia
| | - A A Postnov
- Burdenko Neurosurgery Center, Moscow, Russia
- Moscow Engineering Physics Institute, Moscow, Russia
- Lebedev Physical Institute, Moscow, Russia
| | - I N Pronin
- Burdenko Neurosurgery Center, Moscow, Russia
| |
Collapse
|
38
|
Do YA, Cho SJ, Choi BS, Baik SH, Bae YJ, Sunwoo L, Jung C, Kim JH. Predictive accuracy of T2-FLAIR mismatch sign for the IDH-mutant, 1p/19q noncodeleted low-grade glioma: An updated systematic review and meta-analysis. Neurooncol Adv 2022; 4:vdac010. [PMID: 35198981 PMCID: PMC8859831 DOI: 10.1093/noajnl/vdac010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The T2-fluid-attenuated inversion recovery (FLAIR) mismatch sign, has been considered a highly specific imaging biomarker of IDH-mutant, 1p/19q noncodeleted low-grade glioma. This systematic review and meta-analysis aimed to evaluate the diagnostic performance of T2-FLAIR mismatch sign for prediction of a patient with IDH-mutant, 1p/19q noncodeleted low-grade glioma, and identify the causes responsible for the heterogeneity across the included studies. METHODS A systematic literature search in the Ovid-MEDLINE and EMBASE databases was performed for studies reporting the relevant topic before November 17, 2020. The pooled sensitivity and specificity values with their 95% confidence intervals were calculated using bivariate random-effects modeling. Meta-regression analyses were also performed to determine factors influencing heterogeneity. RESULTS For all the 10 included cohorts from 8 studies, the pooled sensitivity was 40% (95% confidence interval [CI] 28-53%), and the pooled specificity was 100% (95% CI 95-100%). In the hierarchic summary receiver operating characteristic curve, the difference between the 95% confidence and prediction regions was relatively large, indicating heterogeneity among the studies. Higgins I2 statistics demonstrated considerable heterogeneity in sensitivity (I2 = 83.5%) and considerable heterogeneity in specificity (I2 = 95.83%). Among the potential covariates, it seemed that none of factors was significantly associated with study heterogeneity in the joint model. However, the specificity was increased in studies with all the factors based on the differences in the composition of the detailed tumors. CONCLUSIONS The T2-FLAIR mismatch sign is near-perfect specific marker of IDH mutation and 1p/19q noncodeletion.
Collapse
Affiliation(s)
- Yoon Ah Do
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Se Jin Cho
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Byung Se Choi
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Sung Hyun Baik
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Yun Jung Bae
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Leonard Sunwoo
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Cheolkyu Jung
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jae Hyoung Kim
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| |
Collapse
|
39
|
Pinto C, Noronha C, Taipa R, Ramos C. T2-FLAIR mismatch sign: a roadmap of pearls and pitfalls. Br J Radiol 2022; 95:20210825. [PMID: 34618597 PMCID: PMC8722227 DOI: 10.1259/bjr.20210825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
T2-FLAIR mismatch sign has been advocated to be 100% specific for IDH-mutant 1p/19q non-codeleted gliomas (diffuse astrocytomas). However, false positives have been reported in recent works. Loose application of the criteria may lead to erroneous classification, especially by non-trained neuroradiologists. In this pictorial essay, we aim to bring attention to the need for strict criteria for the application of T2-FLAIR mismatch sign and to discuss the potential pitfalls in the application of these criteria. For that, a series of adult brain tumour cases are presented to demonstrate how to apply this radiological sign in the clinical practice.
Collapse
Affiliation(s)
- Catarina Pinto
- Neuroradiology Department, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Carolina Noronha
- Neurosurgery Department, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Ricardo Taipa
- Neuropathology Unit, Department of Neurosciences, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Cristina Ramos
- Neuroradiology Department, Centro Hospitalar Universitário do Porto, Porto, Portugal
| |
Collapse
|
40
|
Li G, Huang R, Fan W, Wang D, Wu F, Zeng F, Yu M, Zhai Y, Chang Y, Pan C, Jiang T, Yan W, Wang H, Zhang W. Galectin-9/TIM-3 as a Key Regulator of Immune Response in Gliomas With Chromosome 1p/19q Codeletion. Front Immunol 2021; 12:800928. [PMID: 34956239 PMCID: PMC8692744 DOI: 10.3389/fimmu.2021.800928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 11/24/2021] [Indexed: 11/13/2022] Open
Abstract
Gliomas with chromosome 1p/19q codeletion were considered a specific tumor entity. This study was designed to reveal the biological function alterations tightly associated with 1p/19q codeletion in gliomas. Clinicopathological and RNA sequencing data from glioma patients were obtained from The Cancer Genome Atlas and Chinese Glioma Genome Atlas databases. Gene set variation analysis was performed to explore the differences in biological functions between glioma subgroups stratified by 1p/19q codeletion status. The abundance of immune cells in each sample was detected using the CIBERSORT analytical tool. Single-cell sequencing data from public databases were analyzed using the t-distributed stochastic neighbor embedding (t-SNE) algorithm, and the findings were verified by in vitro and in vivo experiments and patient samples.We found that the activation of immune and inflammatory responses was tightly associated with 1p/19q codeletion in gliomas. As the most important transcriptional regulator of Galectin-9 in gliomas, the expression level of CCAAT enhancer-binding protein alpha in samples with 1p/19q codeletion was significantly decreased, which led to the downregulation of the immune checkpoints Galectin-9 and TIM-3. These results were validated in three independent datasets. The t-SNE analysis showed that the loss of chromosome 19q was the main reason for the promotion of the antitumor immune response. IHC protein staining, in vitro and in vivo experiments verified the results of bioinformatics analysis. In gliomas, 1p/19q codeletion can promote the antitumor immune response by downregulating the expression levels of the immune checkpoint TIM-3 and its ligand Galectin-9.
Collapse
Affiliation(s)
- Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruoyu Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenhua Fan
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Di Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fan Wu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Mingchen Yu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - You Zhai
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yuanhao Chang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Changqing Pan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Wei Yan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongjun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| |
Collapse
|
41
|
Nuechterlein N, Shapiro LG, Holland EC, Cimino PJ. Machine learning modeling of genome-wide copy number alteration signatures reliably predicts IDH mutational status in adult diffuse glioma. Acta Neuropathol Commun 2021; 9:191. [PMID: 34863298 PMCID: PMC8645099 DOI: 10.1186/s40478-021-01295-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 11/20/2021] [Indexed: 11/17/2022] Open
Abstract
Knowledge of 1p/19q-codeletion and IDH1/2 mutational status is necessary to interpret any investigational study of diffuse gliomas in the modern era. While DNA sequencing is the gold standard for determining IDH mutational status, genome-wide methylation arrays and gene expression profiling have been used for surrogate mutational determination. Previous studies by our group suggest that 1p/19q-codeletion and IDH mutational status can be predicted by genome-wide somatic copy number alteration (SCNA) data alone, however a rigorous model to accomplish this task has yet to be established. In this study, we used SCNA data from 786 adult diffuse gliomas in The Cancer Genome Atlas (TCGA) to develop a two-stage classification system that identifies 1p/19q-codeleted oligodendrogliomas and predicts the IDH mutational status of astrocytic tumors using a machine-learning model. Cross-validated results on TCGA SCNA data showed near perfect classification results. Furthermore, our astrocytic IDH mutation model validated well on four additional datasets (AUC = 0.97, AUC = 0.99, AUC = 0.95, AUC = 0.96) as did our 1p/19q-codeleted oligodendroglioma screen on the two datasets that contained oligodendrogliomas (MCC = 0.97, MCC = 0.97). We then retrained our system using data from these validation sets and applied our system to a cohort of REMBRANDT study subjects for whom SCNA data, but not IDH mutational status, is available. Overall, using genome-wide SCNAs, we successfully developed a system to robustly predict 1p/19q-codeletion and IDH mutational status in diffuse gliomas. This system can assign molecular subtype labels to tumor samples of retrospective diffuse glioma cohorts that lack 1p/19q-codeletion and IDH mutational status, such as the REMBRANDT study, recasting these datasets as validation cohorts for diffuse glioma research.
Collapse
Affiliation(s)
- Nicholas Nuechterlein
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA
| | - Linda G Shapiro
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA
| | - Eric C Holland
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Patrick J Cimino
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, Division of Neuropathology, University of Washington, 325 9th Avenue, Box 359791, Seattle, WA, 98104, USA.
| |
Collapse
|
42
|
Yang X, Lin Y, Xing Z, She D, Su Y, Cao D. Predicting 1p/19q codeletion status using diffusion-, susceptibility-, perfusion-weighted, and conventional MRI in IDH-mutant lower-grade gliomas. Acta Radiol 2021; 62:1657-1665. [PMID: 33222488 DOI: 10.1177/0284185120973624] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Isocitrate dehydrogenase (IDH)-mutant lower-grade gliomas (LGGs) are further classified into two classes: with and without 1p/19q codeletion. IDH-mutant and 1p/19q codeleted LGGs have better prognosis compared with IDH-mutant and 1p/19q non-codeleted LGGs. PURPOSE To evaluate conventional magnetic resonance imaging (cMRI), diffusion-weighted imaging (DWI), susceptibility-weighted imaging (SWI), and dynamic susceptibility contrast perfusion-weighted imaging (DSC-PWI) for predicting 1p/19q codeletion status of IDH-mutant LGGs. MATERIAL AND METHODS We retrospectively reviewed cMRI, DWI, SWI, and DSC-PWI in 142 cases of IDH mutant LGGs with known 1p/19q codeletion status. Features of cMRI, relative ADC (rADC), intratumoral susceptibility signals (ITSSs), and the value of relative cerebral blood volume (rCBV) were compared between IDH-mutant LGGs with and without 1p/19q codeletion. Receiver operating characteristic curve and logistic regression were used to determine diagnostic performances. RESULTS IDH-mutant and 1p/19q non-codeleted LGGs tended to present with the T2/FLAIR mismatch sign and distinct borders (P < 0.001 and P = 0.038, respectively). Parameters of rADC, ITSSs, and rCBVmax were significantly different between the 1p/19q codeleted and 1p/19q non-codeleted groups (P < 0.001, P = 0.017, and P < 0.001, respectively). A combination of cMRI, SWI, DWI, and DSC-PWI for predicting 1p/19q codeletion status in IDH-mutant LGGs resulted in a sensitivity, specificity, positive predictive value, negative predictive value, and an AUC of 80.36%, 78.57%, 83.30%, 75.00%, and 0.88, respectively. CONCLUSION 1p/19q codeletion status of IDH-mutant LGGs can be stratified using cMRI and advanced MRI techniques, including DWI, SWI, and DSC-PWI. A combination of cMRI, rADC, ITSSs, and rCBVmax may improve the diagnostic performance for predicting 1p/19q codeletion status.
Collapse
Affiliation(s)
- Xiefeng Yang
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| | - Yu Lin
- Department of Radiology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, PR China
| | - Zhen Xing
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| | - Dejun She
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| | - Yan Su
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| | - Dairong Cao
- Department of Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| |
Collapse
|
43
|
Abstract
This article reviews recent advances in the use of standard and advanced imaging techniques for diagnosis and treatment of central nervous system (CNS) tumors, including glioma and brain metastasis. Following the recent transition from a histology-based approach in classifying CNS tumors to one that integrates histology with the molecular information of tumor, the approaches for imaging CNS tumors have also been adapted to this new framework. Some challenges related to the diagnosis and treatment of CNS tumors, such as differentiating tumor from treatment-related imaging changes, require further progress to implement advanced imaging for clinical use.
Collapse
Affiliation(s)
- Raymond Y Huang
- Department of Neuroradiology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Whitney B Pope
- Radiology, Section of Neuroradiology, Brain Tumor Imaging, UCLA Medical Center, Los Angeles, CA, USA; Department of Radiological Sciences, David Geffen School of Medicine, University of California-Los Angeles, 924 Westwood Boulevard, Suite 615, Los Angeles, CA 90024, USA; Department of Neurology, David Geffen School of Medicine, University of California-Los Angeles, 924 Westwood Boulevard, Suite 615, Los Angeles, CA 90024, USA
| |
Collapse
|
44
|
Adamou A, Beltsios ET, Papanagiotou P. The T2-FLAIR Mismatch Sign as an Imaging Indicator of IDH-Mutant, 1p/19q Non-Codeleted Lower Grade Gliomas: A Systematic Review and Diagnostic Accuracy Meta-Analysis. Diagnostics (Basel) 2021; 11:diagnostics11091620. [PMID: 34573962 PMCID: PMC8471804 DOI: 10.3390/diagnostics11091620] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 02/01/2023] Open
Abstract
The study's objective was the evaluation of the diagnostic accuracy of the T2-FLAIR mismatch sign in terms of diagnosing IDH-mutant non-codeleted (IDHmut-Noncodel) lower grade gliomas (LGG) of the brain. We searched the MEDLINE, Scopus and Cochrane Central databases. The last database search was performed on 12 April 2021. Studies that met the following were included: MRI scan assessing the presence of T2-FLAIR mismatch sign, and available IDH mutation and 1p/19q codeletion status. The quality of studies was assessed using the QUADAS-2 tool. Twelve studies involving 14 cohorts were included in the quantitative analysis. The diagnostic odds ratio [DOR (95% confidence interval; CI)] was estimated at 34.42 (20.95, 56.56), Pz < 0.01. Pooled sensitivity and specificity (95% CI) were estimated at 40% (31-50%; Pz = 0.05) and 97% (93-99%; Pz < 0.01), respectively. The likelihood ratio (LR; 95% CI) for a positive test was 11.39 (6.10, 21.29; Pz < 0.01) and the LR (95% CI) for a negative test was 0.40 (0.24, 0.65; Pz < 0.01).The T2-FLAIR mismatch sign is a highly specific biomarker for the diagnosis of IDHmut-Noncodel LGGs. However, the test was found positive in some other tumors and had a high number of false negative results. The diagnostic accuracy of the mismatch sign might be improved when combined with further imaging parameters.
Collapse
Affiliation(s)
- Antonis Adamou
- Department of Radiology and Medical Imaging, University of Thessaly, 41110 Larissa, Greece;
| | - Eleftherios T. Beltsios
- Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece;
| | - Panagiotis Papanagiotou
- Department of Diagnostic and Interventional Neuroradiology, Hospital Bremen-Mitte/Bremen-Ost, 28205 Bremen, Germany
- First Department of Radiology, School of Medicine, National & Kapodistrian University of Athens, Areteion Hospital, 11528 Athens, Greece
- Correspondence:
| |
Collapse
|
45
|
Assessing Versatile Machine Learning Models for Glioma Radiogenomic Studies across Hospitals. Cancers (Basel) 2021; 13:cancers13143611. [PMID: 34298824 PMCID: PMC8306149 DOI: 10.3390/cancers13143611] [Citation(s) in RCA: 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: 03/28/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Radiogenomics enables prediction of the status and prognosis of patients using non-invasively obtained imaging data. Current machine learning (ML) methods used in radiogenomics require huge datasets, which involve the handling of large heterogeneous datasets from multiple cohorts/hospitals. In this study, two different glioma datasets were used to test various ML and image pre-processing methods to confirm whether the models trained on one dataset are universally applicable to other datasets. Our result suggested that the ML method that yielded the highest accuracy in a single dataset was likely to be overfitted. We demonstrated that implementation of standardization and dimension reduction procedures prior to classification, enabled the development of ML methods that are less affected by the multiple cohort difference. We advocate using caution in interpreting the results of radiogenomic studies of the training and testing datasets that are small or mixed, with a view to implementing practical ML methods in radiogenomics. Abstract Radiogenomics use non-invasively obtained imaging data, such as magnetic resonance imaging (MRI), to predict critical biomarkers of patients. Developing an accurate machine learning (ML) technique for MRI requires data from hundreds of patients, which cannot be gathered from any single local hospital. Hence, a model universally applicable to multiple cohorts/hospitals is required. We applied various ML and image pre-processing procedures on a glioma dataset from The Cancer Image Archive (TCIA, n = 159). The models that showed a high level of accuracy in predicting glioblastoma or WHO Grade II and III glioma using the TCIA dataset, were then tested for the data from the National Cancer Center Hospital, Japan (NCC, n = 166) whether they could maintain similar levels of high accuracy. Results: we confirmed that our ML procedure achieved a level of accuracy (AUROC = 0.904) comparable to that shown previously by the deep-learning methods using TCIA. However, when we directly applied the model to the NCC dataset, its AUROC dropped to 0.383. Introduction of standardization and dimension reduction procedures before classification without re-training improved the prediction accuracy obtained using NCC (0.804) without a loss in prediction accuracy for the TCIA dataset. Furthermore, we confirmed the same tendency in a model for IDH1/2 mutation prediction with standardization and application of dimension reduction that was also applicable to multiple hospitals. Our results demonstrated that overfitting may occur when an ML method providing the highest accuracy in a small training dataset is used for different heterogeneous data sets, and suggested a promising process for developing an ML method applicable to multiple cohorts.
Collapse
|
46
|
Doig D, Kachramanoglou C, Dumba M, Tona F, Gontsarova A, Limbäck C, Jan W. Characterisation of isocitrate dehydrogenase gene mutant WHO grade 2 and 3 gliomas: MRI predictors of 1p/19q co-deletion and tumour grade. Clin Radiol 2021; 76:785.e9-785.e16. [PMID: 34289936 DOI: 10.1016/j.crad.2021.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/18/2021] [Indexed: 11/28/2022]
Abstract
AIM To identify imaging predictors of molecular subtype and tumour grade in patients with isocitrate dehydrogenase (IDH) gene mutant (IDHmut) World Health Organization (WHO) grade 2 or 3 gliomas. MATERIALS AND METHODS Patients with histologically confirmed WHO grade 2 or 3 IDHmut gliomas between 2016 and 2019 were included in the study. Magnetic resonance imaging (MRI) images were evaluated for the presence or absence of potential imaging predictors of tumour subtype, such as T2/fluid attenuated inversion recovery (FLAIR) signal match, and these factors were examined using regression analysis. On perfusion imaging, the maximum relative cerebral blood volume (rCBVmax) was evaluated as a potential predictor of tumour grade. The performance of two experienced neuroradiologists in correctly predicting tumour type on MRI was evaluated. RESULTS Eighty-five patients were included in the study. The presence of T2/FLAIR signal match >50% of tumour volume (p<0.01) and intratumoural susceptibility (p=0.02) were independent predictors of 1p/19q co-deletion. Mean rCBV max was significantly higher in WHO grade 3 astrocytomas (p=0.04) than WHO grade 2 astrocytomas. The consensus prediction of 1p/19q co-deletion status by two neuroradiologists of tumour was 95% sensitive and 86% specific. CONCLUSION The presence of matched T2/FLAIR signal could be used to identify tumour subtype when biopsy is inconclusive or genetic analysis is unavailable. rCBVmax predicted astrocytoma grade. Experienced neuroradiologists predict tumour subtype with good sensitivity and specificity.
Collapse
Affiliation(s)
- D Doig
- Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK.
| | - C Kachramanoglou
- Department of Imaging, Imperial College Healthcare NHS Trust, London, UK
| | - M Dumba
- Department of Imaging, Imperial College Healthcare NHS Trust, London, UK; Imperial College Faculty of Medicine, London, UK
| | - F Tona
- Department of Imaging, Imperial College Healthcare NHS Trust, London, UK
| | - A Gontsarova
- Department of Imaging, Imperial College Healthcare NHS Trust, London, UK
| | - C Limbäck
- Department of Cellular Pathology, Imperial College Healthcare NHS Trust, London, UK; Imperial College Faculty of Medicine, London, UK
| | - W Jan
- Department of Imaging, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
47
|
Mweempwa A, Rosenthal MA, Dimou J, Drummond KJ, Whittle JR. Perioperative clinical trials for glioma: Raising the bar. J Clin Neurosci 2021; 89:144-150. [PMID: 34119258 DOI: 10.1016/j.jocn.2021.04.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/25/2021] [Indexed: 02/07/2023]
Abstract
Gliomas are a heterogeneous group of primary brain cancers with poor survival despite multimodality therapy that includes surgery, radiation and chemotherapy. Numerous clinical trials have investigated systemic therapies in glioma, but have largely been negative. Multiple factors have contributed to the lack of progress including tumour heterogeneity, the tumour micro-environment and presence of the blood-brain barrier, as well as extrinsic factors relating to trial design, such as the lack of a contemporaneous biopsy at the time of treatment. A number of strategies have been proposed to progress new agents into the clinic. Here, we review the progress of perioperative, including phase 0 and 'window of opportunity', studies and provide recommendations for trial design in the development of new agents for glioma. The incorporation of pre- and post-treatment biopsies in glioma early phase trials will provide valuable pharmacokinetic and pharmacodynamic data and also determine the target or biomarker effect, which will guide further development of new agents. Perioperative 'window of opportunity' studies must use drugs with a recommended-phase-2-dose, known safety profile and adequate blood-brain barrier penetration. Drugs shown to have on-target effects in perioperative trials can then be evaluated further in a larger cohort of patients in an adaptive trial to increase the efficiency of drug development.
Collapse
Affiliation(s)
- Angela Mweempwa
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Mark A Rosenthal
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - James Dimou
- Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC 3050, Australia; Department of Surgery, University of Melbourne, Parkville, VIC 3010, Australia
| | - Katharine J Drummond
- Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC 3050, Australia; Department of Surgery, University of Melbourne, Parkville, VIC 3010, Australia
| | - James R Whittle
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
48
|
Neuroimaging in the Era of the Evolving WHO Classification of Brain Tumors, From the AJR Special Series on Cancer Staging. AJR Am J Roentgenol 2021; 217:3-15. [PMID: 33502214 DOI: 10.2214/ajr.20.25246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The inclusion of molecular and genetic information with histopathologic information defines the framework for brain tumor classification and grading. This framework is reflected in the major restructuring of the WHO brain tumor classification system in 2016 and in numerous subsequent proposed updates reflecting ongoing developments in understanding the impact of tumor genotype on classification and grading. This incorporation of molecular and genetic features improves tumor diagnosis and prediction of tumor behavior and response to treatment. Neuroimaging is essential for the noninvasive assessment of pretreatment tumor grading and for identification and determination of therapeutic efficacy. Use of conventional neuroimaging and physiologic imaging techniques, such as diffusion- and perfusion-weighted MRI, can increase diagnostic confidence before and after treatment. Although the use of neuroimaging to consistently determine tumor genetics is not yet robust, promising developments are on the horizon. Given the complexity of the brain tumor microenvironment, the development and implementation of a standardized reporting system can aid in conveying to radiologists, referring providers, and patients important information about brain tumor response to treatment. The purpose of this article is to review the current state and role of neuroimaging in this continuously evolving field.
Collapse
|
49
|
Jain R, Johnson DR, Patel SH, Castillo M, Smits M, van den Bent MJ, Chi AS, Cahill DP. "Real world" use of a highly reliable imaging sign: "T2-FLAIR mismatch" for identification of IDH mutant astrocytomas. Neuro Oncol 2021; 22:936-943. [PMID: 32064507 DOI: 10.1093/neuonc/noaa041] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AbstractThe T2-FLAIR (fluid attenuated inversion recovery) mismatch sign is an easily detectable imaging sign on routine clinical MRI studies that suggests diagnosis of isocitrate dehydrogenase (IDH)-mutant 1p/19q non-codeleted gliomas. Multiple independent studies show that the T2-FLAIR mismatch sign has near-perfect specificity, but low sensitivity for diagnosing IDH-mutant astrocytomas. Thus, the T2-FLAIR mismatch sign represents a non-invasive radiogenomic diagnostic finding with potential clinical impact. Recently, false positive cases have been reported, many related to variable application of the sign's imaging criteria and differences in image acquisition, as well as to differences in the included patient populations. Here we summarize the imaging criteria for the T2-FLAIR mismatch sign, review similarities and differences between the multiple validation studies, outline strategies to optimize its clinical use, and discuss potential opportunities to refine imaging criteria in order to maximize its impact in glioma diagnostics.
Collapse
Affiliation(s)
- Rajan Jain
- Departments of Radiology and Neurosurgery, New York University Langone Health, New York, New York, USA
| | - Derek R Johnson
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sohil H Patel
- Department of Radiology, University of Virginia Health, Charlottesville, Virginia, USA
| | - Mauricio Castillo
- Department of Radiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | | | | | - Daniel P Cahill
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
50
|
Habib A, Jovanovich N, Hoppe M, Ak M, Mamindla P, R. Colen R, Zinn PO. MRI-Based Radiomics and Radiogenomics in the Management of Low-Grade Gliomas: Evaluating the Evidence for a Paradigm Shift. J Clin Med 2021; 10:1411. [PMID: 33915813 PMCID: PMC8036428 DOI: 10.3390/jcm10071411] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 12/29/2022] Open
Abstract
Low-grade gliomas (LGGs) are tumors that affect mostly adults. These neoplasms are comprised mainly of oligodendrogliomas and diffuse astrocytomas. LGGs remain vexing to current management and therapeutic modalities although they exhibit more favorable survival rates compared with high-grade gliomas (HGGs). The specific genetic subtypes that these tumors exhibit result in variable clinical courses and the need to involve multidisciplinary teams of neurologists, epileptologists, neurooncologists and neurosurgeons. Currently, the diagnosis of an LGG pivots mainly around the preliminary radiological findings and the subsequent definitive surgical diagnosis (via surgical sampling). The introduction of radiomics as a high throughput quantitative imaging technique that allows for improved diagnostic, prognostic and predictive indices has created more interest for such techniques in cancer research and especially in neurooncology (MRI-based classification of LGGs, predicting Isocitrate dehydrogenase (IDH) and Telomerase reverse transcriptase (TERT) promoter mutations and predicting LGG associated seizures). Radiogenomics refers to the linkage of imaging findings with the tumor/tissue genomics. Numerous applications of radiomics and radiogenomics have been described in the clinical context and management of LGGs. In this review, we describe the recently published studies discussing the potential application of radiomics and radiogenomics in LGGs. We also highlight the potential pitfalls of the above-mentioned high throughput computerized techniques and, most excitingly, explore the use of machine learning artificial intelligence technologies as standalone and adjunct imaging tools en route to enhance a personalized MRI-based tumor diagnosis and management plan design.
Collapse
Affiliation(s)
- Ahmed Habib
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA;
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
| | - Nicolina Jovanovich
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
| | - Meagan Hoppe
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
| | - Murat Ak
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
- Department of Diagnostic Radiology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Priyadarshini Mamindla
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
| | - Rivka R. Colen
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
- Department of Diagnostic Radiology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Pascal O. Zinn
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA;
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (N.J.); (M.H.); (M.A.); (P.M.); (R.R.C.)
| |
Collapse
|