101
|
MRI Features and IDH Mutational Status of Grade II Diffuse Gliomas: Impact on Diagnosis and Prognosis. AJR Am J Roentgenol 2017; 210:621-628. [PMID: 29261348 DOI: 10.2214/ajr.17.18457] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Grade II diffuse gliomas (DGs) with isocitrate dehydrogenase (IDH) mutations are associated with better prognosis than their IDH wild-type counterparts. We sought to determine the MRI characteristics associated with IDH mutational status and ascertain whether MRI considered in combination with IDH mutational status can better predict the clinical outcomes of grade II DGs. MATERIALS AND METHODS Preoperative MRI examinations were retrospectively studied for qualitative tumor characteristics, including location, extent, cortical involvement, margin sharpness, cystic component, mineralization or hemorrhage, and contrast enhancement. Quantitative diffusion and perfusion metrics were also assessed. Logistic regression and ROC analyses were used to evaluate the relationship between MRI features and IDH mutational status. The association between IDH mutational status, 1p19q codeletion, MRI features, extent of resection, and clinical outcomes was assessed by Kaplan-Meier and Cox proportional hazards models. RESULTS Of 100 grade II DGs, 78 were IDH mutant and 22 were IDH wild type. IDH wild-type tumors were associated with older age, multifocality, brainstem involvement, lack of cystic change, and a lower apparent diffusion coefficient (ADC). Multivariable regression showed that age older than 45 years as well as low minimum ADC (ADCmin), mean ADC, and maximum ADC values were independently associated with IDH mutational status. Of these, an ADCmin threshold of 0.9 × 10-3 mm2/s or less provided the greatest sensitivity and specificity (91% and 76%, respectively) in defining IDH wild-type grade II DGs. Combining low ADCmin with IDH wild-type status conferred worse outcomes than did IDH wild-type status alone. CONCLUSION IDH wild-type grade II DGs are associated with a lower ADC and poor clinical outcomes. Combining IDH mutational status and ADC may allow more accurate prediction of clinical outcomes for patients with grade II DGs.
Collapse
|
102
|
Delfanti RL, Piccioni DE, Handwerker J, Bahrami N, Krishnan A, Karunamuni R, Hattangadi-Gluth JA, Seibert TM, Srikant A, Jones KA, Snyder VS, Dale AM, White NS, McDonald CR, Farid N. Imaging correlates for the 2016 update on WHO classification of grade II/III gliomas: implications for IDH, 1p/19q and ATRX status. J Neurooncol 2017; 135:601-609. [PMID: 28871469 PMCID: PMC5700844 DOI: 10.1007/s11060-017-2613-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 08/20/2017] [Indexed: 12/12/2022]
Abstract
The 2016 World Health Organization Classification of Tumors of the Central Nervous System incorporates the use of molecular information into the classification of brain tumors, including grade II and III gliomas, providing new prognostic information that cannot be delineated based on histopathology alone. We hypothesized that these genomic subgroups may also have distinct imaging features. A retrospective single institution study was performed on 40 patients with pathologically proven infiltrating WHO grade II/III gliomas with a pre-treatment MRI and molecular data on IDH, chromosomes 1p/19q and ATRX status. Two blinded Neuroradiologists qualitatively assessed MR features. The relationship between each parameter and molecular subgroup (IDH-wildtype; IDH-mutant-1p/19q codeleted-ATRX intact; IDH-mutant-1p/19q intact-ATRX loss) was evaluated with Fisher's exact test. Progression free survival (PFS) was also analyzed. A border that could not be defined on FLAIR was most characteristic of IDH-wildtype tumors, whereas IDH-mutant tumors demonstrated either well-defined or slightly ill-defined borders (p = 0.019). Degree of contrast enhancement and presence of restricted diffusion did not distinguish molecular subgroups. Frontal lobe predominance was associated with IDH-mutant tumors (p = 0.006). The IDH-wildtype subgroup had significantly shorter PFS than the IDH-mutant groups (p < 0.001). No differences in PFS were present when separating by tumor grade. FLAIR border patterns and tumor location were associated with distinct molecular subgroups of grade II/III gliomas. These imaging features may provide fundamental prognostic and predictive information at time of initial diagnostic imaging.
Collapse
Affiliation(s)
- Rachel L Delfanti
- Department of Radiology, University of California, San Diego, 200 West Arbor Drive, La Jolla, CA, 92037, USA.
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, 92037, USA.
| | - David E Piccioni
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Jason Handwerker
- Department of Radiology, University of California, San Diego, 200 West Arbor Drive, La Jolla, CA, 92037, USA
| | - Naeim Bahrami
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, 92037, USA
| | - AnithaPriya Krishnan
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Roshan Karunamuni
- Department of Radiation Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Jona A Hattangadi-Gluth
- Department of Radiation Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Tyler M Seibert
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, 92037, USA
- Department of Radiation Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Ashwin Srikant
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Karra A Jones
- Department of Pathology, University of Iowa Hospitals & Clinics, Iowa City, IA, 52242, USA
| | - Vivian S Snyder
- Department of Pathology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Anders M Dale
- Department of Radiology, University of California, San Diego, 200 West Arbor Drive, La Jolla, CA, 92037, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92037, USA
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Nathan S White
- Department of Radiology, University of California, San Diego, 200 West Arbor Drive, La Jolla, CA, 92037, USA
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Carrie R McDonald
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, 92037, USA
- Department of Radiation Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Nikdokht Farid
- Department of Radiology, University of California, San Diego, 200 West Arbor Drive, La Jolla, CA, 92037, USA
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, 92037, USA
| |
Collapse
|
103
|
Zhou J, Reddy MV, Wilson BKJ, Blair DA, Taha A, Frampton CM, Eiholzer RA, Gan PYC, Ziad F, Thotathil Z, Kirs S, Hung NA, Royds JA, Slatter TL. MR Imaging Characteristics Associate with Tumor-Associated Macrophages in Glioblastoma and Provide an Improved Signature for Survival Prognostication. AJNR Am J Neuroradiol 2017; 39:252-259. [PMID: 29191871 DOI: 10.3174/ajnr.a5441] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 09/07/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE In glioblastoma, tumor-associated macrophages have tumor-promoting properties. This study determined whether routine MR imaging features could predict molecular subtypes of glioblastoma that differ in the content of tumor-associated macrophages. MATERIALS AND METHODS Seven internally derived MR imaging features were assessed in 180 patients, and 25 features from the Visually AcceSAble Rembrandt Images feature set were assessed in 164 patients. Glioblastomas were divided into subtypes based on the telomere maintenance mechanism: alternative lengthening of telomeres positive (ALT+) and negative (ALT-) and the content of tumor-associated macrophages (with [M+] or without [M-] a high content of macrophages). The 3 most frequent subtypes (ALT+/M-, ALT-/M+, and ALT-/M-) were correlated with MR imaging features and clinical parameters. The fourth group (ALT+/M+) did not have enough cases for correlation with MR imaging features. RESULTS Tumors with a regular margin and those lacking a fungating margin, an expansive T1/FLAIR ratio, and reduced ependymal extension were more frequent in the subgroup of ALT+/M- (P < .05). Radiologic necrosis, lack of cystic component (by both criteria), and extensive peritumoral edema were more frequent in ALT-/M+ tumors (P < .05). Multivariate testing with a Cox regression analysis found the cystic imaging feature was additive to tumor subtype, and O6-methylguanine methyltransferase (MGMT) status to predict improved patient survival (P < .05). CONCLUSIONS Glioblastomas with tumor-associated macrophages are associated with routine MR imaging features consistent with these tumors being more aggressive. Inclusion of cystic change with molecular subtypes and MGMT status provided a better estimate of survival.
Collapse
Affiliation(s)
- J Zhou
- From the Departments of Radiology (J.Z., M.V.R., B.K.J.W.) .,Department of Pathology (J.Z., R.A.E., N.A.H., J.A.R., T.L.S.)
| | - M V Reddy
- From the Departments of Radiology (J.Z., M.V.R., B.K.J.W.)
| | - B K J Wilson
- From the Departments of Radiology (J.Z., M.V.R., B.K.J.W.)
| | | | - A Taha
- Neurosurgery (A.T.), Southern District Health Board, Dunedin, New Zealand.,Surgical Sciences (A.T., S.K.), Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - C M Frampton
- Department of Medicine (C.M.F.), University of Otago, Christchurch, New Zealand
| | - R A Eiholzer
- Department of Pathology (J.Z., R.A.E., N.A.H., J.A.R., T.L.S.)
| | | | | | - Z Thotathil
- Medical Oncology (Z.T.), Waikato District Health Board, Hamilton, New Zealand
| | - S Kirs
- Surgical Sciences (A.T., S.K.), Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - N A Hung
- Department of Pathology (J.Z., R.A.E., N.A.H., J.A.R., T.L.S.)
| | - J A Royds
- Department of Pathology (J.Z., R.A.E., N.A.H., J.A.R., T.L.S.)
| | - T L Slatter
- Department of Pathology (J.Z., R.A.E., N.A.H., J.A.R., T.L.S.)
| |
Collapse
|
104
|
Chang K, Bai HX, Zhou H, Su C, Bi WL, Agbodza E, Kavouridis VK, Senders JT, Boaro A, Beers A, Zhang B, Capellini A, Liao W, Shen Q, Li X, Xiao B, Cryan J, Ramkissoon S, Ramkissoon L, Ligon K, Wen PY, Bindra RS, Woo J, Arnaout O, Gerstner ER, Zhang PJ, Rosen BR, Yang L, Huang RY, Kalpathy-Cramer J. Residual Convolutional Neural Network for the Determination of IDH Status in Low- and High-Grade Gliomas from MR Imaging. Clin Cancer Res 2017; 24:1073-1081. [PMID: 29167275 DOI: 10.1158/1078-0432.ccr-17-2236] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/12/2017] [Accepted: 11/16/2017] [Indexed: 01/23/2023]
Abstract
Purpose: Isocitrate dehydrogenase (IDH) mutations in glioma patients confer longer survival and may guide treatment decision making. We aimed to predict the IDH status of gliomas from MR imaging by applying a residual convolutional neural network to preoperative radiographic data.Experimental Design: Preoperative imaging was acquired for 201 patients from the Hospital of University of Pennsylvania (HUP), 157 patients from Brigham and Women's Hospital (BWH), and 138 patients from The Cancer Imaging Archive (TCIA) and divided into training, validation, and testing sets. We trained a residual convolutional neural network for each MR sequence (FLAIR, T2, T1 precontrast, and T1 postcontrast) and built a predictive model from the outputs. To increase the size of the training set and prevent overfitting, we augmented the training set images by introducing random rotations, translations, flips, shearing, and zooming.Results: With our neural network model, we achieved IDH prediction accuracies of 82.8% (AUC = 0.90), 83.0% (AUC = 0.93), and 85.7% (AUC = 0.94) within training, validation, and testing sets, respectively. When age at diagnosis was incorporated into the model, the training, validation, and testing accuracies increased to 87.3% (AUC = 0.93), 87.6% (AUC = 0.95), and 89.1% (AUC = 0.95), respectively.Conclusions: We developed a deep learning technique to noninvasively predict IDH genotype in grade II-IV glioma using conventional MR imaging using a multi-institutional data set. Clin Cancer Res; 24(5); 1073-81. ©2017 AACR.
Collapse
Affiliation(s)
- Ken Chang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Harrison X Bai
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hao Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chang Su
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ena Agbodza
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vasileios K Kavouridis
- Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Boston, Massachusetts
| | - Joeky T Senders
- Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Boston, Massachusetts
| | - Alessandro Boaro
- Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Boston, Massachusetts
| | - Andrew Beers
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Biqi Zhang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Alexandra Capellini
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qin Shen
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jane Cryan
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Shakti Ramkissoon
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lori Ramkissoon
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Keith Ligon
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Patrick Y Wen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut
| | - John Woo
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Omar Arnaout
- Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Paul J Zhang
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Li Yang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Jayashree Kalpathy-Cramer
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
105
|
Miller JJ, Shih HA, Andronesi OC, Cahill DP. Isocitrate dehydrogenase-mutant glioma: Evolving clinical and therapeutic implications. Cancer 2017; 123:4535-4546. [DOI: 10.1002/cncr.31039] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/18/2017] [Accepted: 08/29/2017] [Indexed: 02/04/2023]
Affiliation(s)
- Julie J. Miller
- Pappas Center for Neuro-Oncology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Boston Massachusetts
| | - Helen A. Shih
- Department of Radiation Oncology; Massachusetts General Hospital, Harvard Medical School; Boston Massachusetts
| | - Ovidiu C. Andronesi
- Martinos Center for Biomedical Imaging, Department of Radiology; Massachusetts General Hospital, Harvard Medical School; Boston Massachusetts
| | - Daniel P. Cahill
- Department of Neurosurgery; Massachusetts General Hospital, Harvard Medical School; Boston Massachusetts
| |
Collapse
|
106
|
Tateishi K, Wakimoto H, Cahill DP. IDH1 Mutation and World Health Organization 2016 Diagnostic Criteria for Adult Diffuse Gliomas: Advances in Surgical Strategy. Neurosurgery 2017; 64:134-138. [PMID: 28899049 DOI: 10.1093/neuros/nyx247] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/14/2017] [Indexed: 12/12/2022] Open
Affiliation(s)
- Kensuke Tateishi
- Department of Neurosurgery, Massac-husetts General Hospital Cancer Cen-ter, Harvard Medical School, Boston, Ma-ssachusetts.,Department of Neuro-surgery, Graduate School of Medical Science, Yokohama City University, Yokohama, Japan.,Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massac-husetts General Hospital Cancer Cen-ter, Harvard Medical School, Boston, Ma-ssachusetts.,Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Daniel P Cahill
- Department of Neurosurgery, Massac-husetts General Hospital Cancer Cen-ter, Harvard Medical School, Boston, Ma-ssachusetts.,Translational Neuro-Oncology Laboratory, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
107
|
Zuo M, Li M, Chen N, Yu T, Kong B, Liang R, Wang X, Mao Q, Liu Y. IDH1 status is significantly different between high-grade thalamic and superficial gliomas. Cancer Biomark 2017; 20:183-189. [PMID: 28869450 DOI: 10.3233/cbm-170175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND While major progress has been made in diagnosis and treatment of gliomas based on molecules, molecular features of thalamic glioma have rarely been reported till now. OBJECTIVE IDH1 mutation is important for prognosis of gliomas and represents a distinctive category of glioma. We intended to survey specific molecular abnormalities in high-grade thalamic gliomas (WHO III-IV). METHODS We collected data of 50 and 93 newly diagnosed high-grade thalamic and superficial glioma patients respectively and conducted a comparative analysis of molecular characteristics between them. We analyzed expressions of molecules as follow: IDH1/2, P53, Ki-67, ATRX, PTEN, MMP9 and MGMT by Immunohistochemistry (IHC). Direct gene sequencing was performed to test the IDH1(R 132H) mutation. RESULTS We found a significant difference of IDH1 mutation between those high-grade gliomas, with 92% (46/50) of the thalamic tumors and 71% (66/93) of the superficial gliomas showing IDH1 wild-type (p= 0.004). It also showed that IDH1 mutation in superficial glioblastomas 18.6% (13/70) occurred more than thalamic glioblastomas 2.6% (1/39) (p= 0.017). As to high-grade superficial gliomas, there were 26 patients with IDH1 mutation, which contained 7, 13, and 6 high, moderate and low Ki-67 expression gliomas, respectively. The IDH1 wild-type group (62 patients), was composed of 29, 30, and 3 high, moderate and low Ki-67 expression gliomas, respectively. There was a significant distinction between the IDH1 mutation and Ki-67 expressions (p= 0.024). We also noted that the occurrence of low Ki-67 expressions 23.1% (6/26) in IDH1 mutation group was outnumbered than IDH1 wild-type group 4.8% (3/62) (p= 0.018). In addition, we found PTEN negative correlated with MMP9 negative in thalamic high-grade gliomas, whereas no such difference was found in superficial gliomas (p= 0.016). CONCLUSION The rare occurrence of IDH1 mutant high-grade thalamic gliomas strongly suggested that the high-grade thalamic glioma is another distinct tumor entity as compared to the high-grade superficial gliomas.
Collapse
Affiliation(s)
- Mingrong Zuo
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan, China
| | - Mao Li
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan, China
| | - Ni Chen
- Department of Pathology, Sichuan University, West China Hospital, Chengdu, Sichuan, China
| | - Tianping Yu
- Department of Pathology, Sichuan University, West China Hospital, Chengdu, Sichuan, China
| | - Bing Kong
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan, China
| | - Ruofei Liang
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan, China
| | - Xiang Wang
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan, China
| | - Qing Mao
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan, China
| | - Yanhui Liu
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan, China
| |
Collapse
|
108
|
Youland RS, Kreofsky CR, Schomas DA, Brown PD, Buckner JC, Laack NN. The impact of adjuvant therapy for patients with high-risk diffuse WHO grade II glioma. J Neurooncol 2017; 135:535-543. [PMID: 28836106 DOI: 10.1007/s11060-017-2599-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/18/2017] [Indexed: 11/26/2022]
Abstract
Despite recent randomized, prospective evidence supporting use of RT and chemotherapy (CRT) for high-risk low-grade gliomas (LGG), many patients have historically received RT alone, chemotherapy alone or observation postoperatively. The purpose of this study is to evaluate outcomes for historical treatments in comparison to CRT for high-risk diffuse WHO grade II glioma patients. Records from 309 adults with WHO grade II glioma (1997-2008) eligible for RTOG 9802 (incomplete resection/biopsy or age ≥40 years) were retrospectively reviewed. Kaplan-Meier estimates were used for progression-free survival (PFS) and overall survival (OS). The Cox proportional hazards model was used for estimates of risk ratios for univariate and multivariate analyses. Median follow-up was 10.6 years. Adjuvant treatments included radiotherapy (RT) alone (45%), observation (31%), CRT (21%) and chemotherapy alone (3%). Non-astrocytic histology, TERT promoter mutation, 1p/19q codeletion and extensive resections were associated with improved PFS and OS on univariate analysis (all p < 0.05). IDH mutations and adjuvant CRT was associated with improved PFS (all p < 0.05). On multivariate analysis, histology, molecular grouping and extent of resection were significantly associated with PFS and OS. In addition, multivariate analysis revealed that CRT was associated with improved PFS and OS compared with RT alone, and improved PFS compared with observation. This study confirms the benefit of adding chemotherapy to RT compared with RT alone or observation. These findings emphasize the need for aggressive treatment in patients with high-risk LGG.
Collapse
Affiliation(s)
- Ryan S Youland
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Cole R Kreofsky
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - David A Schomas
- Department of Radiation Oncology, Saint Luke's Cancer Institute, Kansas City, MO, 64111, USA
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jan C Buckner
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nadia N Laack
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
109
|
Abstract
Primary brain tumors, most commonly gliomas, are histopathologically typed and graded as World Health Organization (WHO) grades I-IV according to increasing degrees of malignancy. These grades provide prognostic information and guidance on treatment such as radiation therapy and chemotherapy after surgery. Despite the confirmed value of the WHO grading system, results of a multitude of studies and prospective interventional trials now indicate that tumors with identical morphologic criteria can have highly different outcomes. Molecular markers can allow subtypes of tumors of the same morphologic type and WHO grade to be distinguished and are, therefore, of great interest in personalization of brain tumor treatment. Recent genomic-wide studies have resulted in a far more comprehensive understanding of the genomic alterations in gliomas and provide suggestions for a new molecularly based classification. Magnetic resonance (MR) imaging phenotypes can serve as noninvasive surrogates for tumor genotypes and can provide important information for diagnosis, prognosis, and, eventually, personalized treatment. The newly emerged field of radiogenomics allows specific MR imaging phenotypes to be linked with gene expression profiles. In this article, the authors review the conventional and advanced imaging features of three tumoral genotypes with prognostic and therapeutic consequences: (a) isocitrate dehydrogenase mutation; (b) the combined loss of the short arm of chromosome 1 and the long arm of chromosome 19, or 1p19q codeletion; and (c) methylguanine methyltransferase promoter methylation. © RSNA, 2017.
Collapse
Affiliation(s)
- Marion Smits
- From the Department of Radiology, Erasmus MC University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, the Netherlands (M.S.); and Brain Tumor Center, Erasmus MC Cancer Center, Rotterdam, the Netherlands (M.J.v.d.B.)
| | - Martin J van den Bent
- From the Department of Radiology, Erasmus MC University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, the Netherlands (M.S.); and Brain Tumor Center, Erasmus MC Cancer Center, Rotterdam, the Netherlands (M.J.v.d.B.)
| |
Collapse
|
110
|
Patel SH, Poisson LM, Brat DJ, Zhou Y, Cooper L, Snuderl M, Thomas C, Franceschi AM, Griffith B, Flanders AE, Golfinos JG, Chi AS, Jain R. T2-FLAIR Mismatch, an Imaging Biomarker for IDH and 1p/19q Status in Lower-grade Gliomas: A TCGA/TCIA Project. Clin Cancer Res 2017; 23:6078-6085. [PMID: 28751449 DOI: 10.1158/1078-0432.ccr-17-0560] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/11/2017] [Accepted: 07/19/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Lower-grade gliomas (WHO grade II/III) have been classified into clinically relevant molecular subtypes based on IDH and 1p/19q mutation status. The purpose was to investigate whether T2/FLAIR MRI features could distinguish between lower-grade glioma molecular subtypes.Experimental Design: MRI scans from the TCGA/TCIA lower grade glioma database (n = 125) were evaluated by two independent neuroradiologists to assess (i) presence/absence of homogenous signal on T2WI; (ii) presence/absence of "T2-FLAIR mismatch" sign; (iii) sharp or indistinct lesion margins; and (iv) presence/absence of peritumoral edema. Metrics with moderate-substantial agreement underwent consensus review and were correlated with glioma molecular subtypes. Somatic mutation, DNA copy number, DNA methylation, gene expression, and protein array data from the TCGA lower-grade glioma database were analyzed for molecular-radiographic associations. A separate institutional cohort (n = 82) was analyzed to validate the T2-FLAIR mismatch sign.Results: Among TCGA/TCIA cases, interreader agreement was calculated for lesion homogeneity [κ = 0.234 (0.111-0.358)], T2-FLAIR mismatch sign [κ = 0.728 (0.538-0.918)], lesion margins [κ = 0.292 (0.135-0.449)], and peritumoral edema [κ = 0.173 (0.096-0.250)]. All 15 cases that were positive for the T2-FLAIR mismatch sign were IDH-mutant, 1p/19q non-codeleted tumors (P < 0.0001; PPV = 100%, NPV = 54%). Analysis of the validation cohort demonstrated substantial interreader agreement for the T2-FLAIR mismatch sign [κ = 0.747 (0.536-0.958)]; all 10 cases positive for the T2-FLAIR mismatch sign were IDH-mutant, 1p/19q non-codeleted tumors (P < 0.00001; PPV = 100%, NPV = 76%).Conclusions: Among lower-grade gliomas, T2-FLAIR mismatch sign represents a highly specific imaging biomarker for the IDH-mutant, 1p/19q non-codeleted molecular subtype. Clin Cancer Res; 23(20); 6078-85. ©2017 AACR.
Collapse
Affiliation(s)
- Sohil H Patel
- Department of Radiology and Medical Imaging, University of Virginia Health System, Charlottesville, Virginia.
| | - Laila M Poisson
- Department of Public Health, Henry Ford Health System, Detroit, Michigan
| | - Daniel J Brat
- Department of Pathology and Laboratory Medicine, Winship Cancer Institute at Emory University, Atlanta, Georgia
| | - Yueren Zhou
- Department of Public Health, Henry Ford Health System, Detroit, Michigan
| | - Lee Cooper
- Department of Biomedical Informatics, Emory School of Medicine, Atlanta, Georgia
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University School of Medicine, Atlanta, Georgia
| | - Matija Snuderl
- Department of Pathology, NYU Langone Medical Center, New York, New York
| | - Cheddhi Thomas
- Department of Pathology, NYU Langone Medical Center, New York, New York
| | - Ana M Franceschi
- Department of Radiology, NYU Langone Medical Center, New York, New York
| | - Brent Griffith
- Department of Radiology, Henry Ford Health System, Detroit, Michigan
| | - Adam E Flanders
- Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - John G Golfinos
- Department of Neurosurgery, NYU Langone Medical Center, New York, New York
| | - Andrew S Chi
- Department of Neurosurgery, NYU Langone Medical Center, New York, New York
- Division of Neuro-Oncology, NYU Langone Medical Center, New York, New York
| | - Rajan Jain
- Department of Radiology, NYU Langone Medical Center, New York, New York.
- Department of Neurosurgery, NYU Langone Medical Center, New York, New York
| |
Collapse
|
111
|
Jiang S, Zou T, Eberhart CG, Villalobos MAV, Heo HY, Zhang Y, Wang Y, Wang X, Yu H, Du Y, van Zijl PCM, Wen Z, Zhou J. Predicting IDH mutation status in grade II gliomas using amide proton transfer-weighted (APTw) MRI. Magn Reson Med 2017; 78:1100-1109. [PMID: 28714279 DOI: 10.1002/mrm.26820] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/31/2017] [Accepted: 06/11/2017] [Indexed: 12/24/2022]
Abstract
PURPOSE To assess the amide proton transfer-weighted (APTw) MRI features of isocitrate dehydrogenase (IDH)-wildtype and IDH-mutant grade II gliomas and to test the hypothesis that the APTw signal is a surrogate imaging marker for identifying IDH mutation status preoperatively. METHODS Twenty-seven patients with pathologically confirmed low-grade glioma, who were previously scanned at 3T, were retrospectively analyzed. The Mann-Whitney test was used to evaluate relationships between APTw intensities for IDH-mutant and IDH-wildtype groups, and receiver operator characteristic (ROC) analysis was used to assess the diagnostic performance of APTw. RESULTS Based on histopathology and molecular analysis, seven cases were diagnosed as IDH-wildtype grade II gliomas and 20 cases as IDH-mutant grade II gliomas. The maximum and minimum APTw values, based on multiple regions of interest, as well as the whole-tumor histogram-based mean and 50th percentile APTw values, were significantly higher in the IDH-wildtype gliomas than in the IDH-mutant groups. This corresponded to the areas under the ROC curves of 0.89, 0.76, 0.75, and 0.75, respectively, for the prediction of the IDH mutation status. CONCLUSION IDH-wildtype lesions typically were associated with relatively high APTw signal intensities as compared with IDH-mutant lesions. The APTw signal could be a valuable imaging biomarker by which to identify IDH1 mutation status in grade II gliomas. Magn Reson Med 78:1100-1109, 2017. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Shanshan Jiang
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Radiology, Futian Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Tianyu Zou
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Hye-Young Heo
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yi Zhang
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yu Wang
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xianlong Wang
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hao Yu
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongxing Du
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Peter C M van Zijl
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Zhibo Wen
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinyuan Zhou
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| |
Collapse
|
112
|
Lobanova NV, Shishkina LV, Ryzhova MV, Kobyakov GL, Sycheva RV, Burov SA, Lukyanov AV, Omarova ZR. [Clinical, immunohistochemical, and molecular genetic prognostic factors in adult patients with glioblastoma]. Arkh Patol 2017; 78:10-19. [PMID: 27600777 DOI: 10.17116/patol201678410-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
UNLABELLED Glioblastoma is the most common primary malignant glial tumor of the brain in adult patients. AIM to define the prognostic value of isocitrate dehydrogenase-1 (IDH-1) mutation and methylguanine-DNA methyltransferase (MGMT) methylation status in patients with glioblastoma (GB) and to analyze the impact of clinical data (gender, age, and tumor site), histological variants of the tumor structure, and time to development of recurrences on the course of the disease. SUBJECTS AND METHODS The investigation enrolled 63 GB patients aged 18 to 71 years who had received combined treatment (surgery, chemo- and radiotherapy) at the N.I. Burdenko Research Institute of Neurosurgery, Ministry of Health of the Russian Federation, in the period 2008 to 2011. The investigators performed a morphological examination of all tumor tissue samples and an immunohistochemical examination using anti-IDH-1 R-132 antibody clone («Dianova», Germany) and defined MGMT methylation status by a polymerase chain reaction using the CpGenome DNA Modification Kit («Chemicon International», USA). The data were statistically processed using a package of Statistica 6.0 programs. RESULTS Patient age, time to development of recurrent glioblastoma, mutations in the IDH-1 gene and MGMT were found to be prognostic factors for overall survival among adult patients in this category. CONCLUSION Analysis of clinical findings and identification of molecular genetic aberrations in the tumor cells will be able to elaborate an individual approach to treating patients with glioblastoma in order to increase their survival rates and to improve quality of life.
Collapse
Affiliation(s)
- N V Lobanova
- Central Clinical Military Hospital, Federal Security Service of Russia, Moscow
| | - L V Shishkina
- N.N. Burdenko Research Institute of Neurosurgery, Ministry of Health of Russia, Moscow, Russia
| | - M V Ryzhova
- N.N. Burdenko Research Institute of Neurosurgery, Ministry of Health of Russia, Moscow, Russia
| | - G L Kobyakov
- N.N. Burdenko Research Institute of Neurosurgery, Ministry of Health of Russia, Moscow, Russia
| | - R V Sycheva
- N.N. Burdenko Research Institute of Neurosurgery, Ministry of Health of Russia, Moscow, Russia
| | - S A Burov
- Central Clinical Military Hospital, Federal Security Service of Russia, Moscow
| | - A V Lukyanov
- Central Clinical Military Hospital, Federal Security Service of Russia, Moscow
| | - Zh R Omarova
- Central Clinical Military Hospital, Federal Security Service of Russia, Moscow
| |
Collapse
|
113
|
Johnson DR, Diehn FE, Giannini C, Jenkins RB, Jenkins SM, Parney IF, Kaufmann TJ. Genetically Defined Oligodendroglioma Is Characterized by Indistinct Tumor Borders at MRI. AJNR Am J Neuroradiol 2017; 38:678-684. [PMID: 28126746 DOI: 10.3174/ajnr.a5070] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/10/2016] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE In 2016, the World Health Organization revised the brain tumor classification, making IDH mutation and 1p/19q codeletion the defining features of oligodendroglioma. To determine whether imaging characteristics previously associated with oligodendroglial tumors are still applicable, we evaluated the MR imaging features of genetically defined oligodendrogliomas. MATERIALS AND METHODS One hundred forty-eight adult patients with untreated World Health Organization grade II and III infiltrating gliomas with histologic oligodendroglial morphology, known 1p/19q status, and at least 1 preoperative MR imaging were retrospectively identified. The association of 1p/19q codeletion with tumor imaging characteristics and ADC values was evaluated. RESULTS Ninety of 148 (61%) patients had 1p/19q codeleted tumors, corresponding to genetically defined oligodendroglioma, and 58/148 (39%) did not show 1p/19q codeletion, corresponding to astrocytic tumors. Eighty-three of 90 (92%) genetically defined oligodendrogliomas had noncircumscribed borders, compared with 26/58 (45%) non-1p/19q codeleted tumors with at least partial histologic oligodendroglial morphology (P < .0001). Eighty-nine of 90 (99%) oligodendrogliomas were heterogeneous on T1- and/or T2-weighted imaging. In patients with available ADC values, a lower mean ADC value predicted 1p/19q codeletion (P = .0005). CONCLUSIONS Imaging characteristics of World Health Organization 2016 genetically defined oligodendrogliomas differ from the previously considered characteristics of morphologically defined oligodendrogliomas. We found that genetically defined oligodendrogliomas were commonly poorly circumscribed and were almost always heterogeneous in signal intensity.
Collapse
Affiliation(s)
- D R Johnson
- From the Departments of Radiology (D.R.J., F.E.D., T.J.K.)
| | - F E Diehn
- From the Departments of Radiology (D.R.J., F.E.D., T.J.K.)
| | | | | | | | - I F Parney
- Neurosurgery (I.F.P.), Mayo Clinic, Rochester, Minnesota
| | - T J Kaufmann
- From the Departments of Radiology (D.R.J., F.E.D., T.J.K.)
| |
Collapse
|
114
|
Ryzhova MV, Shaykhaev EG, Kazarova MV, Telysheva EN, Shishkina LV, Shibaeva IV, Shugay SV, Voronina EI, Snigireva GP. [The spectrum of genetic alterations in anaplastic gliomas: and anaplastic oligodendrogliomas]. ZHURNAL VOPROSY NEIROKHIRURGII IMENI N. N. BURDENKO 2017; 81:26-31. [PMID: 29393283 DOI: 10.17116/neiro201781626-31] [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: 06/07/2023]
Abstract
The work explores the molecular genetic features of anaplastic astrocytomas and oligodendrogliomas in a series of 43 cases. The mutational status was studied using domestic chemicals and reagent kits. We revealed clear genetic differences between astrocytic and oligodendroglial tumors and proposed an algorithm to study diagnostic and prognostic markers.
Collapse
Affiliation(s)
- M V Ryzhova
- Burdenko Neurosurgical Institute, Moscow, Russia, 125047
| | - E G Shaykhaev
- Russian Scientific Center of Roentgen Radiology, Moscow, Russia, 117997
| | - M V Kazarova
- Russian Scientific Center of Roentgen Radiology, Moscow, Russia, 117997
| | - E N Telysheva
- Russian Scientific Center of Roentgen Radiology, Moscow, Russia, 117997
| | - L V Shishkina
- Burdenko Neurosurgical Institute, Moscow, Russia, 125047
| | - I V Shibaeva
- Burdenko Neurosurgical Institute, Moscow, Russia, 125047
| | - S V Shugay
- Burdenko Neurosurgical Institute, Moscow, Russia, 125047
| | - E I Voronina
- Novosibirsk State Medical University, Novosibirsk, Russia, 630091
| | - G P Snigireva
- Russian Scientific Center of Roentgen Radiology, Moscow, Russia, 117997
| |
Collapse
|
115
|
Zhang B, Chang K, Ramkissoon S, Tanguturi S, Bi WL, Reardon DA, Ligon KL, Alexander BM, Wen PY, Huang RY. Multimodal MRI features predict isocitrate dehydrogenase genotype in high-grade gliomas. Neuro Oncol 2017; 19:109-117. [PMID: 27353503 PMCID: PMC5193019 DOI: 10.1093/neuonc/now121] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND High-grade gliomas with mutations in the isocitrate dehydrogenase (IDH) gene family confer longer overall survival relative to their IDH-wild-type counterparts. Accurate determination of the IDH genotype preoperatively may have both prognostic and diagnostic value. The current study used a machine-learning algorithm to generate a model predictive of IDH genotype in high-grade gliomas based on clinical variables and multimodal features extracted from conventional MRI. METHODS Preoperative MRIs were obtained for 120 patients with primary grades III (n = 35) and IV (n = 85) glioma in this retrospective study. IDH genotype was confirmed for grade III (32/35, 91%) and IV (22/85, 26%) tumors by immunohistochemistry, spectrometry-based mutation genotyping (OncoMap), or multiplex exome sequencing (OncoPanel). IDH1 and IDH2 mutations were mutually exclusive, and all mutated tumors were collapsed into one IDH-mutated cohort. Cases were randomly assigned to either the training (n = 90) or validation cohort (n = 30). A total of 2970 imaging features were extracted from pre- and postcontrast T1-weighted, T2-weighted, and apparent diffusion coefficient map. Using a random forest algorithm, nonredundant features were integrated with clinical data to generate a model predictive of IDH genotype. RESULTS Our model achieved accuracies of 86% (area under the curve [AUC] = 0.8830) in the training cohort and 89% (AUC = 0.9231) in the validation cohort. Features with the highest predictive value included patient age as well as parametric intensity, texture, and shape features. CONCLUSION Using a machine-learning algorithm, we achieved accurate prediction of IDH genotype in high-grade gliomas with preoperative clinical and MRI features.
Collapse
Affiliation(s)
- Biqi Zhang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Ken Chang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Shakti Ramkissoon
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Shyam Tanguturi
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Wenya Linda Bi
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - David A Reardon
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Keith L Ligon
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Brian M Alexander
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Patrick Y Wen
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| |
Collapse
|
116
|
Mohamed Yusoff AA, Zulfakhar FN, Sul’ain MD, Idris Z, Abdullah JM. Association of The IDH1 C.395G>A (R132H) Mutation with Histological Type in Malay Brain Tumors. Asian Pac J Cancer Prev 2016; 17:5195-5201. [PMID: 28125199 PMCID: PMC5454658 DOI: 10.22034/apjcp.2016.17.12.5195] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Brain tumors, constituting one of the most deadly forms of cancer worldwide, result from the accumulation of multiple genetic and epigenetic alterations in genes and signaling pathways. Isocitrate dehydrogenase enzyme isoform 1 (IDH1) mutations are frequently identified in primary brain tumors and acute myeloid leukemia. Studies on IDH1 gene mutations have been extensively performed in various populations worldwide but not in Malaysia. This work was conducted to study the prevalence of IDH1 c.395G>A (R132H) hotspot mutations in a group of Malaysian patients with brain tumors in order to gain local data for the IDH1 mutation profile in our population. Methods: Mutation analysis of c.395G>A (R132H) of IDH1 was performed in 40 brain tumor specimens by the polymerase chain reaction-restriction fragment length polymorphism method (PCR-RFLP) and then verified by direct sequencing. Associations between the IDH1 c.395G>A (R132H) mutation and clinicopathologic characteristics were also analyzed. Results: The IDH1 c.395G>A (R132H) mutation was detected in 14/40 patients (35%). A significant association was found with histological tumor types, but not with age, gender and race. Conclusions: IDH1 is frequently mutated and associated with histological subtypes in Malay brain tumors.
Collapse
Affiliation(s)
- Abdul Aziz Mohamed Yusoff
- Department of Neurosciences School of Medical Sciences Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan MALAYSIA.
| | | | | | | | | |
Collapse
|
117
|
Frontal glioblastoma multiforme may be biologically distinct from non-frontal and multilobar tumors. J Clin Neurosci 2016; 34:128-132. [PMID: 27593971 DOI: 10.1016/j.jocn.2016.05.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/20/2016] [Indexed: 11/21/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults and carries a grim prognosis. Lobar GBM, notably those localized to the frontal lobe, are generally more amenable to complete surgical resection, and may carry a better prognosis. The biology of differently localized GBM has been reported scarcely in terms of prognostic markers, including isocitrate dehydrogenase 1 (IDH1) mutation and O(6)-methylguanine-methyltransferase (MGMT) methylation. To our knowledge, there has been no evaluation in the literature of different proliferation indexes in different GBM locations in the brain. We performed a retrospective evaluation of our prospectively collected database to assess the rate of IDH1 positivity, MGMT methylation and Ki67 index for GBM located in the frontal lobes alone, lobar GBM in other supra-tentorial lobes and multilobar GBM. IDH1 mutated tumors were localized in the frontal lobes in 50.0%, whereas only 20.3% of IDH1 wild-type tumors were localized in the frontal lobe (p=0.006); MGMT methylated tumors were localized in the frontal lobe in 32.0% of the cases. Only 13.75% of the MGMT unmethylated tumors were localized to the frontal lobe (p=0.005); Tumors with higher Ki67 proliferation index were more likely to be localized in the frontal lobe (40.6% vs. 19.5%, p=0.019). This is the largest cohort of GBM assessed for these purposes in the literature. Frontal lobe GBMs may be intrinsically biologically distinct from GBM in other lobes and from multilobar tumors.
Collapse
|
118
|
Paldor I, Drummond KJ, Kaye AH. IDH1 mutation may not be prognostically favorable in glioblastoma when controlled for tumor location: A case-control study. J Clin Neurosci 2016; 34:117-120. [PMID: 27522495 DOI: 10.1016/j.jocn.2016.05.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 05/18/2016] [Indexed: 11/25/2022]
Abstract
Isocitrate dehydrogenase 1 (IDH1) mutation is a known prognostic factor in glioblastoma multiforme (GBM). It has been well documented that patients with IDH1 mutant (IDH1-mu) GBM have a better outcome compared to patients with IDH1 wild-type (IDH1-WT) GBM. IDH1-mu tumors have been shown to be more commonly located in the frontal lobe, and less likely to be in multiple lobes. It is unclear whether differential location is part of the prognostically favorable profile of these tumors. We performed a case-control study, matching IDH1-mu GBMs to IDH1-WT GBMs that are controlled for age, sex and tumor location. There were 21 IDH1-mu tumors and 21 matched IDH1-WT tumors. Age, sex and tumor location were matched between the two groups. After controlling for the factors described, the IDH1-mu tumors were more likely to be secondary GBM (61.9% secondary vs. 14.3%, p=0.004). There was an insignificant trend towards smaller tumor volume in the IDH1-mu group (28.13±6.56 vs. 41.8±7.33 cm3, p=0.173). Extent of surgical resection was similar in both groups (mean 84.49% vs. 89.89%, p=0.419). There was no survival advantage for IDH1-mu tumors when controlled for location: 25.2months overall survival for IDH1-mu patients and 23.6 for IDH1-WT patients, p=0.794. IDH1 mutation may provide part of its prognostic significance by differential localization of tumor, both making IDH1-mu tumors more amenable to gross total resection and placing these tumors in less eloquent areas, thereby lowering neurological morbidity.
Collapse
Affiliation(s)
- Iddo Paldor
- Department of Neurosurgery, The Royal Melbourne Hospital, 300 Grattan Street, Parkville, VIC 3052, Australia.
| | - Katharine J Drummond
- Department of Neurosurgery, The Royal Melbourne Hospital, 300 Grattan Street, Parkville, VIC 3052, Australia; Department of Surgery, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew H Kaye
- Department of Surgery, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
119
|
Impact of gross total resection in patients with WHO grade III glioma harboring the IDH 1/2 mutation without the 1p/19q co-deletion. J Neurooncol 2016; 129:505-514. [DOI: 10.1007/s11060-016-2201-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 07/04/2016] [Indexed: 10/21/2022]
|
120
|
Hong CS, Hsieh JK, Edwards NA, Ray-Chaudhury A, Zaghloul KA. IDH mutations may not preclude distant, trans-tentorial spread in gliomas: a case report and review of the literature. World J Surg Oncol 2016; 14:53. [PMID: 26911558 PMCID: PMC4766660 DOI: 10.1186/s12957-016-0814-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 02/17/2016] [Indexed: 11/17/2022] Open
Abstract
Background IDH mutations have been demonstrated to confer prolonged survival in patients suffering from gliomas, but the mechanisms underlying the improved prognosis are unclear. While some studies have attributed these observations to an enhanced sensitivity to genotoxic therapies, others have postulated that IDH-mutated gliomas exhibit less aggressive intrinsic biological behavior, including the propensity to invade distant sites. Although most gliomas recur local to the site of initial presentation, some tumors demonstrate distant recurrence, the vast majority of which involve the contralateral hemisphere. Trans-tentorial spread has been described once before, in which a supratentorial glioblastoma was reported to recur infratentorially in the cerebellum. Case presentation We describe a patient who underwent surgical resection, followed by adjuvant radiation and temozolomide of a World Health Organization (WHO) III anaplastic astrocytoma in the right temporal lobe, exhibiting an IDH1 (R132H) mutation. Twenty-two months after surgery, he developed a second lesion, located in the right cerebellum, suspicious for recurrent tumor versus radiation necrosis. A second surgery was performed, and pathology demonstrated recurrent tumor, consistent with IDH1-mutated anaplastic astrocytoma. Conclusions This is the first example of trans-tentorial spread in an IDH-mutated glioma, suggesting that despite improved survival, IDH mutations may not preclude gliomas from exhibiting the ability to invade distant sites of the brain.
Collapse
Affiliation(s)
- Christopher S Hong
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Building 10, Room 3D20, Bethesda, MD, 20892, USA.,The Ohio State University College of Medicine, 410 W. 10th Avenue, Columbus, OH, 43210, USA
| | - Jason K Hsieh
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Building 10, Room 3D20, Bethesda, MD, 20892, USA.,Cleveland Clinic Lerner College of Medicine, 9500 Euclid Ave./NA21, Cleveland, OH, 44195, USA
| | - Nancy A Edwards
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Building 10, Room 3D20, Bethesda, MD, 20892, USA
| | - Abhik Ray-Chaudhury
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Building 10, Room 3D20, Bethesda, MD, 20892, USA
| | - Kareem A Zaghloul
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Building 10, Room 3D20, Bethesda, MD, 20892, USA.
| |
Collapse
|
121
|
Abstract
Imaging is integral to the management of patients with brain tumors. Conventional structural imaging provides exquisite anatomic detail but remains limited in the evaluation of molecular characteristics of intracranial neoplasms. Quantitative and physiologic biomarkers derived from advanced imaging techniques have been increasingly utilized as problem-solving tools to identify glioma grade and assess response to therapy. This chapter provides a comprehensive overview of the imaging strategies used in the clinical assessment of patients with gliomas and describes how novel imaging biomarkers have the potential to improve patient management.
Collapse
Affiliation(s)
- Whitney B Pope
- Radiological Sciences, Ronald Reagan Medical Center, Los Angeles, CA, USA.
| | - Ibrahim Djoukhadar
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Alan Jackson
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| |
Collapse
|
122
|
Fan X, Wang Y, Wang K, Liu S, Liu Y, Ma J, Li S, Jiang T. Anatomical specificity of vascular endothelial growth factor expression in glioblastomas: a voxel-based mapping analysis. Neuroradiology 2015; 58:69-75. [PMID: 26453565 DOI: 10.1007/s00234-015-1602-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/28/2015] [Indexed: 10/22/2022]
Abstract
INTRODUCTION The expression of vascular endothelial growth factor (VEGF) is a common genetic alteration in malignant gliomas and contributes to the angiogenesis of tumors. This study aimed to investigate the anatomical specificity of VEGF expression levels in glioblastomas using voxel-based neuroimaging analysis. METHODS Clinical information, MR scans, and immunohistochemistry stains of 209 patients with glioblastomas were reviewed. All tumor lesions were segmented manually and subsequently registered to standard brain space. Voxel-based regression analysis was performed to correlate the brain regions of tumor involvement with the level of VEGF expression. Brain regions identified as significantly associated with high or low VEGF expression were preserved following permutation correction. RESULTS High VEGF expression was detected in 123 (58.9 %) of the 209 patients. Voxel-based statistical analysis demonstrated that high VEGF expression was more likely in tumors located in the left frontal lobe and the right caudate and low VEGF expression was more likely in tumors that occurred in the posterior region of the right lateral ventricle. CONCLUSION Voxel-based neuroimaging analysis revealed the anatomic specificity of VEGF expression in glioblastoma, which may further our understanding of genetic heterogeneity during tumor origination. This finding provides primary theoretical support for potential future application of customized antiangiogenic therapy.
Collapse
Affiliation(s)
- Xing Fan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 6 Tiantan Xili, Dongcheng District, Beijing, 100050, People's Republic of China
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 6 Tiantan Xili, Dongcheng District, Beijing, 100050, People's Republic of China.,Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, People's Republic of China
| | - Kai Wang
- Department of Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Shuai Liu
- Departments of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yong Liu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Jun Ma
- Department of Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Shaowu Li
- Department of Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 6 Tiantan Xili, Dongcheng District, Beijing, 100050, People's Republic of China. .,Department of Clinical Oncology, Beijing Academy of Critical Illness in Brain, Beijing, People's Republic of China.
| |
Collapse
|
123
|
Wang K, Wang Y, Fan X, Wang J, Li G, Ma J, Ma J, Jiang T, Dai J. Radiological features combined with IDH1 status for predicting the survival outcome of glioblastoma patients. Neuro Oncol 2015; 18:589-97. [PMID: 26409566 DOI: 10.1093/neuonc/nov239] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 08/24/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Radiological characteristics may reflect the biological features of brain tumors and may be associated with genetic alterations that occur in tumorigenesis. This study aimed to investigate the relationship between radiological features and IDH1 status as well as their predictive value for survival of glioblastoma patients. METHODS The clinical information and MR images of 280 patients with histologically confirmed glioblastoma were retrospectively reviewed. The radiological characteristics of tumors were examined on MR images, and the IDH1 status was determined using DNA sequencing for all cases. The Kaplan-Meier method and Cox regression model were used to identify prognostic factors for progression-free and overall survival. RESULTS The IDH1 mutation was associated with longer progression-free survival (P = .022; hazard ratio, 0.602) and overall survival (P = .018; hazard ratio, 0.554). In patients with the IDH1 mutation, tumor contrast enhancement and peritumoral edema indicated worse progression-free survival (P = .015 and P = .024, respectively) and worse overall survival (P = .024 and P = .032, respectively). For tumors with contrast enhancement, multifocal contrast enhancement of the tumor lesion was associated with poor progression-free survival (P = .002) and poor overall survival (P = .010) in patients with wild-type IDH1 tumors. CONCLUSIONS Combining the radiological features and IDH1 status of a tumor allows more accurate prediction of survival outcomes in glioblastoma patients. The complementary roles of genetic changes and radiological features of tumors should be considered in future studies.
Collapse
Affiliation(s)
- Kai Wang
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (K.W., J.M., J.M., J.D.); Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (Y.W., X.F., J.W., T.J.); Department of Pathology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (G.L.); Beijing Neurosurgical Institute, Capital Medical University, Beijing, China (Y.W., X.F., T.J., J.D.); Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China (T.J.)
| | - Yinyan Wang
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (K.W., J.M., J.M., J.D.); Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (Y.W., X.F., J.W., T.J.); Department of Pathology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (G.L.); Beijing Neurosurgical Institute, Capital Medical University, Beijing, China (Y.W., X.F., T.J., J.D.); Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China (T.J.)
| | - Xing Fan
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (K.W., J.M., J.M., J.D.); Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (Y.W., X.F., J.W., T.J.); Department of Pathology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (G.L.); Beijing Neurosurgical Institute, Capital Medical University, Beijing, China (Y.W., X.F., T.J., J.D.); Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China (T.J.)
| | - Jiangfei Wang
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (K.W., J.M., J.M., J.D.); Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (Y.W., X.F., J.W., T.J.); Department of Pathology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (G.L.); Beijing Neurosurgical Institute, Capital Medical University, Beijing, China (Y.W., X.F., T.J., J.D.); Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China (T.J.)
| | - Guilin Li
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (K.W., J.M., J.M., J.D.); Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (Y.W., X.F., J.W., T.J.); Department of Pathology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (G.L.); Beijing Neurosurgical Institute, Capital Medical University, Beijing, China (Y.W., X.F., T.J., J.D.); Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China (T.J.)
| | - Jieling Ma
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (K.W., J.M., J.M., J.D.); Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (Y.W., X.F., J.W., T.J.); Department of Pathology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (G.L.); Beijing Neurosurgical Institute, Capital Medical University, Beijing, China (Y.W., X.F., T.J., J.D.); Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China (T.J.)
| | - Jun Ma
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (K.W., J.M., J.M., J.D.); Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (Y.W., X.F., J.W., T.J.); Department of Pathology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (G.L.); Beijing Neurosurgical Institute, Capital Medical University, Beijing, China (Y.W., X.F., T.J., J.D.); Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China (T.J.)
| | - Tao Jiang
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (K.W., J.M., J.M., J.D.); Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (Y.W., X.F., J.W., T.J.); Department of Pathology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (G.L.); Beijing Neurosurgical Institute, Capital Medical University, Beijing, China (Y.W., X.F., T.J., J.D.); Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China (T.J.)
| | - Jianping Dai
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (K.W., J.M., J.M., J.D.); Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (Y.W., X.F., J.W., T.J.); Department of Pathology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China (G.L.); Beijing Neurosurgical Institute, Capital Medical University, Beijing, China (Y.W., X.F., T.J., J.D.); Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China (T.J.)
| |
Collapse
|
124
|
Sun ZL, Chan AKY, Chen LC, Tang C, Zhang ZY, Ding XJ, Wang Y, Sun CR, Ng HK, Yao Y, Zhou LF. TERT promoter mutated WHO grades II and III gliomas are located preferentially in the frontal lobe and avoid the midline. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:11485-11494. [PMID: 26617880 PMCID: PMC4637696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/28/2015] [Indexed: 06/05/2023]
Abstract
The promoter region of telomerase reverse transcriptase (TERTp) and isocitrate dehydrogenase (IDH) have been regarded as biomarkers with distinct clinical and phenotypic features. Investigated the possible correlations between tumor location and genetic alterations would enhance our understanding of gliomagenesis and heterogeneity of glioma. We examined mutations of TERTp and IDH by direct sequencing and fluorescence in-situ hybridization in a cohort of 225 grades II and III diffuse gliomas. Correlation analysis between molecular markers and tumor locations was performed by Chi-square tests/Fisher's exact test and multivariate logistic regression analysis. We found gliomas in frontal lobe showed higher frequency of TERTp mutation (P=0.0337) and simultaneously mutations of IDH and TERTp (IDH (mut)-TERTp(mut)) (P=0.0281) than frequency of biomarkers mutation of tumors in no-Frontal lobes, while lower frequency of TERTp mutation (P<0.0001) and simultaneously wild type of IDH and TERTp (IDH (wt)-TERTp(wt)) (P<0.0001) in midline than no-midline lobes. Logistic regression analysis indicated that locations of tumors associated with TERTp mutation (OR=0.540, 95% CI 0.324-0.900, P=0.018) and status of combinations of IDH and TERTp (IDH (mut)-TERTp (mut) vs. IDH (wt)-TERTp (wt) OR=0.162, 95% CI 0.075-0.350, P<0.001). In conclusion, grades II and III gliomas harboring TERTp mutation were located preferentially in the frontal lobe and rarely in midline. Association of IDH-TERTp status and tumor location suggests their potential values in molecular classification of grades II and III gliomas.
Collapse
Affiliation(s)
- Ze-Lin Sun
- Department of Neurosurgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
- Department of Neurosurgery, North China University of Science and Technology Affiliated HospitalTangshan 063000, Hebei Province, China
- Neurosurgical Immunology Laboratory of Huashan Hospital, Fudan UniversityShanghai, Chian
| | - Aden Ka-Yin Chan
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong KongHong Kong, China
| | - Ling-Chao Chen
- Department of Neurosurgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
- Neurosurgical Immunology Laboratory of Huashan Hospital, Fudan UniversityShanghai, Chian
| | - Chao Tang
- Department of Neurosurgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
- Neurosurgical Immunology Laboratory of Huashan Hospital, Fudan UniversityShanghai, Chian
| | - Zhen-Yu Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Xiao-Jie Ding
- Department of Neurosurgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Yang Wang
- Department of Neurosurgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
| | - Chong-Ran Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou 310000, China
| | - Ho-Keung Ng
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong KongHong Kong, China
| | - Yu Yao
- Department of Neurosurgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
- Neurosurgical Immunology Laboratory of Huashan Hospital, Fudan UniversityShanghai, Chian
| | - Liang-Fu Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan UniversityShanghai 200040, China
| |
Collapse
|
125
|
Wang YY, Wang K, Li SW, Wang JF, Ma J, Jiang T, Dai JP. Patterns of Tumor Contrast Enhancement Predict the Prognosis of Anaplastic Gliomas with IDH1 Mutation. AJNR Am J Neuroradiol 2015; 36:2023-9. [PMID: 26316565 DOI: 10.3174/ajnr.a4407] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/21/2015] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE It is proposed that isocitrate dehydrogenase 1 (IDH1) mutation predicts the outcome in patients with high-grade glioma. In addition, contrast enhancement on preoperative MR imaging reflects tumor biologic features. Patients with anaplastic glioma with the IDH1 mutation were evaluated by using MR imaging to determine whether tumor enhancement is a prognostic factor and can be used to predict survival. MATERIALS AND METHODS A cohort of 216 patients with histologically confirmed anaplastic glioma was reviewed retrospectively. Tumor contrast-enhancement patterns were classified on the basis of preoperative T1 contrast MR images. Tumor IDH1 status was examined by using RNA sequencing. We used univariate analysis and the multivariate Cox model to evaluate the prognostic value of the IDH1 mutation and tumor contrast-enhancement pattern for progression-free survival and overall survival. RESULTS In all 216 patients, IDH1 mutation was associated with longer progression-free survival (P = .004, hazard ratio = 0.439) and overall survival (P = .002, hazard ratio = 0.406). For patients with IDH1 mutant anaplastic glioma, the absence of contrast enhancement was associated with longer progression-free survival (P = .038, hazard ratio = 0.473) and overall survival (P = .043, hazard ratio = 0.436). Furthermore, we were able to stratify the progression-free survival and overall survival of patients with IDH1 mutation by using the tumor contrast-enhancement patterns (P = .022 and 0.029, respectively; log-rank). CONCLUSIONS Tumor enhancement on postcontrast MR imaging is a valuable prognostic factor for patients with anaplastic glioma and IDH1 mutation. Furthermore, the contrast-enhancement patterns could potentially be used to stratify the survival outcome of such patients.
Collapse
Affiliation(s)
- Y Y Wang
- From the Departments of Neurosurgery (Y.Y.W., J.F.W., T.J.) Beijing Neurosurgical Institute (Y.Y.W., T.J., J.P.D.), Capital Medical University, Beijing, China
| | - K Wang
- Neuroradiology (K.W., S.W.L., J.M., J.P.D.), Beijing Tian Tan Hospital
| | - S W Li
- Neuroradiology (K.W., S.W.L., J.M., J.P.D.), Beijing Tian Tan Hospital
| | - J F Wang
- From the Departments of Neurosurgery (Y.Y.W., J.F.W., T.J.)
| | - J Ma
- Neuroradiology (K.W., S.W.L., J.M., J.P.D.), Beijing Tian Tan Hospital
| | - T Jiang
- From the Departments of Neurosurgery (Y.Y.W., J.F.W., T.J.) Beijing Neurosurgical Institute (Y.Y.W., T.J., J.P.D.), Capital Medical University, Beijing, China Center for Brain Tumor (T.J.), Beijing Institute for Brain Disorders, Beijing, China.
| | - J P Dai
- Neuroradiology (K.W., S.W.L., J.M., J.P.D.), Beijing Tian Tan Hospital Beijing Neurosurgical Institute (Y.Y.W., T.J., J.P.D.), Capital Medical University, Beijing, China
| |
Collapse
|
126
|
Price SJ. Imaging Markers of Isocitrate Dehydrogenase-1 Mutations in Gliomas. Neurosurgery 2015; 62 Suppl 1:166-70. [DOI: 10.1227/neu.0000000000000812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Stephen J. Price
- Neurosurgery Division, Department of Clinical Neurosciences and Wolfson Brain Imaging Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
127
|
Genomic predictors of patterns of progression in glioblastoma and possible influences on radiation field design. J Neurooncol 2015; 124:447-53. [PMID: 26186902 DOI: 10.1007/s11060-015-1858-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/10/2015] [Indexed: 10/23/2022]
Abstract
We present a retrospective investigation of the role of genomics in the prediction of central versus marginal disease progression patterns for glioblastoma (GBM). Between August 2000 and May 2010, 41 patients with GBM and gene expression and methylation data available were treated with radiotherapy with or without concurrent temozolomide. Location of disease progression was categorized as within the high dose (60 Gy) or low dose (46 Gy) volume. Samples were grouped into previously described TCGA genomic groupings: Mesenchymal (m), classical (c), proneural (pn), and neural (n); and were also classified by MGMT-Methylation status and G-Cimp methylation phenotype. Genomic groupings and methylation status were investigated as a possible predictor of disease progression in the high dose region, progression in the low dose region, and time to progression. Based on TCGA category there was no difference in OS (p = 0.26), 60 Gy progression (PN: 71 %, N: 60 %, M: 89 %, C: 83 %, p = 0.19), 46 Gy progression (PN: 57 %, N: 40 %, M: 61 %,C: 50 %, p = 0.8) or time to progression (PN: 9 months, N:15 months, M: 9 months, C: 7 months, p = 0.58). MGMT methylation predicted for improved OS (median 25 vs. 13 months, p = 0.01), improved DFS (median 13 vs. 8 months, p = 0.007) and decreased 60 Gy (p = 0.003) and 46 Gy (p = 0.006) progression. There was a cohort of MGMT methylated patients with late marginal disease progression (4/22 patients, 18 %). TCGA groups demonstrated no difference in survival or progression patterns. MGMT methylation predicted for a statistically significant decrease in in-field and marginal disease progression. There was a cohort of MGMT methylated patients with late marginal progression. Validations of these findings would have implications that could affect radiation field size.
Collapse
|
128
|
Wang Y, Wang K, Li H, Wang J, Wang L, Dai J, Jiang T, Ma J. Identifying the association of contrast enhancement with vascular endothelia growth factor expression in anaplastic gliomas: a volumetric magnetic resonance imaging analysis. PLoS One 2015; 10:e0121380. [PMID: 25823012 PMCID: PMC4379034 DOI: 10.1371/journal.pone.0121380] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 01/31/2015] [Indexed: 01/31/2023] Open
Abstract
Contrast enhancement is a crucial radiologic feature of malignant brain tumors, which are associated with genetic changes of the tumor. The purpose of the current study was to investigate the potential relationship among tumor contrast enhancement with MR imaging, vascular endothelial growth factor (VEGF) expression, and survival outcome in anaplastic gliomas. MR images from 240 patients with histologically confirmed anaplastic gliomas were retrospectively analyzed. The volumes of T2 hyperintense, contrast enhanced regions and necrotic regions on postcontrast T1-weighted images were measured. The ratio of the enhanced volume to necrotic volume was compared between patients with high versus low levels of VEGF expression and was further used in the survival analysis. The volumetric ratio of enhancement to necrosis was significantly higher in patients with low VEGF expression than in those with high VEGF expression (Mann-Whitney, p = 0.009). In addition, the enhancement/necrosis ratio was identified as a significant predictor of progression-free survival (Cox regression model, p = 0.004) and overall survival (Cox regression model, p = 0.006) in the multivariate analysis. These results suggest that the volumetric ratio of enhancement to necrosis could serve as a noninvasive radiographic marker associated with VEGF expression and that this ratio is an independent predictor for progression-free survival and overall survival in patients with anaplastic gliomas.
Collapse
Affiliation(s)
- Yinyan Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Kai Wang
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Hongming Li
- Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jiangfei Wang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Lei Wang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jianping Dai
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- * E-mail: (JM); (TJ)
| | - Jun Ma
- Department of Neuroradiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- * E-mail: (JM); (TJ)
| |
Collapse
|
129
|
Association between molecular alterations and tumor location and MRI characteristics in anaplastic gliomas. Brain Tumor Pathol 2014; 32:99-104. [PMID: 25537428 DOI: 10.1007/s10014-014-0211-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 12/14/2014] [Indexed: 10/24/2022]
Abstract
The aim of this study was to determine if molecular alterations are associated with tumor location and radiological characteristics in anaplastic gliomas. We performed a retrospective analysis of 122 anaplastic gliomas for molecular alterations (IDH1/2 mutations, TP53 mutations, and 1p19q co-deletion) to compare MRI features (location and image characteristics). We observed that IDH mutation is strongly associated with frontal location (P = 0.001). However, 13 tumors not located in the cerebral cortex were IDH intact tumors (P < 0.0001). While IDH mutation and TP53 mutation are significantly associated with AA (p < 0.0001), IDH mutation and 1p19q co-deletion were significantly associated with AO/AOA (p < 0.0001). No tumors with IDH mutation and 1p19q co-deletion infiltrated the temporal lobe (P = 0.003). The tumors with 1p19q co-deletion and histologically diagnosed as AO/AOA were associated with contrast enhancement on MR images (p = 0.007, p = 0.002, respectively) and those with TP53 mutation had a weak association with sharp tumor borders (p = 0.043). MRI features might be useful to predict molecular profiles in anaplastic gliomas.
Collapse
|