101
|
Thuwajit C, Thuwajit P, Jamjantra P, Pairojkul C, Wongkham S, Bhudhisawasdi V, Ono J, Ohta S, Fujimoto K, Izuhara K. Clustering of patients with intrahepatic cholangiocarcinoma based on serum periostin may be predictive of prognosis. Oncol Lett 2017; 14:623-634. [PMID: 28693214 PMCID: PMC5494708 DOI: 10.3892/ol.2017.6250] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/03/2017] [Indexed: 12/13/2022] Open
Abstract
An effective serum biomarker may improve cholangiocarcinoma (CCA) management. Periostin (PN) has been demonstrated to be associated with aggressive CCA. The current study evaluated PN in blood serum for its diagnostic and prognostic potential in patients with CCA. Sera of 68 patients with CCA were collected prior to treatment, and PN levels were measured using an ELISA. Sera from 50 normal controls, 6 patients with benign liver diseases, 2 with hepatocellular carcinoma and 21 with breast cancer were analyzed. Immunohistochemistry of PN in CCA tissues was also investigated. The data were analyzed using the Mann-Whitney U test, Kaplan-Meier log rank tests, Cox proportional hazard regression models and Fisher's exact tests. The median serum PN level in patients with CCA was significantly increased compared with that in healthy controls, patients with benign liver diseases and patients with breast cancer (all P<0.05). Using an optimal threshold value of 94 ng/ml PN, the diagnostic values for CCA compared with other conditions demonstrated a sensitivity level of 0.38 [95% confidence interval (CI), 0.27-0.51], specificity of 0.90 (95% CI, 0.81-0.96), accuracy of 0.66 (95% CI, 0.58-0.74), positive predictive value of 0.76 (95% CI, 0.59-0.89) and negative predictive value of 0.63 (95% CI, 0.53-0.72) (P<0.001). Furthermore, PN stain in stromal fibroblasts in CCA tissues was associated with serum PN levels (P=0.001), and patients with CCA were classified as low (≤94 ng/ml) or high PN (>94 ng/ml) accordingly. High serum and tissue PN levels were significantly associated with reduced survival rate (P<0.001 and P=0.033, respectively). Serum PN was an independent prognostic factor with a hazard ratio of 3.197 (P=0.001). In conclusion, serum PN may be used to divide patients with intrahepatic CCA into high and low PN groups. Elevated serum PN may be utilized as a marker of poor prognosis in patients with CCA.
Collapse
Affiliation(s)
- Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-Noi, Bangkok 10700, Thailand
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-Noi, Bangkok 10700, Thailand
| | - Pranisa Jamjantra
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-Noi, Bangkok 10700, Thailand
| | - Chawalit Pairojkul
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Khon Kaen 40002, Thailand
| | - Sopit Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Khon Kaen 40002, Thailand
| | | | - Junya Ono
- Research and Development Unit, Shino-Test Corporation, Minami-Ku, Sagamihara, Kanagawa 252-0331, Japan
| | - Shoichiro Ohta
- Department of Biomolecular Sciences, Saga Medical School, Saga 849-8501, Japan
| | - Kiminori Fujimoto
- Department of Radiology, Kurume University School of Medicine and Center for Diagnostic Imaging, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Kenji Izuhara
- Department of Biomolecular Sciences, Saga Medical School, Saga 849-8501, Japan
| |
Collapse
|
102
|
Radiogenomics of lower-grade glioma: algorithmically-assessed tumor shape is associated with tumor genomic subtypes and patient outcomes in a multi-institutional study with The Cancer Genome Atlas data. J Neurooncol 2017; 133:27-35. [DOI: 10.1007/s11060-017-2420-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 04/09/2017] [Indexed: 01/15/2023]
|
103
|
Giardino A, Gupta S, Olson E, Sepulveda K, Lenchik L, Ivanidze J, Rakow-Penner R, Patel MJ, Subramaniam RM, Ganeshan D. Role of Imaging in the Era of Precision Medicine. Acad Radiol 2017; 24:639-649. [PMID: 28131497 DOI: 10.1016/j.acra.2016.11.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/07/2016] [Accepted: 11/29/2016] [Indexed: 12/17/2022]
Abstract
Precision medicine is an emerging approach for treating medical disorders, which takes into account individual variability in genetic and environmental factors. Preventive or therapeutic interventions can then be directed to those who will benefit most from targeted interventions, thereby maximizing benefits and minimizing costs and complications. Precision medicine is gaining increasing recognition by clinicians, healthcare systems, pharmaceutical companies, patients, and the government. Imaging plays a critical role in precision medicine including screening, early diagnosis, guiding treatment, evaluating response to therapy, and assessing likelihood of disease recurrence. The Association of University Radiologists Radiology Research Alliance Precision Imaging Task Force convened to explore the current and future role of imaging in the era of precision medicine and summarized its finding in this article. We review the increasingly important role of imaging in various oncological and non-oncological disorders. We also highlight the challenges for radiology in the era of precision medicine.
Collapse
Affiliation(s)
- Angela Giardino
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Supriya Gupta
- Department of Radiology and Imaging, Medical College of Georgia, 1120 15th St, Augusta, GA 30912.
| | - Emmi Olson
- Radiology Resident, University of California San Diego, San Diego, California
| | | | - Leon Lenchik
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jana Ivanidze
- Department of Diagnostic Radiology, Weill Cornell Medicine, New York, New York
| | - Rebecca Rakow-Penner
- Department of Radiology, University of California San Diego, San Diego, California
| | - Midhir J Patel
- Department of Radiology, University of South Florida, Tampa, Florida
| | - Rathan M Subramaniam
- Cyclotron and Molecular Imaging Program, Department of Radiology, UT Southwestern Medical Center, Dallas, Texas
| | | |
Collapse
|
104
|
Incoronato M, Aiello M, Infante T, Cavaliere C, Grimaldi AM, Mirabelli P, Monti S, Salvatore M. Radiogenomic Analysis of Oncological Data: A Technical Survey. Int J Mol Sci 2017; 18:ijms18040805. [PMID: 28417933 PMCID: PMC5412389 DOI: 10.3390/ijms18040805] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 12/18/2022] Open
Abstract
In the last few years, biomedical research has been boosted by the technological development of analytical instrumentation generating a large volume of data. Such information has increased in complexity from basic (i.e., blood samples) to extensive sets encompassing many aspects of a subject phenotype, and now rapidly extending into genetic and, more recently, radiomic information. Radiogenomics integrates both aspects, investigating the relationship between imaging features and gene expression. From a methodological point of view, radiogenomics takes advantage of non-conventional data analysis techniques that reveal meaningful information for decision-support in cancer diagnosis and treatment. This survey is aimed to review the state-of-the-art techniques employed in radiomics and genomics with special focus on analysis methods based on molecular and multimodal probes. The impact of single and combined techniques will be discussed in light of their suitability in correlation and predictive studies of specific oncologic diseases.
Collapse
Affiliation(s)
| | - Marco Aiello
- IRCCS SDN, Via E. Gianturco, 113, 80143 Naples, Italy.
| | | | | | | | | | - Serena Monti
- IRCCS SDN, Via E. Gianturco, 113, 80143 Naples, Italy.
| | | |
Collapse
|
105
|
Lee M, Woo B, Kuo MD, Jamshidi N, Kim JH. Quality of Radiomic Features in Glioblastoma Multiforme: Impact of Semi-Automated Tumor Segmentation Software. Korean J Radiol 2017; 18:498-509. [PMID: 28458602 PMCID: PMC5390619 DOI: 10.3348/kjr.2017.18.3.498] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 12/27/2016] [Indexed: 12/03/2022] Open
Abstract
Objective The purpose of this study was to evaluate the reliability and quality of radiomic features in glioblastoma multiforme (GBM) derived from tumor volumes obtained with semi-automated tumor segmentation software. Materials and Methods MR images of 45 GBM patients (29 males, 16 females) were downloaded from The Cancer Imaging Archive, in which post-contrast T1-weighted imaging and fluid-attenuated inversion recovery MR sequences were used. Two raters independently segmented the tumors using two semi-automated segmentation tools (TumorPrism3D and 3D Slicer). Regions of interest corresponding to contrast-enhancing lesion, necrotic portions, and non-enhancing T2 high signal intensity component were segmented for each tumor. A total of 180 imaging features were extracted, and their quality was evaluated in terms of stability, normalized dynamic range (NDR), and redundancy, using intra-class correlation coefficients, cluster consensus, and Rand Statistic. Results Our study results showed that most of the radiomic features in GBM were highly stable. Over 90% of 180 features showed good stability (intra-class correlation coefficient [ICC] ≥ 0.8), whereas only 7 features were of poor stability (ICC < 0.5). Most first order statistics and morphometric features showed moderate-to-high NDR (4 > NDR ≥1), while above 35% of the texture features showed poor NDR (< 1). Features were shown to cluster into only 5 groups, indicating that they were highly redundant. Conclusion The use of semi-automated software tools provided sufficiently reliable tumor segmentation and feature stability; thus helping to overcome the inherent inter-rater and intra-rater variability of user intervention. However, certain aspects of feature quality, including NDR and redundancy, need to be assessed for determination of representative signature features before further development of radiomics.
Collapse
Affiliation(s)
- Myungeun Lee
- Center for Medical-IT Convergence Technology Research, Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Korea.,Department of Radiology, Seoul National University Hospital, Seoul 03080, Korea
| | - Boyeong Woo
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Suwon 16229, Korea
| | - Michael D Kuo
- Department of Electronic and Computer Engineering, National Chiao Tung University, Hsinchu 300, Taiwan.,Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Neema Jamshidi
- Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jong Hyo Kim
- Center for Medical-IT Convergence Technology Research, Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Korea.,Department of Radiology, Seoul National University Hospital, Seoul 03080, Korea.,Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Suwon 16229, Korea
| |
Collapse
|
106
|
Ellingson BM, Wen PY, Cloughesy TF. Modified Criteria for Radiographic Response Assessment in Glioblastoma Clinical Trials. Neurotherapeutics 2017; 14:307-320. [PMID: 28108885 PMCID: PMC5398984 DOI: 10.1007/s13311-016-0507-6] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Radiographic endpoints including response and progression are important for the evaluation of new glioblastoma therapies. The current RANO criteria was developed to overcome many of the challenges identified with previous guidelines for response assessment, however, significant challenges and limitations remain. The current recommendations build on the strengths of the current RANO criteria, while addressing many of these limitations. Modifications to the current RANO criteria include suggestions for volumetric response evaluation, use contrast enhanced T1 subtraction maps to increase lesion conspicuity, removal of qualitative non-enhancing tumor assessment requirements, use of the post-radiation time point as the baseline for newly diagnosed glioblastoma response assessment, and "treatment-agnostic" response assessment rubrics for identifying pseudoprogression, pseudoresponse, and a confirmed durable response in newly diagnosed and recurrent glioblastoma trials.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.
- Department of Radiological Sciences, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- UCLA Neuro-Oncology Program, University of California Los Angeles, Los Angeles, CA, USA.
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
107
|
Lee G, Lee HY, Ko ES, Jeong WK. Radiomics and imaging genomics in precision medicine. PRECISION AND FUTURE MEDICINE 2017. [DOI: 10.23838/pfm.2017.00101] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
108
|
Spatiotemporal genomic architecture informs precision oncology in glioblastoma. Nat Genet 2017; 49:594-599. [PMID: 28263318 DOI: 10.1038/ng.3806] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 02/10/2017] [Indexed: 12/13/2022]
Abstract
Precision medicine in cancer proposes that genomic characterization of tumors can inform personalized targeted therapies. However, this proposition is complicated by spatial and temporal heterogeneity. Here we study genomic and expression profiles across 127 multisector or longitudinal specimens from 52 individuals with glioblastoma (GBM). Using bulk and single-cell data, we find that samples from the same tumor mass share genomic and expression signatures, whereas geographically separated, multifocal tumors and/or long-term recurrent tumors are seeded from different clones. Chemical screening of patient-derived glioma cells (PDCs) shows that therapeutic response is associated with genetic similarity, and multifocal tumors that are enriched with PIK3CA mutations have a heterogeneous drug-response pattern. We show that targeting truncal events is more efficacious than targeting private events in reducing the tumor burden. In summary, this work demonstrates that evolutionary inference from integrated genomic analysis in multisector biopsies can inform targeted therapeutic interventions for patients with GBM.
Collapse
|
109
|
Kanas VG, Zacharaki EI, Thomas GA, Zinn PO, Megalooikonomou V, Colen RR. Learning MRI-based classification models for MGMT methylation status prediction in glioblastoma. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2017; 140:249-257. [PMID: 28254081 DOI: 10.1016/j.cmpb.2016.12.018] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 12/14/2016] [Accepted: 12/29/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND AND OBJECTIVE The O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation has been shown to be associated with improved outcomes in patients with glioblastoma (GBM) and may be a predictive marker of sensitivity to chemotherapy. However, determination of the MGMT promoter methylation status requires tissue obtained via surgical resection or biopsy. The aim of this study was to assess the ability of quantitative and qualitative imaging variables in predicting MGMT methylation status noninvasively. METHODS A retrospective analysis of MR images from GBM patients was conducted. Multivariate prediction models were obtained by machine-learning methods and tested on data from The Cancer Genome Atlas (TCGA) database. RESULTS The status of MGMT promoter methylation was predicted with an accuracy of up to 73.6%. Experimental analysis showed that the edema/necrosis volume ratio, tumor/necrosis volume ratio, edema volume, and tumor location and enhancement characteristics were the most significant variables in respect to the status of MGMT promoter methylation in GBM. CONCLUSIONS The obtained results provide further evidence of an association between standard preoperative MRI variables and MGMT methylation status in GBM.
Collapse
Affiliation(s)
- Vasileios G Kanas
- Department of Electrical and Computer Engineering, University of Patras, Patras, Greece; Department of Computer Engineering and Informatics, University of Patras, Patras, Greece
| | - Evangelia I Zacharaki
- Department of Computer Engineering and Informatics, University of Patras, Patras, Greece; Center for Visual Computing (CVC), CentraleSupélec, INRIA, Université Paris-Saclay, France.
| | - Ginu A Thomas
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pascal O Zinn
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | | | - Rivka R Colen
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
110
|
Cui Y, Ren S, Tha KK, Wu J, Shirato H, Li R. Volume of high-risk intratumoral subregions at multi-parametric MR imaging predicts overall survival and complements molecular analysis of glioblastoma. Eur Radiol 2017; 27:3583-3592. [PMID: 28168370 DOI: 10.1007/s00330-017-4751-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/10/2017] [Accepted: 01/16/2017] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To develop and validate a volume-based, quantitative imaging marker by integrating multi-parametric MR images for predicting glioblastoma survival, and to investigate its relationship and synergy with molecular characteristics. METHODS We retrospectively analysed 108 patients with primary glioblastoma. The discovery cohort consisted of 62 patients from the cancer genome atlas (TCGA). Another 46 patients comprising 30 from TCGA and 16 internally were used for independent validation. Based on integrated analyses of T1-weighted contrast-enhanced (T1-c) and diffusion-weighted MR images, we identified an intratumoral subregion with both high T1-c and low ADC, and accordingly defined a high-risk volume (HRV). We evaluated its prognostic value and biological significance with genomic data. RESULTS On both discovery and validation cohorts, HRV predicted overall survival (OS) (concordance index: 0.642 and 0.653, P < 0.001 and P = 0.038, respectively). HRV stratified patients within the proneural molecular subtype (log-rank P = 0.040, hazard ratio = 2.787). We observed different OS among patients depending on their MGMT methylation status and HRV (log-rank P = 0.011). Patients with unmethylated MGMT and high HRV had significantly shorter survival (median survival: 9.3 vs. 18.4 months, log-rank P = 0.002). CONCLUSION Volume of the high-risk intratumoral subregion identified on multi-parametric MRI predicts glioblastoma survival, and may provide complementary value to genomic information. KEY POINTS • High-risk volume (HRV) defined on multi-parametric MRI predicted GBM survival. • The proneural molecular subtype tended to harbour smaller HRV than other subtypes. • Patients with unmethylated MGMT and high HRV had significantly shorter survival. • HRV complements genomic information in predicting GBM survival.
Collapse
Affiliation(s)
- Yi Cui
- Department of Radiation Oncology, Stanford University, 1070 Arastradero Rd., Palo Alto, CA, 94304, USA. .,Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education, Hokkaido University, Hokkaido, Japan.
| | - Shangjie Ren
- School of Electrical Engineering and Automation, Tianjin University, Tianjin Shi, China
| | - Khin Khin Tha
- Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education, Hokkaido University, Hokkaido, Japan.,Department of Radiology and Nuclear Medicine, Hokkaido University, Hokkaido, Japan
| | - Jia Wu
- Department of Radiation Oncology, Stanford University, 1070 Arastradero Rd., Palo Alto, CA, 94304, USA
| | - Hiroki Shirato
- Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education, Hokkaido University, Hokkaido, Japan.,Department of Radiology and Nuclear Medicine, Hokkaido University, Hokkaido, Japan
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University, 1070 Arastradero Rd., Palo Alto, CA, 94304, USA.,Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education, Hokkaido University, Hokkaido, Japan
| |
Collapse
|
111
|
Abrol S, Kotrotsou A, Salem A, Zinn PO, Colen RR. Radiomic Phenotyping in Brain Cancer to Unravel Hidden Information in Medical Images. Top Magn Reson Imaging 2017; 26:43-53. [PMID: 28079714 DOI: 10.1097/rmr.0000000000000117] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Radiomics is a new area of research in the field of imaging with tremendous potential to unravel the hidden information in digital images. The scope of radiology has grown exponentially over the last two decades; since the advent of radiomics, many quantitative imaging features can now be extracted from medical images through high-throughput computing, and these can be converted into mineable data that can help in linking imaging phenotypes with clinical data, genomics, proteomics, and other "omics" information. In cancer, radiomic imaging analysis aims at extracting imaging features embedded in the imaging data, which can act as a guide in the disease or cancer diagnosis, staging and planning interventions for treating patients, monitor patients on therapy, predict treatment response, and determine patient outcomes.
Collapse
Affiliation(s)
- Srishti Abrol
- *Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center †Department of Neurosurgery, Baylor College of Medicine ‡Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | |
Collapse
|
112
|
Gevaert O, Echegaray S, Khuong A, Hoang CD, Shrager JB, Jensen KC, Berry GJ, Guo HH, Lau C, Plevritis SK, Rubin DL, Napel S, Leung AN. Predictive radiogenomics modeling of EGFR mutation status in lung cancer. Sci Rep 2017; 7:41674. [PMID: 28139704 PMCID: PMC5282551 DOI: 10.1038/srep41674] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 12/21/2016] [Indexed: 11/18/2022] Open
Abstract
Molecular analysis of the mutation status for EGFR and KRAS are now routine in the management of non-small cell lung cancer. Radiogenomics, the linking of medical images with the genomic properties of human tumors, provides exciting opportunities for non-invasive diagnostics and prognostics. We investigated whether EGFR and KRAS mutation status can be predicted using imaging data. To accomplish this, we studied 186 cases of NSCLC with preoperative thin-slice CT scans. A thoracic radiologist annotated 89 semantic image features of each patient’s tumor. Next, we built a decision tree to predict the presence of EGFR and KRAS mutations. We found a statistically significant model for predicting EGFR but not for KRAS mutations. The test set area under the ROC curve for predicting EGFR mutation status was 0.89. The final decision tree used four variables: emphysema, airway abnormality, the percentage of ground glass component and the type of tumor margin. The presence of either of the first two features predicts a wild type status for EGFR while the presence of any ground glass component indicates EGFR mutations. These results show the potential of quantitative imaging to predict molecular properties in a non-invasive manner, as CT imaging is more readily available than biopsies.
Collapse
Affiliation(s)
- Olivier Gevaert
- Stanford Center for Biomedical Informatics Research, Department of Medicine &Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | | | - Amanda Khuong
- Thoracic and GI Oncology Branch, CCR, National Institutes of Health, National Cancer Institute, Bethesda, MD, USA
| | - Chuong D Hoang
- Thoracic and GI Oncology Branch, CCR, National Institutes of Health, National Cancer Institute, Bethesda, MD, USA
| | - Joseph B Shrager
- Thoracic and GI Oncology Branch, CCR, National Institutes of Health, National Cancer Institute, Bethesda, MD, USA
| | - Kirstin C Jensen
- Department of Pathology, Stanford University Medical Center, Stanford, CA, USA.,Pathology and Laboratory Service of Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Gerald J Berry
- Department of Pathology, Stanford University Medical Center, Stanford, CA, USA
| | - H Henry Guo
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Charles Lau
- Department of Radiology, Stanford University, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | | | - Daniel L Rubin
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Sandy Napel
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Ann N Leung
- Department of Radiology, Stanford University, Stanford, CA, USA
| |
Collapse
|
113
|
Piazza M, Munasinghe J, Murayi R, Edwards N, Montgomery B, Walbridge S, Merrill M, Chittiboina P. Simulating vasogenic brain edema using chronic VEGF infusion. J Neurosurg 2017; 127:905-916. [PMID: 28059647 DOI: 10.3171/2016.9.jns1627] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To study peritumoral brain edema (PTBE), it is necessary to create a model that accurately simulates vasogenic brain edema (VBE) without introducing a complicated tumor environment. PTBE associated with brain tumors is predominantly a result of vascular endothelial growth factor (VEGF) secreted by brain tumors, and VEGF infusion alone can lead to histological blood-brain barrier (BBB) breakdown in the absence of tumor. VBE is intimately linked to BBB breakdown. The authors sought to establish a model for VBE with chronic infusion of VEGF that can be validated by serial in-vivo MRI and histological findings. METHODS Male Fischer rats (n = 182) underwent stereotactic striatal implantation of MRI-safe brain cannulas for chronic infusion of VEGF (2-20 µg/ml). Following a preinfusion phase (4-6 days), the rats were exposed to VEGF or control rat serum albumin (1.5 µl/hr) for as long as 144 hours. Serial MRI was performed during infusion on a high-field (9.4-T) machine at 12-24, 24-36, 48-72, and 120-144 hours. Rat brains were then collected and histological analysis was performed. RESULTS Control animals and animals infused with 2 µg/ml of VEGF experienced no neurological deficits, seizure activity, or abnormal behavior. Animals treated with VEGF demonstrated a significantly larger volume (42.90 ± 3.842 mm3) of T2 hyper-attenuation at 144 hours when compared with the volume (8.585 ± 1.664 mm3) in control animals (mean difference 34.31 ± 4.187 mm3, p < 0.0001, 95% CI 25.74-42.89 mm3). Postcontrast T1 enhancement in the juxtacanalicular region indicating BBB breakdown was observed in rats undergoing infusion with VEGF. At the later time periods (120-144 hrs) the volume of T1 enhancement (34.97 ± 8.99 mm3) was significantly less compared with the region of edema (p < 0.0001). Histologically, no evidence of necrosis or inflammation was observed with VEGF or control infusion. Immunohistochemical analysis demonstrated astrocyte activation, vascular remodeling, and increased claudin-5 expression in juxtacanalicular regions. Aquaporin-4 expression was increased in both control and VEGF animals in the juxtacanalicular regions. CONCLUSIONS The results of this study show that chronic brain infusion of VEGF creates a reliable model of VBE. This model lacks necrosis and inflammation that are characteristic of previous models of VBE. The model allows for a precise investigation into the mechanism of VBE formation. The authors also anticipate that this model will allow for investigation into the mechanism of glucocorticoid action in abrogating VBE, and to test novel therapeutic strategies targeting PTBE.
Collapse
Affiliation(s)
- Martin Piazza
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, and
| | | | - Roger Murayi
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, and
| | - Nancy Edwards
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, and
| | - Blake Montgomery
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, and
| | - Stuart Walbridge
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, and
| | - Marsha Merrill
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, and
| | - Prashant Chittiboina
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, and
| |
Collapse
|
114
|
Huynh E, Coroller TP, Narayan V, Agrawal V, Romano J, Franco I, Parmar C, Hou Y, Mak RH, Aerts HJWL. Associations of Radiomic Data Extracted from Static and Respiratory-Gated CT Scans with Disease Recurrence in Lung Cancer Patients Treated with SBRT. PLoS One 2017; 12:e0169172. [PMID: 28046060 PMCID: PMC5207741 DOI: 10.1371/journal.pone.0169172] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 12/13/2016] [Indexed: 02/06/2023] Open
Abstract
Radiomics aims to quantitatively capture the complex tumor phenotype contained in medical images to associate them with clinical outcomes. This study investigates the impact of different types of computed tomography (CT) images on the prognostic performance of radiomic features for disease recurrence in early stage non-small cell lung cancer (NSCLC) patients treated with stereotactic body radiation therapy (SBRT). 112 early stage NSCLC patients treated with SBRT that had static free breathing (FB) and average intensity projection (AIP) images were analyzed. Nineteen radiomic features were selected from each image type (FB or AIP) for analysis based on stability and variance. The selected FB and AIP radiomic feature sets had 6 common radiomic features between both image types and 13 unique features. The prognostic performances of the features for distant metastasis (DM) and locoregional recurrence (LRR) were evaluated using the concordance index (CI) and compared with two conventional features (tumor volume and maximum diameter). P-values were corrected for multiple testing using the false discovery rate procedure. None of the FB radiomic features were associated with DM, however, seven AIP radiomic features, that described tumor shape and heterogeneity, were (CI range: 0.638-0.676). Conventional features from FB images were not associated with DM, however, AIP conventional features were (CI range: 0.643-0.658). Radiomic and conventional multivariate models were compared between FB and AIP images using cross validation. The differences between the models were assessed using a permutation test. AIP radiomic multivariate models (median CI = 0.667) outperformed all other models (median CI range: 0.601-0.630) in predicting DM. None of the imaging features were prognostic of LRR. Therefore, image type impacts the performance of radiomic models in their association with disease recurrence. AIP images contained more information than FB images that were associated with disease recurrence in early stage NSCLC patients treated with SBRT, which suggests that AIP images may potentially be more optimal for the development of an imaging biomarker.
Collapse
Affiliation(s)
- Elizabeth Huynh
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
- * E-mail:
| | - Thibaud P. Coroller
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Vivek Narayan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Vishesh Agrawal
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - John Romano
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Idalid Franco
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Chintan Parmar
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Ying Hou
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Raymond H. Mak
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Hugo J. W. L. Aerts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
- Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| |
Collapse
|
115
|
Halpenny DF, Plodkowski A, Riely G, Zheng J, Litvak A, Moscowitz C, Ginsberg MS. Radiogenomic evaluation of lung cancer - Are there imaging characteristics associated with lung adenocarcinomas harboring BRAF mutations? Clin Imaging 2016; 42:147-151. [PMID: 28012356 DOI: 10.1016/j.clinimag.2016.11.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/09/2016] [Accepted: 11/23/2016] [Indexed: 02/06/2023]
Abstract
INTRODUCTION We studied computed tomography (CT) features associated with BRAF mutated lung cancer. MATERIALS AND METHODS CT features of BRAF mutated lung cancers were compared to stage matched lesions without BRAF mutation. RESULTS 47 (25%) patients with BRAF mutation and 141 (75%) without BRAF mutation were included. BRAF lesions were most frequently solid 37 (84%), spiculated 22 (50%), and peripheral 37 (84%). No feature of the primary tumor was significantly different between BRAF and non-BRAF groups. BRAF patients were more likely than KRAS patients to have pleural metastases [5 (11%) vs 0 (0%), p=0.045]. CONCLUSION No feature of the primary tumor differentiates BRAF lesions from non-BRAF lesions.
Collapse
Affiliation(s)
- Darragh F Halpenny
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Andrew Plodkowski
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Gregory Riely
- Department of Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Junting Zheng
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Anya Litvak
- Department of Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Chaya Moscowitz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Michelle S Ginsberg
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
116
|
Demerath T, Simon-Gabriel CP, Kellner E, Schwarzwald R, Lange T, Heiland DH, Reinacher P, Staszewski O, Mast H, Kiselev VG, Egger K, Urbach H, Weyerbrock A, Mader I. Mesoscopic imaging of glioblastomas: Are diffusion, perfusion and spectroscopic measures influenced by the radiogenetic phenotype? Neuroradiol J 2016; 30:36-47. [PMID: 27864578 DOI: 10.1177/1971400916678225] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The purpose of this study was to identify markers from perfusion, diffusion, and chemical shift imaging in glioblastomas (GBMs) and to correlate them with genetically determined and previously published patterns of structural magnetic resonance (MR) imaging. Twenty-six patients (mean age 60 years, 13 female) with GBM were investigated. Imaging consisted of native and contrast-enhanced 3D data, perfusion, diffusion, and spectroscopic imaging. In the presence of minor necrosis, cerebral blood volume (CBV) was higher (median ± SD, 2.23% ± 0.93) than in pronounced necrosis (1.02% ± 0.71), pcorr = 0.0003. CBV adjacent to peritumoral fluid-attenuated inversion recovery (FLAIR) hyperintensity was lower in edema (1.72% ± 0.31) than in infiltration (1.91% ± 0.35), pcorr = 0.039. Axial diffusivity adjacent to peritumoral FLAIR hyperintensity was lower in severe mass effect (1.08*10-3 mm2/s ± 0.08) than in mild mass effect (1.14*10-3 mm2/s ± 0.06), pcorr = 0.048. Myo-inositol was positively correlated with a marker for mitosis (Ki-67) in contrast-enhancing tumor, r = 0.5, pcorr = 0.0002. Changed CBV and axial diffusivity, even outside FLAIR hyperintensity, in adjacent normal-appearing matter can be discussed as to be related to angiogenesis pathways and to activated proliferation genes. The correlation between myo-inositol and Ki-67 might be attributed to its binding to cell surface receptors regulating tumorous proliferation of astrocytic cells.
Collapse
Affiliation(s)
- Theo Demerath
- 1 Department of Neuroradiology, Medical Centre-University of Freiburg, Germany.,2 Department of Radiology, University Medical Centre Basel, Switzerland.,3 Faculty of Medicine, University of Freiburg, Germany
| | - Carl Philipp Simon-Gabriel
- 1 Department of Neuroradiology, Medical Centre-University of Freiburg, Germany.,3 Faculty of Medicine, University of Freiburg, Germany
| | - Elias Kellner
- 3 Faculty of Medicine, University of Freiburg, Germany.,4 Medical Physics, Department of Radiology, Medical Centre-University of Freiburg, Germany
| | - Ralf Schwarzwald
- 1 Department of Neuroradiology, Medical Centre-University of Freiburg, Germany.,3 Faculty of Medicine, University of Freiburg, Germany
| | - Thomas Lange
- 3 Faculty of Medicine, University of Freiburg, Germany.,4 Medical Physics, Department of Radiology, Medical Centre-University of Freiburg, Germany
| | - Dieter Henrik Heiland
- 3 Faculty of Medicine, University of Freiburg, Germany.,5 Department of Neurosurgery, Medical Centre-University of Freiburg, Germany
| | - Peter Reinacher
- 3 Faculty of Medicine, University of Freiburg, Germany.,6 Department of Functional and Stereotactic Neurosurgery, Medical Centre-University of Freiburg, Germany
| | - Ori Staszewski
- 3 Faculty of Medicine, University of Freiburg, Germany.,7 Institute of Neuropathology, Medical Centre-University of Freiburg, Germany
| | - Hansjörg Mast
- 1 Department of Neuroradiology, Medical Centre-University of Freiburg, Germany.,3 Faculty of Medicine, University of Freiburg, Germany
| | - Valerij G Kiselev
- 3 Faculty of Medicine, University of Freiburg, Germany.,4 Medical Physics, Department of Radiology, Medical Centre-University of Freiburg, Germany
| | - Karl Egger
- 1 Department of Neuroradiology, Medical Centre-University of Freiburg, Germany.,3 Faculty of Medicine, University of Freiburg, Germany
| | - Horst Urbach
- 1 Department of Neuroradiology, Medical Centre-University of Freiburg, Germany.,3 Faculty of Medicine, University of Freiburg, Germany
| | - Astrid Weyerbrock
- 3 Faculty of Medicine, University of Freiburg, Germany.,5 Department of Neurosurgery, Medical Centre-University of Freiburg, Germany
| | - Irina Mader
- 1 Department of Neuroradiology, Medical Centre-University of Freiburg, Germany.,3 Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
117
|
|
118
|
|
119
|
|
120
|
D'Souza NM, Fang P, Logan J, Yang J, Jiang W, Li J. Combining Radiation Therapy with Immune Checkpoint Blockade for Central Nervous System Malignancies. Front Oncol 2016; 6:212. [PMID: 27774435 PMCID: PMC5053992 DOI: 10.3389/fonc.2016.00212] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/26/2016] [Indexed: 12/14/2022] Open
Abstract
Malignancies of the central nervous system (CNS), particularly glioblastoma and brain metastases from a variety of disease sites, are difficult to treat despite advances in multimodality approaches consisting of surgery, chemotherapy, and radiation therapy (RT). Recent successes of immunotherapeutic strategies including immune checkpoint blockade (ICB) via anti-PD-1 and anti-CTLA-4 antibodies against aggressive cancers, such as melanoma, non-small cell lung cancer, and renal cell carcinoma, have presented an exciting opportunity to translate these strategies for CNS malignancies. Moreover, via both localized cytotoxicity and systemic proinflammatory effects, the role of RT in enhancing antitumor immune response and, therefore, promoting tumor control is being re-examined, with several preclinical and clinical studies demonstrating potential synergistic effect of RT with ICB in the treatment of primary and metastatic CNS tumors. In this review, we highlight the preclinical evidence supporting the immunomodulatory effect of RT and discuss the rationales for its combination with ICB to promote antitumor immune response. We then outline the current clinical experience of combining RT with ICB in the treatment of multiple primary and metastatic brain tumors. Finally, we review advances in characterizing and modifying tumor radioimmunotherapy responses using biomarkers and microRNA (miRNA) that may potentially be used to guide clinical decision-making in the near future.
Collapse
Affiliation(s)
- Neil M D'Souza
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Baylor College of Medicine, Houston, TX, USA
| | - Penny Fang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Jennifer Logan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Jinzhong Yang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Jing Li
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| |
Collapse
|
121
|
Kickingereder P, Bonekamp D, Nowosielski M, Kratz A, Sill M, Burth S, Wick A, Eidel O, Schlemmer HP, Radbruch A, Debus J, Herold-Mende C, Unterberg A, Jones D, Pfister S, Wick W, von Deimling A, Bendszus M, Capper D. Radiogenomics of Glioblastoma: Machine Learning-based Classification of Molecular Characteristics by Using Multiparametric and Multiregional MR Imaging Features. Radiology 2016; 281:907-918. [PMID: 27636026 DOI: 10.1148/radiol.2016161382] [Citation(s) in RCA: 219] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Purpose To evaluate the association of multiparametric and multiregional magnetic resonance (MR) imaging features with key molecular characteristics in patients with newly diagnosed glioblastoma. Materials and Methods Retrospective data evaluation was approved by the local ethics committee, and the requirement to obtain informed consent was waived. Preoperative MR imaging features were correlated with key molecular characteristics within a single-institution cohort of 152 patients with newly diagnosed glioblastoma. Preoperative MR imaging features (n = 31) included multiparametric (anatomic and diffusion-, perfusion-, and susceptibility-weighted images) and multiregional (contrast-enhancing regions and hyperintense regions at nonenhanced fluid-attenuated inversion recovery imaging) information with histogram quantification of tumor volumes, volume ratios, apparent diffusion coefficients, cerebral blood flow, cerebral blood volume, and intratumoral susceptibility signals. Molecular characteristics determined included global DNA methylation subgroups (eg, mesenchymal, RTK I "PGFRA," RTK II "classic"), MGMT promoter methylation status, and hallmark copy number variations (EGFR, PDGFRA, MDM4, and CDK4 amplification; PTEN, CDKN2A, NF1, and RB1 loss). Univariate analyses (voxel-lesion symptom mapping for tumor location, Wilcoxon test for all other MR imaging features) and machine learning models were applied to study the strength of association and discriminative value of MR imaging features for predicting underlying molecular characteristics. Results There was no tumor location predilection for any of the assessed molecular parameters (permutation-adjusted P > .05). Univariate imaging parameter associations were noted for EGFR amplification and CDKN2A loss, with both demonstrating increased Gaussian-normalized relative cerebral blood volume and Gaussian-normalized relative cerebral blood flow values (area under the receiver operating characteristics curve: 63%-69%, false discovery rate-adjusted P < .05). Subjecting all MR imaging features to machine learning-based classification enabled prediction of EGFR amplification status and the RTK II glioblastoma subgroup with a moderate, yet significantly greater, accuracy (63% for EGFR [P < .01], 61% for RTK II [P = .01]) than prediction by chance; prediction accuracy for all other molecular parameters was not significant. Conclusion The authors found associations between established MR imaging features and molecular characteristics, although not of sufficient strength to enable generation of machine learning classification models for reliable and clinically meaningful prediction of molecular characteristics in patients with glioblastoma. © RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Philipp Kickingereder
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - David Bonekamp
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Martha Nowosielski
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Annekathrin Kratz
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Martin Sill
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Sina Burth
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Antje Wick
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Oliver Eidel
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Heinz-Peter Schlemmer
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Alexander Radbruch
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Jürgen Debus
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Christel Herold-Mende
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Andreas Unterberg
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - David Jones
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Stefan Pfister
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Wolfgang Wick
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Andreas von Deimling
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - Martin Bendszus
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| | - David Capper
- From the Department of Neuroradiology, University of Heidelberg Medical Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., S.B., O.E., M.B.); German Cancer Research Center (DKFZ), Department of Radiology, Heidelberg, Germany (D.B., H.P.S., A.R.); Department of Neurology, Medical University Innsbruck, Austria (M.N.); Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany (A.K., A.v.D., D.C.); German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.K., A.v.D., D.C.); Division of Biostatistics, DKFZ, Heidelberg, Germany (M.S.); Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany (M.N., S.B., A.W., W.W.); Department of Radiation Oncology, University of Heidelberg Medical Center, Heidelberg, Germany (J.D.); Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (J.D.); Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (C.H.M.); Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany (A.U.); Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.J., S.P.); German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany (D.J., S.P.); Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany (S.P.); and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany (W.W.)
| |
Collapse
|
122
|
Grossmann P, Gutman DA, Dunn WD, Holder CA, Aerts HJWL. Imaging-genomics reveals driving pathways of MRI derived volumetric tumor phenotype features in Glioblastoma. BMC Cancer 2016; 16:611. [PMID: 27502180 PMCID: PMC4977720 DOI: 10.1186/s12885-016-2659-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 08/01/2016] [Indexed: 12/24/2022] Open
Abstract
Background Glioblastoma (GBM) tumors exhibit strong phenotypic differences that can be quantified using magnetic resonance imaging (MRI), but the underlying biological drivers of these imaging phenotypes remain largely unknown. An Imaging-Genomics analysis was performed to reveal the mechanistic associations between MRI derived quantitative volumetric tumor phenotype features and molecular pathways. Methods One hundred fourty one patients with presurgery MRI and survival data were included in our analysis. Volumetric features were defined, including the necrotic core (NE), contrast-enhancement (CE), abnormal tumor volume assessed by post-contrast T1w (tumor bulk or TB), tumor-associated edema based on T2-FLAIR (ED), and total tumor volume (TV), as well as ratios of these tumor components. Based on gene expression where available (n = 91), pathway associations were assessed using a preranked gene set enrichment analysis. These results were put into context of molecular subtypes in GBM and prognostication. Results Volumetric features were significantly associated with diverse sets of biological processes (FDR < 0.05). While NE and TB were enriched for immune response pathways and apoptosis, CE was associated with signal transduction and protein folding processes. ED was mainly enriched for homeostasis and cell cycling pathways. ED was also the strongest predictor of molecular GBM subtypes (AUC = 0.61). CE was the strongest predictor of overall survival (C-index = 0.6; Noether test, p = 4x10−4). Conclusion GBM volumetric features extracted from MRI are significantly enriched for information about the biological state of a tumor that impacts patient outcomes. Clinical decision-support systems could exploit this information to develop personalized treatment strategies on the basis of noninvasive imaging. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2659-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Patrick Grossmann
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - David A Gutman
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.,Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - William D Dunn
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.,Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Chad A Holder
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Hugo J W L Aerts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
123
|
Wong AJ, Kanwar A, Mohamed AS, Fuller CD. Radiomics in head and neck cancer: from exploration to application. Transl Cancer Res 2016; 5:371-382. [PMID: 30627523 DOI: 10.21037/tcr.2016.07.18] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In the context of clinical oncology, a fundamental goal of radiomics is the extraction of large amounts of quantitative features whose subsequent analysis can be used for decision support towards personalized and actionable cancer care. Head and neck cancers present a unique set of diagnostic and therapeutic challenges by nature of its complex anatomy and heterogeneity. Radiomics holds the potential to address these barriers, but only if as a collective field we direct future effort towards investigating specific oncologic function and oncologic outcomes, with external validation and collaborative multi-institutional efforts to begin standardizing and refining radiomic signatures. Here we present an overview of radiomic texture analysis methods as well as the software infrastructure, review the developments of radiomics in head and neck cancer applications, discuss unmet challenges, and propose key recommendations for moving the field forward.
Collapse
Affiliation(s)
- Andrew J Wong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Aasheesh Kanwar
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,School of Medicine, Texas Tech University Health Science Center, Lubbock, TX, USA
| | - Abdallah S Mohamed
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Clinical Oncology, University of Alexandria, Alexandria, Egypt
| | - Clifton D Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
124
|
Zinn PO, Hatami M, Youssef E, Thomas GA, Luedi MM, Singh SK, Colen RR. Diffusion Weighted Magnetic Resonance Imaging Radiophenotypes and Associated Molecular Pathways in Glioblastoma. Neurosurgery 2016; 63 Suppl 1:127-135. [DOI: 10.1227/neu.0000000000001302] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
125
|
Dunn WD, Aerts HJ, Cooper LA, Holder CA, Hwang SN, Jaffe CC, Brat DJ, Jain R, Flanders AE, Zinn PO, Colen RR, Gutman DA. Assessing the Effects of Software Platforms on Volumetric Segmentation of Glioblastoma. JOURNAL OF NEUROIMAGING IN PSYCHIATRY & NEUROLOGY 2016; 1:64-72. [PMID: 29600296 PMCID: PMC5870135 DOI: 10.17756/jnpn.2016-008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Radiological assessments of biologically relevant regions in glioblastoma have been associated with genotypic characteristics, implying a potential role in personalized medicine. Here, we assess the reproducibility and association with survival of two volumetric segmentation platforms and explore how methodology could impact subsequent interpretation and analysis. METHODS Post-contrast T1- and T2-weighted FLAIR MR images of 67 TCGA patients were segmented into five distinct compartments (necrosis, contrast-enhancement, FLAIR, post contrast abnormal, and total abnormal tumor volumes) by two quantitative image segmentation platforms - 3D Slicer and a method based on Velocity AI and FSL. We investigated the internal consistency of each platform by correlation statistics, association with survival, and concordance with consensus neuroradiologist ratings using ordinal logistic regression. RESULTS We found high correlations between the two platforms for FLAIR, post contrast abnormal, and total abnormal tumor volumes (spearman's r(67) = 0.952, 0.959, and 0.969 respectively). Only modest agreement was observed for necrosis and contrast-enhancement volumes (r(67) = 0.693 and 0.773 respectively), likely arising from differences in manual and automated segmentation methods of these regions by 3D Slicer and Velocity AI/FSL, respectively. Survival analysis based on AUC revealed significant predictive power of both platforms for the following volumes: contrast-enhancement, post contrast abnormal, and total abnormal tumor volumes. Finally, ordinal logistic regression demonstrated correspondence to manual ratings for several features. CONCLUSION Tumor volume measurements from both volumetric platforms produced highly concordant and reproducible estimates across platforms for general features. As automated or semi-automated volumetric measurements replace manual linear or area measurements, it will become increasingly important to keep in mind that measurement differences between segmentation platforms for more detailed features could influence downstream survival or radio genomic analyses.
Collapse
Affiliation(s)
- William D. Dunn
- Departments of Biomedical Informatics and Neurology, Emory
University School of Medicine, Atlanta, GA, USA
| | - Hugo J.W.L. Aerts
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer
Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA,
USA
- Department of Biostatistics & Computational Biology, Dana-Farber
Cancer Institute, Boston, MA, USA
| | - Lee A. Cooper
- Departments of Biomedical Informatics and Neurology, Emory
University School of Medicine, Atlanta, GA, USA
- Department Winship Cancer Institute, Emory University, Atlanta, GA,
USA
- Department Biomedical Engineering, Georgia Institute of
Technology/Emory University, Atlanta, GA, USA
| | - Chad A. Holder
- Department of Radiology and Imaging Sciences, Emory University
School of Medicine, Atlanta, GA, USA
| | - Scott N. Hwang
- Department of Diagnostic Imaging Department, St. Jude
Children’s Research Hospital, Memphis, TN, USA
| | - Carle C. Jaffe
- Department of Radiology, Boston University School of Medicine,
Boston, MA, USA
| | - Daniel J. Brat
- Department of Pathology and Laboratory Medicine, Emory University
School of Medicine, Atlanta, GA, USA
| | - Rajan Jain
- Departments of Radiology and Neurosurgery, NYU School of Medicine,
New York, NY, USA
| | - Adam E. Flanders
- Department of Neuroradiology, Thomas Jefferson University
Hospitals, Philadelphia, PA, USA
| | - Pascal O. Zinn
- Department of Neurosurgery, The University of Texas MD Anderson
Cancer Center, Houston, TX, USA
| | - Rivka R. Colen
- Department of Diagnostic Radiology, The University of Texas MD
Anderson Cancer Center, Houston, TX, USA
| | - David A. Gutman
- Departments of Biomedical Informatics and Neurology, Emory
University School of Medicine, Atlanta, GA, USA
- Department Winship Cancer Institute, Emory University, Atlanta, GA,
USA
| |
Collapse
|
126
|
Heiland DH, Mader I, Schlosser P, Pfeifer D, Carro MS, Lange T, Schwarzwald R, Vasilikos I, Urbach H, Weyerbrock A. Integrative Network-based Analysis of Magnetic Resonance Spectroscopy and Genome Wide Expression in Glioblastoma multiforme. Sci Rep 2016; 6:29052. [PMID: 27350391 PMCID: PMC4924099 DOI: 10.1038/srep29052] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/10/2016] [Indexed: 11/18/2022] Open
Abstract
The goal of this study was to identify correlations between metabolites from proton MR spectroscopy and genetic pathway activity in glioblastoma multiforme (GBM). Twenty patients with primary GBM were analysed by short echo-time chemical shift imaging and genome-wide expression analyses. Weighed Gene Co-Expression Analysis was used for an integrative analysis of imaging and genetic data. N-acetylaspartate, normalised to the contralateral healthy side (nNAA), was significantly correlated to oligodendrocytic and neural development. For normalised creatine (nCr), a group with low nCr was linked to the mesenchymal subtype, while high nCr could be assigned to the proneural subtype. Moreover, clustering of normalised glutamine and glutamate (nGlx) revealed two groups, one with high nGlx being attributed to the neural subtype, and one with low nGlx associated with the classical subtype. Hence, the metabolites nNAA, nCr, and nGlx correlate with a specific gene expression pattern reflecting the previously described subtypes of GBM. Moreover high nNAA was associated with better clinical prognosis, whereas patients with lower nNAA revealed a shorter progression-free survival (PFS).
Collapse
Affiliation(s)
- Dieter Henrik Heiland
- Department of Neurosurgery, Medical Center University of Freiburg, Freiburg, Germany
| | - Irina Mader
- Department of Neuroradiology, Medical Center University of Freiburg, Freiburg, Germany
| | - Pascal Schlosser
- Institute for Medical Biometry and Statistics, Medical Center University of Freiburg, Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center University of Freiburg, Freiburg, Germany
| | - Maria Stella Carro
- Department of Neurosurgery, Medical Center University of Freiburg, Freiburg, Germany
| | - Thomas Lange
- Department of Medical Physics, Diagnostic Radiology, Medical Center University of Freiburg, Freiburg, Germany
| | - Ralf Schwarzwald
- Department of Neuroradiology, Medical Center University of Freiburg, Freiburg, Germany
| | - Ioannis Vasilikos
- Department of Neurosurgery, Medical Center University of Freiburg, Freiburg, Germany
| | - Horst Urbach
- Department of Neuroradiology, Medical Center University of Freiburg, Freiburg, Germany
| | - Astrid Weyerbrock
- Department of Neurosurgery, Medical Center University of Freiburg, Freiburg, Germany
| |
Collapse
|
127
|
Huynh E, Coroller TP, Narayan V, Agrawal V, Hou Y, Romano J, Franco I, Mak RH, Aerts HJWL. CT-based radiomic analysis of stereotactic body radiation therapy patients with lung cancer. Radiother Oncol 2016; 120:258-66. [PMID: 27296412 DOI: 10.1016/j.radonc.2016.05.024] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 05/23/2016] [Accepted: 05/24/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Radiomics uses a large number of quantitative imaging features that describe the tumor phenotype to develop imaging biomarkers for clinical outcomes. Radiomic analysis of pre-treatment computed-tomography (CT) scans was investigated to identify imaging predictors of clinical outcomes in early stage non-small cell lung cancer (NSCLC) patients treated with stereotactic body radiation therapy (SBRT). MATERIALS AND METHODS CT images of 113 stage I-II NSCLC patients treated with SBRT were analyzed. Twelve radiomic features were selected based on stability and variance. The association of features with clinical outcomes and their prognostic value (using the concordance index (CI)) was evaluated. Radiomic features were compared with conventional imaging metrics (tumor volume and diameter) and clinical parameters. RESULTS Overall survival was associated with two conventional features (volume and diameter) and two radiomic features (LoG 3D run low gray level short run emphasis and stats median). One radiomic feature (Wavelet LLH stats range) was significantly prognostic for distant metastasis (CI=0.67, q-value<0.1), while none of the conventional and clinical parameters were. Three conventional and four radiomic features were prognostic for overall survival. CONCLUSION This exploratory analysis demonstrates that radiomic features have potential to be prognostic for some outcomes that conventional imaging metrics cannot predict in SBRT patients.
Collapse
Affiliation(s)
- Elizabeth Huynh
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.
| | - Thibaud P Coroller
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Vivek Narayan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Vishesh Agrawal
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Ying Hou
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - John Romano
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Idalid Franco
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Raymond H Mak
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Hugo J W L Aerts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
128
|
Quantitative Multiparametric MRI Features and PTEN Expression of Peripheral Zone Prostate Cancer: A Pilot Study. AJR Am J Roentgenol 2016; 206:559-65. [PMID: 26901012 DOI: 10.2214/ajr.15.14967] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The objective of our study was to investigate associations between quantitative image features of multiparametric MRI of the prostate and PTEN expression of peripheral zone prostate cancer. MATERIALS AND METHODS A total of 45 peripheral zone cancer foci from 30 patients who had undergone multiparametric prostate MRI before prostatectomy were identified by a genitourinary pathologist and a radiologist who reviewed histologic findings and MR images. Histologic sections of cancer foci underwent immunohistochemical analysis and were scored according to the percentage of tumor-positive cells expressing PTEN as negative (0-20%), mixed (20-80%), or positive (80-100%). Average and 10th percentile apparent diffusion coefficient (ADC) values, skewness of T2-weighted signal intensity histogram, and quantitative perfusion parameters (i.e., forward volume transfer constant [K(trans)], extravascular extracellular volume fraction [ve], and reverse reflux rate constant between the extracellular space and plasma [k(ep)]) from the Tofts model were calculated for each cancer focus. Associations between the quantitative image features and PTEN expression were analyzed with the Spearman rank correlation coefficient (r). RESULTS Analysis of the 45 cancer foci revealed that 21 (47%) were PTEN-positive, 12 (27%) were PTEN-negative, and 12 (27%) were mixed. There was a weak but significant negative correlation between Gleason score and PTEN expression (r = -0.30, p = 0.04) and between k(ep) and PTEN expression (r = -0.35, p = 0.02). There was no significant correlation between other multiparametric MRI features and PTEN expression. CONCLUSION This preliminary study of radiogenomics of peripheral zone prostate cancer revealed weak-but significant-associations between the quantitative dynamic contrast-enhanced MRI feature k(ep) and Gleason score with PTEN expression. These findings warrant further investigation and validation with the aim of using multiparametric MRI to improve risk assessment of patients with prostate cancer.
Collapse
|
129
|
Itakura H, Achrol AS, Mitchell LA, Loya JJ, Liu T, Westbroek EM, Feroze AH, Rodriguez S, Echegaray S, Azad TD, Yeom KW, Napel S, Rubin DL, Chang SD, Harsh GR, Gevaert O. Magnetic resonance image features identify glioblastoma phenotypic subtypes with distinct molecular pathway activities. Sci Transl Med 2016; 7:303ra138. [PMID: 26333934 DOI: 10.1126/scitranslmed.aaa7582] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Glioblastoma (GBM) is the most common and highly lethal primary malignant brain tumor in adults. There is a dire need for easily accessible, noninvasive biomarkers that can delineate underlying molecular activities and predict response to therapy. To this end, we sought to identify subtypes of GBM, differentiated solely by quantitative magnetic resonance (MR) imaging features, that could be used for better management of GBM patients. Quantitative image features capturing the shape, texture, and edge sharpness of each lesion were extracted from MR images of 121 single-institution patients with de novo, solitary, unilateral GBM. Three distinct phenotypic "clusters" emerged in the development cohort using consensus clustering with 10,000 iterations on these image features. These three clusters--pre-multifocal, spherical, and rim-enhancing, names reflecting their image features--were validated in an independent cohort consisting of 144 multi-institution patients with similar tumor characteristics from The Cancer Genome Atlas (TCGA). Each cluster mapped to a unique set of molecular signaling pathways using pathway activity estimates derived from the analysis of TCGA tumor copy number and gene expression data with the PARADIGM (Pathway Recognition Algorithm Using Data Integration on Genomic Models) algorithm. Distinct pathways, such as c-Kit and FOXA, were enriched in each cluster, indicating differential molecular activities as determined by the image features. Each cluster also demonstrated differential probabilities of survival, indicating prognostic importance. Our imaging method offers a noninvasive approach to stratify GBM patients and also provides unique sets of molecular signatures to inform targeted therapy and personalized treatment of GBM.
Collapse
Affiliation(s)
- Haruka Itakura
- Division of Biomedical Informatics, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Achal S Achrol
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Lex A Mitchell
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Joshua J Loya
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Tiffany Liu
- Division of Biomedical Informatics, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Erick M Westbroek
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Abdullah H Feroze
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Scott Rodriguez
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Sebastian Echegaray
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Tej D Azad
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Kristen W Yeom
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Sandy Napel
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Daniel L Rubin
- Division of Biomedical Informatics, Department of Medicine, Stanford University, Stanford, CA 94305, USA. Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Steven D Chang
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Griffith R Harsh
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Olivier Gevaert
- Division of Biomedical Informatics, Department of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
130
|
Mazurowski MA. Radiogenomics: what it is and why it is important. J Am Coll Radiol 2016; 12:862-6. [PMID: 26250979 DOI: 10.1016/j.jacr.2015.04.019] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/20/2015] [Indexed: 01/29/2023]
Abstract
In recent years, a new direction in cancer research has emerged that focuses on the relationship between imaging phenotypes and genomics. This direction is referred to as radiogenomics or imaging genomics. The question that subsequently arises is: What is the practical significance of elucidating this relationship in improving cancer patient outcomes. In this article, I address this question. Although I discuss some limitations of the radiogenomic approach, and describe scenarios in which radiogenomic analysis might not be the best choice, I also argue that radiogenomics will play a significant practical role in cancer research. Specifically, I argue that the significance of radiogenomics is largely related to practical limitations of currently available data that often lack complete characterization of the patients and poor integration of individual datasets. Radiogenomics offers a practical way to leverage limited and incomplete data to generate knowledge that might lead to improved decision making, and as a result, improved patient outcomes.
Collapse
Affiliation(s)
- Maciej A Mazurowski
- Department of Radiology, Duke Cancer Institute, Duke Medical Physics Program, Duke University School of Medicine, Durham, North Carolina.
| |
Collapse
|
131
|
|
132
|
Saha A, Banerjee S, Kurtek S, Narang S, Lee J, Rao G, Martinez J, Bharath K, Rao AUK, Baladandayuthapani V. DEMARCATE: Density-based magnetic resonance image clustering for assessing tumor heterogeneity in cancer. Neuroimage Clin 2016; 12:132-43. [PMID: 27408798 PMCID: PMC4932621 DOI: 10.1016/j.nicl.2016.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/11/2016] [Accepted: 05/25/2016] [Indexed: 01/24/2023]
Abstract
Tumor heterogeneity is a crucial area of cancer research wherein inter- and intra-tumor differences are investigated to assess and monitor disease development and progression, especially in cancer. The proliferation of imaging and linked genomic data has enabled us to evaluate tumor heterogeneity on multiple levels. In this work, we examine magnetic resonance imaging (MRI) in patients with brain cancer to assess image-based tumor heterogeneity. Standard approaches to this problem use scalar summary measures (e.g., intensity-based histogram statistics) that do not adequately capture the complete and finer scale information in the voxel-level data. In this paper, we introduce a novel technique, DEMARCATE (DEnsity-based MAgnetic Resonance image Clustering for Assessing Tumor hEterogeneity) to explore the entire tumor heterogeneity density profiles (THDPs) obtained from the full tumor voxel space. THDPs are smoothed representations of the probability density function of the tumor images. We develop tools for analyzing such objects under the Fisher-Rao Riemannian framework that allows us to construct metrics for THDP comparisons across patients, which can be used in conjunction with standard clustering approaches. Our analyses of The Cancer Genome Atlas (TCGA) based Glioblastoma dataset reveal two significant clusters of patients with marked differences in tumor morphology, genomic characteristics and prognostic clinical outcomes. In addition, we see enrichment of image-based clusters with known molecular subtypes of glioblastoma multiforme, which further validates our representation of tumor heterogeneity and subsequent clustering techniques.
Collapse
Affiliation(s)
- Abhijoy Saha
- Department of Statistics, The Ohio State University, United States
| | - Sayantan Banerjee
- Operations Management and Quantitative Techniques Area, Indian Institute of Management Indore, India
| | - Sebastian Kurtek
- Department of Statistics, The Ohio State University, United States
| | - Shivali Narang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, United States
| | - Joonsang Lee
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, United States
| | - Ganesh Rao
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, United States
| | - Juan Martinez
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, United States
| | - Karthik Bharath
- School of Mathematical Sciences, The University of Nottingham, United Kingdom
| | - Arvind U K Rao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, United States
| | | |
Collapse
|
133
|
Zhou C, Yang Z, Yao Z, Yin B, Pan J, Yu Y, Zhu W, Hua W, Mao Y. Segmentation of peritumoral oedema offers a valuable radiological feature of cerebral metastasis. Br J Radiol 2016; 89:20151054. [PMID: 27119727 DOI: 10.1259/bjr.20151054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Peritumoral oedema (PTO) is commonly observed on MRI in malignant brain tumours including brain metastasis (bMET) and glioblastoma multiforme (GBM). This study aimed to differentiate bMET from GBM by comparing the volume ratio of PTO to tumour lesion (Rvol). METHODS 56 patients with solitary bMET or GBM were enrolled, and MRI was analyzed by a semi-automatic methodology based on MATLAB (Mathworks, Natick, MA). The PTO volume (Voedema) was segmented for quantification using T2 fluid-attenuated inversion-recovery images, while the tumour volume was quantified with enhanced T1 images. The quantitative volume of the tumour, PTO and the ratio of PTO to tumour were interpreted using SPSS(®) (IBM Corp., New York, NY; formerly SPSS Inc., Chicago, IL) by considering different locations and pathologies. RESULTS The tumour volumes of supratentorial GBM, supratentorial bMET (supra-bMET) and infratentorial bMET were 32.22 ± 21.9, 18.45 ± 17.28 and 11.40 ± 5.63 ml, respectively. The corresponding Voedema were 44.08 ± 25.84, 73.20 ± 40.35 and 23.74 ± 7.78 ml, respectively. The Voedema difference between supratentorial and infratentorial lesions is significant (p-value = 0.002). Supra-bMET has a smaller tumour volume (p-value = 0.032), but a larger PTO (p-value = 0.007). The ratio of Voedema to the tumour volume in bMET is statistically higher than that in GBM (p-value = 0.015). The cut-off ratio for identifying bMET from GBM is 3.9, with a specificity and sensitivity of 90.0% and 68.8%, respectively. CONCLUSION Segmentation is an efficient method to quantify irregular PTO. bMET possesses more extensive oedema with smaller tumour volume than does GBM. The Rvol is a valuable index to distinguish bMET from GBM. ADVANCES IN KNOWLEDGE This study presents a new method for the quantitation of PTO to differentiate bMET from GBM.
Collapse
Affiliation(s)
- Chengcheng Zhou
- Department of Neurosurgery, Huashan Hospital, Shanghai, China
| | - Zixiao Yang
- Department of Neurosurgery, Huashan Hospital, Shanghai, China
| | - Zhengwei Yao
- Department of Radiology, Huashan Hospital, Shanghai, China
| | - Bo Yin
- Department of Radiology, Huashan Hospital, Shanghai, China
| | - Jiawei Pan
- Department of Radiology, Huashan Hospital, Shanghai, China
| | - Yang Yu
- Department of Radiology, Huashan Hospital, Shanghai, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai, China
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institute of Brain Science, Fudan University, Shanghai, China.,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
134
|
Rao A, Rao G, Gutman DA, Flanders AE, Hwang SN, Rubin DL, Colen RR, Zinn PO, Jain R, Wintermark M, Kirby JS, Jaffe CC, Freymann J. A combinatorial radiographic phenotype may stratify patient survival and be associated with invasion and proliferation characteristics in glioblastoma. J Neurosurg 2016; 124:1008-17. [PMID: 26473782 PMCID: PMC4990448 DOI: 10.3171/2015.4.jns142732] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Individual MRI characteristics (e.g., volume) are routinely used to identify survival-associated phenotypes for glioblastoma (GBM). This study investigated whether combinations of MRI features can also stratify survival. Furthermore, the molecular differences between phenotype-induced groups were investigated. METHODS Ninety-two patients with imaging, molecular, and survival data from the TCGA (The Cancer Genome Atlas)-GBM collection were included in this study. For combinatorial phenotype analysis, hierarchical clustering was used. Groups were defined based on a cutpoint obtained via tree-based partitioning. Furthermore, differential expression analysis of microRNA (miRNA) and mRNA expression data was performed using GenePattern Suite. Functional analysis of the resulting genes and miRNAs was performed using Ingenuity Pathway Analysis. Pathway analysis was performed using Gene Set Enrichment Analysis. RESULTS Clustering analysis reveals that image-based grouping of the patients is driven by 3 features: volume-class, hemorrhage, and T1/FLAIR-envelope ratio. A combination of these features stratifies survival in a statistically significant manner. A cutpoint analysis yields a significant survival difference in the training set (median survival difference: 12 months, p = 0.004) as well as a validation set (p = 0.0001). Specifically, a low value for any of these 3 features indicates favorable survival characteristics. Differential expression analysis between cutpoint-induced groups suggests that several immune-associated (natural killer cell activity, T-cell lymphocyte differentiation) and metabolism-associated (mitochondrial activity, oxidative phosphorylation) pathways underlie the transition of this phenotype. Integrating data for mRNA and miRNA suggests the roles of several genes regulating proliferation and invasion. CONCLUSIONS A 3-way combination of MRI phenotypes may be capable of stratifying survival in GBM. Examination of molecular processes associated with groups created by this combinatorial phenotype suggests the role of biological processes associated with growth and invasion characteristics.
Collapse
Affiliation(s)
- Arvind Rao
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ganesh Rao
- Department of Neurosurgery, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - David A. Gutman
- Department of Biomedical Informatics, Emory University, Atlanta, Georgia
| | - Adam E. Flanders
- Department of Radiology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Scott N. Hwang
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Daniel L. Rubin
- Department of Radiology, Stanford University, Stanford, California
| | - Rivka R. Colen
- Department of Diagnostic Radiology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Pascal O. Zinn
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Rajan Jain
- Department of Radiology, New York University School of Medicine, New York, New York
| | - Max Wintermark
- Department of Interventional Neuroradiology, Stanford Medical Center, Stanford, California
| | - Justin S. Kirby
- Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland
| | - C. Carl Jaffe
- Department of Radiology, Boston University School of Medicine, Boston, Massachusetts
| | - John Freymann
- Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland
| |
Collapse
|
135
|
Li YM, Suki D, Hess K, Sawaya R. The influence of maximum safe resection of glioblastoma on survival in 1229 patients: Can we do better than gross-total resection? J Neurosurg 2016; 124:977-88. [DOI: 10.3171/2015.5.jns142087] [Citation(s) in RCA: 428] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECT
Glioblastoma multiforme (GBM) is the most common and deadliest primary brain tumor. The value of extent of resection (EOR) in improving survival in patients with GBM has been repeatedly confirmed, with more extensive resections providing added advantages. The authors reviewed the survival of patients with significant EORs and assessed the relative benefit/risk of resecting 100% of the MRI region showing contrast-enhancement with or without additional resection of the surrounding FLAIR abnormality region, and they assessed the relative benefit/risk of performing this additional resection.
METHODS
The study cohort included 1229 patients with histologically verified GBM in whom ≥ 78% resection was achieved at The University of Texas MD Anderson Cancer Center between June 1993 and December 2012. Patients with > 1 tumor and those 80 years old or older were excluded. The survival of patients having 100% removal of the contrast-enhancing tumor, with or without additional resection of the surrounding FLAIR abnormality region, was compared with that of patients undergoing 78% to < 100% EOR of the enhancing mass. Within the first subgroup, the survival durations of patients with and without resection of the surrounding FLAIR abnormality were subsequently compared. The data on patients and their tumor characteristics were collected prospectively. The incidence of 30-day postoperative complications (overall and neurological) was noted.
RESULTS
Complete resection of the T1 contrast-enhancing tumor volume was achieved in 876 patients (71%). The median survival time for these patients (15.2 months) was significantly longer than that for patients undergoing less than complete resection (9.8 months; p < 0.001). This survival advantage was achieved without an increase in the risk of overall or neurological postoperative deficits and after correcting for established prognostic factors including age, Karnofsky Performance Scale score, preoperative contrast-enhancing tumor volume, presence of cyst, and prior treatment status (HR 1.53, 95% CI 1.33–1.77, p < 0.001). The effect remained essentially unchanged when data from previously treated and previously untreated groups of patients were analyzed separately. Additional analyses showed that the resection of ≥ 53.21% of the surrounding FLAIR abnormality beyond the 100% contrast-enhancing resection was associated with a significant prolongation of survival compared with that following less extensive resections (median survival times 20.7 and 15.5 months, respectively; p < 0.001). In the multivariate analysis, the previously treated group with < 53.21% resection had significantly shorter survival than the 3 other groups (that is, previously treated patients who underwent FLAIR resection ≥ 53.21%, previously untreated patients who underwent FLAIR resection < 53.21%, and previously untreated patients who underwent FLAIR resection ≥ 53.21%); the previously untreated group with ≥ 53.21% resection had the longest survival.
CONCLUSIONS
What is believed to be the largest single-center series of GBM patients with extensive tumor resections, this study supports the established association between EOR and survival and presents additional data that pushing the boundary of a conventional 100% resection by the additional removal of a significant portion of the FLAIR abnormality region, when safely feasible, may result in the prolongation of survival without significant increases in overall or neurological postoperative morbidity. Additional supportive evidence is warranted.
Collapse
Affiliation(s)
- Yan Michael Li
- Departments of 1Neurosurgery and
- 2Department of Neurosurgery and Oncology, University of Rochester Medical Center School of Medicine and Dentistry, Rochester, New York
| | | | - Kenneth Hess
- 3Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas; and
| | | |
Collapse
|
136
|
Wu W, Parmar C, Grossmann P, Quackenbush J, Lambin P, Bussink J, Mak R, Aerts HJWL. Exploratory Study to Identify Radiomics Classifiers for Lung Cancer Histology. Front Oncol 2016; 6:71. [PMID: 27064691 PMCID: PMC4811956 DOI: 10.3389/fonc.2016.00071] [Citation(s) in RCA: 256] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 03/14/2016] [Indexed: 01/05/2023] Open
Abstract
Background Radiomics can quantify tumor phenotypic characteristics non-invasively by applying feature algorithms to medical imaging data. In this study of lung cancer patients, we investigated the association between radiomic features and the tumor histologic subtypes (adenocarcinoma and squamous cell carcinoma). Furthermore, in order to predict histologic subtypes, we employed machine-learning methods and independently evaluated their prediction performance. Methods Two independent radiomic cohorts with a combined size of 350 patients were included in our analysis. A total of 440 radiomic features were extracted from the segmented tumor volumes of pretreatment CT images. These radiomic features quantify tumor phenotypic characteristics on medical images using tumor shape and size, intensity statistics, and texture. Univariate analysis was performed to assess each feature’s association with the histological subtypes. In our multivariate analysis, we investigated 24 feature selection methods and 3 classification methods for histology prediction. Multivariate models were trained on the training cohort and their performance was evaluated on the independent validation cohort using the area under ROC curve (AUC). Histology was determined from surgical specimen. Results In our univariate analysis, we observed that fifty-three radiomic features were significantly associated with tumor histology. In multivariate analysis, feature selection methods ReliefF and its variants showed higher prediction accuracy as compared to other methods. We found that Naive Baye’s classifier outperforms other classifiers and achieved the highest AUC (0.72; p-value = 2.3 × 10−7) with five features: Stats_min, Wavelet_HLL_rlgl_lowGrayLevelRunEmphasis, Wavelet_HHL_stats_median, Wavelet_HLL_stats_skewness, and Wavelet_HLH_glcm_clusShade. Conclusion Histological subtypes can influence the choice of a treatment/therapy for lung cancer patients. We observed that radiomic features show significant association with the lung tumor histology. Moreover, radiomics-based multivariate classifiers were independently validated for the prediction of histological subtypes. Despite achieving lower than optimal prediction accuracy (AUC 0.72), our analysis highlights the impressive potential of non-invasive and cost-effective radiomics for precision medicine. Further research in this direction could lead us to optimal performance and therefore to clinical applicability, which could enhance the efficiency and efficacy of cancer care.
Collapse
Affiliation(s)
- Weimiao Wu
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Chintan Parmar
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Research Institute GROW, Maastricht University, Maastricht, Netherlands
| | - Patrick Grossmann
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - John Quackenbush
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Philippe Lambin
- Research Institute GROW, Maastricht University , Maastricht , Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center , Nijmegen , Netherlands
| | - Raymond Mak
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| | - Hugo J W L Aerts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
137
|
Rao A, Manyam G, Rao G, Jain R. Integrative Analysis of mRNA, microRNA, and Protein Correlates of Relative Cerebral Blood Volume Values in GBM Reveals the Role for Modulators of Angiogenesis and Tumor Proliferation. Cancer Inform 2016; 15:29-33. [PMID: 27053917 PMCID: PMC4814129 DOI: 10.4137/cin.s33014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/29/2016] [Accepted: 12/07/2015] [Indexed: 12/12/2022] Open
Abstract
Dynamic susceptibility contrast-enhanced magnetic resonance imaging is routinely used to provide hemodynamic assessment of brain tumors as a diagnostic as well as a prognostic tool. Recently, it was shown that the relative cerebral blood volume (rCBV), obtained from the contrast-enhancing as well as -nonenhancing portion of glioblastoma (GBM), is strongly associated with overall survival. In this study, we aim to characterize the genomic correlates (microRNA, messenger RNA, and protein) of this vascular parameter. This study aims to provide a comprehensive radiogenomic and radioproteomic characterization of the hemodynamic phenotype of GBM using publicly available imaging and genomic data from the Cancer Genome Atlas GBM cohort. Based on this analysis, we identified pathways associated with angiogenesis and tumor proliferation underlying this hemodynamic parameter in GBM.
Collapse
Affiliation(s)
- Arvind Rao
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganiraju Manyam
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganesh Rao
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rajan Jain
- Department of Radiology, NY University School of Medicine, New York, NY, USA
| |
Collapse
|
138
|
Reproducibility of radiomics for deciphering tumor phenotype with imaging. Sci Rep 2016; 6:23428. [PMID: 27009765 PMCID: PMC4806325 DOI: 10.1038/srep23428] [Citation(s) in RCA: 366] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/04/2016] [Indexed: 12/11/2022] Open
Abstract
Radiomics (radiogenomics) characterizes tumor phenotypes based on quantitative image features derived from routine radiologic imaging to improve cancer diagnosis, prognosis, prediction and response to therapy. Although radiomic features must be reproducible to qualify as biomarkers for clinical care, little is known about how routine imaging acquisition techniques/parameters affect reproducibility. To begin to fill this knowledge gap, we assessed the reproducibility of a comprehensive, commonly-used set of radiomic features using a unique, same-day repeat computed tomography data set from lung cancer patients. Each scan was reconstructed at 6 imaging settings, varying slice thicknesses (1.25 mm, 2.5 mm and 5 mm) and reconstruction algorithms (sharp, smooth). Reproducibility was assessed using the repeat scans reconstructed at identical imaging setting (6 settings in total). In separate analyses, we explored differences in radiomic features due to different imaging parameters by assessing the agreement of these radiomic features extracted from the repeat scans reconstructed at the same slice thickness but different algorithms (3 settings in total). Our data suggest that radiomic features are reproducible over a wide range of imaging settings. However, smooth and sharp reconstruction algorithms should not be used interchangeably. These findings will raise awareness of the importance of properly setting imaging acquisition parameters in radiomics/radiogenomics research.
Collapse
|
139
|
Clinical Evaluation of a Fully-automatic Segmentation Method for Longitudinal Brain Tumor Volumetry. Sci Rep 2016; 6:23376. [PMID: 27001047 PMCID: PMC4802217 DOI: 10.1038/srep23376] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/04/2016] [Indexed: 11/16/2022] Open
Abstract
Information about the size of a tumor and its temporal evolution is needed for diagnosis as well as treatment of brain tumor patients. The aim of the study was to investigate the potential of a fully-automatic segmentation method, called BraTumIA, for longitudinal brain tumor volumetry by comparing the automatically estimated volumes with ground truth data acquired via manual segmentation. Longitudinal Magnetic Resonance (MR) Imaging data of 14 patients with newly diagnosed glioblastoma encompassing 64 MR acquisitions, ranging from preoperative up to 12 month follow-up images, was analysed. Manual segmentation was performed by two human raters. Strong correlations (R = 0.83–0.96, p < 0.001) were observed between volumetric estimates of BraTumIA and of each of the human raters for the contrast-enhancing (CET) and non-enhancing T2-hyperintense tumor compartments (NCE-T2). A quantitative analysis of the inter-rater disagreement showed that the disagreement between BraTumIA and each of the human raters was comparable to the disagreement between the human raters. In summary, BraTumIA generated volumetric trend curves of contrast-enhancing and non-enhancing T2-hyperintense tumor compartments comparable to estimates of human raters. These findings suggest the potential of automated longitudinal tumor segmentation to substitute manual volumetric follow-up of contrast-enhancing and non-enhancing T2-hyperintense tumor compartments.
Collapse
|
140
|
Abstract
Glioblastoma is regarded as the most aggressive and most common primary malignant brain tumor in adults. Despite advancements in chemotherapy and radiotherapy, prognosis and overall survival of glioblastoma patients remain dismal. Recently, progresses in genetic profiling have increased our understanding of the underlying heterogenous molecular nature of this aggressive tumor. Several prognostic and predictive molecular biomarkers have been identified that have been linked to patient's survival and response to treatment, respectively. Imaging genomics represents a novel entity in clinical sciences that bidirectionally links imaging features with underlying molecular profile and thus can serve as a surrogate for noninvasive genomic correlation, prediction, and identification.
Collapse
|
141
|
Katrib A, Hsu W, Bui A, Xing Y. "RADIOTRANSCRIPTOMICS": A synergy of imaging and transcriptomics in clinical assessment. QUANTITATIVE BIOLOGY 2016; 4:1-12. [PMID: 28529815 DOI: 10.1007/s40484-016-0061-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Recent advances in quantitative imaging and "omics" technology have generated a wealth of mineable biological "big data". With the push towards a P4 "predictive, preventive, personalized, and participatory" approach to medicine, researchers began integrating complementary tools to further tune existing diagnostic and therapeutic models. The field of radiogenomics has long pioneered such multidisciplinary investigations in neuroscience and oncology, correlating genotypic and phenotypic signatures to study structural and functional changes in relation to altered molecular behavior. Given the innate dynamic nature of complex disorders and the role of environmental and epigenetic factors in pathogenesis, the transcriptome can further elucidate serial modifications undetected at the genome level. We therefore propose "radiotranscriptomics" as a new member of the P4 medicine initiative, combining transcriptome information, including gene expression and isoform variation, and quantitative image annotations.
Collapse
Affiliation(s)
- Amal Katrib
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - William Hsu
- Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alex Bui
- Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yi Xing
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
142
|
Bai HX, Lee AM, Yang L, Zhang P, Davatzikos C, Maris JM, Diskin SJ. Imaging genomics in cancer research: limitations and promises. Br J Radiol 2016; 89:20151030. [PMID: 26864054 DOI: 10.1259/bjr.20151030] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recently, radiogenomics or imaging genomics has emerged as a novel high-throughput method of associating imaging features with genomic data. Radiogenomics has the potential to provide comprehensive intratumour, intertumour and peritumour information non-invasively. This review article summarizes the current state of radiogenomic research in tumour characterization, discusses some of its limitations and promises and projects its future directions. Semi-radiogenomic studies that relate specific gene expressions to imaging features will also be briefly reviewed.
Collapse
Affiliation(s)
- Harrison X Bai
- 1 Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley M Lee
- 1 Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Li Yang
- 2 Department of Neurology, The Second Xiangya Hospital, Changsha, Hunan, China
| | - Paul Zhang
- 3 Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- 1 Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - John M Maris
- 4 Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,5 Abramson Family Cancer Research Institute, PerelmanSchool of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,6 Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sharon J Diskin
- 4 Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,5 Abramson Family Cancer Research Institute, PerelmanSchool of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,6 Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
143
|
Calcagno C, Mulder WJM, Nahrendorf M, Fayad ZA. Systems Biology and Noninvasive Imaging of Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 36:e1-8. [PMID: 26819466 PMCID: PMC4861402 DOI: 10.1161/atvbaha.115.306350] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Claudia Calcagno
- From the Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.C., W.J.M.M., Z.A.F.); Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands (W.J.M.M.); and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (M.N.).
| | - Willem J M Mulder
- From the Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.C., W.J.M.M., Z.A.F.); Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands (W.J.M.M.); and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (M.N.)
| | - Matthias Nahrendorf
- From the Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.C., W.J.M.M., Z.A.F.); Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands (W.J.M.M.); and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (M.N.)
| | - Zahi A Fayad
- From the Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.C., W.J.M.M., Z.A.F.); Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands (W.J.M.M.); and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (M.N.)
| |
Collapse
|
144
|
Blackledge MD, Collins DJ, Koh DM, Leach MO. Rapid development of image analysis research tools: Bridging the gap between researcher and clinician with pyOsiriX. Comput Biol Med 2016; 69:203-12. [PMID: 26773941 PMCID: PMC4761020 DOI: 10.1016/j.compbiomed.2015.12.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/07/2015] [Accepted: 12/03/2015] [Indexed: 01/08/2023]
Abstract
We present pyOsiriX, a plugin built for the already popular dicom viewer OsiriX that provides users the ability to extend the functionality of OsiriX through simple Python scripts. This approach allows users to integrate the many cutting-edge scientific/image-processing libraries created for Python into a powerful DICOM visualisation package that is intuitive to use and already familiar to many clinical researchers. Using pyOsiriX we hope to bridge the apparent gap between basic imaging scientists and clinical practice in a research setting and thus accelerate the development of advanced clinical image processing. We provide arguments for the use of Python as a robust scripting language for incorporation into larger software solutions, outline the structure of pyOsiriX and how it may be used to extend the functionality of OsiriX, and we provide three case studies that exemplify its utility. For our first case study we use pyOsiriX to provide a tool for smooth histogram display of voxel values within a user-defined region of interest (ROI) in OsiriX. We used a kernel density estimation (KDE) method available in Python using the scikit-learn library, where the total number of lines of Python code required to generate this tool was 22. Our second example presents a scheme for segmentation of the skeleton from CT datasets. We have demonstrated that good segmentation can be achieved for two example CT studies by using a combination of Python libraries including scikit-learn, scikit-image, SimpleITK and matplotlib. Furthermore, this segmentation method was incorporated into an automatic analysis of quantitative PET-CT in a patient with bone metastases from primary prostate cancer. This enabled repeatable statistical evaluation of PET uptake values for each lesion, before and after treatment, providing estaimes maximum and median standardised uptake values (SUVmax and SUVmed respectively). Following treatment we observed a reduction in lesion volume, SUVmax and SUVmed for all lesions, in agreement with a reduction in concurrent measures of serum prostate-specific antigen (PSA).
Collapse
Affiliation(s)
- Matthew D Blackledge
- CR-UK Cancer Imaging Centre, Radiotherapy and Imaging Division, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom.
| | - David J Collins
- CR-UK Cancer Imaging Centre, Radiotherapy and Imaging Division, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Dow-Mu Koh
- CR-UK Cancer Imaging Centre, Radiotherapy and Imaging Division, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Martin O Leach
- CR-UK Cancer Imaging Centre, Radiotherapy and Imaging Division, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
145
|
Liu B, Shen X, Pan W. Integrative and regularized principal component analysis of multiple sources of data. Stat Med 2016; 35:2235-50. [PMID: 26756854 DOI: 10.1002/sim.6866] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 09/28/2015] [Accepted: 12/14/2015] [Indexed: 12/14/2022]
Abstract
Integration of data of disparate types has become increasingly important to enhancing the power for new discoveries by combining complementary strengths of multiple types of data. One application is to uncover tumor subtypes in human cancer research in which multiple types of genomic data are integrated, including gene expression, DNA copy number, and DNA methylation data. In spite of their successes, existing approaches based on joint latent variable models require stringent distributional assumptions and may suffer from unbalanced scales (or units) of different types of data and non-scalability of the corresponding algorithms. In this paper, we propose an alternative based on integrative and regularized principal component analysis, which is distribution-free, computationally efficient, and robust against unbalanced scales. The new method performs dimension reduction simultaneously on multiple types of data, seeking data-adaptive sparsity and scaling. As a result, in addition to feature selection for each type of data, integrative clustering is achieved. Numerically, the proposed method compares favorably against its competitors in terms of accuracy (in identifying hidden clusters), computational efficiency, and robustness against unbalanced scales. In particular, compared with a popular method, the new method was competitive in identifying tumor subtypes associated with distinct patient survival patterns when applied to a combined analysis of DNA copy number, mRNA expression, and DNA methylation data in a glioblastoma multiforme study. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Binghui Liu
- School of Mathematics and Statistics, Northeast Normal University, Changchun, 130024, Jilin Province, China.,School of Statistics, University of Minnesota, 224 Church St. S.E., Minneapolis, 55455, MN, U.S.A.,Division of Biostatistics, University of Minnesota, 420 Delaware St. S.E., Minneapolis, 55455, MN, U.S.A
| | - Xiaotong Shen
- School of Statistics, University of Minnesota, 224 Church St. S.E., Minneapolis, 55455, MN, U.S.A
| | - Wei Pan
- Division of Biostatistics, University of Minnesota, 420 Delaware St. S.E., Minneapolis, 55455, MN, U.S.A
| |
Collapse
|
146
|
Histone H3F3A and HIST1H3B K27M mutations define two subgroups of diffuse intrinsic pontine gliomas with different prognosis and phenotypes. Acta Neuropathol 2015; 130:815-27. [PMID: 26399631 PMCID: PMC4654747 DOI: 10.1007/s00401-015-1478-0] [Citation(s) in RCA: 456] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/08/2015] [Accepted: 09/10/2015] [Indexed: 01/15/2023]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is the most severe paediatric solid tumour, with no significant therapeutic progress made in the past 50 years. Recent studies suggest that diffuse midline glioma, H3-K27M mutant, may comprise more than one biological entity. The aim of the study was to determine the clinical and biological variables that most impact their prognosis. Ninety-one patients with classically defined DIPG underwent a systematic stereotactic biopsy and were included in this observational retrospective study. Histone H3 genes mutations were assessed by immunochemistry and direct sequencing, whilst global gene expression profiling and chromosomal imbalances were determined by microarrays. A full description of the MRI findings at diagnosis and at relapse was integrated with the molecular profiling data and clinical outcome. All DIPG but one were found to harbour either a somatic H3-K27M mutation and/or loss of H3K27 trimethylation. We also discovered a novel K27M mutation in HIST2H3C, and a lysine-to-isoleucine substitution (K27I) in H3F3A, also creating a loss of trimethylation. Patients with tumours harbouring a K27M mutation in H3.3 (H3F3A) did not respond clinically to radiotherapy as well, relapsed significantly earlier and exhibited more metastatic recurrences than those in H3.1 (HIST1H3B/C). H3.3-K27M-mutated DIPG have a proneural/oligodendroglial phenotype and a pro-metastatic gene expression signature with PDGFRA activation, while H3.1-K27M-mutated tumours exhibit a mesenchymal/astrocytic phenotype and a pro-angiogenic/hypoxic signature supported by expression profiling and radiological findings. H3K27 alterations appear as the founding event in DIPG and the mutations in the two main histone H3 variants drive two distinct oncogenic programmes with potential specific therapeutic targets.
Collapse
|
147
|
Zussman BM, Ozpinar A, Engh JA. Neuroimaging as a Prognostication Tool for Glioblastoma. Neurosurgery 2015; 77:N14-6. [PMID: 26584320 DOI: 10.1227/01.neu.0000473808.37985.f9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
148
|
Rios Velazquez E, Meier R, Dunn Jr WD, Alexander B, Wiest R, Bauer S, Gutman DA, Reyes M, Aerts HJ. Fully automatic GBM segmentation in the TCGA-GBM dataset: Prognosis and correlation with VASARI features. Sci Rep 2015; 5:16822. [PMID: 26576732 PMCID: PMC4649540 DOI: 10.1038/srep16822] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 10/20/2015] [Indexed: 01/22/2023] Open
Abstract
Reproducible definition and quantification of imaging biomarkers is essential. We evaluated a fully automatic MR-based segmentation method by comparing it to manually defined sub-volumes by experienced radiologists in the TCGA-GBM dataset, in terms of sub-volume prognosis and association with VASARI features. MRI sets of 109 GBM patients were downloaded from the Cancer Imaging archive. GBM sub-compartments were defined manually and automatically using the Brain Tumor Image Analysis (BraTumIA). Spearman's correlation was used to evaluate the agreement with VASARI features. Prognostic significance was assessed using the C-index. Auto-segmented sub-volumes showed moderate to high agreement with manually delineated volumes (range (r): 0.4 - 0.86). Also, the auto and manual volumes showed similar correlation with VASARI features (auto r = 0.35, 0.43 and 0.36; manual r = 0.17, 0.67, 0.41, for contrast-enhancing, necrosis and edema, respectively). The auto-segmented contrast-enhancing volume and post-contrast abnormal volume showed the highest AUC (0.66, CI: 0.55-0.77 and 0.65, CI: 0.54-0.76), comparable to manually defined volumes (0.64, CI: 0.53-0.75 and 0.63, CI: 0.52-0.74, respectively). BraTumIA and manual tumor sub-compartments showed comparable performance in terms of prognosis and correlation with VASARI features. This method can enable more reproducible definition and quantification of imaging based biomarkers and has potential in high-throughput medical imaging research.
Collapse
Affiliation(s)
- Emmanuel Rios Velazquez
- Departments of Radiation Oncology and Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Raphael Meier
- Institute for Surgical Technology and Biomechanics , University of Bern, Switzerland
| | - William D. Dunn Jr
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Brian Alexander
- Departments of Radiation Oncology and Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Roland Wiest
- Support Center for Advanced Neuroimaging (SCAN), Institute for Diagnostic and Interventional Neuroradiology, University Hospital Inselspital and University of Bern, Bern, Switzerland
| | - Stefan Bauer
- Institute for Surgical Technology and Biomechanics , University of Bern, Switzerland
- Support Center for Advanced Neuroimaging (SCAN), Institute for Diagnostic and Interventional Neuroradiology, University Hospital Inselspital and University of Bern, Bern, Switzerland
| | - David A. Gutman
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Mauricio Reyes
- Institute for Surgical Technology and Biomechanics , University of Bern, Switzerland
| | - Hugo J.W.L. Aerts
- Departments of Radiation Oncology and Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Departments of Radiology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
149
|
Sathyan P, Zinn PO, Marisetty AL, Liu B, Kamal MM, Singh SK, Bady P, Lu L, Wani KM, Veo BL, Gumin J, Kassem DH, Robinson F, Weng C, Baladandayuthapani V, Suki D, Colman H, Bhat KP, Sulman EP, Aldape K, Colen RR, Verhaak RGW, Lu Z, Fuller GN, Huang S, Lang FF, Sawaya R, Hegi M, Majumder S. Mir-21-Sox2 Axis Delineates Glioblastoma Subtypes with Prognostic Impact. J Neurosci 2015; 35:15097-15112. [PMID: 26558781 PMCID: PMC4642241 DOI: 10.1523/jneurosci.1265-15.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 08/10/2015] [Accepted: 09/11/2015] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Glioblastoma (GBM) is the most aggressive human brain tumor. Although several molecular subtypes of GBM are recognized, a robust molecular prognostic marker has yet to be identified. Here, we report that the stemness regulator Sox2 is a new, clinically important target of microRNA-21 (miR-21) in GBM, with implications for prognosis. Using the MiR-21-Sox2 regulatory axis, approximately half of all GBM tumors present in the Cancer Genome Atlas (TCGA) and in-house patient databases can be mathematically classified into high miR-21/low Sox2 (Class A) or low miR-21/high Sox2 (Class B) subtypes. This classification reflects phenotypically and molecularly distinct characteristics and is not captured by existing classifications. Supporting the distinct nature of the subtypes, gene set enrichment analysis of the TCGA dataset predicted that Class A and Class B tumors were significantly involved in immune/inflammatory response and in chromosome organization and nervous system development, respectively. Patients with Class B tumors had longer overall survival than those with Class A tumors. Analysis of both databases indicated that the Class A/Class B classification is a better predictor of patient survival than currently used parameters. Further, manipulation of MiR-21-Sox2 levels in orthotopic mouse models supported the longer survival of the Class B subtype. The MiR-21-Sox2 association was also found in mouse neural stem cells and in the mouse brain at different developmental stages, suggesting a role in normal development. Therefore, this mechanism-based classification suggests the presence of two distinct populations of GBM patients with distinguishable phenotypic characteristics and clinical outcomes. SIGNIFICANCE STATEMENT Molecular profiling-based classification of glioblastoma (GBM) into four subtypes has substantially increased our understanding of the biology of the disease and has pointed to the heterogeneous nature of GBM. However, this classification is not mechanism based and its prognostic value is limited. Here, we identify a new mechanism in GBM (the miR-21-Sox2 axis) that can classify ∼50% of patients into two subtypes with distinct molecular, radiological, and pathological characteristics. Importantly, this classification can predict patient survival better than the currently used parameters. Further, analysis of the miR-21-Sox2 relationship in mouse neural stem cells and in the mouse brain at different developmental stages indicates that miR-21 and Sox2 are predominantly expressed in mutually exclusive patterns, suggesting a role in normal neural development.
Collapse
Affiliation(s)
- Pratheesh Sathyan
- Departments of Genetics, Department of Clinical Neurosciences, University Hospital (CHUV BH19-110), Lausanne 1011, Switzerland, and
| | | | | | | | | | | | - Pierre Bady
- Department of Clinical Neurosciences, University Hospital (CHUV BH19-110), Lausanne 1011, Switzerland, and
| | | | | | | | | | | | | | | | | | - Dima Suki
- Neurosurgery, The Brain Tumor Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | | | | | - Erik P Sulman
- Neurosurgery, The University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030
| | | | | | - Roel G W Verhaak
- Bioinformatics, and The Brain Tumor Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Zhimin Lu
- Neuro-Oncology, The Brain Tumor Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Gregory N Fuller
- Pathology, The Brain Tumor Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Suyun Huang
- Neurosurgery, The Brain Tumor Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Frederick F Lang
- Neurosurgery, The Brain Tumor Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Raymond Sawaya
- Neurosurgery, The Brain Tumor Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Monika Hegi
- Department of Clinical Neurosciences, University Hospital (CHUV BH19-110), Lausanne 1011, Switzerland, and
| | - Sadhan Majumder
- Departments of Genetics, Neuro-Oncology, The Brain Tumor Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, The University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030
| |
Collapse
|
150
|
Ghosh P, Tamboli P, Vikram R, Rao A. Imaging-genomic pipeline for identifying gene mutations using three-dimensional intra-tumor heterogeneity features. J Med Imaging (Bellingham) 2015; 2:041009. [PMID: 26839909 DOI: 10.1117/1.jmi.2.4.041009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 09/10/2015] [Indexed: 11/14/2022] Open
Abstract
This paper presents an imaging-genomic pipeline to derive three-dimensional intra-tumor heterogeneity features from contrast-enhanced CT images and correlates them with gene mutation status. The pipeline has been demonstrated using CT scans of patients with clear cell renal cell carcinoma (ccRCC) from The Cancer Genome Atlas. About 15% of ccRCC cases reported have BRCA1-associated protein 1 (BAP1) gene alterations that are associated with high tumor grade and poor prognosis. We hypothesized that BAP1 mutation status can be detected using computationally derived image features. The molecular data pertaining to gene mutation status were obtained from the cBioPortal. Correlation of the image features with gene mutation status was assessed using the Mann-Whitney-Wilcoxon rank-sum test. We also used the random forests classifier in the Waikato Environment for Knowledge Analysis software to assess the predictive ability of the computationally derived image features to discriminate cases with BAP1 mutations for ccRCC. Receiver operating characteristics were obtained using a leave-one-out-cross-validation procedure. Our results show that our model can predict BAP1 mutation status with a high degree of sensitivity and specificity. This framework demonstrates a methodology for noninvasive disease biomarker detection from contrast-enhanced CT images.
Collapse
Affiliation(s)
- Payel Ghosh
- University of Texas MD Anderson Cancer Center, Department of Diagnostic Radiology, 1400 Pressler Street, Unit 1459, Houston, Texas 77030, United States; University of Texas MD Anderson Cancer Center, Department of Bioinformatics and Computational Biology, 1400 Pressler Street, Unit 1410, Houston, Texas 77030, United States
| | - Pheroze Tamboli
- University of Texas MD Anderson Cancer Center , Department of Pathology, 1515 Holcombe Boulevard, Unit 0085, Houston, Texas 77030, United States
| | - Raghu Vikram
- University of Texas MD Anderson Cancer Center , Department of Diagnostic Radiology, 1400 Pressler Street, Unit 1459, Houston, Texas 77030, United States
| | - Arvind Rao
- University of Texas MD Anderson Cancer Center , Department of Bioinformatics and Computational Biology, 1400 Pressler Street, Unit 1410, Houston, Texas 77030, United States
| |
Collapse
|