51
|
Peruzzi P, Valdes PQ, Aghi MK, Berger M, Chiocca EA, Golby AJ. The Evolving Role of Neurosurgical Intervention for Central Nervous System Tumors. Hematol Oncol Clin North Am 2021; 36:63-75. [PMID: 34565649 DOI: 10.1016/j.hoc.2021.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Since its inception, greater than a century ago, neurosurgery has represented the fundamental trait-d'union between clinical management, scientific investigation, and therapeutic advancements in the field of brain tumors. During the years, oncological neurosurgery has evolved as a self-standing subspecialty, due to technical progress, equipment improvement, evolution of therapeutic paradigms, and the progressively crucial role that it plays in the execution of complex therapeutic strategies and modern clinical trials.
Collapse
Affiliation(s)
- Pierpaolo Peruzzi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA 02115, USA.
| | - Pablo Q Valdes
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA 02115, USA
| | - Manish K Aghi
- Department of Neurological Surgery, University of California San Francisco, 505 Parnassus Avenue, San Francisco, CA 94117, USA
| | - Mitchel Berger
- Department of Neurological Surgery, University of California San Francisco, 505 Parnassus Avenue, San Francisco, CA 94117, USA
| | - Ennio Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA 02115, USA
| | - Alexandra J Golby
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA 02115, USA; Department of Radiology, Brigham and Women's Hospital/Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA 02115, USA
| |
Collapse
|
52
|
Wu J, Li C, Gensheimer M, Padda S, Kato F, Shirato H, Wei Y, Schönlieb CB, Price SJ, Jaffray D, Heymach J, Neal JW, Loo BW, Wakelee H, Diehn M, Li R. Radiological tumor classification across imaging modality and histology. NAT MACH INTELL 2021; 3:787-798. [PMID: 34841195 PMCID: PMC8612063 DOI: 10.1038/s42256-021-00377-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Radiomics refers to the high-throughput extraction of quantitative features from radiological scans and is widely used to search for imaging biomarkers for prediction of clinical outcomes. Current radiomic signatures suffer from limited reproducibility and generalizability, because most features are dependent on imaging modality and tumor histology, making them sensitive to variations in scan protocol. Here, we propose novel radiological features that are specially designed to ensure compatibility across diverse tissues and imaging contrast. These features provide systematic characterization of tumor morphology and spatial heterogeneity. In an international multi-institution study of 1,682 patients, we discover and validate four unifying imaging subtypes across three malignancies and two major imaging modalities. These tumor subtypes demonstrate distinct molecular characteristics and prognoses after conventional therapies. In advanced lung cancer treated with immunotherapy, one subtype is associated with improved survival and increased tumor-infiltrating lymphocytes compared with the others. Deep learning enables automatic tumor segmentation and reproducible subtype identification, which can facilitate practical implementation. The unifying radiological tumor classification may inform prognosis and treatment response for precision medicine.
Collapse
Affiliation(s)
- Jia Wu
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic and Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Chao Li
- The Centre for Mathematical Imaging in Healthcare, Department of Pure Mathematics and Mathematical Statistics, University of Cambridge, UK
- Cambridge Brain Tumor Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Michael Gensheimer
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sukhmani Padda
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Fumi Kato
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Hiroki Shirato
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yiran Wei
- Cambridge Brain Tumor Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - Stephen John Price
- Cambridge Brain Tumor Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - David Jaffray
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX, USA
- Office of the Chief Technology and Digital Officer, MD Anderson Cancer Center, Houston, TX, USA
| | - John Heymach
- Department of Thoracic and Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Joel W Neal
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Heather Wakelee
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
53
|
Sakai K. [2. Radiomics of MRI]. Nihon Hoshasen Gijutsu Gakkai Zasshi 2021; 77:866-875. [PMID: 34421076 DOI: 10.6009/jjrt.2021_jsrt_77.8.866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Koji Sakai
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| |
Collapse
|
54
|
Maurer GD, Tichy J, Harter PN, Nöth U, Weise L, Quick-Weller J, Deichmann R, Steinbach JP, Bähr O, Hattingen E. Matching Quantitative MRI Parameters with Histological Features of Treatment-Naïve IDH Wild-Type Glioma. Cancers (Basel) 2021; 13:cancers13164060. [PMID: 34439213 PMCID: PMC8392045 DOI: 10.3390/cancers13164060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 11/16/2022] Open
Abstract
Quantitative MRI allows to probe tissue properties by measuring relaxation times and may thus detect subtle changes in tissue composition. In this work we analyzed different relaxation times (T1, T2, T2* and T2') and histological features in 321 samples that were acquired from 25 patients with newly diagnosed IDH wild-type glioma. Quantitative relaxation times before intravenous application of gadolinium-based contrast agent (GBCA), T1 relaxation time after GBCA as well as the relative difference between T1 relaxation times pre-to-post GBCA (T1rel) were compared with histopathologic features such as the presence of tumor cells, cell and vessel density, endogenous markers for hypoxia and cell proliferation. Image-guided stereotactic biopsy allowed for the attribution of each tissue specimen to its corresponding position in the respective relaxation time map. Compared to normal tissue, T1 and T2 relaxation times and T1rel were prolonged in samples containing tumor cells. The presence of vascular proliferates was associated with higher T1rel values. Immunopositivity for lactate dehydrogenase A (LDHA) involved slightly longer T1 relaxation times. However, low T2' values, suggesting high amounts of deoxyhemoglobin, were found in samples with elevated vessel densities, but not in samples with increased immunopositivity for LDHA. Taken together, some of our observations were consistent with previous findings but the correlation of quantitative MRI and histologic parameters did not confirm all our pathophysiology-based assumptions.
Collapse
Affiliation(s)
- Gabriele D. Maurer
- Senckenberg Institute of Neurooncology, Goethe University Hospital, 60528 Frankfurt am Main, Germany; (J.T.); (J.P.S.); (O.B.)
- Correspondence:
| | - Julia Tichy
- Senckenberg Institute of Neurooncology, Goethe University Hospital, 60528 Frankfurt am Main, Germany; (J.T.); (J.P.S.); (O.B.)
| | - Patrick N. Harter
- Institute of Neurology (Edinger Institute), Goethe University Hospital, 60528 Frankfurt am Main, Germany;
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
| | - Ulrike Nöth
- Brain Imaging Center, Goethe University, 60528 Frankfurt am Main, Germany; (U.N.); (R.D.)
| | - Lutz Weise
- Division of Neurosurgery, Dalhousie University Halifax, Halifax, NS B3H 4R2, Canada;
| | - Johanna Quick-Weller
- Department of Neurosurgery, Goethe University Hospital, 60528 Frankfurt am Main, Germany;
| | - Ralf Deichmann
- Brain Imaging Center, Goethe University, 60528 Frankfurt am Main, Germany; (U.N.); (R.D.)
| | - Joachim P. Steinbach
- Senckenberg Institute of Neurooncology, Goethe University Hospital, 60528 Frankfurt am Main, Germany; (J.T.); (J.P.S.); (O.B.)
| | - Oliver Bähr
- Senckenberg Institute of Neurooncology, Goethe University Hospital, 60528 Frankfurt am Main, Germany; (J.T.); (J.P.S.); (O.B.)
- Department of Neurology, Klinikum Aschaffenburg-Alzenau, 63739 Aschaffenburg, Germany
| | - Elke Hattingen
- Institute of Neuroradiology, Goethe University Hospital, 60528 Frankfurt am Main, Germany;
| |
Collapse
|
55
|
Barajas RF, Politi LS, Anzalone N, Schöder H, Fox CP, Boxerman JL, Kaufmann TJ, Quarles CC, Ellingson BM, Auer D, Andronesi OC, Ferreri AJM, Mrugala MM, Grommes C, Neuwelt EA, Ambady P, Rubenstein JL, Illerhaus G, Nagane M, Batchelor TT, Hu LS. Consensus recommendations for MRI and PET imaging of primary central nervous system lymphoma: guideline statement from the International Primary CNS Lymphoma Collaborative Group (IPCG). Neuro Oncol 2021; 23:1056-1071. [PMID: 33560416 PMCID: PMC8248856 DOI: 10.1093/neuonc/noab020] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Advanced molecular and pathophysiologic characterization of primary central nervous system lymphoma (PCNSL) has revealed insights into promising targeted therapeutic approaches. Medical imaging plays a fundamental role in PCNSL diagnosis, staging, and response assessment. Institutional imaging variation and inconsistent clinical trial reporting diminishes the reliability and reproducibility of clinical response assessment. In this context, we aimed to: (1) critically review the use of advanced positron emission tomography (PET) and magnetic resonance imaging (MRI) in the setting of PCNSL; (2) provide results from an international survey of clinical sites describing the current practices for routine and advanced imaging, and (3) provide biologically based recommendations from the International PCNSL Collaborative Group (IPCG) on adaptation of standardized imaging practices. The IPCG provides PET and MRI consensus recommendations built upon previous recommendations for standardized brain tumor imaging protocols (BTIP) in primary and metastatic disease. A biologically integrated approach is provided to addresses the unique challenges associated with the imaging assessment of PCNSL. Detailed imaging parameters facilitate the adoption of these recommendations by researchers and clinicians. To enhance clinical feasibility, we have developed both “ideal” and “minimum standard” protocols at 3T and 1.5T MR systems that will facilitate widespread adoption.
Collapse
Affiliation(s)
- Ramon F Barajas
- Department of Radiology, Neuroradiology Section, Oregon Health & Science University, Portland Oregon, USA.,Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cancer Institute Translational Oncology Program, Oregon Health & Science University, Portland, Oregon, USA
| | - Letterio S Politi
- Humanitas University and Humanitas Research and Clinical Center - IRCCS, Milan, Italy.,Boston Children's Hospital, Boston, Massachusetts, USA
| | - Nicoletta Anzalone
- Neuroradiology Unit, IRCCS San Raffaele Hospital and Vita-Salute University, Milan, Italy
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Christopher P Fox
- Department of Clinical Haematology, Nottingham University Hospitals NHS Trust, School of Medicine, University of Nottingham, Nottingham, UK
| | - Jerrold L Boxerman
- Department of Diagnostic Imaging, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | | | - C Chad Quarles
- Department of Neuroimaging Research & Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, California, USA.,Departments of Radiological Sciences, Psychiatry, and Biobehavioral Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, California, USA
| | - Dorothee Auer
- Versus Arthritis Pain Centre, University of Nottingham, Nottingham, UK.,NIHR Nottingham Biomedical Research Centre, Queen's Medical Centre, University of Nottingham, Nottingham, UK.,Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ovidiu C Andronesi
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Andres J M Ferreri
- Lymphoma Unit, Department of Onco-Hematology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maciej M Mrugala
- Department of Medicine, Division of Hematology and Oncology, Mayo Clinic Cancer Center, Phoenix, Arizona, USA.,Department of Neurology, Mayo Clinic, Phoenix, Arizona, USA
| | - Christian Grommes
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Neurology, Weill Cornell Medical School, New York, New York, USA
| | - Edward A Neuwelt
- Blood-Brain Barrier Program, Oregon Health & Science University, Portland, Oregon, USA.,Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA.,Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon, USA.,Portland Veterans Affairs Medical Center, Portland, Oregon, USA
| | - Prakash Ambady
- Blood-Brain Barrier Program, Oregon Health & Science University, Portland, Oregon, USA.,Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - James L Rubenstein
- Division of Hematology/Oncology, University of California, San Francisco, California, USA.,Department of Medicine, University of California, San Francisco, California, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Gerald Illerhaus
- Clinic of Hematology, Oncology and Palliative Care, Klinikum Stuttgart, Stuttgart, Germany
| | - Motoo Nagane
- Department of Neurosurgery, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Tracy T Batchelor
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Leland S Hu
- Department of Radiology, Neuroradiology Division, Mayo Clinic, Phoenix, Arizona, USA
| |
Collapse
|
56
|
Muster RH, Young JS, Woo PYM, Morshed RA, Warrier G, Kakaizada S, Molinaro AM, Berger MS, Hervey-Jumper SL. The Relationship Between Stimulation Current and Functional Site Localization During Brain Mapping. Neurosurgery 2021; 88:1043-1050. [PMID: 33289525 DOI: 10.1093/neuros/nyaa364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/24/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Gliomas are often in close proximity to functional regions of the brain; therefore, electrocortical stimulation (ECS) mapping is a common technique utilized during glioma resection to identify functional areas. Stimulation-induced seizure (SIS) remains the most common reason for aborted procedures. Few studies have focused on oncological factors impacting cortical stimulation thresholds. OBJECTIVE To examine oncological factors thought to impact stimulation threshold in order to understand whether a linear relationship exists between stimulation current and number of functional cortical sites identified. METHODS We retrospectively reviewed single-institution prospectively collected brain mapping data of patients with dominant hemisphere gliomas. Comparisons of stimulation threshold were made using t-tests and ANOVAs. Associations between oncologic factors and stimulation threshold were made using multivariate regressions. The association between stimulation current and number of positive sites was made using a Poisson model. RESULTS Of the 586 patients included in the study, SIS occurred in 3.92% and the rate of SIS events differed by cortical location (frontal 8.5%, insular 1.6%, parietal 1.3%, and temporal 2.8%; P = .009). Stimulation current was lower when mapping frontal cortex (P = .002). Stimulation current was not associated with tumor plus peritumor edema volume, world health organization) (WHO grade, histology, or isocitrate dehydrogenase (IDH) mutation status but was associated with tumor volume within the frontal lobe (P = .018). Stimulation current was not associated with number of positive sites identified during ECS mapping (P = .118). CONCLUSION SISs are rare but serious events during ECS mapping. SISs are most common when mapping the frontal lobe. Greater stimulation current is not associated with the identification of more cortical functional sites during glioma surgery.
Collapse
Affiliation(s)
- Rachel H Muster
- School of Medicine, University of California, San Francisco, San Francisco, San Francisco, California
| | - Jacob S Young
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Peter Y M Woo
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Ramin A Morshed
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Gayathri Warrier
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Sofia Kakaizada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Shawn L Hervey-Jumper
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| |
Collapse
|
57
|
Pasquini L, Di Napoli A, Napolitano A, Lucignani M, Dellepiane F, Vidiri A, Villani V, Romano A, Bozzao A. Glioblastoma radiomics to predict survival: Diffusion characteristics of surrounding nonenhancing tissue to select patients for extensive resection. J Neuroimaging 2021; 31:1192-1200. [PMID: 34231927 DOI: 10.1111/jon.12903] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Glioblastoma (GBM) is an aggressive primary CNS neoplasm with poor overall survival (OS) despite standard of care. On MRI, GBM is usually characterized by an enhancing portion (CET) (surgery target) and a nonenhancing surrounding (NET). Extent of resection is a long debated issue in GBM, with recent evidence suggesting that both CET and NET should be resected in <65 years old patients, regardless of other risk factors (i.e., molecular biomarkers). Our aim was to test a radiomic model for patient survival stratification in <65 years old patients, by analyzing MRI features of NET, to aid tumor resection. METHODS Sixty-eight <65 years old GBM patients, with extensive CET resection, were selected. Resection was evaluated by manually segmenting CET on volumetric T1-weighted MRI pre and postsurgery (within 72 h). All patients underwent the same treatment protocol including chemoradiation. NET radiomic features were extracted with a custom version of Pyradiomics. Feature selection was performed with principal component analysis (PCA) and its effect on survival tested with Cox regression model. Twelve months OS discrimination was tested by t-test followed by logistic regression. Statistical significance was set at p<0.05. The most relevant features were identified from the component matrix. RESULTS Five PCA components (PC1-5) explained 90% of the variance. PC5 resulted significant in the Cox model (p = 0.002; exp(B) = 0.686), at t-test (p = 0.002) and logistic regression analysis (p = 0.006). Apparent diffusion coefficient (ADC)-based features were the most significant for patient survival stratification. CONCLUSIONS ADC radiomic features on NET predict survival after standard therapy and could be used to improve patient selection for more extensive surgery.
Collapse
Affiliation(s)
- Luca Pasquini
- Neuroradiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, USA.,Neuroradiology Unit, NESMOS Department, Sant'Andrea Hospital, La Sapienza University, Rome, Italy
| | - Alberto Di Napoli
- Neuroradiology Unit, NESMOS Department, Sant'Andrea Hospital, La Sapienza University, Rome, Italy.,Radiology Department, Castelli Romani Hospital, Rome, Italy
| | - Antonio Napolitano
- Medical Physics Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Martina Lucignani
- Medical Physics Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesco Dellepiane
- Neuroradiology Unit, NESMOS Department, Sant'Andrea Hospital, La Sapienza University, Rome, Italy
| | - Antonello Vidiri
- Radiology and Diagnostic Imaging Department, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Veronica Villani
- Neuro-Oncology Unit, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Andrea Romano
- Neuroradiology Unit, NESMOS Department, Sant'Andrea Hospital, La Sapienza University, Rome, Italy
| | - Alessandro Bozzao
- Neuroradiology Unit, NESMOS Department, Sant'Andrea Hospital, La Sapienza University, Rome, Italy
| |
Collapse
|
58
|
Kim MM, Sun Y, Aryal MP, Parmar HA, Piert M, Rosen B, Mayo CS, Balter JM, Schipper M, Gabel N, Briceño EM, You D, Heth J, Al-Holou W, Umemura Y, Leung D, Junck L, Wahl DR, Lawrence TS, Cao Y. A Phase 2 Study of Dose-intensified Chemoradiation Using Biologically Based Target Volume Definition in Patients With Newly Diagnosed Glioblastoma. Int J Radiat Oncol Biol Phys 2021; 110:792-803. [PMID: 33524546 PMCID: PMC8920120 DOI: 10.1016/j.ijrobp.2021.01.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 01/14/2021] [Accepted: 01/21/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE We hypothesized that dose-intensified chemoradiation therapy targeting adversely prognostic hypercellular (TVHCV) and hyperperfused (TVCBV) tumor volumes would improve outcomes in patients with glioblastoma. METHODS AND MATERIALS This single-arm, phase 2 trial enrolled adult patients with newly diagnosed glioblastoma. Patients with a TVHCV/TVCBV >1 cm3, identified using high b-value diffusion-weighted magnetic resonance imaging (MRI) and dynamic contrast-enhanced perfusion MRI, were treated over 30 fractions to 75 Gy to the TVHCV/TVCBV with temozolomide. The primary objective was to estimate improvement in 12-month overall survival (OS) versus historical control. Secondary objectives included evaluating the effect of 3-month TVHCV/TVCBV reduction on OS using Cox proportional-hazard regression and characterizing coverage (95% isodose line) of metabolic tumor volumes identified using correlative 11C-methionine positron emission tomography. Clinically meaningful change was assessed for quality of life by the European Organisation for the Research and Treatment of Cancer Quality of Life Questionnaire C30, for symptom burden by the MD Anderson Symptom Inventory for brain tumor, and for neurocognitive function (NCF) by the Controlled Oral Word Association Test, the Trail Making Test, parts A and B, and the Hopkins Verbal Learning Test-Revised. RESULTS Between 2016 and 2018, 26 patients were enrolled. Initial patients were boosted to TVHCV alone, and 13 patients were boosted to both TVHCV/TVCBV. Gross or subtotal resection was performed in 87% of patients; 22% were O6-methylguanine-DNA methyltransferase (MGMT) methylated. With 26-month follow-up (95% CI, 19-not reached), the 12-month OS rate among patients boosted to the combined TVHCV/TVCBV was 92% (95% CI, 78%-100%; P = .03) and the median OS was 20 months (95% CI, 18-not reached); the median OS for the whole study cohort was 20 months (95% CI, 14-29 months). Patients whose 3-month TVHCV/TVCBV decreased to less than the median volume (3 cm3) had superior OS (29 vs 12 months; P = .02). Only 5 patients had central or in-field failures, and 93% (interquartile range, 59%-100%) of the 11C-methionine metabolic tumor volumes received high-dose coverage. Late grade 3 neurologic toxicity occurred in 2 patients. Among non-progressing patients, 1-month and 7-month deterioration in quality of life, symptoms, and NCF were similar in incidence to standard therapy. CONCLUSIONS Dose intensification against hypercellular/hyperperfused tumor regions in glioblastoma yields promising OS with favorable outcomes for NCF, symptom burden, and quality of life, particularly among patients with greater tumor reduction 3 months after radiation therapy.
Collapse
Affiliation(s)
- Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.
| | - Yilun Sun
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Madhava P Aryal
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Hemant A Parmar
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Morand Piert
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Benjamin Rosen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Charles S Mayo
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - James M Balter
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Matthew Schipper
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Nicolette Gabel
- Department of Physical Medicine and Rehabilitation, University of Michigan, Ann Arbor, Michigan
| | - Emily M Briceño
- Department of Physical Medicine and Rehabilitation, University of Michigan, Ann Arbor, Michigan
| | - Daekeun You
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Jason Heth
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan
| | - Wajd Al-Holou
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan
| | - Yoshie Umemura
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Denise Leung
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Larry Junck
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Yue Cao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Radiology, University of Michigan, Ann Arbor, Michigan; Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
59
|
van Dijken BRJ, Schuuring B, Jeltema HR, van Laar PJ, Enting RH, Dierckx RAJO, Stormezand GN, van der Hoorn A. Ventricle contact may be associated with higher 11C methionine PET uptake in glioblastoma. Neuroradiology 2021; 64:247-252. [PMID: 34114063 PMCID: PMC8789691 DOI: 10.1007/s00234-021-02742-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/30/2021] [Indexed: 11/11/2022]
Abstract
Purpose Ventricle contact is associated with a worse prognosis and more aggressive tumor characteristics in glioblastoma (GBM). This is hypothesized to be a result of neural stem cells located around the lateral ventricles, in the subventricular zone. 11C Methionine positron emission tomography (metPET) is an indicator for increased proliferation, as it shows uptake of methionine, an amino acid needed for protein synthesis. This study is the first to study metPET characteristics of GBM in relation to ventricle contact. Methods A total of 12 patients with IDH wild-type GBM were included. Using MRI, the following regions were determined: primary tumor (defined as contrast enhancing lesion on T1) and peritumoral edema (defined as edema visible on FLAIR excluding the enhancement). PET parameters in these areas were extracted using PET fused with MRI imaging. Parameters extracted from the PET included maximum and mean tumor-to-normal ratio (TNRmax and TNRmean) and metabolic tumor volume (MTV). Results TNRmean of the primary tumor showed significantly higher values for the ventricle-contacting group compared to that for the non-contacting group (4.44 vs 2.67, p = 0.030). Other metPET parameters suggested higher values for the ventricle-contacting group, but these differences did not reach statistical significance. Conclusion GBM with ventricle contact demonstrated a higher methionine uptake and might thus have increased proliferation compared with GBM without ventricle contact. This might explain survival differences and should be considered in treatment decisions. Supplementary Information The online version contains supplementary material available at 10.1007/s00234-021-02742-7.
Collapse
Affiliation(s)
- Bart R J van Dijken
- Department of Radiology, Medical Imaging Center (MIC), University Medical Center Groningen, P.O. Box 30.001, 9700 RB, Groningen, The Netherlands.
| | - Bram Schuuring
- Department of Radiology, Medical Imaging Center (MIC), University Medical Center Groningen, P.O. Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Hanne-Rinck Jeltema
- Department of Neurosurgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter Jan van Laar
- Department of Radiology, Hospital Group Twente, Almelo and Hengelo, The Netherlands
| | - Roelien H Enting
- Department of Neurology, University Medical Center Groningen, Groningen, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine, Medical Imaging Center (MIC), University Medical Center Groningen, Groningen, The Netherlands
| | - Gilles N Stormezand
- Department of Nuclear Medicine, Medical Imaging Center (MIC), University Medical Center Groningen, Groningen, The Netherlands
| | - Anouk van der Hoorn
- Department of Radiology, Medical Imaging Center (MIC), University Medical Center Groningen, P.O. Box 30.001, 9700 RB, Groningen, The Netherlands
| |
Collapse
|
60
|
Azab MA, Ghozy S, Hassanein SF, Azzam AY. Specific Preoperative Dynamic Contrast-Enhanced MRI Semi-quantitative Markers Can Correlate With Vascularity in Specific Areas of Glioblastoma Tissue and Predict Recurrence. Cureus 2021; 13:e15528. [PMID: 34277164 PMCID: PMC8269995 DOI: 10.7759/cureus.15528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Glioblastoma is one of the most aggressive tumours despite all advanced therapies. We aimed to investigate the correlation between qualitative markers of dynamic contrast-enhanced magnetic resonance imaging and vascularity in different tumour regions and elucidate their potential in predicting recurrence. Methods: Radiological markers of vascularity as wash-in rate, washout rate, and capillary time to peak in different single tumour regions were extracted for all glioblastoma patients before being surgically resected using preoperative dynamic contrast-enhanced MRI (DCE-MRI). Tissue samples were obtained from different intratumoral regions and peritumoral oedema and evaluated for the vascular endothelial growth factor (VEGF). Results: Two hundred sixty individuals were included in the final analysis, with 180 dead ones and 80 survivors. Radio- and chemo-therapy were received by all surviving patients and 77.8% (n= 140) of the dead ones. The mean time to peak, in seconds, was longest at the peritumoral oedema region (71.7±23.5), followed by the tumour's necrotic centre (50.0±28.5) and its periphery (2.9±1.8). The expression of VEGF at the peritumoral edema region was inversely correlated to the washout rate at the periphery (r= -0.66; P-value= 0.014) and positively correlated to peritumoral TTP (r= 0.94; P-value< 0.001). Conclusion: Using DCE-MRI, VEGF expression may be used as a non-invasive marker to estimate tumour grade for clinical diagnosis and treatment. Moreover, the risk of glioblastoma recurrence could be determined by evaluating the washout rate at the tumour's periphery. Further large-scale studies are needed to validate the results and to have concrete evidence.
Collapse
Affiliation(s)
- Mohammed A Azab
- Department of Biochemistry, Boise State University, Boise, USA.,Neurological Surgery, Cairo University Hospital, Cairo, EGY
| | - Sherief Ghozy
- Neurological Surgery, Faculty of Medicine, Mansoura University, Mansoura, EGY
| | | | - Ahmed Y Azzam
- Neurological Surgery, October 6 University, 6th of October City, EGY
| |
Collapse
|
61
|
Genovesi LA, Puttick S, Millar A, Kojic M, Ji P, Lagendijk AK, Brighi C, Bonder CS, Adolphe C, Wainwright BJ. Patient-derived orthotopic xenograft models of medulloblastoma lack a functional blood-brain barrier. Neuro Oncol 2021; 23:732-742. [PMID: 33258962 DOI: 10.1093/neuonc/noaa266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Novel targeted therapies for children diagnosed with medulloblastoma (MB), the most common malignant pediatric brain tumor, are urgently required. A major hurdle in the development of effective therapies is the impaired delivery of systemic therapies to tumor cells due to a specialized endothelial blood-brain barrier (BBB). Accordingly, the integrity of the BBB is an essential consideration in any preclinical model used for assessing novel therapeutics. This study sought to assess the functional integrity of the BBB in several preclinical mouse models of MB. METHODS Dynamic contrast enhancement magnetic resonance imaging (MRI) was used to evaluate blood-brain-tumor barrier (BBTB) permeability in a murine genetically engineered mouse model (GEMM) of Sonic Hedgehog (SHH) MB, patient-derived orthotopic xenograft models of MB (SHH and Gp3), and orthotopic transplantation of GEMM tumor cells, enabling a comparison of the direct effects of transplantation on the integrity of the BBTB. Immunofluorescence analysis was performed to compare the structural and subcellular features of tumor-associated vasculature in all models. RESULTS Contrast enhancement was observed in all transplantation models of MB. No contrast enhancement was observed in the GEMM despite significant tumor burden. Cellular analysis of BBTB integrity revealed aberrancies in all transplantation models, correlating to the varying levels of BBTB permeability observed by MRI in these models. CONCLUSIONS These results highlight functional differences in the integrity of the BBTB and tumor vessel phenotype between commonly utilized preclinical models of MB, with important implications for the preclinical evaluation of novel therapeutic agents for MB.
Collapse
Affiliation(s)
- Laura A Genovesi
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Simon Puttick
- Probing Biosystems Future Science Platform, Commonwealth Scientific and Industrial Research Organization, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Amanda Millar
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Marija Kojic
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Pengxiang Ji
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Anne K Lagendijk
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Caterina Brighi
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland, Australia
| | - Claudine S Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Christelle Adolphe
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Brandon J Wainwright
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
62
|
Abstract
The 2016 World Health Organization brain tumor classification is based on genomic and molecular profile of tumor tissue. These characteristics have improved understanding of the brain tumor and played an important role in treatment planning and prognostication. There is an ongoing effort to develop noninvasive imaging techniques that provide insight into tissue characteristics at the cellular and molecular levels. This article focuses on the molecular characteristics of gliomas, transcriptomic subtypes, and radiogenomic studies using semantic and radiomic features. The limitations and future directions of radiogenomics as a standalone diagnostic tool also are discussed.
Collapse
Affiliation(s)
- Chaitra Badve
- Department of Radiology, Division of Neuroradiology, University Hospitals Cleveland Medical Center, BSH 5056, 11100 Euclid Avenue, Cleveland, OH 44106, USA.
| | - Sangam Kanekar
- Department of Radiology and Neurology, Division of Neuroradiology, Penn State College of Medicine, Penn State Milton Hershey Medical Center, Mail Code H066 500, University Drive, Hershey, PA 17033, USA
| |
Collapse
|
63
|
Quantitative mapping of individual voxels in the peritumoral region of IDH-wildtype glioblastoma to distinguish between tumor infiltration and edema. J Neurooncol 2021; 153:251-261. [PMID: 33905055 DOI: 10.1007/s11060-021-03762-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 04/20/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE The peritumoral region (PTR) in glioblastoma (GBM) represents a combination of infiltrative tumor and vasogenic edema, which are indistinguishable on magnetic resonance imaging (MRI). We developed a radiomic signature by using imaging data from low grade glioma (LGG) (marker of tumor) and PTR of brain metastasis (BM) (marker of edema) and applied it on the GBM PTR to generate probabilistic maps. METHODS 270 features were extracted from T1-weighted, T2-weighted, and apparent diffusion coefficient maps in over 3.5 million voxels of LGG (36 segments) and BM (45 segments) scanned in a 1.5T MRI. A support vector machine classifier was used to develop the radiomics model from approximately 50% voxels (downsampled to 10%) and validated with the remaining. The model was applied to over 575,000 voxels of the PTR of 10 patients with GBM to generate a quantitative map using Platt scaling (infiltrative tumor vs. edema). RESULTS The radiomics model had an accuracy of 0.92 and 0.79 in the training and test set, respectively (LGG vs. BM). When extrapolated on the GBM PTR, 9 of 10 patients had a higher percentage of voxels with a tumor-like signature over radiological recurrence areas. In 7 of 10 patients, the areas under curves (AUC) were > 0.50 confirming a positive correlation. Including all the voxels from the GBM patients, the infiltration signature had an AUC of 0.61 to predict recurrence. CONCLUSION A radiomic signature can demarcate areas of microscopic tumors from edema in the PTR of GBM, which correlates with areas of future recurrence.
Collapse
|
64
|
Cluceru J, Nelson SJ, Wen Q, Phillips JJ, Shai A, Molinaro AM, Alcaide-Leon P, Olson MP, Nair D, LaFontaine M, Chunduru P, Villanueva-Meyer JE, Cha S, Chang SM, Berger MS, Lupo JM. Recurrent tumor and treatment-induced effects have different MR signatures in contrast enhancing and non-enhancing lesions of high-grade gliomas. Neuro Oncol 2021; 22:1516-1526. [PMID: 32319527 DOI: 10.1093/neuonc/noaa094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Differentiating treatment-induced injury from recurrent high-grade glioma is an ongoing challenge in neuro-oncology, in part due to lesion heterogeneity. This study aimed to determine whether different MR features were relevant for distinguishing recurrent tumor from the effects of treatment in contrast-enhancing lesions (CEL) and non-enhancing lesions (NEL). METHODS This prospective study analyzed 291 tissue samples (222 recurrent tumor, 69 treatment-effect) with known coordinates on imaging from 139 patients who underwent preoperative 3T MRI and surgery for a suspected recurrence. 8 MR parameter values were tested from perfusion-weighted, diffusion-weighted, and MR spectroscopic imaging at each tissue sample location for association with histopathological outcome using generalized estimating equation models for CEL and NEL tissue samples. Individual cutoff values were evaluated using receiver operating characteristic curve analysis with 5-fold cross-validation. RESULTS In tissue samples obtained from CEL, elevated relative cerebral blood volume (rCBV) was associated with the presence of recurrent tumor pathology (P < 0.03), while increases in normalized choline (nCho) and choline-to-NAA index (CNI) were associated with the presence of recurrent tumor pathology in NEL tissue samples (P < 0.008). A mean CNI cutoff value of 2.7 had the highest performance, resulting in mean sensitivity and specificity of 0.61 and 0.81 for distinguishing treatment-effect from recurrent tumor within the NEL. CONCLUSION Although our results support prior work that underscores the utility of rCBV in distinguishing the effects of treatment from recurrent tumor within the contrast enhancing lesion, we found that metabolic parameters may be better at differentiating recurrent tumor from treatment-related changes in the NEL of high-grade gliomas.
Collapse
Affiliation(s)
- Julia Cluceru
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Qiuting Wen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Joanna J Phillips
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California.,Department of Neurological Surgery, University of California San Francisco, San Francisco, California.,Department of Pathology, University of California San Francisco, San Francisco, California
| | - Anny Shai
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Paula Alcaide-Leon
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Marram P Olson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Devika Nair
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Marisa LaFontaine
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Pranathi Chunduru
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Susan M Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| |
Collapse
|
65
|
Distinct Cerebrovascular Reactivity Patterns for Brain Radiation Necrosis. Cancers (Basel) 2021; 13:cancers13081840. [PMID: 33924308 PMCID: PMC8069508 DOI: 10.3390/cancers13081840] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/31/2021] [Accepted: 04/09/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Current imaging-based discrimination between radiation necrosis versus recurrent glioblastoma contrast-enhancing lesions remains imprecise but is paramount for prognostic and therapeutic evaluation. We examined whether patients with radiation necrosis exhibit distinct patterns of blood oxygenation-level dependent fMRI cerebrovascular reactivity (BOLD-CVR) as the first step to better distinguishing patients with radiation necrosis from recurrent glioblastoma compared with patients with newly diagnosed glioblastoma before surgery and radiotherapy. Methods: Eight consecutive patients with primary and secondary brain tumors and a multidisciplinary clinical and radiological diagnosis of radiation necrosis, and fourteen patients with a first diagnosis of glioblastoma underwent BOLD-CVR mapping. For all these patients, the contrast-enhancing lesion was derived from high-resolution T1-weighted MRI and rendered the volume-of-interest (VOI). From this primary VOI, additional 3 mm concentric expanding VOIs up to 30 mm were created for a detailed perilesional BOLD-CVR tissue analysis between the two groups. Receiver operating characteristic curves assessed the discriminative properties of BOLD-CVR for both groups. Results: Mean intralesional BOLD-CVR values were markedly lower in radiation necrosis than in glioblastoma contrast-enhancing lesions (0.001 ± 0.06 vs. 0.057 ± 0.05; p = 0.04). Perilesionally, a characteristic BOLD-CVR pattern was observed for radiation necrosis and glioblastoma patients, with an improvement of BOLD-CVR values in the radiation necrosis group and persisting lower perilesional BOLD-CVR values in glioblastoma patients. The ROC analysis discriminated against both groups when these two parameters were analyzed together (area under the curve: 0.85, 95% CI: 0.65-1.00). Conclusions: In this preliminary analysis, distinctive intralesional and perilesional BOLD-cerebrovascular reactivity patterns are found for radiation necrosis.
Collapse
|
66
|
Pasquini L, Napolitano A, Tagliente E, Dellepiane F, Lucignani M, Vidiri A, Ranazzi G, Stoppacciaro A, Moltoni G, Nicolai M, Romano A, Di Napoli A, Bozzao A. Deep Learning Can Differentiate IDH-Mutant from IDH-Wild GBM. J Pers Med 2021; 11:290. [PMID: 33918828 PMCID: PMC8069494 DOI: 10.3390/jpm11040290] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022] Open
Abstract
Isocitrate dehydrogenase (IDH) mutant and wildtype glioblastoma multiforme (GBM) often show overlapping features on magnetic resonance imaging (MRI), representing a diagnostic challenge. Deep learning showed promising results for IDH identification in mixed low/high grade glioma populations; however, a GBM-specific model is still lacking in the literature. Our aim was to develop a GBM-tailored deep-learning model for IDH prediction by applying convoluted neural networks (CNN) on multiparametric MRI. We selected 100 adult patients with pathologically demonstrated WHO grade IV gliomas and IDH testing. MRI sequences included: MPRAGE, T1, T2, FLAIR, rCBV and ADC. The model consisted of a 4-block 2D CNN, applied to each MRI sequence. Probability of IDH mutation was obtained from the last dense layer of a softmax activation function. Model performance was evaluated in the test cohort considering categorical cross-entropy loss (CCEL) and accuracy. Calculated performance was: rCBV (accuracy 83%, CCEL 0.64), T1 (accuracy 77%, CCEL 1.4), FLAIR (accuracy 77%, CCEL 1.98), T2 (accuracy 67%, CCEL 2.41), MPRAGE (accuracy 66%, CCEL 2.55). Lower performance was achieved on ADC maps. We present a GBM-specific deep-learning model for IDH mutation prediction, with a maximal accuracy of 83% on rCBV maps. Highest predictivity achieved on perfusion images possibly reflects the known link between IDH and neoangiogenesis through the hypoxia inducible factor.
Collapse
Affiliation(s)
- Luca Pasquini
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
- Neuroradiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Antonio Napolitano
- Medical Physics Department, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy; (E.T.); (M.L.)
| | - Emanuela Tagliente
- Medical Physics Department, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy; (E.T.); (M.L.)
| | - Francesco Dellepiane
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| | - Martina Lucignani
- Medical Physics Department, Bambino Gesù Children’s Hospital, IRCCS, Piazza di Sant’Onofrio, 4, 00165 Rome, Italy; (E.T.); (M.L.)
| | - Antonello Vidiri
- Radiology and Diagnostic Imaging Department, Regina Elena National Cancer Institute, IRCCS, Via Elio Chianesi 53, 00144 Rome, Italy;
| | - Giulio Ranazzi
- Surgical Pathology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (G.R.); (A.S.)
| | - Antonella Stoppacciaro
- Surgical Pathology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (G.R.); (A.S.)
| | - Giulia Moltoni
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| | - Matteo Nicolai
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| | - Andrea Romano
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| | - Alberto Di Napoli
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| | - Alessandro Bozzao
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, Via di Grottarossa 1035, 00189 Rome, Italy; (L.P.); (F.D.); (G.M.); (M.N.); (A.R.); (A.D.N.); (A.B.)
| |
Collapse
|
67
|
d’Este SH, Nielsen MB, Hansen AE. Visualizing Glioma Infiltration by the Combination of Multimodality Imaging and Artificial Intelligence, a Systematic Review of the Literature. Diagnostics (Basel) 2021; 11:diagnostics11040592. [PMID: 33806195 PMCID: PMC8067218 DOI: 10.3390/diagnostics11040592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 03/23/2021] [Indexed: 01/14/2023] Open
Abstract
The aim of this study was to systematically review the literature concerning the integration of multimodality imaging with artificial intelligence methods for visualization of tumor cell infiltration in glioma patients. The review was performed in accordance with the preferred reporting items for systematic reviews and meta-analysis (PRISMA) guidelines. The literature search was conducted in PubMed, Embase, The Cochrane Library and Web of Science and yielded 1304 results. 14 studies were included in the qualitative analysis. The reference standard for tumor infiltration was either histopathology or recurrence on image follow-up. Critical assessment was performed according to the Quality Assessment of Diagnostic Accuracy Studies (QUADAS2). All studies concluded their findings to be of significant value for future clinical practice. Diagnostic test accuracy reached an area under the curve of 0.74–0.91 reported in six studies. There was no consensus with regard to included image modalities, models or training and test strategies. The integration of artificial intelligence with multiparametric imaging shows promise for visualizing tumor cell infiltration in glioma patients. This approach can possibly optimize surgical resection margins and help provide personalized radiotherapy planning.
Collapse
Affiliation(s)
- Sabrina Honoré d’Este
- Department of Diagnostic Radiology, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark; (M.B.N.); (A.E.H.)
- Correspondence:
| | - Michael Bachmann Nielsen
- Department of Diagnostic Radiology, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark; (M.B.N.); (A.E.H.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Adam Espe Hansen
- Department of Diagnostic Radiology, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark; (M.B.N.); (A.E.H.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
68
|
Hong JH, Kang S, Sa JK, Park G, Oh YT, Kim TH, Yin J, Kim SS, D'Angelo F, Koo H, You Y, Park S, Kwon HJ, Kim CI, Ryu H, Lin W, Park EJ, Kim YJ, Park MJ, Kim H, Kim MS, Chung S, Park CK, Park SH, Kang YH, Kim JH, Saya H, Nakano I, Gwak HS, Yoo H, Lee J, Hur EM, Shi B, Nam DH, Iavarone A, Lee SH, Park JB. Modulation of Nogo receptor 1 expression orchestrates myelin-associated infiltration of glioblastoma. Brain 2021; 144:636-654. [PMID: 33479772 DOI: 10.1093/brain/awaa408] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/25/2020] [Accepted: 09/17/2020] [Indexed: 01/19/2023] Open
Abstract
As the clinical failure of glioblastoma treatment is attributed by multiple components, including myelin-associated infiltration, assessment of the molecular mechanisms underlying such process and identification of the infiltrating cells have been the primary objectives in glioblastoma research. Here, we adopted radiogenomic analysis to screen for functionally relevant genes that orchestrate the process of glioma cell infiltration through myelin and promote glioblastoma aggressiveness. The receptor of the Nogo ligand (NgR1) was selected as the top candidate through Differentially Expressed Genes (DEG) and Gene Ontology (GO) enrichment analysis. Gain and loss of function studies on NgR1 elucidated its underlying molecular importance in suppressing myelin-associated infiltration in vitro and in vivo. The migratory ability of glioblastoma cells on myelin is reversibly modulated by NgR1 during differentiation and dedifferentiation process through deubiquitinating activity of USP1, which inhibits the degradation of ID1 to downregulate NgR1 expression. Furthermore, pimozide, a well-known antipsychotic drug, upregulates NgR1 by post-translational targeting of USP1, which sensitizes glioma stem cells to myelin inhibition and suppresses myelin-associated infiltration in vivo. In primary human glioblastoma, downregulation of NgR1 expression is associated with highly infiltrative characteristics and poor survival. Together, our findings reveal that loss of NgR1 drives myelin-associated infiltration of glioblastoma and suggest that novel therapeutic strategies aimed at reactivating expression of NgR1 will improve the clinical outcome of glioblastoma patients.
Collapse
Affiliation(s)
- Jun-Hee Hong
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
- Department of Clinical Research, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Sangjo Kang
- Department of Clinical Research, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Jason K Sa
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Gunwoo Park
- Department of Clinical Research, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Young Taek Oh
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York 10032, USA
| | - Tae Hoon Kim
- Department of Clinical Research, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Jinlong Yin
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
- Henan and Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Sung Soo Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Fulvio D'Angelo
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York 10032, USA
| | - Harim Koo
- Department of Clinical Research, Research Institute and Hospital, National Cancer Center, Goyang, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea
| | - Yeonhee You
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Saewhan Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Hyung Joon Kwon
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Chan Il Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Haseo Ryu
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Weiwei Lin
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Eun Jung Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Youn-Jae Kim
- Division of Translational Science, Research Institute, National Cancer Center, Goyang, Korea
| | - Myung-Jin Park
- Divisions of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hyunggee Kim
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Korea
| | - Mi-Suk Kim
- Department of Neurosurgery and Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul, Korea
| | - Chul-Kee Park
- Neurosurgery, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Hye Park
- Department of Pathology Seoul National University College of Medicine, Seoul, Korea
| | - Yun Hee Kang
- Eulji Biomedical Science Research Institute, Eulji University School of Medicine, Daejeon 34824, Korea
| | - Jong Heon Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Hideyuki Saya
- Division of Gene Regulation, IAMR, Keio University School of Medicine, Tokyo, Japan
| | - Ichiro Nakano
- Research and Development Center for Precision Medicine, Tsukuba University, Japan
| | - Ho-Shin Gwak
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Heon Yoo
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Jeongwu Lee
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eun-Mi Hur
- Department of Neuroscience, Collage of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Bingyang Shi
- Henan and Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Do-Hyun Nam
- Department of Neurosurgery and Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea
| | - Antonio Iavarone
- Institute for Cancer Genetics, Department of Pathology and Neurology, Columbia University Medical Center, New York, 10032 New York, USA
| | - Seung-Hoon Lee
- Department of Neurosurgery, Eulji University School of Medicine, Daejeon 34824, Korea
| | - Jong Bae Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
- Department of Clinical Research, Research Institute and Hospital, National Cancer Center, Goyang, Korea
- Henan and Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, China
| |
Collapse
|
69
|
Hu LS, Wang L, Hawkins-Daarud A, Eschbacher JM, Singleton KW, Jackson PR, Clark-Swanson K, Sereduk CP, Peng S, Wang P, Wang J, Baxter LC, Smith KA, Mazza GL, Stokes AM, Bendok BR, Zimmerman RS, Krishna C, Porter AB, Mrugala MM, Hoxworth JM, Wu T, Tran NL, Swanson KR, Li J. Uncertainty quantification in the radiogenomics modeling of EGFR amplification in glioblastoma. Sci Rep 2021; 11:3932. [PMID: 33594116 PMCID: PMC7886858 DOI: 10.1038/s41598-021-83141-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
Radiogenomics uses machine-learning (ML) to directly connect the morphologic and physiological appearance of tumors on clinical imaging with underlying genomic features. Despite extensive growth in the area of radiogenomics across many cancers, and its potential role in advancing clinical decision making, no published studies have directly addressed uncertainty in these model predictions. We developed a radiogenomics ML model to quantify uncertainty using transductive Gaussian Processes (GP) and a unique dataset of 95 image-localized biopsies with spatially matched MRI from 25 untreated Glioblastoma (GBM) patients. The model generated predictions for regional EGFR amplification status (a common and important target in GBM) to resolve the intratumoral genetic heterogeneity across each individual tumor-a key factor for future personalized therapeutic paradigms. The model used probability distributions for each sample prediction to quantify uncertainty, and used transductive learning to reduce the overall uncertainty. We compared predictive accuracy and uncertainty of the transductive learning GP model against a standard GP model using leave-one-patient-out cross validation. Additionally, we used a separate dataset containing 24 image-localized biopsies from 7 high-grade glioma patients to validate the model. Predictive uncertainty informed the likelihood of achieving an accurate sample prediction. When stratifying predictions based on uncertainty, we observed substantially higher performance in the group cohort (75% accuracy, n = 95) and amongst sample predictions with the lowest uncertainty (83% accuracy, n = 72) compared to predictions with higher uncertainty (48% accuracy, n = 23), due largely to data interpolation (rather than extrapolation). On the separate validation set, our model achieved 78% accuracy amongst the sample predictions with lowest uncertainty. We present a novel approach to quantify radiogenomics uncertainty to enhance model performance and clinical interpretability. This should help integrate more reliable radiogenomics models for improved medical decision-making.
Collapse
Affiliation(s)
- Leland S Hu
- Department of Radiology, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA. .,School of Computing, Informatics, and Decision Systems Engineering, Arizona State University, 699 S Mill Ave, Tempe, AZ, 85281, USA. .,Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic Arizona, 5777 East Mayo Blvd, Support Services Building Suite 2-700, Phoenix, AZ, 85054, USA.
| | - Lujia Wang
- Department of Radiology, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA.,School of Computing, Informatics, and Decision Systems Engineering, Arizona State University, 699 S Mill Ave, Tempe, AZ, 85281, USA.,Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic Arizona, 5777 East Mayo Blvd, Support Services Building Suite 2-700, Phoenix, AZ, 85054, USA
| | - Andrea Hawkins-Daarud
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic Arizona, 5777 East Mayo Blvd, Support Services Building Suite 2-700, Phoenix, AZ, 85054, USA
| | - Jennifer M Eschbacher
- Department of Pathology, Barrow Neurological Institute-St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - Kyle W Singleton
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic Arizona, 5777 East Mayo Blvd, Support Services Building Suite 2-700, Phoenix, AZ, 85054, USA
| | - Pamela R Jackson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic Arizona, 5777 East Mayo Blvd, Support Services Building Suite 2-700, Phoenix, AZ, 85054, USA
| | - Kamala Clark-Swanson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic Arizona, 5777 East Mayo Blvd, Support Services Building Suite 2-700, Phoenix, AZ, 85054, USA
| | - Christopher P Sereduk
- Department of Neurosurgery, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA.,Department of Cancer Biology, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Sen Peng
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
| | - Panwen Wang
- Department of Quantitative Health Sciences, Center for Individualized Medicine, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Junwen Wang
- Department of Quantitative Health Sciences, Center for Individualized Medicine, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Leslie C Baxter
- Department of Neuropsychology, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Kris A Smith
- Department of Neurosurgery, Barrow Neurological Institute-St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - Gina L Mazza
- Department of Quantitative Health Sciences, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Ashley M Stokes
- Department of Imaging Research, Barrow Neurological Institute-St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - Bernard R Bendok
- Department of Neurosurgery, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Richard S Zimmerman
- Department of Neurosurgery, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Chandan Krishna
- Department of Neurosurgery, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Alyx B Porter
- Department of Neuro-Oncology, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Maciej M Mrugala
- Department of Neuro-Oncology, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Joseph M Hoxworth
- Department of Radiology, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Teresa Wu
- Department of Radiology, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA.,School of Computing, Informatics, and Decision Systems Engineering, Arizona State University, 699 S Mill Ave, Tempe, AZ, 85281, USA
| | - Nhan L Tran
- Department of Neurosurgery, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA.,Department of Cancer Biology, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Kristin R Swanson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic Arizona, 5777 East Mayo Blvd, Support Services Building Suite 2-700, Phoenix, AZ, 85054, USA.,Department of Neurosurgery, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Jing Li
- Department of Radiology, Mayo Clinic Arizona, 5777 E. Mayo Blvd, Phoenix, AZ, 85054, USA.,School of Computing, Informatics, and Decision Systems Engineering, Arizona State University, 699 S Mill Ave, Tempe, AZ, 85281, USA.,Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic Arizona, 5777 East Mayo Blvd, Support Services Building Suite 2-700, Phoenix, AZ, 85054, USA
| |
Collapse
|
70
|
Abstract
PURPOSE OF REVIEW Radiogenomics is a growing field that has garnered immense interest over the past decade, owing to its numerous applications in the field of oncology and its potential value in improving patient outcomes. Current applications have only begun to delve into the potential of radiogenomics, and particularly in interventional oncology, there is room for development and increased value of these applications. RECENT FINDINGS The field of interventional oncology (IO) has seen valuable radiogenomic applications, from prediction of response to locoregional therapies in hepatocellular carcinoma to identification of genetic mutations in non-small cell lung cancer. Future directions that can increase the value of radiogenomics include applications that address tumor heterogeneity, predict immune responsiveness of tumors, and differentiate between oligoprogression and early widespread progression, among others. Radiogenomics, whether in terms of methodologies or applications, is still in the early stages of development and far from maturation. Future applications, particularly in the field of interventional oncology, will allow realization of its full potential.
Collapse
|
71
|
Gates EDH, Weinberg JS, Prabhu SS, Lin JS, Hamilton J, Hazle JD, Fuller GN, Baladandayuthapani V, Fuentes DT, Schellingerhout D. Estimating Local Cellular Density in Glioma Using MR Imaging Data. AJNR Am J Neuroradiol 2021; 42:102-108. [PMID: 33243897 PMCID: PMC7814791 DOI: 10.3174/ajnr.a6884] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/22/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND PURPOSE Increased cellular density is a hallmark of gliomas, both in the bulk of the tumor and in areas of tumor infiltration into surrounding brain. Altered cellular density causes altered imaging findings, but the degree to which cellular density can be quantitatively estimated from imaging is unknown. The purpose of this study was to discover the best MR imaging and processing techniques to make quantitative and spatially specific estimates of cellular density. MATERIALS AND METHODS We collected stereotactic biopsies in a prospective imaging clinical trial targeting untreated patients with gliomas at our institution undergoing their first resection. The data included preoperative MR imaging with conventional anatomic, diffusion, perfusion, and permeability sequences and quantitative histopathology on biopsy samples. We then used multiple machine learning methodologies to estimate cellular density using local intensity information from the MR images and quantitative cellular density measurements at the biopsy coordinates as the criterion standard. RESULTS The random forest methodology estimated cellular density with R 2 = 0.59 between predicted and observed values using 4 input imaging sequences chosen from our full set of imaging data (T2, fractional anisotropy, CBF, and area under the curve from permeability imaging). Limiting input to conventional MR images (T1 pre- and postcontrast, T2, and FLAIR) yielded slightly degraded performance (R2 = 0.52). Outputs were also reported as graphic maps. CONCLUSIONS Cellular density can be estimated with moderate-to-strong correlations using MR imaging inputs. The random forest machine learning model provided the best estimates. These spatially specific estimates of cellular density will likely be useful in guiding both diagnosis and treatment.
Collapse
Affiliation(s)
- E D H Gates
- From the Departments of Imaging Physics (E.D.H.G., J.S.L., J.D.H., D.T.F.)
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (E.D.H.G.), Houston, Texas
| | | | | | - J S Lin
- From the Departments of Imaging Physics (E.D.H.G., J.S.L., J.D.H., D.T.F.)
- Baylor College of Medicine (J.S.L.), Houston, Texas
- Department of Bioengineering (J.S.L.), Rice University, Houston, Texas
| | - J Hamilton
- Neuroradiology (J.H., D.S.)
- Radiology Partners (J.H.), Houston, Texas
| | - J D Hazle
- From the Departments of Imaging Physics (E.D.H.G., J.S.L., J.D.H., D.T.F.)
| | | | - V Baladandayuthapani
- Department of Computational Medicine and Bioinformatics (V.B.), University of Michigan School of Public Health, Ann Arbor, Michigan
| | - D T Fuentes
- From the Departments of Imaging Physics (E.D.H.G., J.S.L., J.D.H., D.T.F.)
| | - D Schellingerhout
- Neuroradiology (J.H., D.S.)
- Cancer Systems Imaging (D.S.), University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
72
|
Riva M, Lopci E, Gay LG, Nibali MC, Rossi M, Sciortino T, Castellano A, Bello L. Advancing Imaging to Enhance Surgery: From Image to Information Guidance. Neurosurg Clin N Am 2021; 32:31-46. [PMID: 33223024 DOI: 10.1016/j.nec.2020.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Conventional magnetic resonance imaging (cMRI) has an established role as a crucial disease parameter in the multidisciplinary management of glioblastoma, guiding diagnosis, treatment planning, assessment, and follow-up. Yet, cMRI cannot provide adequate information regarding tissue heterogeneity and the infiltrative extent beyond the contrast enhancement. Advanced magnetic resonance imaging and PET and newer analytical methods are transforming images into data (radiomics) and providing noninvasive biomarkers of molecular features (radiogenomics), conveying enhanced information for improving decision making in surgery. This review analyzes the shift from image guidance to information guidance that is relevant for the surgical treatment of glioblastoma.
Collapse
Affiliation(s)
- Marco Riva
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Festa del Perdono 7, Milan 20122, Italy; IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy.
| | - Egesta Lopci
- Unit of Nuclear Medicine, Humanitas Clinical and Research Center - IRCCS, Via Manzoni 56, Rozzano, Milan 20089, Italy. https://twitter.com/LopciEgesta
| | - Lorenzo G Gay
- IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy; Department of Oncology and Hemato-Oncology, Via Festa del Perdono 7, Milan 20122, Italy
| | - Marco Conti Nibali
- IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy; Department of Oncology and Hemato-Oncology, Via Festa del Perdono 7, Milan 20122, Italy. https://twitter.com/dr_mcn
| | - Marco Rossi
- IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy; Department of Oncology and Hemato-Oncology, Via Festa del Perdono 7, Milan 20122, Italy
| | - Tommaso Sciortino
- IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy; Department of Oncology and Hemato-Oncology, Via Festa del Perdono 7, Milan 20122, Italy
| | - Antonella Castellano
- Neuroradiology Unit and CERMAC, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan 20123, Italy. https://twitter.com/antocastella
| | - Lorenzo Bello
- IRCCS Istituto Ortopedico Galeazzi, U.O. Neurochirurgia Oncologica, Milan, Italy; Department of Oncology and Hemato-Oncology, Via Festa del Perdono 7, Milan 20122, Italy
| |
Collapse
|
73
|
Hu LS, Brat DJ, Bloch O, Ramkissoon S, Lesser GJ. The Practical Application of Emerging Technologies Influencing the Diagnosis and Care of Patients With Primary Brain Tumors. Am Soc Clin Oncol Educ Book 2020; 40:1-12. [PMID: 32324425 DOI: 10.1200/edbk_280955] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the past decade, a variety of new and innovative technologies has led to important advances in the diagnosis and management of patients with primary malignant brain tumors. New approaches to surgical navigation and tumor localization, advanced imaging to define tumor biology and treatment response, and the widespread adoption of a molecularly defined integrated diagnostic paradigm that complements traditional histopathologic diagnosis continue to impact the day-to-day care of these patients. In the neuro-oncology clinic, discussions with patients about the role of tumor treating fields (TTFields) and the incorporation of next-generation sequencing (NGS) data into therapeutic decision-making are now a standard practice. This article summarizes newer applications of technology influencing the pathologic, neuroimaging, neurosurgical, and medical management of patients with malignant primary brain tumors.
Collapse
Affiliation(s)
- Leland S Hu
- Neuroradiology Section, Department of Radiology, Mayo Clinic, Phoenix, AZ
| | - Daniel J Brat
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Orin Bloch
- Department of Neurologic Surgery, UC Davis Comprehensive Cancer Center, Sacramento, CA
| | - Shakti Ramkissoon
- Foundation Medicine, Inc., Morrisville, NC.,Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC.,Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Glenn J Lesser
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC
| |
Collapse
|
74
|
Wan Y, Rahmat R, Price SJ. Deep learning for glioblastoma segmentation using preoperative magnetic resonance imaging identifies volumetric features associated with survival. Acta Neurochir (Wien) 2020; 162:3067-3080. [PMID: 32662042 PMCID: PMC7593295 DOI: 10.1007/s00701-020-04483-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/02/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Measurement of volumetric features is challenging in glioblastoma. We investigate whether volumetric features derived from preoperative MRI using a convolutional neural network-assisted segmentation is correlated with survival. METHODS Preoperative MRI of 120 patients were scored using Visually Accessible Rembrandt Images (VASARI) features. We trained and tested a multilayer, multi-scale convolutional neural network on multimodal brain tumour segmentation challenge (BRATS) data, prior to testing on our dataset. The automated labels were manually edited to generate ground truth segmentations. Network performance for our data and BRATS data was compared. Multivariable Cox regression analysis corrected for multiple testing using the false discovery rate was performed to correlate clinical and imaging variables with overall survival. RESULTS Median Dice coefficients in our sample were (1) whole tumour 0.94 (IQR, 0.82-0.98) compared to 0.91 (IQR, 0.83-0.94 p = 0.012), (2) FLAIR region 0.84 (IQR, 0.63-0.95) compared to 0.81 (IQR, 0.69-0.8 p = 0.170), (3) contrast-enhancing region 0.91 (IQR, 0.74-0.98) compared to 0.83 (IQR, 0.78-0.89 p = 0.003) and (4) necrosis region were 0.82 (IQR, 0.47-0.97) compared to 0.67 (IQR, 0.42-0.81 p = 0.005). Contrast-enhancing region/tumour core ratio (HR 4.73 [95% CI, 1.67-13.40], corrected p = 0.017) and necrotic core/tumour core ratio (HR 8.13 [95% CI, 2.06-32.12], corrected p = 0.011) were independently associated with overall survival. CONCLUSION Semi-automated segmentation of glioblastoma using a convolutional neural network trained on independent data is robust when applied to routine clinical data. The segmented volumes have prognostic significance.
Collapse
|
75
|
Kang Y, Hong EK, Rhim JH, Yoo RE, Kang KM, Yun TJ, Kim JH, Sohn CH, Park SW, Choi SH. Prognostic Value of Dynamic Contrast-Enhanced MRI-Derived Pharmacokinetic Variables in Glioblastoma Patients: Analysis of Contrast-Enhancing Lesions and Non-Enhancing T2 High-Signal Intensity Lesions. Korean J Radiol 2020; 21:707-716. [PMID: 32410409 PMCID: PMC7231611 DOI: 10.3348/kjr.2019.0629] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/31/2019] [Accepted: 02/09/2020] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE To evaluate pharmacokinetic variables from contrast-enhancing lesions (CELs) and non-enhancing T2 high signal intensity lesions (NE-T2HSILs) on dynamic contrast-enhanced (DCE) magnetic resonance (MR) imaging for predicting progression-free survival (PFS) in glioblastoma (GBM) patients. MATERIALS AND METHODS Sixty-four GBM patients who had undergone preoperative DCE MR imaging and received standard treatment were retrospectively included. We analyzed the pharmacokinetic variables of the volume transfer constant (Ktrans) and volume fraction of extravascular extracellular space within the CEL and NE-T2HSIL of the entire tumor. Univariate and multivariate Cox regression analyses were performed using preoperative clinical characteristics, pharmacokinetic variables of DCE MR imaging, and postoperative molecular biomarkers to predict PFS. RESULTS The increased mean Ktrans of the CEL, increased 95th percentile Ktrans of the CELs, and absence of methylated O⁶-methylguanine-DNA methyltransferase promoter were relevant adverse variables for PFS in the univariate analysis (p = 0.041, p = 0.032, and p = 0.083, respectively). The Kaplan-Meier survival curves demonstrated that PFS was significantly shorter in patients with a mean Ktrans of the CEL > 0.068 and 95th percentile Ktrans of the CEL>0.223 (log-rank p = 0.038 and p = 0.041, respectively). However, only mean Ktrans of the CEL was significantly associated with PFS (p = 0.024; hazard ratio, 553.08; 95% confidence interval, 2.27-134756.74) in the multivariate Cox proportional hazard analysis. None of the pharmacokinetic variables from NE-T2HSILs were significantly related to PFS. CONCLUSION Among the pharmacokinetic variables extracted from CELs and NE-T2HSILs on preoperative DCE MR imaging, the mean Ktrans of CELs exhibits potential as a useful imaging predictor of PFS in GBM patients.
Collapse
Affiliation(s)
- Yeonah Kang
- Department of Radiology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea.,Department of Radiology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Eun Kyoung Hong
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Hyo Rhim
- Department of Radiology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Roh Eul Yoo
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Tae Jin Yun
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Hoon Kim
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Chul Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Sun Won Park
- Department of Radiology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.
| | - Seung Hong Choi
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
76
|
Radiomic analysis of magnetic resonance fingerprinting in adult brain tumors. Eur J Nucl Med Mol Imaging 2020; 48:683-693. [PMID: 32979059 DOI: 10.1007/s00259-020-05037-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE This is a radiomics study investigating the ability of texture analysis of MRF maps to improve differentiation between intra-axial adult brain tumors and to predict survival in the glioblastoma cohort. METHODS Magnetic resonance fingerprinting (MRF) acquisition was performed on 31 patients across 3 groups: 17 glioblastomas, 6 low-grade gliomas, and 8 metastases. Using regions of interest for the solid tumor and peritumoral white matter on T1 and T2 maps, second-order texture features were calculated from gray-level co-occurrence matrices and gray-level run length matrices. Selected features were compared across the three tumor groups using Wilcoxon rank-sum test. Receiver operating characteristic curve analysis was performed for each feature. Kaplan-Meier method was used for survival analysis with log rank tests. RESULTS Low-grade gliomas and glioblastomas had significantly higher run percentage, run entropy, and information measure of correlation 1 on T1 than metastases (p < 0.017). The best separation of all three tumor types was seen utilizing inverse difference normalized and homogeneity values for peritumoral white matter in both T1 and T2 maps (p < 0.017). In solid tumor T2 maps, lower values in entropy and higher values of maximum probability and high-gray run emphasis were associated with longer survival in glioblastoma patients (p < 0.05). Several texture features were associated with longer survival in glioblastoma patients on peritumoral white matter T1 maps (p < 0.05). CONCLUSION Texture analysis of MRF-derived maps can improve our ability to differentiate common adult brain tumors by characterizing tumor heterogeneity, and may have a role in predicting outcomes in patients with glioblastoma.
Collapse
|
77
|
Yuan T, Ying J, Zuo Z, Jin L, Gui S, Gao Z, Li G, Wang R, Zhang Y, Li C. Contrahemispheric Cortex Predicts Survival and Molecular Markers in Patients With Unilateral High-Grade Gliomas. Front Oncol 2020; 10:953. [PMID: 32793462 PMCID: PMC7390929 DOI: 10.3389/fonc.2020.00953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/15/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Malignant high-grade gliomas are characterized by infiltration and destruction of surrounding brain tissue. Alterations in the contrahemispheric brain structure and their roles that may offer prognostically valuable information have not been investigated in high-grade gliomas. Methods: In total, 153 patients with unilateral glioma (low-grade, n = 77; high-grade, n = 76) and 115 healthy controls (HCs) were recruited and scanned with 3-D T1 imaging. The gray matter (GM), white matter (WM), and cerebrospinal fluid (CSF) volume in the contrahemisphere were examined. Partial correlation, logistic regression, and multivariate Cox's regression analyses were performed. Results: The contrahemispheric GM volume (CHGMV) in the high-grade glioma patients was significantly decreased compared to that in the HCs/low-grade gliomas (one-way ANOVA, Bonferroni corrected, p < 0.05). The CHGMV is significantly correlated with the WHO grade (r = -0.22, p < 0.05) and contrast-enhanced volume (r = -0.33, p < 0.01). In the high-grade gliomas, the binary logistic regression revealed that the CHGMV can independently predict isocitrate dehydrogenase 1 (IDH1) and P53 mutations. The survival curves revealed that the patients with a low CHGMV had a shorter overall survival (OS) than the patients with a high CHGMV (p = 0.001). The multivariate Cox's regression analysis showed that a low CHGMV can independently predict unfavorable OS with a hazard ratio of 2.883 (p = 0.035). Conclusions: Volume of the contrahemispheric cortex can be potentially used in clinical practice as an imaging biomarker to predict survival and molecular markers in patients with unilateral high-grade gliomas.
Collapse
Affiliation(s)
- Taoyang Yuan
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jianyou Ying
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhentao Zuo
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Lu Jin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhixian Gao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guilin Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Rui Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Beijing Institute for Brain Disorders Brain Tumor Center, Beijing, China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders Brain Tumor Center, Beijing, China
| |
Collapse
|
78
|
Gates EDH, Lin JS, Weinberg JS, Hamilton J, Prabhu SS, Hazle JD, Fuller GN, Baladandayuthapani V, Fuentes D, Schellingerhout D. Guiding the first biopsy in glioma patients using estimated Ki-67 maps derived from MRI: conventional versus advanced imaging. Neuro Oncol 2020; 21:527-536. [PMID: 30657997 DOI: 10.1093/neuonc/noz004] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Undersampling of gliomas at first biopsy is a major clinical problem, as accurate grading determines all subsequent treatment. We submit a technological solution to reduce the problem of undersampling by estimating a marker of tumor proliferation (Ki-67) using MR imaging data as inputs, against a stereotactic histopathology gold standard. METHODS MR imaging was performed with anatomic, diffusion, permeability, and perfusion sequences, in untreated glioma patients in a prospective clinical trial. Stereotactic biopsies were harvested from each patient immediately prior to surgical resection. For each biopsy, an imaging description (23 parameters) was developed, and the Ki-67 index was recorded. Machine learning models were built to estimate Ki-67 from imaging inputs, and cross validation was undertaken to determine the error in estimates. The best model was used to generate graphical maps of Ki-67 estimates across the whole brain. RESULTS Fifty-two image-guided biopsies were collected from 23 evaluable patients. The random forest algorithm best modeled Ki-67 with 4 imaging inputs (T2-weighted, fractional anisotropy, cerebral blood flow, Ktrans). It predicted the Ki-67 expression levels with a root mean square (RMS) error of 3.5% (R2 = 0.75). A less accurate predictive result (RMS error 5.4%, R2 = 0.50) was found using conventional imaging only. CONCLUSION Ki-67 can be predicted to clinically useful accuracies using clinical imaging data. Advanced imaging (diffusion, perfusion, and permeability) improves predictive accuracy over conventional imaging alone. Ki-67 predictions, displayed as graphical maps, could be used to guide biopsy, resection, and/or radiation in the care of glioma patients.
Collapse
Affiliation(s)
- Evan D H Gates
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center (UT MDACC), Houston, Texas.,UT MDACC UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Jonathan S Lin
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center (UT MDACC), Houston, Texas.,Baylor College of Medicine, Houston, Texas.,Department of Bioengineering, Rice University, Houston, Texas
| | | | - Jackson Hamilton
- Department of Diagnostic Radiology, UT MDACC, Houston, Texas.,Radiology Partners, Houston, Texas
| | | | - John D Hazle
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center (UT MDACC), Houston, Texas
| | | | | | - David Fuentes
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center (UT MDACC), Houston, Texas
| | - Dawid Schellingerhout
- Department of Diagnostic Radiology, UT MDACC, Houston, Texas.,Department of Cancer Systems Imaging, UT MDACC, Houston, Texas
| |
Collapse
|
79
|
John F, Bosnyák E, Robinette NL, Amit-Yousif AJ, Barger GR, Shah KD, Michelhaugh SK, Klinger NV, Mittal S, Juhász C. Multimodal imaging-defined subregions in newly diagnosed glioblastoma: impact on overall survival. Neuro Oncol 2020; 21:264-273. [PMID: 30346623 DOI: 10.1093/neuonc/noy169] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Although glioblastomas are heterogeneous brain-infiltrating tumors, their treatment is mostly focused on the contrast-enhancing tumor mass. In this study, we combined conventional MRI, diffusion-weighted imaging (DWI), and amino acid PET to explore imaging-defined glioblastoma subregions and evaluate their potential prognostic value. METHODS Contrast-enhanced T1, T2/fluid attenuated inversion recovery (FLAIR) MR images, apparent diffusion coefficient (ADC) maps from DWI, and alpha-[11C]-methyl-L-tryptophan (AMT)-PET images were analyzed in 30 patients with newly diagnosed glioblastoma. Five tumor subregions were identified based on a combination of MRI contrast enhancement, T2/FLAIR signal abnormalities, and AMT uptake on PET. ADC and AMT uptake tumor/contralateral normal cortex (T/N) ratios in these tumor subregions were correlated, and their prognostic value was determined. RESULTS A total of 115 MRI/PET-defined subregions were analyzed. Most tumors showed not only a high-AMT uptake (T/N ratio > 1.65, N = 27) but also a low-uptake subregion (N = 21) within the contrast-enhancing tumor mass. High AMT uptake extending beyond contrast enhancement was also common (N = 25) and was associated with low ADC (r = -0.40, P = 0.05). Higher AMT uptake in the contrast-enhancing tumor subregions was strongly prognostic for overall survival (hazard ratio: 7.83; 95% CI: 1.98-31.02, P = 0.003), independent of clinical and molecular genetic prognostic variables. Nonresected high-AMT uptake subregions predicted the sites of tumor progression on posttreatment PET performed in 10 patients. CONCLUSIONS Glioblastomas show heterogeneous amino acid uptake with high-uptake regions often extending into non-enhancing brain with high cellularity; nonresection of these predict the site of posttreatment progression. High tryptophan uptake values in MRI contrast-enhancing tumor subregions are a strong, independent imaging marker for longer overall survival.
Collapse
Affiliation(s)
- Flóra John
- Department of Pediatrics, Wayne State University, Detroit, Michigan.,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, Michigan
| | - Edit Bosnyák
- Department of Pediatrics, Wayne State University, Detroit, Michigan.,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, Michigan
| | - Natasha L Robinette
- Department of Oncology, Wayne State University, Detroit, Michigan.,Department of Radiology, Wayne State University, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| | - Alit J Amit-Yousif
- Department of Oncology, Wayne State University, Detroit, Michigan.,Department of Radiology, Wayne State University, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| | - Geoffrey R Barger
- Department of Neurology, Wayne State University, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| | - Keval D Shah
- Department of Neurology, Wayne State University, Detroit, Michigan
| | | | | | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University, Detroit, Michigan.,Department of Oncology, Wayne State University, Detroit, Michigan.,Department of Biomedical Engineering, Wayne State University, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| | - Csaba Juhász
- Department of Pediatrics, Wayne State University, Detroit, Michigan.,Department of Neurology, Wayne State University, Detroit, Michigan.,Department of Neurosurgery, Wayne State University, Detroit, Michigan.,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| |
Collapse
|
80
|
Hoxworth JM, Eschbacher JM, Gonzales AC, Singleton KW, Leon GD, Smith KA, Stokes AM, Zhou Y, Mazza GL, Porter AB, Mrugala MM, Zimmerman RS, Bendok BR, Patra DP, Krishna C, Boxerman JL, Baxter LC, Swanson KR, Quarles CC, Schmainda KM, Hu LS. Performance of Standardized Relative CBV for Quantifying Regional Histologic Tumor Burden in Recurrent High-Grade Glioma: Comparison against Normalized Relative CBV Using Image-Localized Stereotactic Biopsies. AJNR Am J Neuroradiol 2020; 41:408-415. [PMID: 32165359 DOI: 10.3174/ajnr.a6486] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/23/2019] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Perfusion MR imaging measures of relative CBV can distinguish recurrent tumor from posttreatment radiation effects in high-grade gliomas. Currently, relative CBV measurement requires normalization based on user-defined reference tissues. A recently proposed method of relative CBV standardization eliminates the need for user input. This study compares the predictive performance of relative CBV standardization against relative CBV normalization for quantifying recurrent tumor burden in high-grade gliomas relative to posttreatment radiation effects. MATERIALS AND METHODS We recruited 38 previously treated patients with high-grade gliomas (World Health Organization grades III or IV) undergoing surgical re-resection for new contrast-enhancing lesions concerning for recurrent tumor versus posttreatment radiation effects. We recovered 112 image-localized biopsies and quantified the percentage of histologic tumor content versus posttreatment radiation effects for each sample. We measured spatially matched normalized and standardized relative CBV metrics (mean, median) and fractional tumor burden for each biopsy. We compared relative CBV performance to predict tumor content, including the Pearson correlation (r), against histologic tumor content (0%-100%) and the receiver operating characteristic area under the curve for predicting high-versus-low tumor content using binary histologic cutoffs (≥50%; ≥80% tumor). RESULTS Across relative CBV metrics, fractional tumor burden showed the highest correlations with tumor content (0%-100%) for normalized (r = 0.63, P < .001) and standardized (r = 0.66, P < .001) values. With binary cutoffs (ie, ≥50%; ≥80% tumor), predictive accuracies were similar for both standardized and normalized metrics and across relative CBV metrics. Median relative CBV achieved the highest area under the curve (normalized = 0.87, standardized = 0.86) for predicting ≥50% tumor, while fractional tumor burden achieved the highest area under the curve (normalized = 0.77, standardized = 0.80) for predicting ≥80% tumor. CONCLUSIONS Standardization of relative CBV achieves similar performance compared with normalized relative CBV and offers an important step toward workflow optimization and consensus methodology.
Collapse
Affiliation(s)
- J M Hoxworth
- From the Departments of Radiology (J.M.H., Y.Z., L.S.H.)
| | | | | | - K W Singleton
- Precision Neurotherapeutics Lab (K.W.S., G.D.L., B.R.B., K.R.S.), Mayo Clinic in Arizona, Phoenix, Arizona
| | - G D Leon
- Precision Neurotherapeutics Lab (K.W.S., G.D.L., B.R.B., K.R.S.), Mayo Clinic in Arizona, Phoenix, Arizona
| | - K A Smith
- Keller Center for Imaging Innovation (A.M.S.), Barrow Neurological Institute, Phoenix, Arizona
| | - A M Stokes
- Keller Center for Imaging Innovation (A.M.S.), Barrow Neurological Institute, Phoenix, Arizona
| | - Y Zhou
- From the Departments of Radiology (J.M.H., Y.Z., L.S.H.)
| | - G L Mazza
- Department of Health Sciences Research (G.L.M.), Division of Biomedical Statistics and Informatics, Mayo Clinic Scottsdale, Scottsdale, Arizona
| | | | | | | | - B R Bendok
- Precision Neurotherapeutics Lab (K.W.S., G.D.L., B.R.B., K.R.S.), Mayo Clinic in Arizona, Phoenix, Arizona
| | - D P Patra
- Departments of Neurosurgery (D.P.P.)
| | | | - J L Boxerman
- Department of Diagnostic Imaging (J.L.B.), Rhode Island Hospital, Providence, Rhode Island
| | - L C Baxter
- Neuropsychology (L.C.B.), Mayo Clinic Hospital, Phoenix, Arizona
| | - K R Swanson
- Precision Neurotherapeutics Lab (K.W.S., G.D.L., B.R.B., K.R.S.), Mayo Clinic in Arizona, Phoenix, Arizona
| | | | - K M Schmainda
- Department of Radiology (K.M.S.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - L S Hu
- From the Departments of Radiology (J.M.H., Y.Z., L.S.H.)
| |
Collapse
|
81
|
Hu LS, Hawkins-Daarud A, Wang L, Li J, Swanson KR. Imaging of intratumoral heterogeneity in high-grade glioma. Cancer Lett 2020; 477:97-106. [PMID: 32112907 DOI: 10.1016/j.canlet.2020.02.025] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/19/2022]
Abstract
High-grade glioma (HGG), and particularly Glioblastoma (GBM), can exhibit pronounced intratumoral heterogeneity that confounds clinical diagnosis and management. While conventional contrast-enhanced MRI lacks the capability to resolve this heterogeneity, advanced MRI techniques and PET imaging offer a spectrum of physiologic and biophysical image features to improve the specificity of imaging diagnoses. Published studies have shown how integrating these advanced techniques can help better define histologically distinct targets for surgical and radiation treatment planning, and help evaluate the regional heterogeneity of tumor recurrence and response assessment following standard adjuvant therapy. Application of texture analysis and machine learning (ML) algorithms has also enabled the emerging field of radiogenomics, which can spatially resolve the regional and genetically distinct subpopulations that coexist within a single GBM tumor. This review focuses on the latest advances in neuro-oncologic imaging and their clinical applications for the assessment of intratumoral heterogeneity.
Collapse
Affiliation(s)
- Leland S Hu
- Department of Radiology, Mayo Clinic Arizona, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA.
| | - Andrea Hawkins-Daarud
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, 5777 East Mayo Blvd, Support, Services Building Suite 2-700, Phoenix, AZ, 85054, USA.
| | - Lujia Wang
- School of Computing, Informatics, and Decision Systems Engineering, Arizona State University, 699 S Mill Ave, Tempe, AZ, 85281, USA.
| | - Jing Li
- School of Computing, Informatics, and Decision Systems Engineering, Arizona State University, 699 S Mill Ave, Tempe, AZ, 85281, USA.
| | - Kristin R Swanson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, 5777 East Mayo Blvd, Support, Services Building Suite 2-700, Phoenix, AZ, 85054, USA.
| |
Collapse
|
82
|
Kersch CN, Claunch CJ, Ambady P, Bucher E, Schwartz DL, Barajas RF, Iliff JJ, Risom T, Heiser L, Muldoon LL, Korkola JE, Gray JW, Neuwelt EA. Transcriptional signatures in histologic structures within glioblastoma tumors may predict personalized drug sensitivity and survival. Neurooncol Adv 2020; 2:vdaa093. [PMID: 32904984 PMCID: PMC7462280 DOI: 10.1093/noajnl/vdaa093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Glioblastoma is a rapidly fatal brain cancer that exhibits extensive intra- and intertumoral heterogeneity. Improving survival will require the development of personalized treatment strategies that can stratify tumors into subtypes that differ in therapeutic vulnerability and outcomes. Glioblastoma stratification has been hampered by intratumoral heterogeneity, limiting our ability to compare tumors in a consistent manner. Here, we develop methods that mitigate the impact of intratumoral heterogeneity on transcriptomic-based patient stratification. METHODS We accessed open-source transcriptional profiles of histological structures from 34 human glioblastomas from the Ivy Glioblastoma Atlas Project. Principal component and correlation network analyses were performed to assess sample inter-relationships. Gene set enrichment analysis was used to identify enriched biological processes and classify glioblastoma subtype. For survival models, Cox proportional hazards regression was utilized. Transcriptional profiles from 156 human glioblastomas were accessed from The Cancer Genome Atlas to externally validate the survival model. RESULTS We showed that intratumoral histologic architecture influences tumor classification when assessing established subtyping and prognostic gene signatures, and that indiscriminate sampling can produce misleading results. We identified the cellular tumor as a glioblastoma structure that can be targeted for transcriptional analysis to more accurately stratify patients by subtype and prognosis. Based on expression from cellular tumor, we created an improved risk stratification gene signature. CONCLUSIONS Our results highlight that biomarker performance for diagnostics, prognostics, and prediction of therapeutic response can be improved by analyzing transcriptional profiles in pure cellular tumor, which is a critical step toward developing personalized treatment for glioblastoma.
Collapse
Affiliation(s)
- Cymon N Kersch
- Department of Neurology, Blood-Brain Barrier Program, Oregon Health and Science University, Portland, Oregon, USA
| | - Cheryl J Claunch
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Prakash Ambady
- Department of Neurology, Blood-Brain Barrier Program, Oregon Health and Science University, Portland, Oregon, USA
| | - Elmar Bucher
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Daniel L Schwartz
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, Oregon, USA
- Department of Neurology, Layton Aging and Alzheimer’s Disease Center, Oregon Health and Science University, Portland, Oregon, USA
| | - Ramon F Barajas
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, Oregon, USA
- Department of Radiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Jeffrey J Iliff
- Department of Neurology and Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | - Tyler Risom
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Laura Heiser
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Leslie L Muldoon
- Department of Neurology, Blood-Brain Barrier Program, Oregon Health and Science University, Portland, Oregon, USA
| | - James E Korkola
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Joe W Gray
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Edward A Neuwelt
- Department of Neurology, Blood-Brain Barrier Program, Oregon Health and Science University, Portland, Oregon, USA
- Department of Neurosurgery, Oregon Health and Science University, Portland, Oregon, USA
- Office of Research and Development, Department of Veterans Affairs Medical Center, Portland, Oregon, USA
| |
Collapse
|
83
|
Design, synthesis and cytotoxicity of the antitumor agent 1-azabicycles for chemoresistant glioblastoma cells. Invest New Drugs 2019; 38:1257-1271. [PMID: 31838735 DOI: 10.1007/s10637-019-00877-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/05/2019] [Indexed: 10/25/2022]
Abstract
Twelve multi-functional pyrrolizidinones, indolizidinones and pyrroliazepinones were prepared from formal aza-[3 + 2] and aza-[3 + 3] cycloadditions of five- to seven-membered heterocyclic enaminones as diverse ambident electrophiles. The antitumor activity of these alkaloid-like compounds was investigated through an initial screening performed on human glioblastoma multiform (GBM) cell lines (GL-15, U251), on murine glioma cells line (C6) and on normal glial cells. Of the compounds tested, the new pyrrolo[1,2a]azepinone, [ethyl (3-oxo-1,2-diphenyl-6,7,8,9-tetrahydro-3H-pyrrolo[1,2a]azepin-9a(5H)-yl)acetate] or (Compound-13) exhibited selective cytotoxic effects on GBM-temozolomide resistant cells. Compound-13 exerted dose-dependent cytotoxic activity by promoting arrest of cells in the G0/G1 phase of the cell cycle in the first 24 h. The apoptotic effect observed was in a time-dependent manner. Anti-migratory effect promoted by the treatment with compound-13 was also observed. Moreover, healthy mixed glial cell cultures from rat brain exhibited no cytotoxicity effect upon exposure to compound-13. Thus, the present study paves the way for the use of compound-13 as novel antitumor scaffold candidate for glioma cell therapy.
Collapse
|
84
|
Diffusion- and perfusion-weighted MRI radiomics model may predict isocitrate dehydrogenase (IDH) mutation and tumor aggressiveness in diffuse lower grade glioma. Eur Radiol 2019; 30:2142-2151. [PMID: 31828414 DOI: 10.1007/s00330-019-06548-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/11/2019] [Accepted: 10/24/2019] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To determine whether diffusion- and perfusion-weighted MRI-based radiomics features can improve prediction of isocitrate dehydrogenase (IDH) mutation and tumor aggressiveness in lower grade gliomas (LGGs) METHODS: Radiomics features (n = 6472) were extracted from multiparametric MRI including conventional MRI, apparent diffusion coefficient (ADC), and normalized cerebral blood volume, acquired on 127 LGG patients with determined IDH mutation status and grade (WHO II or III). Radiomics models were constructed using machine learning-based feature selection and generalized linear model classifiers. Segmentation stability was calculated between two readers using concordance correlation coefficients (CCCs). Diagnostic performance to predict IDH mutation and tumor grade was compared between the multiparametric and conventional MRI radiomics models using the area under the receiver operating characteristics curve (AUC). The models were tested using a temporally independent validation set (n = 28). RESULTS The multiparametric MRI radiomics model was optimized with a random forest feature selector, with segmentation stability of a CCC threshold of 0.8. For IDH mutation, multiparametric MR radiomics showed similar performance (AUC 0.795) to the conventional radiomics model (AUC 0.729). In tumor grading, multiparametric model with ADC features showed higher performance (AUC 0.932) than the conventional model (AUC 0.555). The independent validation set showed the same trend with AUCs of 0.747 for IDH prediction and 0.819 for tumor grading with multiparametric MRI radiomics model. CONCLUSION Multiparametric MRI radiomics model showed improved diagnostic performance in tumor grading and comparable diagnostic performance in IDH mutation status, with ADC features playing a significant role. KEY POINTS • The multiparametric MRI radiomics model was comparable with conventional MRI radiomics model in predicting IDH mutation. • The multiparametric MRI radiomics model outperformed conventional MRI in glioma grading. • Apparent diffusion coefficient played an important role in glioma grading and predicting IDH mutation status using radiomics.
Collapse
|
85
|
Gill BJA, Wu X, Khan FA, Sosunov AA, Liou JY, Dovas A, Eissa TL, Banu MA, Bateman LM, McKhann GM, Canoll P, Schevon C. Ex vivo multi-electrode analysis reveals spatiotemporal dynamics of ictal behavior at the infiltrated margin of glioma. Neurobiol Dis 2019; 134:104676. [PMID: 31731042 PMCID: PMC8147009 DOI: 10.1016/j.nbd.2019.104676] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/22/2019] [Accepted: 11/11/2019] [Indexed: 01/02/2023] Open
Abstract
The purpose of this study is to develop a platform in which the cellular and molecular underpinnings of chronic focal neocortical lesional epilepsy can be explored and use it to characterize seizure-like events (SLEs) in an ex vivo model of infiltrating high-grade glioma. Microelectrode arrays were used to study electrophysiologic changes in ex vivo acute brain slices from a PTEN/p53 deleted, PDGF-B driven mouse model of high-grade glioma. Electrode locations were co-registered to the underlying histology to ascertain the influence of the varying histologic landscape on the observed electrophysiologic changes. Peritumoral, infiltrated, and tumor sites were sampled in tumor-bearing slices. Following the addition of zero Mg2+ solution, all three histologic regions in tumor-bearing slices showed significantly greater increases in firing rates when compared to the control sites. Tumor-bearing slices demonstrated increased proclivity for SLEs, with 40 events in tumor-bearing slices and 5 events in control slices (p-value = .0105). Observed SLEs were characterized by either low voltage fast (LVF) onset patterns or short bursts of repetitive widespread, high amplitude low frequency discharges. Seizure foci comprised areas from all three histologic regions. The onset electrode was found to be at the infiltrated margin in 50% of cases and in the peritumoral region in 36.9% of cases. These findings reveal a landscape of histopathologic and electrophysiologic alterations associated with ictogenesis and spread of tumor-associated seizures.
Collapse
Affiliation(s)
- Brian J A Gill
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA.
| | - Xiaoping Wu
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Farhan A Khan
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Alexander A Sosunov
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Jyun-You Liou
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Athanassios Dovas
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Tahra L Eissa
- Department of Applied Mathematics, University of Colorado at Boulder, Boulder, CO, USA
| | - Matei A Banu
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Lisa M Bateman
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Guy M McKhann
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Catherine Schevon
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
86
|
Ellingson BM, Abrey LE, Nelson SJ, Kaufmann TJ, Garcia J, Chinot O, Saran F, Nishikawa R, Henriksson R, Mason WP, Wick W, Butowski N, Ligon KL, Gerstner ER, Colman H, de Groot J, Chang S, Mellinghoff I, Young RJ, Alexander BM, Colen R, Taylor JW, Arrillaga-Romany I, Mehta A, Huang RY, Pope WB, Reardon D, Batchelor T, Prados M, Galanis E, Wen PY, Cloughesy TF. Validation of postoperative residual contrast-enhancing tumor volume as an independent prognostic factor for overall survival in newly diagnosed glioblastoma. Neuro Oncol 2019; 20:1240-1250. [PMID: 29660006 DOI: 10.1093/neuonc/noy053] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background In the current study, we pooled imaging data in newly diagnosed glioblastoma (GBM) patients from international multicenter clinical trials, single institution databases, and multicenter clinical trial consortiums to identify the relationship between postoperative residual enhancing tumor volume and overall survival (OS). Methods Data from 1511 newly diagnosed GBM patients from 5 data sources were included in the current study: (i) a single institution database from UCLA (N = 398; Discovery); (ii) patients from the Ben and Cathy Ivy Foundation for Early Phase Clinical Trials Network Radiogenomics Database (N = 262 from 8 centers; Confirmation); (iii) the chemoradiation placebo arm from an international phase III trial (AVAglio; N = 394 from 120 locations in 23 countries; Validation); (iv) the experimental arm from AVAglio examining chemoradiation plus bevacizumab (N = 404 from 120 locations in 23 countries; Exploratory Set 1); and (v) an Alliance (N0874) phase I/II trial of vorinostat plus chemoradiation (N = 53; Exploratory Set 2). Postsurgical, residual enhancing disease was quantified using T1 subtraction maps. Multivariate Cox regression models were used to determine influence of clinical variables, O6-methylguanine-DNA methyltransferase (MGMT) status, and residual tumor volume on OS. Results A log-linear relationship was observed between postoperative, residual enhancing tumor volume and OS in newly diagnosed GBM treated with standard chemoradiation. Postoperative tumor volume is a prognostic factor for OS (P < 0.01), regardless of therapy, age, and MGMT promoter methylation status. Conclusion Postsurgical, residual contrast-enhancing disease significantly negatively influences survival in patients with newly diagnosed GBM treated with chemoradiation with or without concomitant experimental therapy.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | | - Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), San Francisco, California, USA
| | | | | | - Olivier Chinot
- Aix-Marseille University, AP-HM, Service de Neuro-Oncologie, CHU Timone, Marseille, France
| | - Frank Saran
- The Royal Marsden NHS Foundation Trust, Sutton, UK
| | | | - Roger Henriksson
- Regional Cancer Center Stockholm, Stockholm, Sweden and Umeå University, Umeå, Sweden
| | | | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Nicholas Butowski
- Department of Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | - Keith L Ligon
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Howard Colman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - John de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Susan Chang
- Department of Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | | | - Robert J Young
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Brian M Alexander
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Rivka Colen
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennie W Taylor
- Department of Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | | | - Arnav Mehta
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Whitney B Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - David Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Tracy Batchelor
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Michael Prados
- Department of Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | - Evanthia Galanis
- Department of Molecular Medicine, Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA.,Department of Oncology, Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
87
|
Metabolic Tumor Volume Response Assessment Using (11)C-Methionine Positron Emission Tomography Identifies Glioblastoma Tumor Subregions That Predict Progression Better Than Baseline or Anatomic Magnetic Resonance Imaging Alone. Adv Radiat Oncol 2019; 5:53-61. [PMID: 32051890 PMCID: PMC7004943 DOI: 10.1016/j.adro.2019.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/05/2019] [Indexed: 02/08/2023] Open
Abstract
Purpose To evaluate whether response assessment of newly diagnosed glioblastoma at 3 months using 11C-methionine-positron emission tomography (MET-PET) is better associated with patient outcome compared with baseline MET-PET or anatomic magnetic resonance imaging alone. Methods and Materials Patients included were participants in a phase I/II trial of dose-escalated chemoradiation based on anatomic magnetic resonance imaging. Automated segmentation of metabolic tumor volume (MTV) was performed at a threshold of 1.5 times mean cerebellar uptake. Progression-free (PFS) and overall survival were estimated with the Kaplan-Meier method and compared with log-rank tests. Multivariate analysis for PFS and overall survival was performed using Cox proportional hazards, and spatial overlap between imaging and recurrence volumes were analyzed. Results Among 37 patients, 15 had gross total resection, of whom 10 (67%) had residual MTV, 16 subtotal resection, and 6 biopsy alone. Median radiation therapy dose was 75 Gy (range, 66-81). Median baseline T1 Gd-enhanced tumor volume (GTV-Gd) was 38.0 cm3 (range, 8.0-81.5). Median pre-CRT MTV was 4.9 cm3 (range, 0-43.8). Among 25 patients with 3-month MET-PET, MTV was only 2.4 cm3 (range, 0.004-18.0) in patients with uptake. Patients with MTV = 0 cm3 at 3 months had superior PFS (18.2 vs 10.1 months, P = .03). On multivariate analysis, larger 3-month MTV (hazard ratio [HR] 2.4, 95% confidence interval [CI], 1.4-4.3, P = .03), persistent MET-PET subvolume (overlap of pre-CRT and 3 month MTV; HR 2.0, 95% CI, 1.2-3.4, P = .06), and increase in MTV (HR 1.8, 95% CI, 1.1-3.1, P = .09) were the only imaging factors significant for worse PFS. GTV-Gd at recurrence encompassed 97% of the persistent MET-PET subvolume (interquartile range 72%-100%), versus 71% (interquartile range 39%-93%) of baseline MTV, 54% of baseline GTV-Gd (18%-87%), and 78% of 3-month MTV (47%-95%). Conclusions The majority of patients with apparent gross total resection of glioblastoma have measurable postoperative MTV. Total and persisting MTV 3 months post-CRT were significant predictors of PFS, and persistent MET-PET subvolume was the strongest predictor for localizing tumor recurrence.
Collapse
|
88
|
White ML, Zhang Y, Yu F, Shonka N, Aizenberg MR, Adapa P, Kazmi SAJ. Post-operative perfusion and diffusion MR imaging and tumor progression in high-grade gliomas. PLoS One 2019; 14:e0213905. [PMID: 30883579 PMCID: PMC6422263 DOI: 10.1371/journal.pone.0213905] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/04/2019] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Perfusion and diffusion magnetic resonance imaging (MRI) provide important biomarkers for brain tumor analysis. Our aim was to investigate if regions of increased perfusion or tumor with restricted diffusion on the immediate post-operative MRI examination would be predictive of time to tumor progression in patients with high-grade gliomas. MATERIALS AND METHODS Twenty-three patients with high-grade gliomas were retrospectively analyzed. We measured the perfusion at the resection area and evaluated the presence or absence of the restricted diffusion in residual tumor masses. The associations of the perfusion, diffusion and contrast enhancement (delayed static enhancement (DSE)) characteristics with time to tumor progression were statistically calculated. We also evaluated if the location of the tumor progression was concordant to the areas of the elevated perfusion, tumor type restricted diffusion and enhancement. RESULTS Patients with >200 days to progression are more likely to have no elevated relative cerebral blood volume (rCBV) ratio (p = 0.0004), no tumor restriction (p = 0.024), and no DSE (p = 0.052). The elevated mean rCBV ratio (p<0.001) and tumor type restricted diffusion (p = 0.002) were significantly associated with a higher risk of progression. All cases with rCBV ratio of >1.5 progressed in 275 days or earlier. Tumors tended to progress at the area where patients with post-operative MRIs showed elevated perfusion (p = 0.006), tumor-type restricted diffusion (p = 0.005) and DSE (p = 0.008). CONCLUSIONS Post-operative analysis of rCBV, tumor type restricted diffusion and enhancement characteristics are predictive of time to progression, risk of progression and where tumor progression is likely to occur.
Collapse
Affiliation(s)
- Matthew L. White
- Radiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| | - Yan Zhang
- Radiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Fang Yu
- Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Nicole Shonka
- Internal Medicine Division of Oncology & Hematology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Michele R. Aizenberg
- Neurosurgery, the University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Pavani Adapa
- South Texas Radiology Group, San Antonio, Texas, United States of America
| | - Syed A. Jaffar Kazmi
- Anatomic Pathology, Geisinger Medical Center, Danville, Pennsylvania, United States of America
| |
Collapse
|
89
|
Hu LS, Yoon H, Eschbacher JM, Baxter LC, Dueck AC, Nespodzany A, Smith KA, Nakaji P, Xu Y, Wang L, Karis JP, Hawkins-Daarud AJ, Singleton KW, Jackson PR, Anderies BJ, Bendok BR, Zimmerman RS, Quarles C, Porter-Umphrey AB, Mrugala MM, Sharma A, Hoxworth JM, Sattur MG, Sanai N, Koulemberis PE, Krishna C, Mitchell JR, Wu T, Tran NL, Swanson KR, Li J. Accurate Patient-Specific Machine Learning Models of Glioblastoma Invasion Using Transfer Learning. AJNR Am J Neuroradiol 2019; 40:418-425. [PMID: 30819771 DOI: 10.3174/ajnr.a5981] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 12/13/2018] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE MR imaging-based modeling of tumor cell density can substantially improve targeted treatment of glioblastoma. Unfortunately, interpatient variability limits the predictive ability of many modeling approaches. We present a transfer learning method that generates individualized patient models, grounded in the wealth of population data, while also detecting and adjusting for interpatient variabilities based on each patient's own histologic data. MATERIALS AND METHODS We recruited patients with primary glioblastoma undergoing image-guided biopsies and preoperative imaging, including contrast-enhanced MR imaging, dynamic susceptibility contrast MR imaging, and diffusion tensor imaging. We calculated relative cerebral blood volume from DSC-MR imaging and mean diffusivity and fractional anisotropy from DTI. Following image coregistration, we assessed tumor cell density for each biopsy and identified corresponding localized MR imaging measurements. We then explored a range of univariate and multivariate predictive models of tumor cell density based on MR imaging measurements in a generalized one-model-fits-all approach. We then implemented both univariate and multivariate individualized transfer learning predictive models, which harness the available population-level data but allow individual variability in their predictions. Finally, we compared Pearson correlation coefficients and mean absolute error between the individualized transfer learning and generalized one-model-fits-all models. RESULTS Tumor cell density significantly correlated with relative CBV (r = 0.33, P < .001), and T1-weighted postcontrast (r = 0.36, P < .001) on univariate analysis after correcting for multiple comparisons. With single-variable modeling (using relative CBV), transfer learning increased predictive performance (r = 0.53, mean absolute error = 15.19%) compared with one-model-fits-all (r = 0.27, mean absolute error = 17.79%). With multivariate modeling, transfer learning further improved performance (r = 0.88, mean absolute error = 5.66%) compared with one-model-fits-all (r = 0.39, mean absolute error = 16.55%). CONCLUSIONS Transfer learning significantly improves predictive modeling performance for quantifying tumor cell density in glioblastoma.
Collapse
Affiliation(s)
- L S Hu
- From the Department of Radiology (L.S.H., J.M.H., J.R.M., T.W., J.L.)
| | - H Yoon
- Arizona State University (H.Y., Y.X., L.W., T.W., J.L.), Tempe, Arizona
| | | | | | - A C Dueck
- Department of Biostatistics (A.C.D.), Mayo Clinic in Arizona, Scottsdale, Arizona
| | | | | | - P Nakaji
- Neurosurgery (K.A.S., P.N., N.S.)
| | - Y Xu
- Arizona State University (H.Y., Y.X., L.W., T.W., J.L.), Tempe, Arizona
| | - L Wang
- Arizona State University (H.Y., Y.X., L.W., T.W., J.L.), Tempe, Arizona
| | | | - A J Hawkins-Daarud
- Precision Neurotherapeutics Lab (A.J.H.-D., K.W.S., P.R.J, B.R.B., K.R.S.)
| | - K W Singleton
- Precision Neurotherapeutics Lab (A.J.H.-D., K.W.S., P.R.J, B.R.B., K.R.S.)
| | - P R Jackson
- Precision Neurotherapeutics Lab (A.J.H.-D., K.W.S., P.R.J, B.R.B., K.R.S.)
| | - B J Anderies
- Department of Neurosurgery (B.J.A., B.R.B., R.S.Z., M.G.S., P.E.K., C.K., K.R.S.)
| | - B R Bendok
- Precision Neurotherapeutics Lab (A.J.H.-D., K.W.S., P.R.J, B.R.B., K.R.S.).,Department of Neurosurgery (B.J.A., B.R.B., R.S.Z., M.G.S., P.E.K., C.K., K.R.S.)
| | - R S Zimmerman
- Department of Neurosurgery (B.J.A., B.R.B., R.S.Z., M.G.S., P.E.K., C.K., K.R.S.)
| | - C Quarles
- Neuroimaging Research (C.Q.), Barrow Neurological Institute, Phoenix, Arizona
| | | | - M M Mrugala
- Department of Neuro-Oncology (A.B.P.-U., M.M.M., A.S.)
| | - A Sharma
- Department of Neuro-Oncology (A.B.P.-U., M.M.M., A.S.)
| | - J M Hoxworth
- From the Department of Radiology (L.S.H., J.M.H., J.R.M., T.W., J.L.)
| | - M G Sattur
- Department of Neurosurgery (B.J.A., B.R.B., R.S.Z., M.G.S., P.E.K., C.K., K.R.S.)
| | - N Sanai
- Neurosurgery (K.A.S., P.N., N.S.)
| | - P E Koulemberis
- Department of Neurosurgery (B.J.A., B.R.B., R.S.Z., M.G.S., P.E.K., C.K., K.R.S.)
| | - C Krishna
- Department of Neurosurgery (B.J.A., B.R.B., R.S.Z., M.G.S., P.E.K., C.K., K.R.S.)
| | - J R Mitchell
- From the Department of Radiology (L.S.H., J.M.H., J.R.M., T.W., J.L.).,H. Lee Moffitt Cancer Center and Research Institute (J.R.M.), Tampa, Florida
| | - T Wu
- From the Department of Radiology (L.S.H., J.M.H., J.R.M., T.W., J.L.).,Arizona State University (H.Y., Y.X., L.W., T.W., J.L.), Tempe, Arizona
| | - N L Tran
- Department of Cancer Biology (N.L.T.), Mayo Clinic in Arizona, Phoenix, Arizona
| | - K R Swanson
- Precision Neurotherapeutics Lab (A.J.H.-D., K.W.S., P.R.J, B.R.B., K.R.S.).,Department of Neurosurgery (B.J.A., B.R.B., R.S.Z., M.G.S., P.E.K., C.K., K.R.S.)
| | - J Li
- From the Department of Radiology (L.S.H., J.M.H., J.R.M., T.W., J.L.).,Arizona State University (H.Y., Y.X., L.W., T.W., J.L.), Tempe, Arizona
| |
Collapse
|
90
|
Differentiation of Glioblastoma and Solitary Brain Metastasis by Gradient of Relative Cerebral Blood Volume in the Peritumoral Brain Zone Derived from Dynamic Susceptibility Contrast Perfusion Magnetic Resonance Imaging. J Comput Assist Tomogr 2019; 43:13-17. [PMID: 30015801 DOI: 10.1097/rct.0000000000000771] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The purpose of our study was to evaluate the efficacy of the relative cerebral blood volume (rCBV) gradient in the peritumoral brain zone (PBZ)-the difference in the rCBV values from the area closest to the enhancing lesion to the area closest to the healthy white matter-in differentiating glioblastoma (GB) from solitary brain metastasis (MET). METHODS A 3.0-T magnetic resonance imaging (MRI) machine was used to perform dynamic susceptibility contrast perfusion MRI (DSC-MRI) on 43 patients with a solitary brain tumor (24 GB, 19 MET). The rCBV ratios were acquired by DSC-MRI data in 3 regions of the PBZ (near the enhancing tumor, G1; intermediate distance from the enhancing tumor, G2; far from the enhancing tumor, G3). The maximum rCBV ratios in the PBZ (rCBVp) and the enhancing tumor were also calculated, respectively. The perfusion parameters were evaluated using the nonparametric Mann-Whitney test. The sensitivity, specificity, accuracy, and the area under the receiver operating characteristic curve were identified. RESULTS The rCBVp ratios and rCBV gradient in the PBZ were significantly higher in GB compared with MET (P < 0.05 for both rCBVp ratios and rCBV gradient). The threshold values of 0.50 or greater for rCBVp ratios provide sensitivity and specificity of 57.69% and 79.17%, respectively, for differentiation of GB from MET. Compared with rCBVp ratios, rCBV gradient had higher sensitivity (94.44%) and specificity (91.67%) using the threshold value of greater than 0.06. CONCLUSIONS The parameter of rCBV gradient derived from DSC-MRI in the PBZ seems to be the most efficient parameter to differentiate GB from METs.
Collapse
|
91
|
Förster A, Brehmer S, Seiz-Rosenhagen M, Mildenberger I, Giordano FA, Wenz H, Reuss D, Hänggi D, Groden C. Heterogeneity of glioblastoma with gliomatosis cerebri growth pattern on diffusion and perfusion MRI. J Neurooncol 2018; 142:103-109. [PMID: 30565029 DOI: 10.1007/s11060-018-03068-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/30/2018] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND PURPOSE Gliomatosis cerebri (GC) is a rare growth pattern of glioblastoma whose diffuse nature is reflected by unspecific, relatively uniform findings on conventional MRI. In the present study we sought to evaluate the additional value of diffusion (DWI) and perfusion weighted (PWI) MRI for a more detailed characterization. METHODS We analyzed the MRI findings in patients with histologically proven glioblastoma with GC growth pattern with a specific emphasis on T2 lesion pattern, volume, relative apparent diffusion coefficient (rACD), and relative cerebral blood volume (rCBV) and compared these to age-/gender-matched patients with localized glioblastoma. RESULTS Overall, 16 patients (median age 59.5 years, 4 male) were included in the study. Of these, 8 patients had a glioblastoma with GC growth pattern, and 8 a classical localized growth pattern. While the median rADC (1.27 [IQR 1.12-1.41]) within the T2 lesion was significant lower in glioblastoma with GC growth pattern compared to localized glioblastoma (1.74 [IQR 1.45-1.96]; p = 0.003), the median T2 lesion volume and rCBV within the T2 lesion did not differ significantly. Furthermore, six patients with glioblastoma with GC growth pattern showed focal areas with significantly reduced rADC (p = 0.043), and/or increased rCBV (p = 0.028). CONCLUSIONS Lower rADC in glioblastoma with GC growth pattern might reflect the diffuse tumor cell infiltration whereas focal areas with decreased rADC and/or increased rCBV probably indicate high tumor cell density and/or abnormal tumor vessels which may be useful for biopsy guidance.
Collapse
Affiliation(s)
- Alex Förster
- Department of Neuroradiology, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Stefanie Brehmer
- Department of Neurosurgery, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Marcel Seiz-Rosenhagen
- Department of Neurosurgery, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Iris Mildenberger
- Department of Neurology, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Frank A Giordano
- Department of Radiation Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Holger Wenz
- Department of Neuroradiology, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - David Reuss
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Hänggi
- Department of Neurosurgery, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christoph Groden
- Department of Neuroradiology, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
92
|
Coburger J, Wirtz CR. Fluorescence guided surgery by 5-ALA and intraoperative MRI in high grade glioma: a systematic review. J Neurooncol 2018; 141:533-546. [PMID: 30488293 DOI: 10.1007/s11060-018-03052-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/13/2018] [Indexed: 11/25/2022]
Abstract
PURPOSE Fluorescence guided surgery by 5-aminolevulinic acid (5-ALA) and intraoperative MRI (iMRI) are currently the most important intraoperative imaging techniques in high grade glioma (HGG) surgery. Few comparative studies exist for these techniques. This review aims to systematically compare 5-ALA and iMRI assisted surgery based on the current literature and discuss the potential impact of a combined use of both techniques. METHODS A systematic literature search based on preferred reporting items for systematic reviews and meta-analysis was performed concerning accuracy of tumor detection; extent of resection; neurological deficits (ND); Quality of life (QoL); usability and combined use of both techniques. Original clinical articles on HGG published until March 31st were screened. RESULTS 169 publications were screened, 81 were eligible and 22 were finally included in the review using. Overall, there is evidence that both imaging techniques improve gross total resection rate in non-eloquent lesions. Imaging results do not correlate at the border zone of a HGG. 5-ALA and contrast-enhanced iMRI seem to have a supplementary effect in tumor detection. Overall, both imaging techniques alone or combined do not seem to increase rate of permanent ND or decrease QoL in HGG surgery when used with intraoperative monitoring/mapping. CONCLUSION Based on the currently available literature no superiority of one technique over the other can be found in the most important outcome parameters. Based on the available information a combined use of 5-ALA and iMRI seems very promising to achieve a resection beyond gadolinium-enhancement. However, only low quality of evidence exists for this approach.
Collapse
Affiliation(s)
- Jan Coburger
- Department of Neurosurgery, University of Ulm, Campus Günzburg, Ludwig-Heilmeyerstr. 2, 89321, Günzburg, Germany.
| | - Christian Rainer Wirtz
- Department of Neurosurgery, University of Ulm, Campus Günzburg, Ludwig-Heilmeyerstr. 2, 89321, Günzburg, Germany
| |
Collapse
|
93
|
Kazerooni AF, Nabil M, Zadeh MZ, Firouznia K, Azmoudeh-Ardalan F, Frangi AF, Davatzikos C, Rad HS. Characterization of active and infiltrative tumorous subregions from normal tissue in brain gliomas using multiparametric MRI. J Magn Reson Imaging 2018; 48:938-950. [PMID: 29412496 PMCID: PMC6081259 DOI: 10.1002/jmri.25963] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 01/20/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Targeted localized biopsies and treatments for diffuse gliomas rely on accurate identification of tissue subregions, for which current MRI techniques lack specificity. PURPOSE To explore the complementary and competitive roles of a variety of conventional and quantitative MRI methods for distinguishing subregions of brain gliomas. STUDY TYPE Prospective. POPULATION Fifty-one tissue specimens were collected using image-guided localized biopsy surgery from 10 patients with newly diagnosed gliomas. FIELD STRENGTH/SEQUENCE Conventional and quantitative MR images consisting of pre- and postcontrast T1 w, T2 w, T2 -FLAIR, T2 -relaxometry, DWI, DTI, IVIM, and DSC-MRI were acquired preoperatively at 3T. ASSESSMENT Biopsy specimens were histopathologically attributed to glioma tissue subregion categories of active tumor (AT), infiltrative edema (IE), and normal tissue (NT) subregions. For each tissue sample, a feature vector comprising 15 MRI-based parameters was derived from preoperative images and assessed by a machine learning algorithm to determine the best multiparametric feature combination for characterizing the tissue subregions. STATISTICAL TESTS For discrimination of AT, IE, and NT subregions, a one-way analysis of variance (ANOVA) test and for pairwise tissue subregion differentiation, Tukey honest significant difference, and Games-Howell tests were applied (P < 0.05). Cross-validated feature selection and classification methods were implemented for identification of accurate multiparametric MRI parameter combination. RESULTS After exclusion of 17 tissue specimens, 34 samples (AT = 6, IE = 20, and NT = 8) were considered for analysis. Highest accuracies and statistically significant differences for discrimination of IE from NT and AT from NT were observed for diffusion-based parameters (AUCs >90%), and the perfusion-derived parameter as the most accurate feature in distinguishing IE from AT. A combination of "CBV, MD, T2 _ISO, FLAIR" parameters showed high diagnostic performance for identification of the three subregions (AUC ∼90%). DATA CONCLUSION Integration of a few quantitative along with conventional MRI parameters may provide a potential multiparametric imaging biomarker for predicting the histopathologically proven glioma tissue subregions. LEVEL OF EVIDENCE 2 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2018;48:938-950.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- Quantitative MR Imaging and Spectroscopy Group, Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahnaz Nabil
- Department of Statistics, Faculty of Mathematical Science, University of Guilan, Rasht, Iran
| | - Mehdi Zeinali Zadeh
- Department of Neurological Surgery, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Kavous Firouznia
- Advanced Diagnostic and Interventional Radiology Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Azmoudeh-Ardalan
- Department of Pathology, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Alejandro F. Frangi
- Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK
| | - Christos Davatzikos
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hamidreza Saligheh Rad
- Quantitative MR Imaging and Spectroscopy Group, Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
94
|
Bowden SG, Han SJ. The Evolving Role of the Oncologic Neurosurgeon: Looking Beyond Extent of Resection in the Modern Era. Front Oncol 2018; 8:406. [PMID: 30319971 PMCID: PMC6167541 DOI: 10.3389/fonc.2018.00406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/06/2018] [Indexed: 11/13/2022] Open
Abstract
Neurosurgeons have played an essential role in glioma management and research for over a century. While the past twenty years have played witness to many exciting developments in glioma biology, diagnosis, and classification, relatively few novel, effective treatment strategies have been introduced. The role of neurosurgery in glioma management has been clarified, with a large body of evidence in support of maximal safe resection. However, neurosurgeons have also played a critical role in translational research during this period. The development of new MRI technologies has benefited greatly from validation with stereotactically-targeted human tissue. Careful banking of surgically acquired tissue was key to the development of a new classification scheme for glioma. Similarly, we have garnered a considerably deeper understanding of molecular and genetic properties of glioma through analysis of large surgical specimens. As our classification schemes become more sophisticated, incorporating targeted tissue sampling into the development of novel treatment strategies becomes essential. Such ex vivo analysis could be instrumental in determining mechanisms of treatment failure or success. Modern tumor neurosurgeons should consider themselves surgical neuro-oncologists, with engagement in translational research essential to furthering the field and improving outlooks for our patients.
Collapse
Affiliation(s)
- Stephen G Bowden
- Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| | - Seunggu Jude Han
- Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
95
|
Qin L, Li X, Li A, Cheng S, Qu J, Reinshagen K, Hu J, Himes N, Lu G, Xu X, Young GS. Clinical Validation of Automatable Gaussian Normalized CBV in Brain Tumor Analysis: Superior Reproducibility and Slightly Better Association with Survival than Current Standard Manual Normal Appearing White Matter Normalization. Transl Oncol 2018; 11:1398-1405. [PMID: 30216765 PMCID: PMC6138997 DOI: 10.1016/j.tranon.2018.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/23/2018] [Accepted: 07/30/2018] [Indexed: 10/28/2022] Open
Abstract
PURPOSE To validate Gaussian normalized cerebral blood volume (GN-nCBV) by association with overall survival (OS) in newly diagnosed glioblastoma patients and compare this association with current standard white matter normalized cerebral blood volume (WN-nCBV). METHODS We retrieved spin-echo echo-planar dynamic susceptibility contrast MRI acquired after maximal resection and prior to radiation therapy between 2006 and 2011 in 51 adult patients (28 male, 23 female; age 23-87 years) with newly diagnosed glioblastoma. Software code was developed in house to perform Gaussian normalization of CBV to the standard deviation of the whole brain CBV. Three expert readers manually selected regions of interest in tumor and normal-appearing white matter on CBV maps. Receiver operating characteristics (ROC) curves associating nCBV with 15-month OS were calculated for both GN-nCBV and WN-nCBV. Reproducibility and interoperator variability were compared using within-subject coefficient of variation (wCV) and intraclass correlation coefficients (ICCs). RESULTS GN-nCBV ICC (≥0.82) and wCV (≤21%) were superior to WN-nCBV ICC (0.54-0.55) and wCV (≥46%). The area under the ROC curve analysis demonstrated both GN-nCBV and WN-nCBV to be good predictors of OS, but GN-nCBV was consistently superior, although the difference was not statistically significant. CONCLUSION GN-nCBV has a slightly better association with clinical gold standard OS than conventional WM-nCBV in our glioblastoma patient cohort. This equivalent or superior validity, combined with the advantages of higher reproducibility, lower interoperator variability, and easier automation, makes GN-nCBV superior to WM-nCBV for clinical and research use in glioma patients. We recommend widespread adoption and incorporation of GN-nCBV into commercial dynamic susceptibility contrast processing software.
Collapse
Affiliation(s)
- Lei Qin
- Dana-Farber Cancer Institute, Department of Imaging, Boston, MA, USA; Harvard Medical School, Department of Radiology, Boston, MA, USA
| | - Xiang Li
- Brigham and Women's Hospital, Department of Radiology, Boston, MA, USA; Affiliated Cancer Hospital of Zhengzhou University, Department of Radiology, Zhengzhou, Henan, China
| | - Angie Li
- Brigham and Women's Hospital, Department of Radiology, Boston, MA, USA; The Robert Larner, M.D. College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Suchun Cheng
- Dana-Farber Cancer Institute, Department of Biostatistics and Computational Biology, Boston, MA, USA
| | - Jinrong Qu
- Brigham and Women's Hospital, Department of Radiology, Boston, MA, USA; Affiliated Cancer Hospital of Zhengzhou University, Department of Radiology, Zhengzhou, Henan, China
| | - Katherine Reinshagen
- Harvard Medical School, Department of Radiology, Boston, MA, USA; Brigham and Women's Hospital, Department of Radiology, Boston, MA, USA; Massachusetts Eye and Ear Infirmary, Department of Radiology, Boston, MA, USA
| | - Jiani Hu
- Dana-Farber Cancer Institute, Department of Biostatistics and Computational Biology, Boston, MA, USA
| | - Nathan Himes
- Brigham and Women's Hospital, Department of Radiology, Boston, MA, USA; Medical Imaging of Lehigh Valley, Lehigh Valley Hospital, Allentown, PA, USA
| | - Gao Lu
- Brigham and Women's Hospital, Department of Radiology, Boston, MA, USA; Peking Union Medical College Hospital, Department of Neurosurgery, Beijing, China
| | - Xiaoyin Xu
- Peking Union Medical College Hospital, Department of Neurosurgery, Beijing, China; Peking Union Medical College Hospital, Department of Neurosurgery, Beijing, China
| | - Geoffrey S Young
- Dana-Farber Cancer Institute, Department of Imaging, Boston, MA, USA; Harvard Medical School, Department of Radiology, Boston, MA, USA; Brigham and Women's Hospital, Department of Radiology, Boston, MA, USA.
| |
Collapse
|
96
|
She D, Liu J, Xing Z, Zhang Y, Cao D, Zhang Z. MR Imaging Features of Anaplastic Pleomorphic Xanthoastrocytoma Mimicking High-Grade Astrocytoma. AJNR Am J Neuroradiol 2018; 39:1446-1452. [PMID: 29903923 DOI: 10.3174/ajnr.a5701] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/18/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE Anaplastic pleomorphic xanthoastrocytoma, which has been recently defined as a distinct entity in the 2016 World Health Organization classification, may exhibit aggressive clinical behavior and relatively worse prognosis than pleomorphic xanthoastrocytoma. This study aimed to investigate whether there were any differences in MR imaging characteristics between these 2 tumors. MATERIALS AND METHODS This retrospective study included 9 patients with anaplastic pleomorphic xanthoastrocytoma and 10 patients with pleomorphic xanthoastrocytoma who underwent MR imaging before an operation. DWI was performed in 17 patients (8 with anaplastic pleomorphic xanthoastrocytoma, 9 with pleomorphic xanthoastrocytoma); and DSC-PWI, in 9 patients (5 with anaplastic pleomorphic xanthoastrocytoma, 4 with pleomorphic xanthoastrocytoma). Demographics, conventional imaging characteristics (location, size, cystic degeneration, enhancement, peritumoral edema, and leptomeningeal contact), minimum relative ADC ratio, and maximum relative CBV ratio were evaluated between the anaplastic pleomorphic xanthoastrocytoma and pleomorphic xanthoastrocytoma groups. RESULTS Anaplastic pleomorphic xanthoastrocytoma was more likely to demonstrate high-grade features than pleomorphic xanthoastrocytoma, including greater maximum tumor diameter (4.7 ± 0.6 cm versus 3.1 ± 1.1 cm, P = .001), more frequent heterogeneous contrast enhancement of solid portions (88.9% versus 20.0%, P = .01), more obvious peritumoral edema (2.3 ± 0.9 cm versus 1.0 ± 0.9 cm, P = .008), lower minimum relative ADC on DWI (1.0 ± 0.2 versus 1.5 ± 0.4, P = .008), and higher maximum relative CBV on DSC-PWI (2.6 ± 0.8 versus 1.6 ± 0.2, P = .036). CONCLUSIONS Anaplastic pleomorphic xanthoastrocytomas often have more aggressive MR imaging features mimicking high-grade astrocytomas than pleomorphic xanthoastrocytomas. DWI and DSC-PWI might be useful in the characterization and differentiation of anaplastic pleomorphic xanthoastrocytoma and pleomorphic xanthoastrocytoma.
Collapse
Affiliation(s)
- D She
- From the Departments of Radiology (D.S., J.L., Z.X., D.C.)
| | - J Liu
- From the Departments of Radiology (D.S., J.L., Z.X., D.C.)
| | - Z Xing
- From the Departments of Radiology (D.S., J.L., Z.X., D.C.)
| | - Y Zhang
- Pathology (Y.Z.), First Affiliated Hospital of Fujian Medical University, Fuzhou, P.R. China
| | - D Cao
- From the Departments of Radiology (D.S., J.L., Z.X., D.C.)
| | - Z Zhang
- Siemens Healthcare Ltd (Z.Z.), Shanghai, P.R. China
| |
Collapse
|
97
|
Citak-Er F, Firat Z, Kovanlikaya I, Ture U, Ozturk-Isik E. Machine-learning in grading of gliomas based on multi-parametric magnetic resonance imaging at 3T. Comput Biol Med 2018; 99:154-160. [PMID: 29933126 DOI: 10.1016/j.compbiomed.2018.06.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/10/2018] [Accepted: 06/11/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVE The objective of this study was to assess the contribution of multi-parametric (mp) magnetic resonance imaging (MRI) quantitative features in the machine learning-based grading of gliomas with a multi-region-of-interests approach. MATERIALS AND METHODS Forty-three patients who were newly diagnosed as having a glioma were included in this study. The patients were scanned prior to any therapy using a standard brain tumor magnetic resonance (MR) imaging protocol that included T1 and T2-weighted, diffusion-weighted, diffusion tensor, MR perfusion and MR spectroscopic imaging. Three different regions-of-interest were drawn for each subject to encompass tumor, immediate tumor periphery, and distant peritumoral edema/normal. The normalized mp-MRI features were used to build machine-learning models for differentiating low-grade gliomas (WHO grades I and II) from high grades (WHO grades III and IV). In order to assess the contribution of regional mp-MRI quantitative features to the classification models, a support vector machine-based recursive feature elimination method was applied prior to classification. RESULTS A machine-learning model based on support vector machine algorithm with linear kernel achieved an accuracy of 93.0%, a specificity of 86.7%, and a sensitivity of 96.4% for the grading of gliomas using ten-fold cross validation based on the proposed subset of the mp-MRI features. CONCLUSION In this study, machine-learning based on multiregional and multi-parametric MRI data has proven to be an important tool in grading glial tumors accurately even in this limited patient population. Future studies are needed to investigate the use of machine learning algorithms for brain tumor classification in a larger patient cohort.
Collapse
Affiliation(s)
- Fusun Citak-Er
- Department of Computer Programming, Pîrî Reis University, Istanbul, Turkey; Department of Biotechnology, Yeditepe University, Istanbul, Turkey.
| | - Zeynep Firat
- Department of Radiology, Yeditepe University Hospital, Istanbul, Turkey
| | - Ilhami Kovanlikaya
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Ugur Ture
- Department of Neurosurgery, Yeditepe University Hospital, Istanbul, Turkey
| | - Esin Ozturk-Isik
- Biomedical Engineering Institute, Boğaziçi University, Istanbul, Turkey
| |
Collapse
|
98
|
Luks TL, McKnight TR, Jalbert LE, Williams A, Neill E, Lobo KA, Persson AI, Perry A, Phillips JJ, Molinaro AM, Chang SM, Nelson SJ. Relationship of In Vivo MR Parameters to Histopathological and Molecular Characteristics of Newly Diagnosed, Nonenhancing Lower-Grade Gliomas. Transl Oncol 2018; 11:941-949. [PMID: 29883968 PMCID: PMC6041571 DOI: 10.1016/j.tranon.2018.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/02/2018] [Accepted: 05/08/2018] [Indexed: 11/05/2022] Open
Abstract
The goal of this research was to elucidate the relationship between WHO 2016 molecular classifications of newly diagnosed, nonenhancing lower grade gliomas (LrGG), tissue sample histopathology, and magnetic resonance (MR) parameters derived from diffusion, perfusion, and 1H spectroscopic imaging from the tissue sample locations and the entire tumor. A total of 135 patients were scanned prior to initial surgery, with tumor cellularity scores obtained from 88 image-guided tissue samples. MR parameters were obtained from corresponding sample locations, and histograms of normalized MR parameters within the T2 fluid-attenuated inversion recovery lesion were analyzed in order to evaluate differences between subgroups. For tissue samples, higher tumor scores were related to increased normalized apparent diffusion coefficient (nADC), lower fractional anisotropy (nFA), lower cerebral blood volume (nCBV), higher choline (nCho), and lower N-acetylaspartate (nNAA). Within the T2 lesion, higher tumor grade was associated with higher nADC, lower nFA, and higher Cho to NAA index. Pathological analysis confirmed that diffusion and metabolic parameters increased and perfusion decreased with tumor cellularity. This information can be used to select targets for tissue sampling and to aid in making decisions about treating residual disease.
Collapse
Affiliation(s)
- Tracy L Luks
- Department of Radiology and Biomedical Imaging, University of California San Francisco.
| | | | - Llewellyn E Jalbert
- Department of Radiology and Biomedical Imaging, University of California San Francisco
| | - Aurelia Williams
- Department of Radiology and Biomedical Imaging, University of California San Francisco
| | - Evan Neill
- Department of Radiology and Biomedical Imaging, University of California San Francisco
| | - Khadjia A Lobo
- Department of Radiology and Biomedical Imaging, University of California San Francisco
| | | | - Arie Perry
- Department of Neurology, University of California San Francisco
| | - Joanna J Phillips
- Department of Pathology, University of California San Francisco; Department of Neurological Surgery, University of California San Francisco
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco; Department of Epidemiology and Biostatistics, University of California San Francisco
| | - Susan M Chang
- Department of Neurological Surgery, University of California San Francisco
| | - Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California San Francisco
| |
Collapse
|
99
|
Pala A, Reske SN, Eberhardt N, Scheuerle A, König R, Schmitz B, Beer AJ, Wirtz CR, Coburger J. Diagnostic accuracy of intraoperative perfusion-weighted MRI and 5-aminolevulinic acid in relation to contrast-enhanced intraoperative MRI and 11C-methionine positron emission tomography in resection of glioblastoma: a prospective study. Neurosurg Rev 2018; 42:471-479. [PMID: 29808321 DOI: 10.1007/s10143-018-0987-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/06/2018] [Accepted: 05/21/2018] [Indexed: 10/14/2022]
Abstract
The aim of our study was to compare depicted pre-, intra-, and postoperative tumor volume of met-PET, perfusion-weighed MRI (PWI), and Gd-DTPA MRI. Further, to assess their sensitivity and specificity in correlation with histopathological specimen. Inclusion criteria of the prospective study were histological confirmed glioblastoma (GB), age > 18, and eligible for gross total resection (GTR). Met-PET was performed before and after surgery. Gd-DTPA MRI and PWI were performed before, during, and after surgery. A combined 5-aminolevulinic acid (5-ALA) and iMRI-guided surgery was performed. Volumetric analysis was evaluated for all imaging modalities except for 5-ALA. A total of 59 navigated biopsies were taken. Sensitivity and specificity were calculated for Gd-DTPA MRI, PWI, met-PET, and 5-ALA according to the histology of specimen. Met-PET depicted significantly larger tumor volume before surgery (p = 0.01) compared to PWI and Gd-DTPI MRI. We found no significant difference in tumor volume between met-PET and PWI after surgery (p = 0.059). Both PWI and met-PET showed significantly larger tumor volume after surgery when compared to Gd-DTPA (p = 0.018 and p = 0.003, respectively). Intraoperative PWI reading was impaired in 33.3% due to artifacts. Met-PET showed the highest sensitivity for detection of GB with 95%. The lowest sensitivity was found with Gd-DTPA MRI (50%), while 5-ALA and intraoperative PWI showed similar results (69 and 67%). Met-Pet is the imaging modality with the highest sensitivity to detect a residual tumor in GB. Intraoperative PWI seems to have a synergistic effect to Gd-DTPA and 5-ALA. However, its value may be limited by artifacts. Both pre- and intraoperative PWI cannot substitute met-PET in tumor detection.
Collapse
Affiliation(s)
- Andrej Pala
- Department of Neurosurgery, University of Ulm, Ludwig-Heilmeyerstr. 2, 89312, Günzburg, Germany.
| | - Sven N Reske
- Department of Nuclear Medicine, University of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Nina Eberhardt
- Department of Nuclear Medicine, University of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Angelika Scheuerle
- Department of Neuropathology, University of Ulm, Ludwig-Heilmeyerstr. 2, 89312, Günzburg, Germany
| | - Ralph König
- Department of Neurosurgery, University of Ulm, Ludwig-Heilmeyerstr. 2, 89312, Günzburg, Germany
| | - Bernd Schmitz
- Department of Neuroradiology, University of Ulm, Ludwig-Heilmeyerstr. 2, 89312, Günzburg, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, University of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Christian R Wirtz
- Department of Neurosurgery, University of Ulm, Ludwig-Heilmeyerstr. 2, 89312, Günzburg, Germany
| | - Jan Coburger
- Department of Neurosurgery, University of Ulm, Ludwig-Heilmeyerstr. 2, 89312, Günzburg, Germany
| |
Collapse
|
100
|
Mughal AA, Zhang L, Fayzullin A, Server A, Li Y, Wu Y, Glass R, Meling T, Langmoen IA, Leergaard TB, Vik-Mo EO. Patterns of Invasive Growth in Malignant Gliomas-The Hippocampus Emerges as an Invasion-Spared Brain Region. Neoplasia 2018; 20:643-656. [PMID: 29793116 PMCID: PMC6030235 DOI: 10.1016/j.neo.2018.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 03/07/2018] [Accepted: 04/02/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND: Widespread infiltration of tumor cells into surrounding brain parenchyma is a hallmark of malignant gliomas, but little data exist on the overall invasion pattern of tumor cells throughout the brain. METHODS: We have studied the invasive phenotype of malignant gliomas in two invasive mouse models and patients. Tumor invasion patterns were characterized in a patient-derived xenograft mouse model using brain-wide histological analysis and magnetic resonance (MR) imaging. Findings were histologically validated in a cdkn2a−/− PDGF-β lentivirus-induced mouse glioblastoma model. Clinical verification of the results was obtained by analysis of MR images of malignant gliomas. RESULTS: Histological analysis using human-specific cellular markers revealed invasive tumors with a non-radial invasion pattern. Tumors cells accumulated in structures located far from the transplant site, such as the optic white matter and pons, whereas certain adjacent regions were spared. As such, the hippocampus was remarkably free of infiltrating tumor cells despite the extensive invasion of surrounding regions. Similarly, MR images of xenografted mouse brains displayed tumors with bihemispheric pathology, while the hippocampi appeared relatively normal. In patients, most malignant temporal lobe gliomas were located lateral to the collateral sulcus. Despite widespread pathological fluid-attenuated inversion recovery signal in the temporal lobe, 74% of the “lateral tumors” did not show signs of involvement of the amygdalo-hippocampal complex. CONCLUSIONS: Our data provide clear evidence for a compartmental pattern of invasive growth in malignant gliomas. The observed invasion patterns suggest the presence of preferred migratory paths, as well as intra-parenchymal boundaries that may be difficult for glioma cells to traverse supporting the notion of compartmental growth. In both mice and human patients, the hippocampus appears to be a brain region that is less prone to tumor invasion.
Collapse
Affiliation(s)
- Awais A Mughal
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research, Oslo University Hospital, Oslo, Norway; Department of Neurosurgery, Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; SFI-CAST-Cancer Stem Cell Innovation Center, Oslo University Hospital, Oslo, Norway.
| | - Lili Zhang
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Artem Fayzullin
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research, Oslo University Hospital, Oslo, Norway; Department of Neurosurgery, Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Andres Server
- Section of Neuroradiology, Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Yuping Li
- Neurosurgical Research, Ludwig-Maximilian University of Munich, Munich, Germany
| | - Yingxi Wu
- Neurosurgical Research, Ludwig-Maximilian University of Munich, Munich, Germany
| | - Rainer Glass
- Neurosurgical Research, Ludwig-Maximilian University of Munich, Munich, Germany
| | - Torstein Meling
- Department of Neurosurgery, Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Iver A Langmoen
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research, Oslo University Hospital, Oslo, Norway; Department of Neurosurgery, Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; SFI-CAST-Cancer Stem Cell Innovation Center, Oslo University Hospital, Oslo, Norway; Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, Norway
| | - Trygve B Leergaard
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Einar O Vik-Mo
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research, Oslo University Hospital, Oslo, Norway; Department of Neurosurgery, Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; SFI-CAST-Cancer Stem Cell Innovation Center, Oslo University Hospital, Oslo, Norway; Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, Norway
| |
Collapse
|