1
|
Galldiks N, Kaufmann TJ, Vollmuth P, Lohmann P, Smits M, Veronesi MC, Langen KJ, Rudà R, Albert NL, Hattingen E, Law I, Hutterer M, Soffietti R, Vogelbaum MA, Wen PY, Weller M, Tonn JC. Challenges, limitations, and pitfalls of PET and advanced MRI in patients with brain tumors: A report of the PET/RANO group. Neuro Oncol 2024; 26:1181-1194. [PMID: 38466087 PMCID: PMC11226881 DOI: 10.1093/neuonc/noae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Indexed: 03/12/2024] Open
Abstract
Brain tumor diagnostics have significantly evolved with the use of positron emission tomography (PET) and advanced magnetic resonance imaging (MRI) techniques. In addition to anatomical MRI, these modalities may provide valuable information for several clinical applications such as differential diagnosis, delineation of tumor extent, prognostication, differentiation between tumor relapse and treatment-related changes, and the evaluation of response to anticancer therapy. In particular, joint recommendations of the Response Assessment in Neuro-Oncology (RANO) Group, the European Association of Neuro-oncology, and major European and American Nuclear Medicine societies highlighted that the additional clinical value of radiolabeled amino acids compared to anatomical MRI alone is outstanding and that its widespread clinical use should be supported. For advanced MRI and its steadily increasing use in clinical practice, the Standardization Subcommittee of the Jumpstarting Brain Tumor Drug Development Coalition provided more recently an updated acquisition protocol for the widely used dynamic susceptibility contrast perfusion MRI. Besides amino acid PET and perfusion MRI, other PET tracers and advanced MRI techniques (e.g. MR spectroscopy) are of considerable clinical interest and are increasingly integrated into everyday clinical practice. Nevertheless, these modalities have shortcomings which should be considered in clinical routine. This comprehensive review provides an overview of potential challenges, limitations, and pitfalls associated with PET imaging and advanced MRI techniques in patients with gliomas or brain metastases. Despite these issues, PET imaging and advanced MRI techniques continue to play an indispensable role in brain tumor management. Acknowledging and mitigating these challenges through interdisciplinary collaboration, standardized protocols, and continuous innovation will further enhance the utility of these modalities in guiding optimal patient care.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Germany
| | | | - Philipp Vollmuth
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
| | - Marion Smits
- Department of Radiology and Nuclear Medicine and Brain Tumour Center, Erasmus MC, Rotterdam, The Netherlands
| | - Michael C Veronesi
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilians-University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elke Hattingen
- Goethe University, Department of Neuroradiology, University Hospital Frankfurt, Frankfurt, Germany
| | - Ian Law
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Markus Hutterer
- Department of Neurology with Acute Geriatrics, Saint John of God Hospital, Linz, Austria
| | - Riccardo Soffietti
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Michael A Vogelbaum
- Department of Neuro-Oncology and Neurosurgery, Moffit Cancer Center, Tampa, Florida, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, and University Hospital of Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Joerg-Christian Tonn
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurosurgery, University Hospital of Munich (LMU), Munich, Germany
| |
Collapse
|
2
|
Khalaj K, Jacobs MA, Zhu JJ, Esquenazi Y, Hsu S, Tandon N, Akhbardeh A, Zhang X, Riascos R, Kamali A. The Use of Apparent Diffusion Coefficient Values for Differentiating Bevacizumab-Related Cytotoxicity from Tumor Recurrence and Radiation Necrosis in Glioblastoma. Cancers (Basel) 2024; 16:2440. [PMID: 39001500 PMCID: PMC11240552 DOI: 10.3390/cancers16132440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/16/2024] Open
Abstract
OBJECTIVES Glioblastomas (GBM) are the most common primary invasive neoplasms of the brain. Distinguishing between lesion recurrence and different types of treatment related changes in patients with GBM remains challenging using conventional MRI imaging techniques. Therefore, accurate and precise differentiation between true progression or pseudoresponse is crucial in deciding on the appropriate course of treatment. This retrospective study investigated the potential of apparent diffusion coefficient (ADC) map values derived from diffusion-weighted imaging (DWI) as a noninvasive method to increase diagnostic accuracy in treatment response. METHODS A cohort of 21 glioblastoma patients (mean age: 59.2 ± 11.8, 12 Male, 9 Female) that underwent treatment with bevacizumab were selected. The ADC values were calculated from the DWI images obtained from a standardized brain protocol across 1.5-T and 3-T MRI scanners. Ratios were calculated for rADC values. Lesions were classified as bevacizumab-induced cytotoxicity based on characteristic imaging features (well-defined regions of restricted diffusion with persistent diffusion restriction over the course of weeks without tissue volume loss and absence of contrast enhancement). The rADC value was compared to these values in radiation necrosis and recurrent lesions, which were concluded in our prior study. The nonparametric Wilcoxon signed rank test with p < 0.05 was used for significance. RESULTS The mean ± SD age of the selected patients was 59.2 ± 11.8. ADC values and corresponding mean rADC values for bevacizumab-induced cytotoxicity were 248.1 ± 67.2 and 0.39 ± 0.10, respectively. These results were compared to the ADC values and corresponding mean rADC values of tumor progression and radiation necrosis. Significant differences between rADC values were observed in all three groups (p < 0.001). Bevacizumab-induced cytotoxicity had statistically significant lower ADC values compared to both tumor recurrence and radiation necrosis. CONCLUSION The study demonstrates the potential of ADC values as noninvasive imaging biomarkers for differentiating recurrent glioblastoma from radiation necrosis and bevacizumab-induced cytotoxicity.
Collapse
Affiliation(s)
- Kamand Khalaj
- Department of Diagnostic and Interventional Imaging, UTHealth Houston, Houston, TX 77030, USA
| | - Michael A Jacobs
- Department of Diagnostic and Interventional Imaging, UTHealth Houston, Houston, TX 77030, USA
- The Department of Radiology and Oncology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Computer Science, Rice University, Houston, TX 77005, USA
| | - Jay-Jiguang Zhu
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, UTHealth Houston, Houston, TX 77030, USA
| | - Yoshua Esquenazi
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, UTHealth Houston, Houston, TX 77030, USA
| | - Sigmund Hsu
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, UTHealth Houston, Houston, TX 77030, USA
| | - Nitin Tandon
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, UTHealth Houston, Houston, TX 77030, USA
| | - Alireza Akhbardeh
- Department of Diagnostic and Interventional Imaging, UTHealth Houston, Houston, TX 77030, USA
| | - Xu Zhang
- Division of Clinical and Translational Sciences, Department of Internal Medicine, UTHealth, Houston, TX 77030, USA
| | - Roy Riascos
- Department of Diagnostic and Interventional Imaging, UTHealth Houston, Houston, TX 77030, USA
| | - Arash Kamali
- Department of Diagnostic and Interventional Imaging, UTHealth Houston, Houston, TX 77030, USA
| |
Collapse
|
3
|
Kambe A, Kitao S, Ochiai R, Hosoya T, Fujii S, Kurosaki M. The utility of arterial spin labeling imaging for predicting prognosis after a recurrence of high-grade glioma in patients under bevacizumab treatment. J Neurooncol 2024; 166:175-183. [PMID: 38165552 DOI: 10.1007/s11060-023-04550-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 12/20/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND AND PURPOSE Currently, the antiangiogenic agent bevacizumab (BVZ) is used as a treatment option for high-grade glioma (HGG) patients. However, BVZ restores disruptions of the blood-brain barrier, which leads to the disappearance of contrast enhancement during radiological examinations and therefore complicates evaluations of treatment efficacy. This study aimed to investigate the radio-morphological features of recurrent lesions that newly appeared under BVZ therapy, as well as the utility of arterial spin labeling (ASL) perfusion imaging for evaluating treatment response and prognosis in HGG patients receiving BVZ. METHODS Thirty-two patients (20 males, 12 females; age range, 35-84 years) with HGG who experienced a recurrence under BVZ therapy were enrolled. We measured the relative cerebral blood flow (rCBF) values of each recurrent lesion using ASL, and retrospectively investigated the correlation between rCBF values and prognosis. RESULTS The optimal rCBF cut-off value for predicting prognosis was defined as 1.67 using receiver operating characteristic curve analysis. The patients in the rCBF < 1.67 group had significantly longer overall survival (OS) and post-progression survival (PPS) than those in the rCBF ≥ 1.67 group (OS: 34.0 months vs. 13.0 months, p = 0.03 and PPS: 13.0 months vs. 6.0 months, p < 0.001, respectively). CONCLUSION The ASL-derived rCBF values of recurrent lesions may serve as an effective imaging biomarker for prognosis in HGG patients undergoing BVZ therapy. Low rCBF values may indicate that BVZ efficacy is sustainable, which will influence BVZ treatment strategies in HGG patients.
Collapse
Affiliation(s)
- Atsushi Kambe
- Department of Brain and Neurosciences, Division of Neurosurgery, Faculty of Medicine, Tottori University, Tottori, Japan.
| | - Shinichiro Kitao
- Department of Multidisciplinary Internal Medicine, Division of Radiology, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Ryoya Ochiai
- Department of Multidisciplinary Internal Medicine, Division of Radiology, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Tomohiro Hosoya
- Department of Brain and Neurosciences, Division of Neurosurgery, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Shinya Fujii
- Department of Multidisciplinary Internal Medicine, Division of Radiology, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Masamichi Kurosaki
- Department of Brain and Neurosciences, Division of Neurosurgery, Faculty of Medicine, Tottori University, Tottori, Japan
| |
Collapse
|
4
|
Lawrence LSP, Chan RW, Chen H, Stewart J, Ruschin M, Theriault A, Myrehaug S, Detsky J, Maralani PJ, Tseng CL, Soliman H, Jane Lim-Fat M, Das S, Stanisz GJ, Sahgal A, Lau AZ. Diffusion-weighted imaging on an MRI-linear accelerator to identify adversely prognostic tumour regions in glioblastoma during chemoradiation. Radiother Oncol 2023; 188:109873. [PMID: 37640160 DOI: 10.1016/j.radonc.2023.109873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/12/2023] [Accepted: 08/20/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND AND PURPOSE Survival in glioblastoma might be extended by escalating the radiotherapy dose to treatment-resistant tumour and adapting to tumour changes. Diffusion-weighted imaging (DWI) on MRI-linear accelerators (MR-Linacs) could be used to identify a dose escalation target, but its prognostic value must be demonstrated. The purpose of this study was to determine whether MR-Linac DWI can assess treatment response in glioblastoma and whether changes in DWI show greater prognostic value than changes in the contrast-enhancing gross tumour volume (GTV). MATERIALS AND METHODS Seventy-five patients with glioblastoma were treated with chemoradiotherapy, of which 32 were treated on a 1.5 T MRI-linear accelerator (MR-Linac). Patients were imaged with simulation MRI scanners (MR-sim) at treatment planning and weeks 2, 4, and 10 after treatment start. Twenty-eight patients had additional MR-Linac DWI sequences. Cox modelling was used to evaluate the correlation of overall and progression-free survival (OS and PFS) with clinical variables and volumetric changes in the GTV and low-ADC regions (ADC < 1.25 µm2/ms within GTV). RESULTS In total, 479 MR-Linac DWI and 289 MR-sim DWI datasets were analyzed. MR-Linac low-ADC changes between weeks 2 and 5 inclusive were prognostic for OS (hazard ratio lower limits ≥ 1.2, p-values ≤ 0.02). MR-sim low-ADC changes showed greater correlation with OS and PFS than GTV changes (e.g., OS hazard ratio at week 2 was 3.4 (p <0.001) for low-ADC versus 2.0 (p = 0.022) for GTV). CONCLUSION MR-Linac DWI can measure low-ADC tumour volumes that correlate with OS and PFS better than contrast-enhancing GTV. Low-ADC regions could serve as dose escalation targets.
Collapse
Affiliation(s)
| | - Rachel W Chan
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Hanbo Chen
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - James Stewart
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Mark Ruschin
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Aimee Theriault
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Sten Myrehaug
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Jay Detsky
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Pejman J Maralani
- Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Chia-Lin Tseng
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Hany Soliman
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Mary Jane Lim-Fat
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Sunit Das
- Keenan Chair in Surgery, St. Michael's Hospital, Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada
| | - Greg J Stanisz
- Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Neurosurgery and Paediatric Neurosurgery, Medical University, Lublin, Poland
| | - Arjun Sahgal
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Angus Z Lau
- Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
Breda-Yepes M, Rodríguez-Hernández LA, Gómez-Figueroa E, Mondragón-Soto MG, Arellano-Flores G, Hernández-Hernández A, Rodríguez-Rubio HA, Martínez P, Reyes-Moreno I, Álvaro-Heredia JA, Gutiérrez Aceves GA, Villanueva-Castro E, Sangrador-Deitos MV, Alonso-Vanegas M, Guerrero-Juárez V, González-Aguilar A. Relative cerebral blood volume as response predictor in the treatment of recurrent glioblastoma with anti-angiogenic therapy. Clin Neurol Neurosurg 2023; 233:107904. [PMID: 37499302 DOI: 10.1016/j.clineuro.2023.107904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/08/2023] [Accepted: 07/16/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Glioblastoma is one of the most common brain tumors in adult populations, usually carrying a poor prognosis. While several studies have researched the impact of anti-angiogenic therapies, especially anti-VEFG treatments in glioblastoma, few have attempted to assess its progress using imaging studies. PURPOSE We attempted to analyze whether relative cerebral blood volume (rCBV) from dynamic susceptibility-weighted contrast-enhanced MRI (DSC-MRI) could predict response in patients with glioblastoma undergoing Bevacizumab (BVZ) treatment. METHODS We performed a retrospective study evaluating patients with recurrent glioblastoma receiving anti-angiogenic therapy with BVZ between 2012 and 2017 in our institution. Patients were scheduled for routine MRIs at baseline and first-month follow-up visits. Studies were processed for DSC-MRI, cT1, and FLAIR images, from which relative cerebral blood volume measurements were obtained. We assessed patient response using the Response Assessment in Neuro-Oncology (RANO) working group criteria and overall survival. RESULTS 40 patients were included in the study and were classified as Bevacizumab responders and non-responders. The average rCBV before treatment was 4.5 for both groups, and average rCBV was 2.5 for responders and 5.4 for non-responders. ROC curve set a cutoff point of 3.7 for rCBV predictive of response to BVZ. Cox Multivariate analysis only showed rCBV as a predictive factor of OS. CONCLUSION A statistically significant difference was found in rCBV between patients who responded and those who did not respond to BVZ treatment. rCBV may be a low-cost and effective marker to assess response to Bevacizumab treatment in GBM.
Collapse
Affiliation(s)
- Michele Breda-Yepes
- Department of Neurosurgery, National Institute of Neurology and Neurosurgery, Mexico
| | | | | | | | | | | | | | - Pablo Martínez
- Department of Neurosurgery, National Institute of Neurology and Neurosurgery, Mexico
| | | | - Juan A Álvaro-Heredia
- Department of Neurosurgery, National Institute of Neurology and Neurosurgery, Mexico
| | | | | | | | - Mario Alonso-Vanegas
- Department of Neurosurgery, National Institute of Neurology and Neurosurgery, Mexico
| | | | - Alberto González-Aguilar
- The American British Cowdray (ABC) Medical Center, Mexico City, Mexico; Department of Neuro-Oncology, National Institute of Neurology and Neurosurgery, Mexico; Emergency Department, National Institute of Neurology and Neurosurgery, Mexico.
| |
Collapse
|
6
|
Yearley AG, Goedmakers CMW, Panahi A, Doucette J, Rana A, Ranganathan K, Smith TR. FDA-approved machine learning algorithms in neuroradiology: A systematic review of the current evidence for approval. Artif Intell Med 2023; 143:102607. [PMID: 37673576 DOI: 10.1016/j.artmed.2023.102607] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 09/08/2023]
Abstract
Over the past decade, machine learning (ML) and artificial intelligence (AI) have become increasingly prevalent in the medical field. In the United States, the Food and Drug Administration (FDA) is responsible for regulating AI algorithms as "medical devices" to ensure patient safety. However, recent work has shown that the FDA approval process may be deficient. In this study, we evaluate the evidence supporting FDA-approved neuroalgorithms, the subset of machine learning algorithms with applications in the central nervous system (CNS), through a systematic review of the primary literature. Articles covering the 53 FDA-approved algorithms with applications in the CNS published in PubMed, EMBASE, Google Scholar and Scopus between database inception and January 25, 2022 were queried. Initial searches identified 1505 studies, of which 92 articles met the criteria for extraction and inclusion. Studies were identified for 26 of the 53 neuroalgorithms, of which 10 algorithms had only a single peer-reviewed publication. Performance metrics were available for 15 algorithms, external validation studies were available for 24 algorithms, and studies exploring the use of algorithms in clinical practice were available for 7 algorithms. Papers studying the clinical utility of these algorithms focused on three domains: workflow efficiency, cost savings, and clinical outcomes. Our analysis suggests that there is a meaningful gap between the FDA approval of machine learning algorithms and their clinical utilization. There appears to be room for process improvement by implementation of the following recommendations: the provision of compelling evidence that algorithms perform as intended, mandating minimum sample sizes, reporting of a predefined set of performance metrics for all algorithms and clinical application of algorithms prior to widespread use. This work will serve as a baseline for future research into the ideal regulatory framework for AI applications worldwide.
Collapse
Affiliation(s)
- Alexander G Yearley
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA; Computational Neuroscience Outcomes Center (CNOC), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA.
| | - Caroline M W Goedmakers
- Computational Neuroscience Outcomes Center (CNOC), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA; Department of Neurosurgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Armon Panahi
- The George Washington University School of Medicine and Health Sciences, 2300 I St NW, Washington, DC 20052, USA
| | - Joanne Doucette
- Computational Neuroscience Outcomes Center (CNOC), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA; School of Pharmacy, MCPHS University, 179 Longwood Ave, Boston, MA 02115, USA
| | - Aakanksha Rana
- Computational Neuroscience Outcomes Center (CNOC), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA; Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Kavitha Ranganathan
- Division of Plastic Surgery, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, USA
| | - Timothy R Smith
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA; Computational Neuroscience Outcomes Center (CNOC), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| |
Collapse
|
7
|
Werner JM, Wollring MM, Tscherpel C, Rosen EK, Werr L, Stetter I, Rueß D, Ruge MI, Brunn A, Al Shughri A, Kabbasch C, Fink GR, Langen KJ, Galldiks N. Multimodal imaging findings in patients with glioblastoma with extensive coagulative necrosis related to regorafenib. Neuro Oncol 2023; 25:1193-1195. [PMID: 36960770 PMCID: PMC10237410 DOI: 10.1093/neuonc/noad051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023] Open
Affiliation(s)
- Jan-Michael Werner
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Michael M Wollring
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Caroline Tscherpel
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Elena K Rosen
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lisa Werr
- Department of Experimental Pediatric Oncology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Isabelle Stetter
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Daniel Rueß
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne,Germany
| | - Maximilian I Ruge
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne,Germany
| | - Anna Brunn
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Abdulkader Al Shughri
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Christoph Kabbasch
- Institure of Radiology, Division of Neuroradiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne,Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Cologne, Germany
| |
Collapse
|
8
|
Salans M, Houri J, Karunamuni R, Hopper A, Delfanti R, Seibert TM, Bahrami N, Sharifzadeh Y, McDonald C, Dale A, Moiseenko V, Farid N, Hattangadi-Gluth JA. The relationship between radiation dose and bevacizumab-related imaging abnormality in patients with brain tumors: A voxel-wise normal tissue complication probability (NTCP) analysis. PLoS One 2023; 18:e0279812. [PMID: 36800342 PMCID: PMC9937457 DOI: 10.1371/journal.pone.0279812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/15/2022] [Indexed: 02/18/2023] Open
Abstract
PURPOSE Bevacizumab-related imaging abnormality (BRIA), appearing as areas of restricted diffusion on magnetic resonance imaging (MRI) and representing atypical coagulative necrosis pathologically, has been observed in patients with brain tumors receiving radiotherapy and bevacizumab. We investigated the role of cumulative radiation dose in BRIA development in a voxel-wise analysis. METHODS Patients (n = 18) with BRIA were identified. All had high-grade gliomas or brain metastases treated with radiotherapy and bevacizumab. Areas of BRIA were segmented semi-automatically on diffusion-weighted MRI with apparent diffusion coefficient (ADC) images. To avoid confounding by possible tumor, hypoperfusion was confirmed with perfusion imaging. ADC images and radiation dose maps were co-registered to a high-resolution T1-weighted MRI and registration accuracy was verified. Voxel-wise normal tissue complication probability analyses were performed using a logistic model analyzing the relationship between cumulative voxel equivalent total dose in 2 Gy fractions (EQD2) and BRIA development at each voxel. Confidence intervals for regression model predictions were estimated with bootstrapping. RESULTS Among 18 patients, 39 brain tumors were treated. Patients received a median of 4.5 cycles of bevacizumab and 1-4 radiation courses prior to BRIA appearance. Most (64%) treated tumors overlapped with areas of BRIA. The median proportion of each BRIA region of interest volume overlapping with tumor was 98%. We found a dose-dependent association between cumulative voxel EQD2 and the relative probability of BRIA (β0 = -5.1, β1 = 0.03 Gy-1, γ = 1.3). CONCLUSIONS BRIA is likely a radiation dose-dependent phenomenon in patients with brain tumors receiving bevacizumab and radiotherapy. The combination of radiation effects and tumor microenvironmental factors in potentiating BRIA in this population should be further investigated.
Collapse
Affiliation(s)
- Mia Salans
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Jordan Houri
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, United States of America
- Carl E. Ravin Advanced Imaging Laboratories, Duke University, Durham, North Carolina, United States of America
| | - Roshan Karunamuni
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Austin Hopper
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Rachel Delfanti
- Department of Radiology, University of California San Diego, La Jolla, California, United States of America
| | - Tyler M. Seibert
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, United States of America
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Naeim Bahrami
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Yasamin Sharifzadeh
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Carrie McDonald
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, United States of America
- Department of Psychiatry, University of California San Diego, La Jolla, California, United States of America
| | - Anders Dale
- Department of Radiology, University of California San Diego, La Jolla, California, United States of America
- Department of Psychiatry, University of California San Diego, La Jolla, California, United States of America
| | - Vitali Moiseenko
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Nikdokht Farid
- Department of Radiology, University of California San Diego, La Jolla, California, United States of America
| | - Jona A. Hattangadi-Gluth
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
9
|
Puac-Polanco P, Zakhari N, Miller J, McComiskey D, Thornhill RE, Jansen GH, Nair VJ, Nguyen TB. Diagnostic Accuracy of Centrally Restricted Diffusion Sign in Cerebral Metastatic Disease: Differentiating Radiation Necrosis from Tumor Recurrence. Can Assoc Radiol J 2023; 74:100-109. [PMID: 35848632 DOI: 10.1177/08465371221115341] [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: 01/11/2023] Open
Abstract
Purpose: The centrally restricted diffusion sign of diffusion-weighted imaging (DWI) is associated with radiation necrosis (RN) in treated gliomas. Our goal was to evaluate its diagnostic accuracy to distinguish RN from tumor recurrence (TR) in treated brain metastases. Methods: Retrospective study of consecutive patients with brain metastases who developed a newly centrally necrotic lesion after radiotherapy (RT). One reader placed regions of interest (ROI) in the enhancing solid lesion and the non-enhancing central necrosis on the apparent diffusion coefficient (ADC) map. Two readers qualitatively assessed the presence of the centrally restricted diffusion sign. The final diagnosis was made by histopathology (n = 39) or imaging follow-up (n = 2). Differences between groups were assessed by Fisher's exact or Mann-Whitney U tests. Diagnostic accuracy and inter-reader agreement were evaluated using receiver operating characteristic (ROC) curve analysis and kappa scores. Results: Forty-one lesions (32 predominant RN; 9 predominant TR) were analyzed. An ADC value ≤ 1220 × 10-6 mm2/s (sensitivity 74%, specificity 89%, area under the curve [AUC] .85 [95% confidence interval {CI}, .70-.94] P < .0001) from the necrosis and an ADC necrosis/enhancement ratio ≤1.37 (sensitivity 74%, specificity 89%, AUC .82 [95% CI, .67-.93] P < .0001) provided the highest performance for RN diagnosis. The qualitative centrally restricted diffusion sign had a sensitivity of 69% (95% CI, .50-.83), specificity of 77% (95% CI, .40-.96), and a moderate (k = .49) inter-reader agreement for RN diagnosis. Conclusions: Radiation necrosis is associated with lower ADC values in the central necrosis than TR. A moderate interobserver agreement might limit the qualitative assessment of the centrally restricted diffusion sign.
Collapse
Affiliation(s)
- Paulo Puac-Polanco
- Department of Radiology, Radiation Oncology and Medical Physics, 6363University of Ottawa, Ottawa, ON, Canada
| | - Nader Zakhari
- Department of Radiology, Radiation Oncology and Medical Physics, 6363University of Ottawa, Ottawa, ON, Canada
| | - Jacob Miller
- Department of Radiology, Radiation Oncology and Medical Physics, 6363University of Ottawa, Ottawa, ON, Canada
| | - David McComiskey
- Department of Radiology, Radiation Oncology and Medical Physics, 6363University of Ottawa, Ottawa, ON, Canada
| | - Rebecca E Thornhill
- Department of Radiology, Radiation Oncology and Medical Physics, 6363University of Ottawa, Ottawa, ON, Canada
| | - Gerard H Jansen
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, 6363University of Ottawa, Ottawa, ON, Canada
| | - Vimoj J Nair
- Department of Radiology, Radiation Oncology and Medical Physics, 6363University of Ottawa, Ottawa, ON, Canada.,The Ottawa Hospital Research Institute (OHRI)
| | - Thanh Binh Nguyen
- Department of Radiology, Radiation Oncology and Medical Physics, 6363University of Ottawa, Ottawa, ON, Canada.,The Ottawa Hospital Research Institute (OHRI)
| |
Collapse
|
10
|
Han RH, Johanns TM, Roberts KF, Tao Y, Luo J, Ye Z, Sun P, Blum J, Lin TH, Song SK, Kim AH. Diffusion basis spectrum imaging as an adjunct to conventional MRI leads to earlier diagnosis of high-grade glioma tumor progression versus treatment effect. Neurooncol Adv 2023; 5:vdad050. [PMID: 37215950 PMCID: PMC10195207 DOI: 10.1093/noajnl/vdad050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
Background Following chemoradiotherapy for high-grade glioma (HGG), it is often challenging to distinguish treatment changes from true tumor progression using conventional MRI. The diffusion basis spectrum imaging (DBSI) hindered fraction is associated with tissue edema or necrosis, which are common treatment-related changes. We hypothesized that DBSI hindered fraction may augment conventional imaging for earlier diagnosis of progression versus treatment effect. Methods Adult patients were prospectively recruited if they had a known histologic diagnosis of HGG and completed standard-of-care chemoradiotherapy. DBSI and conventional MRI data were acquired longitudinally beginning 4 weeks post-radiation. Conventional MRI and DBSI metrics were compared with respect to their ability to diagnose progression versus treatment effect. Results Twelve HGG patients were enrolled between August 2019 and February 2020, and 9 were ultimately analyzed (5 progression, 4 treatment effect). Within new or enlarging contrast-enhancing regions, DBSI hindered fraction was significantly higher in the treatment effect group compared to progression group (P = .0004). Compared to serial conventional MRI alone, inclusion of DBSI would have led to earlier diagnosis of either progression or treatment effect in 6 (66.7%) patients by a median of 7.7 (interquartile range = 0-20.1) weeks. Conclusions In the first longitudinal prospective study of DBSI in adult HGG patients, we found that in new or enlarging contrast-enhancing regions following therapy, DBSI hindered fraction is elevated in cases of treatment effect compared to those with progression. Hindered fraction map may be a valuable adjunct to conventional MRI to distinguish tumor progression from treatment effect.
Collapse
Affiliation(s)
- Rowland H Han
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tanner M Johanns
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kaleigh F Roberts
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yu Tao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jingqin Luo
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zezhong Ye
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Peng Sun
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jacob Blum
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tsen-Hsuan Lin
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sheng-Kwei Song
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Albert H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
11
|
Agarwal A, Desai A, Gupta V, Vibhute P. Bevacizumab-induced Coagulative Necrosis with Restricted Diffusion. Radiol Imaging Cancer 2022; 4:e220089. [PMID: 36112037 PMCID: PMC9530776 DOI: 10.1148/rycan.220089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/07/2022] [Accepted: 08/26/2022] [Indexed: 06/15/2023]
|
12
|
Li AY, Iv M. Conventional and Advanced Imaging Techniques in Post-treatment Glioma Imaging. FRONTIERS IN RADIOLOGY 2022; 2:883293. [PMID: 37492665 PMCID: PMC10365131 DOI: 10.3389/fradi.2022.883293] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/06/2022] [Indexed: 07/27/2023]
Abstract
Despite decades of advancement in the diagnosis and therapy of gliomas, the most malignant primary brain tumors, the overall survival rate is still dismal, and their post-treatment imaging appearance remains very challenging to interpret. Since the limitations of conventional magnetic resonance imaging (MRI) in the distinction between recurrence and treatment effect have been recognized, a variety of advanced MR and functional imaging techniques including diffusion-weighted imaging (DWI), diffusion tensor imaging (DTI), perfusion-weighted imaging (PWI), MR spectroscopy (MRS), as well as a variety of radiotracers for single photon emission computed tomography (SPECT) and positron emission tomography (PET) have been investigated for this indication along with voxel-based and more quantitative analytical methods in recent years. Machine learning and radiomics approaches in recent years have shown promise in distinguishing between recurrence and treatment effect as well as improving prognostication in a malignancy with a very short life expectancy. This review provides a comprehensive overview of the conventional and advanced imaging techniques with the potential to differentiate recurrence from treatment effect and includes updates in the state-of-the-art in advanced imaging with a brief overview of emerging experimental techniques. A series of representative cases are provided to illustrate the synthesis of conventional and advanced imaging with the clinical context which informs the radiologic evaluation of gliomas in the post-treatment setting.
Collapse
Affiliation(s)
- Anna Y. Li
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael Iv
- Division of Neuroimaging and Neurointervention, Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
13
|
Bernstock JD, Gary SE, Klinger N, Valdes PA, Ibn Essayed W, Olsen HE, Chagoya G, Elsayed G, Yamashita D, Schuss P, Gessler FA, Peruzzi PP, Bag A, Friedman GK. Standard clinical approaches and emerging modalities for glioblastoma imaging. Neurooncol Adv 2022; 4:vdac080. [PMID: 35821676 PMCID: PMC9268747 DOI: 10.1093/noajnl/vdac080] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary adult intracranial malignancy and carries a dismal prognosis despite an aggressive multimodal treatment regimen that consists of surgical resection, radiation, and adjuvant chemotherapy. Radiographic evaluation, largely informed by magnetic resonance imaging (MRI), is a critical component of initial diagnosis, surgical planning, and post-treatment monitoring. However, conventional MRI does not provide information regarding tumor microvasculature, necrosis, or neoangiogenesis. In addition, traditional MRI imaging can be further confounded by treatment-related effects such as pseudoprogression, radiation necrosis, and/or pseudoresponse(s) that preclude clinicians from making fully informed decisions when structuring a therapeutic approach. A myriad of novel imaging modalities have been developed to address these deficits. Herein, we provide a clinically oriented review of standard techniques for imaging GBM and highlight emerging technologies utilized in disease characterization and therapeutic development.
Collapse
Affiliation(s)
- Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Sam E Gary
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Neil Klinger
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Pablo A Valdes
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Walid Ibn Essayed
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Hannah E Olsen
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Gustavo Chagoya
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Galal Elsayed
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Daisuke Yamashita
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Patrick Schuss
- Department of Neurosurgery, Unfallkrankenhaus Berlin , Berlin, Germany
| | | | - Pier Paolo Peruzzi
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Asim Bag
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital , Memphis, TN USA
| | - Gregory K Friedman
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham , Birmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham , AL, USA
| |
Collapse
|
14
|
Lombardi G, Spimpolo A, Berti S, Campi C, Anglani MG, Simeone R, Evangelista L, Causin F, Zorzi G, Gorgoni G, Caccese M, Padovan M, Zagonel V, Cecchin D. PET/MR in recurrent glioblastoma patients treated with regorafenib: [ 18F]FET and DWI-ADC for response assessment and survival prediction. Br J Radiol 2022; 95:20211018. [PMID: 34762492 PMCID: PMC8722234 DOI: 10.1259/bjr.20211018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Objective: The use of regorafenib in recurrent glioblastoma patients has been recently approved by the Italian Medicines Agency (AIFA) and added to the National Comprehensive Cancer Network (NCCN) 2020 guidelines as a preferred regimen. Given its complex effects at the molecular level, the most appropriate imaging tools to assess early response to treatment is still a matter of debate. Diffusion-weighted imaging and O-(2-18F-fluoroethyl)-L-tyrosine positron emission tomography ([18F]FET PET) are promising methodologies providing additional information to the currently used RANO criteria. The aim of this study was to evaluate the variations in diffusion-weighted imaging/apparent diffusion coefficient (ADC) and [18F]FET PET-derived parameters in patients who underwent PET/MR at both baseline and after starting regorafenib. Methods: We retrospectively reviewed 16 consecutive GBM patients who underwent [18F]FET PET/MR before and after two cycles of regorafenib. Patients were sorted into stable (SD) or progressive disease (PD) categories in accordance with RANO criteria. We were also able to analyze four SD patients who underwent a third PET/MR after another four cycles of regorafenib. [18F]FET uptake greater than 1.6 times the mean background activity was used to define an area to be superimposed on an ADC map at baseline and after treatment. Several metrics were then derived and compared. Log-rank test was applied for overall survival analysis. Results: Percentage difference in FET volumes correlates with the corresponding percentage difference in ADC (R = 0.54). Patients with a twofold increase in FET after regorafenib showed a significantly higher increase in ADC pathological volume than the remaining subjects (p = 0.0023). Kaplan–Meier analysis, performed to compare the performance in overall survival prediction, revealed that the percentage variations of FET- and ADC-derived metrics performed at least as well as RANO criteria (p = 0.02, p = 0.024 and p = 0.04 respectively) and in some cases even better. TBR Max and TBR mean are not able to accurately predict overall survival. Conclusion In recurrent glioblastoma patients treated with regorafenib, [18F]FET and ADC metrics, are able to predict overall survival and being obtained from completely different measures as compared to RANO, could serve as semi-quantitative independent biomarkers of response to treatment. Advances in knowledge Simultaneous evaluation of [18F]FET and ADC metrics using PET/MR allows an early and reliable identification of response to treatment and predict overall survival.
Collapse
Affiliation(s)
- Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology - IRCCS, Padua, Italy
| | - Alessandro Spimpolo
- Nuclear Medicine Unit, Department of Medicine - DIMED, Padua University Hospital, Padua, Italy
| | - Sara Berti
- Nuclear Medicine Unit, Department of Medicine - DIMED, Padua University Hospital, Padua, Italy
| | - Cristina Campi
- Department of Mathematics, University of Genoa, Genoa, Italy
| | | | - Rossella Simeone
- Nuclear Medicine Unit, Department of Medicine - DIMED, Padua University Hospital, Padua, Italy
| | - Laura Evangelista
- Nuclear Medicine Unit, Department of Medicine - DIMED, Padua University Hospital, Padua, Italy
| | - Francesco Causin
- Neuroradiology Unit, Azienda Ospedaliera di Padova, Padua, Italy
| | - Giovanni Zorzi
- Department of Neurosciences (DNS), University of Padua, Padua, Italy
| | - Giancarlo Gorgoni
- Radiopharmacy, Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy
| | - Mario Caccese
- Department of Oncology, Oncology 1, Veneto Institute of Oncology - IRCCS, Padua, Italy
| | - Marta Padovan
- Department of Oncology, Oncology 1, Veneto Institute of Oncology - IRCCS, Padua, Italy
| | - Vittorina Zagonel
- Department of Oncology, Oncology 1, Veneto Institute of Oncology - IRCCS, Padua, Italy
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine - DIMED, Padua University Hospital, Padua, Italy
| |
Collapse
|
15
|
Hagiwara A, Oughourlian TC, Cho NS, Schlossman J, Wang C, Yao J, Raymond C, Everson R, Patel K, Mareninov S, Rodriguez FJ, Salamon N, Pope WB, Nghiemphu PL, Liau LM, Prins RM, Cloughesy TF, Ellingson BM. Diffusion MRI is an early biomarker of overall survival benefit in IDH wild-type recurrent glioblastoma treated with immune checkpoint inhibitors. Neuro Oncol 2021; 24:1020-1028. [PMID: 34865129 DOI: 10.1093/neuonc/noab276] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Diffusion MRI estimates of the apparent diffusion coefficient (ADC) have been shown to be useful in predicting treatment response in patients with glioblastoma (GBM), with ADC elevations indicating tumor cell death. We aimed to investigate whether the ADC values measured before and after treatment with immune checkpoint inhibitors (ICIs) and the changes in these ADC values could predict overall survival (OS) in patients with recurrent IDH wild-type GBM. METHODS Forty-four patients who met the following inclusion criteria were included in this retrospective study: (i) diagnosed with recurrent IDH wild-type GBM and treated with either pembrolizumab or nivolumab and (ii) availability of diffusion data on pre- and post-ICI MRI. Tumor volume and the median relative ADC (rADC) with respect to the normal-appearing white matter within the enhancing tumor were calculated. RESULTS Median OS among all patients was 8.1 months (range, 1.0-22.5 months). Log-rank test revealed that higher post-treatment rADC was associated with a significantly longer OS (median, 10.3 months for rADC ≧ 1.63 versus 6.1 months for rADC < 1.63; P = 0.02), whereas tumor volume, pre-treatment rADC, and changes in rADC after treatment were not significantly associated with OS. Cox regression analysis revealed that post-treatment rADC significantly influenced OS (P = 0.02, univariate analysis), even after controlling for age and sex (P =0.01, multivariate analysis), and additionally controlling for surgery after ICI treatment (P = 0.045, multivariate analysis). CONCLUSIONS Elevated post-treatment rADC may be an early imaging biomarker for OS benefits in GBM patients receiving ICI treatment.
Collapse
Affiliation(s)
- Akifumi Hagiwara
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Talia C Oughourlian
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Neuroscience Interdepartmental PhD Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Nicholas S Cho
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA, USA.,Medical Scientist Training Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jacob Schlossman
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chencai Wang
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA, USA
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Richard Everson
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kunal Patel
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sergey Mareninov
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Fausto J Rodriguez
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Noriko Salamon
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Whitney B Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Phioanh L Nghiemphu
- UCLA Neuro-Oncology Program, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert M Prins
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA, USA.,UCLA Neuro-Oncology Program, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
16
|
Matsuno A, Takaya Y, Chen H, TEN H, Hoya K, Jiang CL, Oyama KI, Onoda K. Evaluation of the malignant potential of gliomas using diffusion-weighted and gadolinium-enhanced magnetic resonance imaging. BRAIN SCIENCE ADVANCES 2021. [DOI: 10.26599/bsa.2021.9050023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
17
|
Abstract
PURPOSE OF REVIEW This review aims to cover current MRI techniques for assessing treatment response in brain tumors, with a focus on radio-induced lesions. RECENT FINDINGS Pseudoprogression and radionecrosis are common radiological entities after brain tumor irradiation and are difficult to distinguish from real progression, with major consequences on daily patient care. To date, shortcomings of conventional MRI have been largely recognized but morphological sequences are still used in official response assessment criteria. Several complementary advanced techniques have been proposed but none of them have been validated, hampering their clinical use. Among advanced MRI, brain perfusion measures increase diagnostic accuracy, especially when added with spectroscopy and susceptibility-weighted imaging. However, lack of reproducibility, because of several hard-to-control variables, is still a major limitation for their standardization in routine protocols. Amide Proton Transfer is an emerging molecular imaging technique that promises to offer new metrics by indirectly quantifying intracellular mobile proteins and peptide concentration. Preliminary studies suggest that this noncontrast sequence may add key biomarkers in tumor evaluation, especially in posttherapeutic settings. SUMMARY Benefits and pitfalls of conventional and advanced imaging on posttreatment assessment are discussed and the potential added value of APT in this clinicoradiological evolving scenario is introduced.
Collapse
Affiliation(s)
- Lucia Nichelli
- Department of Neuroradiology, Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière-Charles Foix
- Sorbonne Université, INSERM, CNRS, Assistance Publique-Hôpitaux de Paris, Institut du Cerveau et de la Moelle épinière, boulevard de l’Hôpital, Paris
| | - Stefano Casagranda
- Department of Research & Innovation, Olea Medical, avenue des Sorbiers, La Ciotat, France
| |
Collapse
|
18
|
Mansour M, Vitale V, Lombardi G, Riva G, Pancheri F, Zanusso M. Modification of MRI pattern of high-grade glioma pseudoprogression in regorafenib therapy. J Med Imaging Radiat Oncol 2021; 66:414-418. [PMID: 34169667 DOI: 10.1111/1754-9485.13267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 05/27/2021] [Indexed: 11/30/2022]
Abstract
Pseudoprogression (PP) is a diagnostic dilemma in the follow-up of brain high grade gliomas (HGG), and the introduction of new therapies has further complicated its identification in Magnetic Resonance Imaging (MRI). We report a case of pseudoprogression after intraoperative radiotherapy (ioRT) and Regorafenib therapy in a patient with anaplastic astrocytoma recurrence. A 65-year-old man, treated in August 2017 for a right frontal anaplastic astrocytoma, with surgical resection and following radiotherapy and Temozolomide, in October 2019 was again treated for peri-surgical bed recurrence with resection and ioRT followed by Regorafenib therapy, interrupted in February 2020, after the onset of adverse reactions. MRI examination showed a large irregular alteration posterior to the surgical bed, T2 weighted hypointense featuring strong diffusion restriction (low ADC values), with an irregular contrast-enhancement (CE) pattern, and surrounded by a vast vasogenic oedema; Dynamic Susceptibility Contrast (DSC) perfusion imaging (PWI) showed no increase of relative cerebral blood volume (rCBV). Particularly, lesion appeared markedly hypointense and dusty-like on susceptibility weighted images (SWI) probably due to a constant hemorrhagic diapedesis promoted by Regorafenib. Therefore, pseudoprogression was suspected. Follow-up MRI exams showed gradual reduction of SWI and CE abnormalities, but a persistent DWI restriction. Unfortunately, the last MRI control showed a secondary cerebellar localisation of the disease. New therapies are changing MRI pattern in HGG imaging and this case underlines how a multimodality approach is increasingly necessary. In particular, when using anti-VEGF drugs, SWI can have a crucial role in identifying therapy-related haemorrhagic changes.
Collapse
Affiliation(s)
- Mariam Mansour
- Neuroradiology Unit, San Bortolo Hospital, AULSS 8 Berica, Vicenza, Italy
| | - Valerio Vitale
- Neuroradiology Unit, San Bortolo Hospital, AULSS 8 Berica, Vicenza, Italy
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Giulio Riva
- Pathology Unit, San Bortolo Hospital, AULSS 8 Berica, Vicenza, Italy
| | | | - Mariano Zanusso
- Neurosurgery Unit, San Bortolo Hospital, AULSS 8 Berica, Vicenza, Italy
| |
Collapse
|
19
|
Hu R, Hoch MJ. Application of Diffusion Weighted Imaging and Diffusion Tensor Imaging in the Pretreatment and Post-treatment of Brain Tumor. Radiol Clin North Am 2021; 59:335-347. [PMID: 33926681 DOI: 10.1016/j.rcl.2021.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Diffusion MR imaging exploits the diffusion properties of water to generate contrast between normal tissue and pathology. Diffusion is an essential component of nearly all brain tumor MR imaging examinations. This review covers the important clinical applications of diffusion weighted imaging in the pretreatment diagnosis and grading of brain tumors and assessment of treatment response. Diffusion imaging improves the accuracy of identifying treatment-related effects that may mimic tumor improvement or worsening. Fiber tractography models of eloquent white matter pathways are generated using diffusion tensor imaging. A practical and concise tractography guide is provided for anyone new to preoperative surgical mapping.
Collapse
Affiliation(s)
- Ranliang Hu
- Department of Radiology & Imaging Sciences, Emory University, Emory University Hospital, 1364 Clifton Road, BG 20, Atlanta, GA 30322, USA
| | - Michael J Hoch
- Department of Radiology, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, Suite 130, Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Overcast WB, Davis KM, Ho CY, Hutchins GD, Green MA, Graner BD, Veronesi MC. Advanced imaging techniques for neuro-oncologic tumor diagnosis, with an emphasis on PET-MRI imaging of malignant brain tumors. Curr Oncol Rep 2021; 23:34. [PMID: 33599882 PMCID: PMC7892735 DOI: 10.1007/s11912-021-01020-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW This review will explore the latest in advanced imaging techniques, with a focus on the complementary nature of multiparametric, multimodality imaging using magnetic resonance imaging (MRI) and positron emission tomography (PET). RECENT FINDINGS Advanced MRI techniques including perfusion-weighted imaging (PWI), MR spectroscopy (MRS), diffusion-weighted imaging (DWI), and MR chemical exchange saturation transfer (CEST) offer significant advantages over conventional MR imaging when evaluating tumor extent, predicting grade, and assessing treatment response. PET performed in addition to advanced MRI provides complementary information regarding tumor metabolic properties, particularly when performed simultaneously. 18F-fluoroethyltyrosine (FET) PET improves the specificity of tumor diagnosis and evaluation of post-treatment changes. Incorporation of radiogenomics and machine learning methods further improve advanced imaging. The complementary nature of combining advanced imaging techniques across modalities for brain tumor imaging and incorporating technologies such as radiogenomics has the potential to reshape the landscape in neuro-oncology.
Collapse
Affiliation(s)
- Wynton B. Overcast
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N University Blvd. Room 0663, Indianapolis, IN 46202 USA
| | - Korbin M. Davis
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N University Blvd. Room 0663, Indianapolis, IN 46202 USA
| | - Chang Y. Ho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Goodman Hall, 355 West 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - Gary D. Hutchins
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Research 2 Building (R2), Room E124, 920 W. Walnut Street, Indianapolis, IN 46202-5181 USA
| | - Mark A. Green
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Research 2 Building (R2), Room E124, 920 W. Walnut Street, Indianapolis, IN 46202-5181 USA
| | - Brian D. Graner
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Goodman Hall, 355 West 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - Michael C. Veronesi
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Research 2 Building (R2), Room E174, 920 W. Walnut Street, Indianapolis, IN 46202-5181 USA
| |
Collapse
|
21
|
Pollack M, Keating K, Wissinger E, Jackson L, Sarnes E, Cuffel B. Transforming approaches to treating TRK fusion cancer: historical comparison of larotrectinib and histology-specific therapies. Curr Med Res Opin 2021; 37:59-70. [PMID: 33148054 DOI: 10.1080/03007995.2020.1847057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The results from basket trials utilized to gain regulatory approval of tumor-agnostic therapies can be difficult to interpret without the context of a comparator arm. We describe the role and efficacy of histology-based treatments to provide a historical comparison with larotrectinib. METHODS A systematic literature review (SLR) was conducted on the clinical outcomes of current histology-based standard of care treatments used in non-small cell lung cancer, colorectal cancer, thyroid cancer, gliomas, soft tissue sarcoma, salivary gland cancer, and infantile fibrosarcoma (7 of the 21 tumor histologies in the larotrectinib trials). The review focused on advanced stage/metastatic disease to make a historical comparison with larotrectinib. RESULTS Larotrectinib provides positive outcomes in both adult and pediatric patients with advanced or metastatic solid tumors known to harbor NTRK gene fusions across a wide range of tumor types. Although the numbers of patients per tumor type are limited, the results of this historical comparison demonstrated that larotrectinib is an efficacious treatment option when naïvely indirectly compared with historical treatments across all 7 reviewed tumor types, especially in comparison to later lines of therapy. CONCLUSIONS Utilizing larotrectinib as a case example across these types of historical comparisons shows that larotrectinib provides positive efficacy outcomes in TRK fusion cancer across tumor histologies known to harbor NTRK gene fusions that may be preferable to historical treatments.
Collapse
Affiliation(s)
| | - Karen Keating
- Bayer Healthcare Pharmaceuticals Inc, Whippany, NJ, USA
| | | | - Louis Jackson
- Bayer Healthcare Pharmaceuticals Inc, Whippany, NJ, USA
| | | | - Brian Cuffel
- Bayer Healthcare Pharmaceuticals Inc, Whippany, NJ, USA
| |
Collapse
|
22
|
Gonçalves FG, Chawla S, Mohan S. Emerging MRI Techniques to Redefine Treatment Response in Patients With Glioblastoma. J Magn Reson Imaging 2020; 52:978-997. [PMID: 32190946 DOI: 10.1002/jmri.27105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most common and most malignant primary brain tumor. Despite aggressive multimodal treatment, its prognosis remains poor. Even with continuous developments in MRI, which has provided us with newer insights into the diagnosis and understanding of tumor biology, response assessment in the posttherapy setting remains challenging. We believe that the integration of additional information from advanced neuroimaging techniques can further improve the diagnostic accuracy of conventional MRI. In this article, we review the utility of advanced neuroimaging techniques such as diffusion-weighted imaging, diffusion tensor imaging, perfusion-weighted imaging, proton magnetic resonance spectroscopy, and chemical exchange saturation transfer in characterizing and evaluating treatment response in patients with glioblastoma. We will also discuss the existing challenges and limitations of using these techniques in clinical settings and possible solutions to avoiding pitfalls in study design, data acquisition, and analysis for future studies. LEVEL OF EVIDENCE: 2 TECHNICAL EFFICACY STAGE: 3 J. Magn. Reson. Imaging 2020;52:978-997.
Collapse
Affiliation(s)
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
23
|
Leao DJ, Craig PG, Godoy LF, Leite CC, Policeni B. Response Assessment in Neuro-Oncology Criteria for Gliomas: Practical Approach Using Conventional and Advanced Techniques. AJNR Am J Neuroradiol 2020; 41:10-20. [PMID: 31857322 PMCID: PMC6975322 DOI: 10.3174/ajnr.a6358] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/29/2019] [Indexed: 01/08/2023]
Abstract
The Response Assessment in Neuro-Oncology criteria were developed as an objective tool for radiologic assessment of treatment response in high-grade gliomas. Imaging plays a critical role in the management of the patient with glioma, from initial diagnosis to posttreatment follow-up, which can be particularly challenging for radiologists. Interpreting findings after surgery, radiation, and chemotherapy requires profound knowledge about the tumor biology, as well as the peculiar changes expected to ensue as a consequence of each treatment technique. In this article, we discuss the imaging findings associated with tumor progression, tumor response, pseudoprogression, and pseudoresponse according to the Response Assessment in Neuro-Oncology criteria for high-grade and lower-grade gliomas. We describe relevant practical issues when evaluating patients with glioma, such as the need for imaging in the first 48 hours, the radiation therapy planning and isodose curves, the significance of T2/FLAIR hyperintense lesions, the impact of the timing for the evaluation after radiation therapy, and the definition of progressive disease on the histologic specimen. We also illustrate the correlation among the findings on conventional MR imaging with advanced techniques, such as perfusion, diffusion-weighted imaging, spectroscopy, and amino acid PET. Because many of the new lesions represent a mixture of tumor cells and tissue with radiation injury, the radiologist aims to identify the predominant component of the lesion and categorize the findings according to Response Assessment in Neuro-Oncology criteria so that the patient can receive the best treatment.
Collapse
Affiliation(s)
- D J Leao
- From the Cancer Hospital of Federal University of Uberlandia (D.J.L.), Uberlandia, Brazil
| | - P G Craig
- Department of Radiology, (P.G.C., B.P.), University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - L F Godoy
- Department of Diagnostic Radiology (L.F.G.), Hospital Sirio-Libanes, Sao Paulo, Brazil
- Department of Neuroradiology (L.F.G., C.C.L.), Faculdade de Medicina Instituto de Radiologia, Universidade de Sao Paulo Neuroradiology, Sao Paulo, Brazil
| | - C C Leite
- Department of Neuroradiology (L.F.G., C.C.L.), Faculdade de Medicina Instituto de Radiologia, Universidade de Sao Paulo Neuroradiology, Sao Paulo, Brazil
| | - B Policeni
- Department of Radiology, (P.G.C., B.P.), University of Iowa Hospitals and Clinics, Iowa City, Iowa
| |
Collapse
|
24
|
Zeiner PS, Kinzig M, Divé I, Maurer GD, Filipski K, Harter PN, Senft C, Bähr O, Hattingen E, Steinbach JP, Sörgel F, Voss M, Steidl E, Ronellenfitsch MW. Regorafenib CSF Penetration, Efficacy, and MRI Patterns in Recurrent Malignant Glioma Patients. J Clin Med 2019; 8:jcm8122031. [PMID: 31766326 PMCID: PMC6947028 DOI: 10.3390/jcm8122031] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/28/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022] Open
Abstract
(1) Background: The phase 2 Regorafenib in Relapsed Glioblastoma (REGOMA) trial indicated a survival benefit for patients with first recurrence of a glioblastoma when treated with the multikinase inhibitor regorafenib (REG) instead of lomustine. The aim of this retrospective study was to investigate REG penetration to cerebrospinal fluid (CSF), treatment efficacy, and effects on magnetic resonance imaging (MRI) in patients with recurrent high-grade gliomas. (2) Methods: Patients were characterized by histology, adverse events, steroid treatment, overall survival (OS), and MRI growth pattern. REG and its two active metabolites were quantified by liquid chromatography/tandem mass spectrometry in patients’ serum and CSF. (3) Results: 21 patients mainly with IDH-wildtype glioblastomas who had been treated with REG were retrospectively identified. Thirteen CFS samples collected from 3 patients of the cohort were available for pharmacokinetic testing. CSF levels of REG and its metabolites were significantly lower than in serum. Follow-up MRI was available in 19 patients and showed progressive disease (PD) in all but 2 patients. Two distinct MRI patterns were identified: 7 patients showed classic PD with progression of contrast enhancing lesions, whereas 11 patients showed a T2-dominant MRI pattern characterized by a marked reduction of contrast enhancement. Median OS was significantly better in patients with a T2-dominant growth pattern (10 vs. 27 weeks respectively, p = 0.003). Diffusion restrictions were observed in 13 patients. (4) Conclusion: REG and its metabolites were detectable in CSF. A distinct MRI pattern that might be associated with an improved OS was observed in half of the patient cohort. Treatment response in the total cohort was poor.
Collapse
Affiliation(s)
- Pia S. Zeiner
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Martina Kinzig
- IBMP—Institute for Biomedical and Pharmaceutical Research, 90562 Nürnberg-Heroldsberg, Germany; (M.K.); (F.S.)
| | - Iris Divé
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Gabriele D. Maurer
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
| | - Katharina Filipski
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Institute of Neurology (Edinger-Institute), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Patrick N. Harter
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
- Institute of Neurology (Edinger-Institute), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Christian Senft
- Department of Neurosurgery, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany;
| | - Oliver Bähr
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- Department of Neurology, Klinikum Aschaffenburg-Alzenau, 63739 Aschaffenburg, Germany
| | - Elke Hattingen
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Department of Neuroradiology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Joachim P. Steinbach
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Fritz Sörgel
- IBMP—Institute for Biomedical and Pharmaceutical Research, 90562 Nürnberg-Heroldsberg, Germany; (M.K.); (F.S.)
- Institute of Pharmacology, University Duisburg-Essen, 45141 Essen, Germany
| | - Martin Voss
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Eike Steidl
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Department of Neuroradiology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Michael W. Ronellenfitsch
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
- Correspondence: ; Tel.: +49-69-6301-87711; Fax: +49-69-6301-87713
| |
Collapse
|
25
|
Lee CY, Kalra A, Spampinato MV, Tabesh A, Jensen JH, Helpern JA, de Fatima Falangola M, Van Horn MH, Giglio P. Early assessment of recurrent glioblastoma response to bevacizumab treatment by diffusional kurtosis imaging: a preliminary report. Neuroradiol J 2019; 32:317-327. [PMID: 31282311 DOI: 10.1177/1971400919861409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
PURPOSE The purpose of this preliminary study is to apply diffusional kurtosis imaging to assess the early response of recurrent glioblastoma to bevacizumab treatment. METHODS This prospective cohort study included 10 patients who had been diagnosed with recurrent glioblastoma and scheduled to receive bevacizumab treatment. Diffusional kurtosis images were obtained from all the patients 0-7 days before (pre-bevacizumab) and 28 days after (post-bevacizumab) initiating bevacizumab treatment. The mean, 10th, and 90th percentile values were derived from the histogram of diffusional kurtosis imaging metrics in enhancing and non-enhancing lesions, selected on post-contrast T1-weighted and fluid-attenuated inversion recovery images. Correlations of imaging measures with progression-free survival and overall survival were evaluated using Spearman's rank correlation coefficient. The significance level was set at P < 0.05. RESULTS Higher pre-bevacizumab non-enhancing lesion volume was correlated with poor overall survival (r = -0.65, P = 0.049). Higher post-bevacizumab mean diffusivity and axial diffusivity (D∥, D∥10% and D∥90%) in non-enhancing lesions were correlated with poor progression-free survival (r = -0.73, -0.83, -0.71 and -0.85; P < 0.05). Lower post-bevacizumab axial kurtosis (K∥10%) in non-enhancing lesions was correlated with poor progression-free survival (r = 0.81, P = 0.008). CONCLUSIONS This preliminary study demonstrates that diffusional kurtosis imaging metrics allow the detection of tissue changes 28 days after initiating bevacizumab treatment and that they may provide information about tumor progression.
Collapse
Affiliation(s)
- Chu-Yu Lee
- 1 Department of Radiology and Radiological Science, Medical University of South Carolina, USA.,2 Center for Biomedical Imaging, Medical University of South Carolina, USA
| | - Amandeep Kalra
- 3 Department of Neuroscience, Medical University of South Carolina, USA.,4 Sarah Cannon Cancer Institute, USA
| | - Maria V Spampinato
- 1 Department of Radiology and Radiological Science, Medical University of South Carolina, USA.,2 Center for Biomedical Imaging, Medical University of South Carolina, USA
| | - Ali Tabesh
- 1 Department of Radiology and Radiological Science, Medical University of South Carolina, USA.,2 Center for Biomedical Imaging, Medical University of South Carolina, USA
| | - Jens H Jensen
- 1 Department of Radiology and Radiological Science, Medical University of South Carolina, USA.,2 Center for Biomedical Imaging, Medical University of South Carolina, USA.,3 Department of Neuroscience, Medical University of South Carolina, USA
| | - Joseph A Helpern
- 1 Department of Radiology and Radiological Science, Medical University of South Carolina, USA.,2 Center for Biomedical Imaging, Medical University of South Carolina, USA.,3 Department of Neuroscience, Medical University of South Carolina, USA.,5 Department of Neurology, Medical University of South Carolina, USA
| | - Maria de Fatima Falangola
- 1 Department of Radiology and Radiological Science, Medical University of South Carolina, USA.,2 Center for Biomedical Imaging, Medical University of South Carolina, USA.,3 Department of Neuroscience, Medical University of South Carolina, USA
| | - Mark H Van Horn
- 1 Department of Radiology and Radiological Science, Medical University of South Carolina, USA.,2 Center for Biomedical Imaging, Medical University of South Carolina, USA
| | - Pierre Giglio
- 3 Department of Neuroscience, Medical University of South Carolina, USA.,6 Department of Neurology, The Ohio State University Wexner Medical Center, USA
| |
Collapse
|
26
|
Nilsson M, Englund E, Szczepankiewicz F, van Westen D, Sundgren PC. Imaging brain tumour microstructure. Neuroimage 2018; 182:232-250. [DOI: 10.1016/j.neuroimage.2018.04.075] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 04/27/2018] [Accepted: 04/30/2018] [Indexed: 01/18/2023] Open
|
27
|
Miyoshi F, Shinohara Y, Kambe A, Kuya K, Murakami A, Kurosaki M, Ogawa T. Utility of intravoxel incoherent motion magnetic resonance imaging and arterial spin labeling for recurrent glioma after bevacizumab treatment. Acta Radiol 2018; 59:1372-1379. [PMID: 29471670 DOI: 10.1177/0284185118759707] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background Detecting recurrence of glioma on magnetic resonance imaging (MRI) is getting more and more important, especially after administration of new anti-tumor agent. However, it is still hard to identify. Purpose To examine the utility of intravoxel incoherent motion (IVIM) MRI and arterial spin labeling-cerebral blood flow (ASL-CBF) for recurrent glioma after initiation of bevacizumab (BEV) treatment. Material and Methods Thirteen patients (7 men, 6 women; age range = 41-82 years) with glioma (high grade, n = 11; low grade, n = 2) were enrolled in the study. IVIM parameters including apparent diffusion coefficient (ADC), true diffusion coefficient (D), and perfusion fraction (f) were obtained with 14 different b-values. We identified tumor progression during BEV therapy by MRI monitoring consisting of diffusion-weighted imaging (DWI), fluid-attenuated inversion recovery (FLAIR) imaging, and contrast-enhanced T1-weighted (CE-T1W) imaging by measuring tumor area. We also measured each parameter of IVIM and ASL-CBF, and calculated relative ADC (rADC), relative D (rD), relative f (rf), and relative CBF (rCBF) by obtaining the ratio between each area and the contralateral cerebral white matter. We calculated the rate of change (Δ) by subtracting values from those from the preceding MRI study, and obtained Spearman's rank correlation coefficient (rs). Results Tumor progression was identified in nine patients (high grade, n = 7; low grade, n = 2). Negative correlations were identified between ΔrD and ΔDWI area (rs = -0.583), and between ΔrD and ΔCE-T1W imaging area (rs = -0.605). Conclusion Tumor progression after BEV treatment can be identified by decreasing rD.
Collapse
Affiliation(s)
- Fuminori Miyoshi
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Yuki Shinohara
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Atsushi Kambe
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Keita Kuya
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Atsushi Murakami
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Masamichi Kurosaki
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Toshihide Ogawa
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
28
|
Pope WB, Brandal G. Conventional and advanced magnetic resonance imaging in patients with high-grade glioma. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2018; 62:239-253. [PMID: 29696946 DOI: 10.23736/s1824-4785.18.03086-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Magnetic resonance imaging is integral to the care of patients with high-grade gliomas. Anatomic detail can be acquired with conventional structural imaging, but newer approaches also add capabilities to interrogate image-derived physiologic and molecular characteristics of central nervous system neoplasms. These advanced imaging techniques are increasingly employed to generate biomarkers that better reflect tumor burden and therapy response. The following is an overview of current strategies based on advanced magnetic resonance imaging that are used in the assessment of high-grade glioma patients with an emphasis on how novel imaging biomarkers can potentially advance patient care.
Collapse
Affiliation(s)
- Whitney B Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA, USA -
| | - Garth Brandal
- Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
29
|
Goyal P, Tenenbaum M, Gupta S, Kochar PS, Bhatt AA, Mangla M, Kumar Y, Mangla R. Survival prediction based on qualitative MRI diffusion signature in patients with recurrent high grade glioma treated with bevacizumab. Quant Imaging Med Surg 2018; 8:268-279. [PMID: 29774180 DOI: 10.21037/qims.2018.04.05] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Bevacizumab was approved by the FDA for the treatment of recurrent or progressive glioblastoma (GBM). Imaging responses are typically assessed by gadolinium-enhanced MRI. We sought to determine the significance of qualitative diffusion signature (manifest as variable degree of dark signal) on ADC maps in recurrent gliomas after treatment with bevacizumab. Methods We performed an institutional review board (IRB) approved retrospective study on patients who underwent MRI of the brain after 8 weeks of receiving bevacizumab for recurrent glioma. Patients were divided into three groups based on qualitative diffusion signature: (I) lesion not bright on diffusion weighted imaging (DWI) suggestive of no restricted diffusion (FDR0); (II) lesion bright on DWI with corresponding homogenous dark signal on apparent diffusion coefficient (ADC) maps suggestive of focal restricted diffusion likely due to bevacizumab induced necrosis (FDRn); and (III) lesion bright on DWI with corresponding homogenous faint dark signal on ADC maps suggestive of focal restricted diffusion likely due to viable tumor or heterogeneous spectrum of dark and faint dark signals on ADC maps suggestive of focal restricted diffusion likely due to viable tumor surrounding the bevacizumab induced necrosis (FDRt). Results Based on the qualitative signal on diffusion weighted sequences after bevacizumab therapy, total number of patients in group (I) were 14 (36%), in group (II) were 17 (44%); and in group (III) were 8 (20%). The median overall survival (OS) from the time of recurrence in patients belonging to group (II) was 364 days vs. 183 days for those with group (I) vs. 298 days for group (III). On simultaneous comparison of survival differences in all three groups by Kaplan-Meier analysis, group (II) was significant in predicting survival with P values for the log-rank tests <0.033. Conclusions In patients with recurrent glioma treated with bevacizumab, the presence of homogenous dark signal (FDRn) on ADC maps at 8 weeks follow-up MRI correlated with a longer survival. Thus, use of this qualitative diffusion signature in adjunct to contrast enhanced MRI may have the widest potential impact on routine clinical care for patients with recurrent high-grade gliomas. However, prospective studies analysing its predictive value are warranted.
Collapse
Affiliation(s)
- Pradeep Goyal
- Department of Radiology, St. Vincent's Medical Center, Bridgeport, CT, USA
| | - Mary Tenenbaum
- Department of Radiology, UMMS-Baystate Regional Campus, Springfield, MA, USA.,Department of Radiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Sonali Gupta
- Department of Medicine, St. Vincent's Medical Center, Bridgeport, CT, USA
| | - Puneet S Kochar
- Department of Radiology, Yale New Haven Health Bridgeport Hospital, Bridgeport, CT, USA
| | - Alok A Bhatt
- Department of Radiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Manisha Mangla
- Department of Public Health, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yogesh Kumar
- Department of Radiology, Columbia University at Bassett Healthcare, Cooperstown, NY, USA
| | - Rajiv Mangla
- Department of Radiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Radiology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
30
|
Gahrmann R, van den Bent M, van der Holt B, Vernhout RM, Taal W, Vos M, de Groot JC, Beerepoot LV, Buter J, Flach ZH, Hanse M, Jasperse B, Smits M. Comparison of 2D (RANO) and volumetric methods for assessment of recurrent glioblastoma treated with bevacizumab-a report from the BELOB trial. Neuro Oncol 2018; 19:853-861. [PMID: 28204639 DOI: 10.1093/neuonc/now311] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background The current method for assessing progressive disease (PD) in glioblastoma is according to the Response Assessment in Neuro-Oncology (RANO) criteria. Bevacizumab-treated patients may show pseudo-response on postcontrast T1-weighted (T1w) MRI, and a more infiltrative non-enhancing growth pattern on T2w/fluid attenuated inversion recovery (FLAIR) images. We investigated whether the RANO criteria remain the method of choice for assessing bevacizumab-treated recurrent glioblastoma when compared with various volumetric methods. Methods Patients with assessable MRI data from the BELOB trial (n = 148) were included. Patients were treated with bevacizumab, lomustine, or both. At first and second radiological follow-up (6 and 12 wk), PD was determined using the 2D RANO criteria and various volumetric methods based on enhancing tumor only and enhancing plus non-enhancing tumor. Differences in overall survival (OS) between PD and non-PD patients were assessed with the log-rank test and a Cox model. Hazard ratios (HRs) and their 95% CIs were determined. Results For all patients together, all methods (except subtraction of non-enhancing from enhancing volume at first follow-up) showed significant differences in OS between PD and non-PD patients (P < .001). The largest risk increase for death in case of PD at both first and second follow-up was found with the RANO criteria: HR = 2.81 (95% CI, 1.92-4.10) and HR = 2.80 (95% CI, 1.75-4.49), respectively. In the bevacizumab-treated patients, all methods assessed showed significant differences in OS between PD and non-PD patients. There were no significant differences between methods. Conclusions In the first 12 weeks, volumetric methods did not provide significant improvement over the RANO criteria as a posttreatment prognostic marker.
Collapse
Affiliation(s)
- Renske Gahrmann
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Martin van den Bent
- Brain Tumor Center at Erasmus MC Cancer Institute Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bronno van der Holt
- Clinical Trial Center, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - René Michel Vernhout
- Clinical Trial Center, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Walter Taal
- Brain Tumor Center at Erasmus MC Cancer Institute Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maaike Vos
- Department of Neurology, Medical Center Haaglanden, The Hague, The Netherlands
| | - Jan Cees de Groot
- Department of Radiology, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Jan Buter
- Department of Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Monique Hanse
- Department of Neurology, Catharina Hospital, Eindhoven, The Netherlands
| | - Bas Jasperse
- Department of Radiology, Antoni van Leeuwenhoek ziekenhuis, Amsterdam, The Netherlands
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
31
|
Thomas A, Rosenblum M, Karimi S, DeAngelis LM, Omuro A, Kaley TJ. Radiographic patterns of recurrence and pathologic correlation in malignant gliomas treated with bevacizumab. CNS Oncol 2018; 7:7-13. [PMID: 29388793 PMCID: PMC6001559 DOI: 10.2217/cns-2017-0025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Interpretation of MRI abnormalities in patients with malignant gliomas (MG) treated with bevacizumab is challenging. Recent reports describe quantitative analyses of diffusion-weighted imaging abnormalities not available in standard clinical settings, to differentiate tumor recurrence from treatment necrosis. We retrospectively reviewed bevacizumab treated MG patients who underwent surgery or autopsy to correlate radiographic recurrence patterns with pathologic findings. 32 patients with MG (26 glioblastoma, three anaplastic astrocytoma and three anaplastic oligodendroglioma) were identified. Recurrence patterns: local enhancing (n = 23), distant enhancing (n = 1), nonenhancing (n = 7) and leptomeningeal (n = 1). Histology: tumor (n = 25), mixed tumor/necrosis (n = 5) and all necrosis (n = 2). On diffusion-weighted imaging, 5/32 had restricted diffusion (three mixed and two necrosis). Irrespective of radiographic recurrence pattern, tumor was found in 94% of cases. Restricted diffusion correlated with necrosis.
Collapse
Affiliation(s)
- Alissa Thomas
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.,Department of Neurology, University of Vermont, 11 Colchester Avenue, Burlington, VT 05401, USA
| | - Marc Rosenblum
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Sasan Karimi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Lisa M DeAngelis
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Antonio Omuro
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Thomas J Kaley
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
32
|
Iv M, Yoon BC, Heit JJ, Fischbein N, Wintermark M. Current Clinical State of Advanced Magnetic Resonance Imaging for Brain Tumor Diagnosis and Follow Up. Semin Roentgenol 2018; 53:45-61. [DOI: 10.1053/j.ro.2017.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
33
|
Abstract
Magnetic resonance imaging (MRI) is the cornerstone for evaluating patients with brain masses such as primary and metastatic tumors. Important challenges in effectively detecting and diagnosing brain metastases and in accurately characterizing their subsequent response to treatment remain. These difficulties include discriminating metastases from potential mimics such as primary brain tumors and infection, detecting small metastases, and differentiating treatment response from tumor recurrence and progression. Optimal patient management could be benefited by improved and well-validated prognostic and predictive imaging markers, as well as early response markers to identify successful treatment prior to changes in tumor size. To address these fundamental needs, newer MRI techniques including diffusion and perfusion imaging, MR spectroscopy, and positron emission tomography (PET) tracers beyond traditionally used 18-fluorodeoxyglucose are the subject of extensive ongoing investigations, with several promising avenues of added value already identified. These newer techniques provide a wealth of physiologic and metabolic information that may supplement standard MR evaluation, by providing the ability to monitor and characterize cellularity, angiogenesis, perfusion, pH, hypoxia, metabolite concentrations, and other critical features of malignancy. This chapter reviews standard and advanced imaging of brain metastases provided by computed tomography, MRI, and amino acid PET, focusing on potential biomarkers that can serve as problem-solving tools in the clinical management of patients with brain metastases.
Collapse
Affiliation(s)
- Whitney B Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States.
| |
Collapse
|
34
|
Hassanzadeh C, Rao YJ, Chundury A, Rowe J, Ponisio MR, Sharma A, Miller-Thomas M, Tsien CI, Ippolito JE. Multiparametric MRI and [ 18F]Fluorodeoxyglucose Positron Emission Tomography Imaging Is a Potential Prognostic Imaging Biomarker in Recurrent Glioblastoma. Front Oncol 2017; 7:178. [PMID: 28868256 PMCID: PMC5563320 DOI: 10.3389/fonc.2017.00178] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/03/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose/objectives Multiparametric advanced MR and [18F]fluorodeoxyglucose (FDG)-positron emission tomography (PET) imaging may be important biomarkers for prognosis as well for distinguishing recurrent glioblastoma multiforme (GBM) from treatment-related changes. Methods/materials We retrospectively evaluated 30 patients treated with chemoradiation for GBM and underwent advanced MR and FDG-PET for confirmation of tumor progression. Multiparametric MRI and FDG-PET imaging metrics were evaluated for their association with 6-month overall (OS) and progression-free survival (PFS) based on pathological, radiographic, and clinical criteria. Results 17 males and 13 females were treated between 2001 and 2014, and later underwent FDG-PET at suspected recurrence. Baseline FDG-PET and MRI imaging was obtained at a median of 7.5 months [interquartile range (IQR) 3.7–12.4] following completion of chemoradiation. Median follow-up after FDG-PET imaging was 10 months (IQR 7.2–13.0). Receiver-operator characteristic curve analysis identified that lesions characterized by a ratio of the SUVmax to the normal contralateral brain (SUVmax/NB index) >1.5 and mean apparent diffusion coefficient (ADC) value of ≤1,400 × 10−6 mm2/s correlated with worse 6-month OS and PFS. We defined three patient groups that predicted the probability of tumor progression: SUVmax/NB index >1.5 and ADC ≤1,400 × 10−6 mm2/s defined high-risk patients (n = 7), SUVmax/NB index ≤1.5 and ADC >1,400 × 10−6 mm2/s defined low-risk patients (n = 11), and intermediate-risk (n = 12) defined the remainder of the patients. Median OS following the time of the FDG-PET scan for the low, intermediate, and high-risk groups were 23.5, 10.5, and 3.8 months (p < 0.01). Median PFS were 10.0, 4.4, and 1.9 months (p = 0.03). Rates of progression at 6-months in the low, intermediate, and high-risk groups were 36, 67, and 86% (p = 0.04). Conclusion Recurrent GBM in the molecular era is associated with highly variable outcomes. Multiparametric MR and FDG-PET biomarkers may provide a clinically relevant, non-invasive and cost-effective method of predicting prognosis and improving clinical decision making in the treatment of patients with suspected tumor recurrence.
Collapse
Affiliation(s)
- Comron Hassanzadeh
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, United States.,Department of Genetics, Washington University in St. Louis, St. Louis, MO, United States
| | - Yuan James Rao
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, United States
| | - Anupama Chundury
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, United States
| | - Jackson Rowe
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, United States
| | - Maria Rosana Ponisio
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, United States
| | - Akash Sharma
- Department of Radiology, Mayo Clinic Florida, Jacksonville, FL, United States
| | - Michelle Miller-Thomas
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, United States
| | - Christina I Tsien
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, United States
| | - Joseph E Ippolito
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, United States.,Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
35
|
Bevacizumab for malignant gliomas: current indications, mechanisms of action and resistance, and markers of response. Brain Tumor Pathol 2017; 34:62-77. [PMID: 28386777 DOI: 10.1007/s10014-017-0284-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/27/2017] [Indexed: 12/21/2022]
Abstract
Vascular endothelial growth factor (VEGF) is an attractive target of antiangiogenic therapy in glioblastomas. Bevacizumab (Bev), a humanized anti-VEGF antibody, is associated with the improvement of progression-free survival and performance status in patients with glioblastoma. However, randomized trials uniformly suggest that these favorable clinical effects of Bev do not translate into an overall survival benefit. The mechanisms of action of Bev appear to include the inhibition of tumor angiogenesis, as well as indirect effects such as the depletion of niches for glioma stem cells and stimulation of antitumor immunity. Although several molecules/pathways have been reported to mediate adaptation and resistance to Bev, including the activation of alternative pro-angiogenic pathways, the resistance mechanisms have not been fully elucidated; for example, the mechanism that reinduces tumor hypoxia remains unclarified. The identification of imaging characteristics or biomarkers predicting the response to Bev, as well as the better understanding of the mechanisms of action and resistance, is crucial to improve the overall clinical outcome and optimize individual therapy. In this article, the authors review the results of important clinical trials/studies, the current understanding of the mechanisms of action and resistance, and the knowledge of imaging characteristics and biomarkers predicting the response to Bev.
Collapse
|
36
|
Abstract
The imaging of treated gliomas is complicated by a variety of treatment related effects, which can falsely simulate disease improvement or progression. Distinguishing between disease progression and treatment effects is difficult with standard MR imaging pulse sequences and added specificity can be gained by the addition of advanced imaging techniques.
Collapse
Affiliation(s)
- Mark F Dalesandro
- Department of Radiology, Harborview Medical Center, University of Washington, Box 357115, 1959 Northeast Pacific Street, NW011, Seattle, WA 98195-7115, USA
| | - Jalal B Andre
- Department of Radiology, Harborview Medical Center, University of Washington, Box 357115, 1959 Northeast Pacific Street, NW011, Seattle, WA 98195-7115, USA.
| |
Collapse
|
37
|
Apparent diffusion coefficient changes predict survival after intra-arterial bevacizumab treatment in recurrent glioblastoma. Neuroradiology 2017; 59:499-505. [PMID: 28343250 DOI: 10.1007/s00234-017-1820-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 03/14/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Superselective intra-arterial cerebral infusion (SIACI) of bevacizumab (BV) has emerged as a novel therapy in the treatment of recurrent glioblastoma (GB). This study assessed the use of apparent diffusion coefficient (ADC) in predicting length of survival after SIACI BV and overall survival in patients with recurrent GB. METHODS Sixty-five patients from a cohort enrolled in a phase I/II trial of SIACI BV for treatment of recurrent GB were retrospectively included in this analysis. MR imaging with a diffusion-weighted (DWI) sequence was performed before and after treatment. ROIs were manually delineated on ADC maps corresponding to the enhancing and non-enhancing portions of the tumor. Cox and logistic regression analyses were performed to determine which ADC values best predicted survival. RESULTS The change in minimum ADC in the enhancing portion of the tumor after SIACI BV therapy was associated with an increased risk of death (hazard ratio = 2.0, 95% confidence interval(CI) [1.04-3.79], p = 0.038), adjusting for age, tumor size, BV dose, and prior IV BV treatments. Similarly, the change in ADC after SIACI BV therapy was associated with greater likelihood of surviving less than 1 year after therapy (odds ratio = 7.0, 95% CI [1.08-45.7], p = 0.04). Having previously received IV BV was associated with increased risk of death (OR 18, 95% CI [1.8-180.0], p = 0.014). CONCLUSION In patients with recurrent GB treated with SIACI BV, the change in ADC value after treatment is predictive of overall survival.
Collapse
|
38
|
Jena A, Taneja S, Jha A, Damesha NK, Negi P, Jadhav GK, Verma SM, Sogani SK. Multiparametric Evaluation in Differentiating Glioma Recurrence from Treatment-Induced Necrosis Using Simultaneous 18F-FDG-PET/MRI: A Single-Institution Retrospective Study. AJNR Am J Neuroradiol 2017; 38:899-907. [PMID: 28341716 DOI: 10.3174/ajnr.a5124] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/21/2016] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Differentiating glioma recurrence from treatment-induced necrosis can be a challenge on conventional imaging. This study aimed to assess the diagnostic performance of each functional MR imaging and PET parameter derived by using simultaneous FDG-PET/MR imaging individually and in combination in the evaluation of suspected glioma recurrence. MATERIALS AND METHODS Thirty-five treated glioma patients with 41 enhancing lesions (World Health Organization grade II = 9, III = 13, IV = 19) on MR imaging after an operation followed by radiation therapy and/or chemotherapy formed part of this study. Using PET/MR imaging, we calculated the normalized mean relative CBV, mean ADC, Cho/Cr, and maximum and mean target-to-background ratios. Statistical analysis was performed to determine the diagnostic performance of each parameter by receiver operating characteristic analysis individually and in combination with multivariate receiver operating characteristic analysis for the detection of glioma recurrence. Histopathology or clinicoradiologic follow-up was considered the criterion standard. RESULTS Of 35 patients, 25 (30 lesions) were classified as having a recurrence and 10 (11 lesions) patients as having treatment-induced necrosis. Parameters like rCBVmean (mean relative CBV), ADCmean, Cho/Cr, and maximum and mean target-to-background ratios were statistically significant in the detection of recurrent lesions with an accuracy of 77.5%, 78.0%, 90.9%, 87.8%, and 87.8%, respectively. On multivariate receiver operating characteristic analysis, the combination of all 3 MR imaging parameters resulted in an area under the curve of 0.913 ± 0.053. Furthermore, an area under the curve of 0.935 ± 0.046 was obtained when MR imaging parameters (ADCmean and Cho/Cr) were combined with the PET parameter (mean target-to-background ratio), demonstrating an increase in diagnostic accuracy. CONCLUSIONS Simultaneous PET/MR imaging with FDG offers correlative and synergistic multiparametric assessment of glioma recurrence with increased accuracy and clinical utility.
Collapse
Affiliation(s)
- A Jena
- From the PET SUITE (A. Jena, S.T., A. Jha, P.N.)
| | - S Taneja
- From the PET SUITE (A. Jena, S.T., A. Jha, P.N.)
| | - A Jha
- From the PET SUITE (A. Jena, S.T., A. Jha, P.N.)
| | - N K Damesha
- Neurosurgery (N.K.D., S.K.S.), Indraprastha Apollo Hospitals, Sarita Vihar, New Delhi, India
| | - P Negi
- From the PET SUITE (A. Jena, S.T., A. Jha, P.N.)
| | - G K Jadhav
- Departments of Molecular Imaging and Nuclear Medicine, Radiation Oncology (G.K.J., S.M.V.)
| | - S M Verma
- Departments of Molecular Imaging and Nuclear Medicine, Radiation Oncology (G.K.J., S.M.V.)
| | - S K Sogani
- Neurosurgery (N.K.D., S.K.S.), Indraprastha Apollo Hospitals, Sarita Vihar, New Delhi, India
| |
Collapse
|
39
|
Lequin M, Hendrikse J. Advanced MR Imaging in Pediatric Brain Tumors, Clinical Applications. Neuroimaging Clin N Am 2017; 27:167-190. [DOI: 10.1016/j.nic.2016.08.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
40
|
Advanced MRI assessment to predict benefit of anti-programmed cell death 1 protein immunotherapy response in patients with recurrent glioblastoma. Neuroradiology 2017; 59:135-145. [PMID: 28070598 DOI: 10.1007/s00234-016-1769-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/18/2016] [Indexed: 12/22/2022]
Abstract
INTRODUCTION We describe the imaging findings encountered in GBM patients receiving immune checkpoint blockade and assess the potential of quantitative MRI biomarkers to differentiate patients who derive therapeutic benefit from those who do not. METHODS A retrospective analysis was performed on longitudinal MRIs obtained on recurrent GBM patients enrolled on clinical trials. Among 10 patients with analyzable data, bidirectional diameters were measured on contrast enhanced T1 (pGd-T1WI) and volumes of interest (VOI) representing measurable abnormality suggestive of tumor were selected on pGdT1WI (pGdT1 VOI), FLAIR-T2WI (FLAIR VOI), and ADC maps. Intermediate ADC (IADC) VOI represented voxels within the FLAIR VOI having ADC in the range of highly cellular tumor (0.7-1.1 × 10-3 mm2/s) (IADC VOI). Therapeutic benefit was determined by tissue pathology and survival on trial. IADC VOI, pGdT1 VOI, FLAIR VOI, and RANO assessment results were correlated with patient benefit. RESULTS Five patients were deemed to have received therapeutic benefit and the other five patients did not. The average time on trial for the benefit group was 194 days, as compared to 81 days for the no benefit group. IADC VOI correlated well with the presence or absence of clinical benefit in 10 patients. Furthermore, pGd VOI, FLAIR VOI, and RANO assessment correlated less well with response. CONCLUSION MRI reveals an initial increase in volumes of abnormal tissue with contrast enhancement, edema, and intermediate ADC suggesting hypercellularity within the first 0-6 months of immunotherapy. Subsequent stabilization and improvement in IADC VOI appear to better predict ultimate therapeutic benefit from these agents than conventional imaging.
Collapse
|
41
|
Nguyen HS, Milbach N, Hurrell SL, Cochran E, Connelly J, Bovi JA, Schultz CJ, Mueller WM, Rand SD, Schmainda KM, LaViolette PS. Progressing Bevacizumab-Induced Diffusion Restriction Is Associated with Coagulative Necrosis Surrounded by Viable Tumor and Decreased Overall Survival in Patients with Recurrent Glioblastoma. AJNR Am J Neuroradiol 2016; 37:2201-2208. [PMID: 27492073 DOI: 10.3174/ajnr.a4898] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/23/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND PURPOSE Patients with recurrent glioblastoma often exhibit regions of diffusion restriction following the initiation of bevacizumab therapy. Studies suggest that these regions represent either diffusion-restricted necrosis or hypercellular tumor. This study explored postmortem brain specimens and a population analysis of overall survival to determine the identity and implications of such lesions. MATERIALS AND METHODS Postmortem examinations were performed on 6 patients with recurrent glioblastoma on bevacizumab with progressively growing regions of diffusion restriction. ADC values were extracted from regions of both hypercellular tumor and necrosis. A receiver operating characteristic analysis was performed to define optimal ADC thresholds for differentiating tissue types. A retrospective population study was also performed comparing the overall survival of 64 patients with recurrent glioblastoma treated with bevacizumab. Patients were separated into 3 groups: no diffusion restriction, diffusion restriction that appeared and progressed within 5 months of bevacizumab initiation, and delayed or stable diffusion restriction. An additional analysis was performed assessing tumor O6-methylguanine-DNA-methyltransferase methylation. RESULTS The optimal ADC threshold for differentiation of hypercellularity and necrosis was 0.736 × 10-3mm2/s. Progressively expanding diffusion restriction was pathologically confirmed to be coagulative necrosis surrounded by viable tumor. Progressive lesions were associated with the worst overall survival, while stable lesions showed the greatest overall survival (P < .05). Of the 40% of patients with O6-methylguanine-DNA-methyltransferase methylated tumors, none developed diffusion-restricted lesions. CONCLUSIONS Progressive diffusion-restricted lesions were pathologically confirmed to be coagulative necrosis surrounded by viable tumor and associated with decreased overall survival. Stable lesions were, however, associated with increased overall survival. All lesions were associated with O6-methylguanine-DNA-methyltransferase unmethylated tumors.
Collapse
Affiliation(s)
- H S Nguyen
- From the Departments of Neurosurgery (H.S.N., W.M.M.)
| | - N Milbach
- Radiology (N.M., S.L.H., S.D.R., K.M.S., P.S.L.)
| | - S L Hurrell
- Radiology (N.M., S.L.H., S.D.R., K.M.S., P.S.L.)
| | | | | | - J A Bovi
- Radiation Oncology (J.A.B., C.J.S.)
| | | | - W M Mueller
- From the Departments of Neurosurgery (H.S.N., W.M.M.)
| | - S D Rand
- Radiology (N.M., S.L.H., S.D.R., K.M.S., P.S.L.)
| | - K M Schmainda
- Radiology (N.M., S.L.H., S.D.R., K.M.S., P.S.L.)
- Biophysics (K.M.S., P.S.L.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - P S LaViolette
- Radiology (N.M., S.L.H., S.D.R., K.M.S., P.S.L.)
- Biophysics (K.M.S., P.S.L.), Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
42
|
|
43
|
Brunsing RL, Schenker-Ahmed NM, White NS, Parsons JK, Kane C, Kuperman J, Bartsch H, Kader AK, Rakow-Penner R, Seibert TM, Margolis D, Raman SS, McDonald CR, Farid N, Kesari S, Hansel D, Shabaik A, Dale AM, Karow DS. Restriction spectrum imaging: An evolving imaging biomarker in prostate MRI. J Magn Reson Imaging 2016; 45:323-336. [PMID: 27527500 DOI: 10.1002/jmri.25419] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/25/2016] [Indexed: 12/28/2022] Open
Abstract
Restriction spectrum imaging (RSI) is a novel diffusion-weighted MRI technique that uses the mathematically distinct behavior of water diffusion in separable microscopic tissue compartments to highlight key aspects of the tissue microarchitecture with high conspicuity. RSI can be acquired in less than 5 min on modern scanners using a surface coil. Multiple field gradients and high b-values in combination with postprocessing techniques allow the simultaneous resolution of length-scale and geometric information, as well as compartmental and nuclear volume fraction filtering. RSI also uses a distortion correction technique and can thus be fused to high resolution T2-weighted images for detailed localization, which improves delineation of disease extension into critical anatomic structures. In this review, we discuss the acquisition, postprocessing, and interpretation of RSI for prostate MRI. We also summarize existing data demonstrating the applicability of RSI for prostate cancer detection, in vivo characterization, localization, and targeting. LEVEL OF EVIDENCE 5 J. Magn. Reson. Imaging 2017;45:323-336.
Collapse
Affiliation(s)
- Ryan L Brunsing
- Department of Radiology, University of California San Diego, San Diego, California, USA
| | | | - Nathan S White
- Department of Radiology, University of California San Diego, San Diego, California, USA
| | - J Kellogg Parsons
- Department of Surgery, University of California San Diego, San Diego, California, USA
| | - Christopher Kane
- Department of Surgery, University of California San Diego, San Diego, California, USA
| | - Joshua Kuperman
- Department of Radiology, University of California San Diego, San Diego, California, USA
| | - Hauke Bartsch
- Department of Radiology, University of California San Diego, San Diego, California, USA
| | - Andrew Karim Kader
- Department of Surgery, University of California San Diego, San Diego, California, USA
| | - Rebecca Rakow-Penner
- Department of Radiology, University of California San Diego, San Diego, California, USA
| | - Tyler M Seibert
- Department of Radiation Medicine, University of California San Diego, San Diego, California, USA
| | - Daniel Margolis
- Department of Radiology, University of California Los Angeles, Los Angeles, California, USA
| | - Steven S Raman
- Department of Radiology, University of California Los Angeles, Los Angeles, California, USA
| | - Carrie R McDonald
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Nikdokht Farid
- Department of Radiology, University of California San Diego, San Diego, California, USA
| | - Santosh Kesari
- Department of Translational Neuro-Oncology and Neurotherapeutics, Pacific Neuroscience Institute and John Wayne Cancer Institute at Providence Saint John's Health Center, Los Angeles, California, USA
| | - Donna Hansel
- Department of Pathology, University of California San Diego, San Diego, California, USA
| | - Ahmed Shabaik
- Department of Pathology, University of California San Diego, San Diego, California, USA
| | - Anders M Dale
- Department of Radiology, University of California San Diego, San Diego, California, USA.,Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - David S Karow
- Department of Radiology, University of California San Diego, San Diego, California, USA
| |
Collapse
|
44
|
Chiang GC, Galla N, Ferraro R, Kovanlikaya I. The Added Prognostic Value of Metabolic Tumor Size on FDG-PET at First Suspected Recurrence of Glioblastoma Multiforme. J Neuroimaging 2016; 27:243-247. [DOI: 10.1111/jon.12386] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/11/2016] [Accepted: 07/12/2016] [Indexed: 11/29/2022] Open
Affiliation(s)
- Gloria C. Chiang
- Department of Radiology, Division of Neuroradiology, Weill Cornell Medical College; NewYork-Presbyterian Hospital; New York NY
| | - Naveen Galla
- Department of Radiology, Division of Neuroradiology, Weill Cornell Medical College; NewYork-Presbyterian Hospital; New York NY
| | - Richard Ferraro
- Department of Radiology, Division of Neuroradiology, Weill Cornell Medical College; NewYork-Presbyterian Hospital; New York NY
| | - Ilhami Kovanlikaya
- Department of Radiology, Division of Neuroradiology, Weill Cornell Medical College; NewYork-Presbyterian Hospital; New York NY
| |
Collapse
|
45
|
Rees J, Bradford R, Brandner S, Fersht N, Jäger R, Wilson E. Neuro-Oncology. Neurology 2016. [DOI: 10.1002/9781118486160.ch21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Jeremy Rees
- National Hospital for Neurology & Neurosurgery
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
This review covers important topics relating to the imaging evaluation of glioblastoma multiforme after therapy. An overview of the Macdonald and Response Assessment in Neuro-Oncology criteria as well as important questions and limitations regarding their use are provided. Pseudoprogression and pseudoresponse as well as the use of advanced magnetic resonance imaging techniques such as perfusion, diffusion, and spectroscopy in the evaluation of the posttherapeutic brain are also reviewed.
Collapse
|
47
|
Abstract
Imaging is integral to the management of patients with brain tumors. Conventional structural imaging provides exquisite anatomic detail but remains limited in the evaluation of molecular characteristics of intracranial neoplasms. Quantitative and physiologic biomarkers derived from advanced imaging techniques have been increasingly utilized as problem-solving tools to identify glioma grade and assess response to therapy. This chapter provides a comprehensive overview of the imaging strategies used in the clinical assessment of patients with gliomas and describes how novel imaging biomarkers have the potential to improve patient management.
Collapse
Affiliation(s)
- Whitney B Pope
- Radiological Sciences, Ronald Reagan Medical Center, Los Angeles, CA, USA.
| | - Ibrahim Djoukhadar
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Alan Jackson
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| |
Collapse
|
48
|
Wen Q, Jalilian L, Lupo JM, Li Y, Roy R, Molinaro AM, Chang SM, Prados M, Butowski N, Clarke J, Nelson SJ. Association of Diffusion and Anatomic Imaging Parameters with Survival for Patients with Newly Diagnosed Glioblastoma Participating in Two Different Clinical Trials. Transl Oncol 2015; 8:446-55. [PMID: 26692525 PMCID: PMC4700297 DOI: 10.1016/j.tranon.2015.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/14/2015] [Accepted: 10/02/2015] [Indexed: 01/20/2023] Open
Abstract
PURPOSE: To evaluate the time course and association with survival of anatomic lesion volumes and diffusion imaging parameters for patients with newly diagnosed glioblastoma who were treated with radiation and concurrently with either temozolomide and enzastaurin (TMZ+enza cohort) or temozolomide, erlotonib, and bevaciumab (TMZ+erl+bev cohort). MATERIALS AND METHODS: Regions of interest corresponding to the contrast-enhancing and hyperintense lesions on T2-weighted images were generated. Diffusion-weighted images were processed to provide maps of apparent diffusion coefficient, fractional anisotropy, and longitudinal and radial eigenvalues. Histograms of diffusion values were generated and summary statistics calculated. Cox proportional hazards models were employed to assess the association of representative imaging parameters with survival with adjustments for age, Karnofsky performance status, and extent of resection. RESULTS: Although progression-free survival was significantly longer for the TMZ+erl+bev cohort (12.8 vs 7.3 months), there was no significant difference in overall survival between the two populations (17.0 vs 17.8 months). The median contrast-enhancing lesion volumes decreased from 6.3 to 1.9 cm3 from baseline to the postradiotherapy scan for patients in the TMZ+enza cohort and from 2.8 to 0.9cm3 for the TMZ+erl+bev cohort. Changes in the T2 lesion volumes were only significant for the latter cohort (26.5 to 11.9 cm3). The median apparent diffusion coefficient and related diffusion parameters were significantly increased for the TMZ+enza cohort (1054 to 1225 μm2/s). More of the anatomic parameters were associated with survival for the TMZ+enza cohort, whereas more diffusion parameters were associated with survival for the TMZ+erl+bev cohort. CONCLUSION: The early changes in anatomic and diffusion imaging parameters and their association with survival reflected differences in the mechanisms of action of the treatments that were being given. This suggests that integrating diffusion metrics and anatomic lesion volumes into the Response Assessment in Neuro-Oncology criteria would assist in interpreting treatment-induced changes and predicting outcome in patients with newly diagnosed glioblastoma who are receiving such combination treatments.
Collapse
Affiliation(s)
- Qiuting Wen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States; UCSF/UCB Joint Graduate Group in Bioengineering, University of California, San Francisco, San Francisco, CA United States
| | - Laleh Jalilian
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States
| | - Yan Li
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States
| | - Ritu Roy
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA United States
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA United States
| | - Susan M Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Michael Prados
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Jennifer Clarke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States; UCSF/UCB Joint Graduate Group in Bioengineering, University of California, San Francisco, San Francisco, CA United States; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA United States.
| |
Collapse
|
49
|
Zhang M, Gulotta B, Thomas A, Kaley T, Karimi S, Gavrilovic I, Woo KM, Zhang Z, Arevalo-Perez J, Holodny AI, Rosenblum M, Young RJ. Large-volume low apparent diffusion coefficient lesions predict poor survival in bevacizumab-treated glioblastoma patients. Neuro Oncol 2015; 18:735-43. [PMID: 26538618 DOI: 10.1093/neuonc/nov268] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 10/01/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Glioblastomas treated with bevacizumab may develop low-signal apparent diffusion coefficient (low-ADC) lesions, which may reflect increased tumor cellularity or atypical necrosis. The purpose of this study was to examine the relationship between low-ADC lesions and overall survival (OS). We hypothesized that growing low-ADC lesions would be associated with shorter OS. METHODS We retrospectively identified 52 patients treated with bevacizumab for the first (n = 42, 81%) or later recurrence of primary glioblastoma, who had low-ADC lesions and 2 post-bevacizumab scans ≤90 days apart. Low-ADC lesion volumes were measured, and normalized 5th percentile histogram low-ADC values were recorded. Using OS as the primary endpoint, semiparametric Cox models were fitted to ascertain univariate and multivariate hazard ratios (HRs) with significance at P = .05. RESULTS Median OS was 9.1 months (95% CI = 7.2-14.3). At the second post-bevacizumab scan, the volume of the low-ADC lesion (median: 12.94 cm(3)) was inversely associated with OS, with larger volumes predicting shorter OS (HR = 1.014 [95% CI = 1.003-1.025], P = .009). The percent change in low-ADC volume (median: 6.8%) trended toward increased risk of death with growing volumes (P = .08). Normalized 5th percentile low-ADC value and its percent change were not associated with OS (P > .51). Also correlated with shorter OS were the pre-bevacizumab nonenhancing volume (P = .025), the first post-bevacizumab enhancing volume (P = .040), and the second post-bevacizumab enhancing volume (P = .004). CONCLUSIONS The volume of low-ADC lesions at the second post-bevacizumab scan predicted shorter OS. This suggests that low-ADC lesions may be considered important imaging markers and included in treatment decision algorithms.
Collapse
Affiliation(s)
- Myron Zhang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| | - Bryanna Gulotta
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| | - Alissa Thomas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| | - Thomas Kaley
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| | - Sasan Karimi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| | - Igor Gavrilovic
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| | - Kaitlin M Woo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| | - Zhigang Zhang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| | - Julio Arevalo-Perez
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| | - Andrei I Holodny
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| | - Marc Rosenblum
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| | - Robert J Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (M.Z., B.G., S.K., J.A.-P., A.I.H., R.J.Y.); Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (A.T., T.K., I.G.); Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York (K.M.W., Z.Z.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York (T.K., S.K., I.G., A.I.H., M.R., R.J.Y.)
| |
Collapse
|
50
|
Shiroishi MS, Boxerman JL, Pope WB. Physiologic MRI for assessment of response to therapy and prognosis in glioblastoma. Neuro Oncol 2015; 18:467-78. [PMID: 26364321 DOI: 10.1093/neuonc/nov179] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 08/01/2015] [Indexed: 02/06/2023] Open
Abstract
Aside from bidimensional measurements from conventional contrast-enhanced MRI, there are no validated or FDA-qualified imaging biomarkers for high-grade gliomas. However, advanced functional MRI techniques, including perfusion- and diffusion-weighted MRI, have demonstrated much potential for determining prognosis, predicting therapeutic response, and assessing early treatment response. They may also prove useful for differentiating pseudoprogression from true progression after temozolomide chemoradiation and pseudoresponse from true response after anti-angiogenic therapy. This review will highlight recent developments using these techniques and emphasize the need for technical standardization and validation in prospective studies in order for these methods to become incorporated into standard-of-care imaging for brain tumor patients.
Collapse
Affiliation(s)
- Mark S Shiroishi
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California (M.S.S.); Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.); Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California (W.B.P.)
| | - Jerrold L Boxerman
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California (M.S.S.); Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.); Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California (W.B.P.)
| | - Whitney B Pope
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California (M.S.S.); Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.); Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California (W.B.P.)
| |
Collapse
|