201
|
Cho N, Wang C, Raymond C, Kaprealian T, Ji M, Salamon N, Pope WB, Nghiemphu PL, Lai A, Cloughesy TF, Ellingson BM. Diffusion MRI changes in the anterior subventricular zone following chemoradiation in glioblastoma with posterior ventricular involvement. J Neurooncol 2020; 147:643-652. [PMID: 32239430 DOI: 10.1007/s11060-020-03460-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/14/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION There is growing evidence that the subventricular zone (SVZ) plays a key role in glioblastoma (GBM) tumorigenesis. However, little is known regarding how the SVZ, which is a harbor for adult neural stem cells, may be influenced by chemoradiation. The current diffusion-weighted imaging (DWI) study explored ipsilateral and contralateral alterations in the anterior SVZ in GBM patients with posterior enhancing lesions following chemoradiation. METHODS Forty GBM patients with tumor involvement in the posterior SVZ (mean age = 57 ± 10; left-hemisphere N = 25; right-hemisphere N = 15) were evaluated using DWI before and after chemoradiation. Regions-of-interest were drawn on the ipsilesional and contralesional anterior SVZ on apparent diffusion coefficient (ADC) maps for both timepoints. ADC histogram analysis was performed by modeling a bimodal, double Gaussian distribution to obtain ADCL, defined as the mean of the lower Gaussian distribution. RESULTS The ipsilesional SVZ had lower ADCL values compared to the contralesional SVZ before treatment (mean difference = 0.025 μm2/ms; P = 0.007). Following chemoradiation, these changes were no longer observed (mean difference = 0.0025 μm2/ms; P > 0.5), as ADCL values of the ipsilesional SVZ increased (mean difference = 0.026 μm2/ms; P = 0.037). An increase in ipsilesional ADCL was associated with shorter progression-free (P = 0.0119) and overall survival (P = 0.0265). CONCLUSIONS These preliminary observations suggest baseline asymmetry as well as asymmetric changes in the SVZ proximal (ipsilesional) to the tumor with respect to contralesional SVZ regions may be present in GBM, potentially implicating this region in tumorigenesis and/or treatment resistance.
Collapse
Affiliation(s)
- Nicholas Cho
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, 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
- Medical Scientist Training Program, 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, David Geffen School of Medicine, 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
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, 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
| | - Tania Kaprealian
- Department of Radiation Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Matthew Ji
- Department of Neurology, 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
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Albert Lai
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, 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, David Geffen School of Medicine, 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.
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Departments of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.
| |
Collapse
|
202
|
Magnetic resonance imaging for brain stereotactic radiotherapy : A review of requirements and pitfalls. Strahlenther Onkol 2020; 196:444-456. [PMID: 32206842 PMCID: PMC7182639 DOI: 10.1007/s00066-020-01604-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/03/2020] [Indexed: 12/29/2022]
Abstract
Due to its superior soft tissue contrast, magnetic resonance imaging (MRI) is essential for many radiotherapy treatment indications. This is especially true for treatment planning in intracranial tumors, where MRI has a long-standing history for target delineation in clinical practice. Despite its routine use, care has to be taken when selecting and acquiring MRI studies for the purpose of radiotherapy treatment planning. Requirements on MRI are particularly demanding for intracranial stereotactic radiotherapy, where accurate imaging has a critical role in treatment success. However, MR images acquired for routine radiological assessment are frequently unsuitable for high-precision stereotactic radiotherapy as the requirements for imaging are significantly different for radiotherapy planning and diagnostic radiology. To assure that optimal imaging is used for treatment planning, the radiation oncologist needs proper knowledge of the most important requirements concerning the use of MRI in brain stereotactic radiotherapy. In the present review, we summarize and discuss the most relevant issues when using MR images for target volume delineation in intracranial stereotactic radiotherapy.
Collapse
|
203
|
Lesueur P, Kao W, Leconte A, Geffrelot J, Lequesne J, Lacroix J, Brachet PE, Hrab I, Royer P, Clarisse B, Stefan D. Stereotactic radiotherapy on brain metastases with recent hemorrhagic signal: STEREO-HBM, a two-step phase 2 trial. BMC Cancer 2020; 20:147. [PMID: 32087691 PMCID: PMC7036220 DOI: 10.1186/s12885-020-6569-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 01/21/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Brain metastases often occur in cancer evolution. They are not only responsible for death but also for disorders affecting the quality of life and the cognitive functions. Management of brain metastases usually consists in multi-modality treatments, including neurosurgery, whole brain radiotherapy (WBRT), and more recently radiosurgery (SRS) or fractionated stereotactic radiotherapy (FSRT), systemic treatment (chemotherapy or targeted therapy), combined or not with corticosteroids. Almost 20% of brain metastases can present recent (within 15 days) bleeding signs on neuro-imagery. In these conditions, WBRT is the usual treatment. Yet, patients may benefit from a more aggressive strategy with SRT or FSRT. However, these options were suspected to possibly major the risk of brain haemorrhage, although no scientifically proven. Radiation oncologists therefore usually remain reluctant to deliver SRS/FSRT for bleeding brain metastases. It is therefore challenging to establish a standard of care for the treatment of bleeding brain metastases. We propose a phase II trial to simultaneously assess safety and efficacy of FSRT to manage brain metastases with hemorrhagic signal. METHODS The STEREO-HBM study is a multicenter two-step non-randomised phase II trial addressing patients with at least one bleeding brain metastasis out of a maximum of 3 brain metastases. Each brain metastasis will be treated with 30 Gy in 3 fractions for 1 week. The main endpoint is based on both safety and efficacy endpoints as proposed by Bryant and Day's design. Safety endpoint is defined as the rate of bleeding complications 4 months post-FSRT while efficacy endpoint is defined as the 6-month local control rate. Multi-modal MRI will be used to assess intra-tumoral hemorrhagic events before and after treatment. Patients' quality of life will also be assessed. DISCUSSION Management of bleeding brain metastases is still debated and poorly explored in clinical trials. There is sparse and weak data on the signification of pretreatment intra-tumour haemorrhagic signs or on the risk of brain bleeding complications after FSRT. We expect this first prospective phase 2 trial in this particular setting will allow to clarify the place of FSRT to optimally manage bleeding brain metastases. TRIAL REGISTRATION NCT03696680, registered October, 4, 2018. PROTOCOL VERSION Version 2.1 dated from 2018/11/09.
Collapse
Affiliation(s)
- Paul Lesueur
- Radiation Oncology Department, Centre François Baclesse, F-14000 Caen, France
- Normandy University, F-14000 Caen, France
| | - William Kao
- Radiation Oncology Department, Centre François Baclesse, F-14000 Caen, France
| | - Alexandra Leconte
- Clinical Research Department, Centre François Baclesse, F-14000 Caen, France
| | - Julien Geffrelot
- Radiation Oncology Department, Centre François Baclesse, F-14000 Caen, France
| | - Justine Lequesne
- Clinical Research Department, Centre François Baclesse, F-14000 Caen, France
| | - Joëlle Lacroix
- Radiology Department, Centre François Baclesse, F-14000 Caen, France
| | - Pierre-Emmanuel Brachet
- Clinical Research Department, Centre François Baclesse, F-14000 Caen, France
- Medical Oncology Department, Centre François Baclesse, F-14000 Caen, France
| | - Ioana Hrab
- Medical Oncology Department, Centre François Baclesse, F-14000 Caen, France
| | - Philippe Royer
- Radiation Oncology Department, Institut de Cancérologie de Lorraine, F-54000 Vandœuvre-lès-Nancy, France
| | - Bénédicte Clarisse
- Clinical Research Department, Centre François Baclesse, F-14000 Caen, France
| | - Dinu Stefan
- Radiation Oncology Department, Centre François Baclesse, F-14000 Caen, France
- Radiation Oncology Department, Centre François Baclesse, 3 Avenue du Général Harris, F-14076 Caen Cedex 05, France
| |
Collapse
|
204
|
Duan S, Zhu Y, Liu F, Xin SX. Numerical Experiments on the Contrast Capability of Magnetic Resonance Electrical Property Tomography. Magn Reson Med Sci 2020; 19:77-85. [PMID: 31019159 PMCID: PMC7067912 DOI: 10.2463/mrms.mp.2018-0167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Purpose: Magnetic resonance electrical property tomography (MR EPT) is a technique for non-invasively obtaining the electric property (EP) distribution of biological tissues, with a promising potential for application in the early detection of tumors. However, the contrast capability (CC) of this technique has not been fully studied. This work aims to theoretically explore the CC for detecting the variation of EP values and the size of the imaging region. Methods: A simulation scheme was specifically designed to evaluate the CC of MR EPT. The simulation study has the advantage that the magnetic field can be accurately obtained. EP maps of the designed phantom embedded with target regions of designated various sizes and EPs were reconstructed using the homogeneous Helmholtz equation based on B1+ with different signal-to-noise ratios (SNRs). The CC was estimated by determining the smallest detectable EP contrast when the target region size was as large as the Laplacian kernel and the smallest detectable target region size when the EP contrast was the same as the difference between healthy and malignant tissues in the brain, based on the reconstructed EP maps. Results: Using noise free B1+, the smallest detectable contrastσ and contrastεr were 1% and 3%, respectively, and the smallest detectable target region size was 1 mesh unit (the base unit size used in the simulation) for conductivity and relative permittivity. The smallest detectable EP contrast and target region size were decreased as the B1+ SNR increased. Conclusion: The CC of MR EPT was related with the SNR of the magnetic field. A small EP contrast and size were necessary for detection at a high-SNR magnetic field. Obtaining a high-SNR magnetic field is important for improving the CC of MR EPT.
Collapse
Affiliation(s)
- Song Duan
- Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Yurong Zhu
- Department of Biomedical Engineering, Southern Medical University
| | - Feng Liu
- School of Information Technology and Electrical Engineering, University of Queensland
| | - Sherman Xuegang Xin
- School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre
| |
Collapse
|
205
|
Ellingson BM. On the promise of artificial intelligence for standardizing radiographic response assessment in gliomas. Neuro Oncol 2020; 21:1346-1347. [PMID: 31504809 DOI: 10.1093/neuonc/noz162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
206
|
Stringfield O, Arrington JA, Johnston SK, Rognin NG, Peeri NC, Balagurunathan Y, Jackson PR, Clark-Swanson KR, Swanson KR, Egan KM, Gatenby RA, Raghunand N. Multiparameter MRI Predictors of Long-Term Survival in Glioblastoma Multiforme. ACTA ACUST UNITED AC 2020; 5:135-144. [PMID: 30854451 PMCID: PMC6403044 DOI: 10.18383/j.tom.2018.00052] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Standard-of-care multiparameter magnetic resonance imaging (MRI) scans of the brain were used to objectively subdivide glioblastoma multiforme (GBM) tumors into regions that correspond to variations in blood flow, interstitial edema, and cellular density. We hypothesized that the distribution of these distinct tumor ecological "habitats" at the time of presentation will impact the course of the disease. We retrospectively analyzed initial MRI scans in 2 groups of patients diagnosed with GBM, a long-term survival group comprising subjects who survived >36 month postdiagnosis, and a short-term survival group comprising subjects who survived ≤19 month postdiagnosis. The single-institution discovery cohort contained 22 subjects in each group, while the multi-institution validation cohort contained 15 subjects per group. MRI voxel intensities were calibrated, and tumor voxels clustered on contrast-enhanced T1-weighted and fluid-attenuated inversion-recovery (FLAIR) images into 6 distinct "habitats" based on low- to medium- to high-contrast enhancement and low-high signal on FLAIR scans. Habitat 6 (high signal on calibrated contrast-enhanced T1-weighted and FLAIR sequences) comprised a significantly higher volume fraction of tumors in the long-term survival group (discovery cohort, 35% ± 6.5%; validation cohort, 34% ± 4.8%) compared with tumors in the short-term survival group (discovery cohort, 17% ± 4.5%, P < .03; validation cohort, 16 ± 4.0%, P < .007). Of the 6 distinct MRI-defined habitats, the fractional tumor volume of habitat 6 at diagnosis was significantly predictive of long- or short-term survival. We discuss a possible mechanistic basis for this association and implications for habitat-driven adaptive therapy of GBM.
Collapse
Affiliation(s)
| | - John A Arrington
- Departments of Diagnostic & Interventional Radiology.,Department of Oncologic Sciences, University of S Florida, Tampa, FL
| | - Sandra K Johnston
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ.,Department of Radiology, University of Washington, Seattle, WA; and
| | | | - Noah C Peeri
- Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL
| | | | - Pamela R Jackson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ
| | - Kamala R Clark-Swanson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ
| | - Kristin R Swanson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ
| | - Kathleen M Egan
- Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL.,Department of Oncologic Sciences, University of S Florida, Tampa, FL
| | - Robert A Gatenby
- Departments of Diagnostic & Interventional Radiology.,Department of Oncologic Sciences, University of S Florida, Tampa, FL
| | - Natarajan Raghunand
- Cancer Physiology, and.,Department of Oncologic Sciences, University of S Florida, Tampa, FL
| |
Collapse
|
207
|
Korshoej AR, Lukacova S, Lassen-Ramshad Y, Rahbek C, Severinsen KE, Guldberg TL, Mikic N, Jensen MH, Cortnum SOS, von Oettingen G, Sørensen JCH. OptimalTTF-1: Enhancing tumor treating fields therapy with skull remodeling surgery. A clinical phase I trial in adult recurrent glioblastoma. Neurooncol Adv 2020; 2:vdaa121. [PMID: 33215088 PMCID: PMC7660275 DOI: 10.1093/noajnl/vdaa121] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Preclinical studies suggest that skull remodeling surgery (SR-surgery) increases the dose of tumor treating fields (TTFields) in glioblastoma (GBM) and prevents wasteful current shunting through the skin. SR-surgery introduces minor skull defects to focus the cancer-inhibiting currents toward the tumor and increase the treatment dose. This study aimed to test the safety and feasibility of this concept in a phase I setting. METHODS Fifteen adult patients with the first recurrence of GBM were treated with personalized SR-surgery, TTFields, and physician's choice oncological therapy. The primary endpoint was toxicity and secondary endpoints included standard efficacy outcomes. RESULTS SR-surgery resulted in a mean skull defect area of 10.6 cm2 producing a median TTFields enhancement of 32% (range 25-59%). The median TTFields treatment duration was 6.8 months and the median compliance rate 90%. Patients received either bevacizumab, bevacizumab/irinotecan, or temozolomide rechallenge. We observed 71 adverse events (AEs) of grades 1 (52%), 2 (35%), and 3 (13%). There were no grade 4 or 5 AEs or intervention-related serious AEs. Six patients experienced minor TTFields-induced skin rash. The median progression-free survival (PFS) was 4.6 months and the PFS rate at 6 months was 36%. The median overall survival (OS) was 15.5 months and the OS rate at 12 months was 55%. CONCLUSIONS TTFields therapy combined with SR-surgery and medical oncological treatment is safe and nontoxic and holds the potential to improve the outcome for GBM patients through focal dose enhancement in the tumor.
Collapse
Affiliation(s)
- Anders Rosendal Korshoej
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Slavka Lukacova
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Christian Rahbek
- Department of Neuroradiology, Aarhus University Hospital, Aarhus Denmark
| | | | | | - Nikola Mikic
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | | | | | | |
Collapse
|
208
|
Penas-Prado M, Wu J, Cahill DP, Brat DJ, Costello JF, Kluetz PG, Cairncross JG, van den Bent M, Verhaak RGW, Aboud O, Burger P, Chang SM, Cordova C, Huang RY, Rowe LS, Taphoorn MJB, Gilbert MR, Armstrong TS. Proceedings of the Comprehensive Oncology Network Evaluating Rare CNS Tumors (NCI-CONNECT) Oligodendroglioma Workshop. Neurooncol Adv 2019; 2:vdz048. [PMID: 33289010 DOI: 10.1093/noajnl/vdz048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Oligodendroglioma is a rare primary central nervous system (CNS) tumor with highly variable outcome and for which therapy is usually not curative. At present, little is known regarding the pathways involved with progression of oligodendrogliomas or optimal biomarkers for stratifying risk. Developing new therapies for this rare cancer is especially challenging. To overcome these challenges, the neuro-oncology community must be particularly innovative, seeking multi-institutional and international collaborations, and establishing partnerships with patients and advocacy groups thereby ensuring that each patient enrolled in a study is as informative as possible. Methods The mission of the National Cancer Institute's NCI-CONNECT program is to address the challenges and unmet needs in rare CNS cancer research and treatment by connecting patients, health care providers, researchers, and advocacy organizations to work in partnership. On November 19, 2018, the program convened a workshop on oligodendroglioma, one of the 12 rare CNS cancers included in its initial portfolio. The purpose of this workshop was to discuss scientific progress and regulatory challenges in oligodendroglioma research and develop a call to action to advance research and treatment for this cancer. Results The recommendations of the workshop include a multifaceted and interrelated approach covering: biology and preclinical models, data sharing and advanced molecular diagnosis and imaging; clinical trial design; and patient outreach and engagement. Conclusions The NCI-CONNECT program is well positioned to address challenges in oligodendroglioma care and research in collaboration with other stakeholders and is developing a list of action items for future initiatives.
Collapse
Affiliation(s)
| | - Jing Wu
- Neuro-Oncology Branch/National Cancer Institute, Bethesda, Maryland
| | - Daniel P Cahill
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel J Brat
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Joseph F Costello
- Department of Neurological Surgery, University of California, San Francisco
| | - Paul G Kluetz
- Oncology Center of Excellence, U.S. Food and Drug Administration, Washington DC
| | | | | | - Roel G W Verhaak
- Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | - Orwa Aboud
- Neuro-Oncology Branch/National Cancer Institute, Bethesda, Maryland.,Brain Tumor Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Peter Burger
- Neuropathology Division, Johns Hopkins, Baltimore, Maryland
| | - Susan M Chang
- Department of Neurological Surgery, University of California, San Francisco
| | - Christine Cordova
- Neuro-Oncology Branch/National Cancer Institute, Bethesda, Maryland.,NYU School of Medicine, Laura and Isaac Perlmutter Cancer Center, New York, NY
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lindsay S Rowe
- Radiation Oncology Branch/National Cancer Institute, Bethesda, Maryland
| | - Martin J B Taphoorn
- Leiden University Medical Center and Haaglanden Medical Center, The Hague, The Netherlands
| | - Mark R Gilbert
- Neuro-Oncology Branch/National Cancer Institute, Bethesda, Maryland
| | | | | |
Collapse
|
209
|
Thomas RP, Nagpal S, Iv M, Soltys SG, Bertrand S, Pelpola JS, Ball R, Yang J, Sundaram V, Lavezo J, Born D, Vogel H, Brown JM, Recht LD. Macrophage Exclusion after Radiation Therapy (MERT): A First in Human Phase I/II Trial using a CXCR4 Inhibitor in Glioblastoma. Clin Cancer Res 2019; 25:6948-6957. [PMID: 31537527 PMCID: PMC6891194 DOI: 10.1158/1078-0432.ccr-19-1421] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/17/2019] [Accepted: 09/11/2019] [Indexed: 01/18/2023]
Abstract
PURPOSE Preclinical studies have demonstrated that postirradiation tumor revascularization is dependent on a stromal cell-derived factor-1 (SDF-1)/C-X-C chemokine receptor type 4 (CXCR4)-driven process in which myeloid cells are recruited from bone marrow. Blocking this axis results in survival improvement in preclinical models of solid tumors, including glioblastoma (GBM). We conducted a phase I/II study to determine the safety and efficacy of Macrophage Exclusion after Radiation Therapy (MERT) using the reversible CXCR4 inhibitor plerixafor in patients with newly diagnosed glioblastoma. PATIENTS AND METHODS We enrolled nine patients in the phase I study and an additional 20 patients in phase II using a modified toxicity probability interval (mTPI) design. Plerixafor was continuously infused intravenously via a peripherally inserted central catheter (PICC) line for 4 consecutive weeks beginning at day 35 of conventional treatment with concurrent chemoradiation. Blood serum samples were obtained for pharmacokinetic analysis. Additional studies included relative cerebral blood volume (rCBV) analysis using MRI and histopathology analysis of recurrent tumors. RESULTS Plerixafor was well tolerated with no drug-attributable grade 3 toxicities observed. At the maximum dose of 400 μg/kg/day, biomarker analysis found suprathreshold plerixafor serum levels and an increase in plasma SDF-1 levels. Median overall survival was 21.3 months [95% confidence interval (CI), 15.9-NA] with a progression-free survival of 14.5 months (95% CI, 11.9-NA). MRI and histopathology support the mechanism of action to inhibit postirradiation tumor revascularization. CONCLUSIONS Infusion of the CXCR4 inhibitor plerixafor was well tolerated as an adjunct to standard chemoirradiation in patients with newly diagnosed GBM and improves local control of tumor recurrences.
Collapse
Affiliation(s)
- Reena P Thomas
- Department of Neurology, Division of Neuro Oncology, Stanford, California.
| | - Seema Nagpal
- Department of Neurology, Division of Neuro Oncology, Stanford, California
| | - Michael Iv
- Department of Radiology, Division of Neuro Radiology, Stanford, California
| | | | - Sophie Bertrand
- Department of Neurology, Division of Neuro Oncology, Stanford, California
| | - Judith S Pelpola
- Department of Neurology, Division of Neuro Oncology, Stanford, California
| | - Robyn Ball
- Department of Medicine, Quantitative Sciences Unit, Stanford, California
| | - Jaden Yang
- Department of Medicine, Quantitative Sciences Unit, Stanford, California
| | - Vandana Sundaram
- Department of Medicine, Quantitative Sciences Unit, Stanford, California
| | - Jonathan Lavezo
- Department of Pathology, Division of Neuro Pathology, Stanford University, Stanford, California
| | - Donald Born
- Department of Pathology, Division of Neuro Pathology, Stanford University, Stanford, California
| | - Hannes Vogel
- Department of Pathology, Division of Neuro Pathology, Stanford University, Stanford, California
| | - J Martin Brown
- Department of Neurology, Division of Neuro Oncology, Stanford, California
| | - Lawrence D Recht
- Department of Neurology, Division of Neuro Oncology, Stanford, California
| |
Collapse
|
210
|
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: 3.5] [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
|
211
|
Rebsamen M, Knecht U, Reyes M, Wiest R, Meier R, McKinley R. Divide and Conquer: Stratifying Training Data by Tumor Grade Improves Deep Learning-Based Brain Tumor Segmentation. Front Neurosci 2019; 13:1182. [PMID: 31749678 PMCID: PMC6848279 DOI: 10.3389/fnins.2019.01182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/18/2019] [Indexed: 11/13/2022] Open
Abstract
It is a general assumption in deep learning that more training data leads to better performance, and that models will learn to generalize well across heterogeneous input data as long as that variety is represented in the training set. Segmentation of brain tumors is a well-investigated topic in medical image computing, owing primarily to the availability of a large publicly-available dataset arising from the long-running yearly Multimodal Brain Tumor Segmentation (BraTS) challenge. Research efforts and publications addressing this dataset focus predominantly on technical improvements of model architectures and less on properties of the underlying data. Using the dataset and the method ranked third in the BraTS 2018 challenge, we performed experiments to examine the impact of tumor type on segmentation performance. We propose to stratify the training dataset into high-grade glioma (HGG) and low-grade glioma (LGG) subjects and train two separate models. Although we observed only minor gains in overall mean dice scores by this stratification, examining case-wise rankings of individual subjects revealed statistically significant improvements. Compared to a baseline model trained on both HGG and LGG cases, two separately trained models led to better performance in 64.9% of cases (p < 0.0001) for the tumor core. An analysis of subjects which did not profit from stratified training revealed that cases were missegmented which had poor image quality, or which presented clinically particularly challenging cases (e.g., underrepresented subtypes such as IDH1-mutant tumors), underlining the importance of such latent variables in the context of tumor segmentation. In summary, we found that segmentation models trained on the BraTS 2018 dataset, stratified according to tumor type, lead to a significant increase in segmentation performance. Furthermore, we demonstrated that this gain in segmentation performance is evident in the case-wise ranking of individual subjects but not in summary statistics. We conclude that it may be useful to consider the segmentation of brain tumors of different types or grades as separate tasks, rather than developing one tool to segment them all. Consequently, making this information available for the test data should be considered, potentially leading to a more clinically relevant BraTS competition.
Collapse
Affiliation(s)
- Michael Rebsamen
- Support Center for Advanced Neuroimaging (SCAN), University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Urspeter Knecht
- Institute for Surgical Technology and Biomechanics, University of Bern, Bern, Switzerland
| | - Mauricio Reyes
- Healthcare Imaging A.I. Lab, Insel Data Science Center, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Roland Wiest
- Support Center for Advanced Neuroimaging (SCAN), University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Raphael Meier
- Support Center for Advanced Neuroimaging (SCAN), University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Richard McKinley
- Support Center for Advanced Neuroimaging (SCAN), University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
212
|
Chang K, Beers AL, Bai HX, Brown JM, Ly KI, Li X, Senders JT, Kavouridis VK, Boaro A, Su C, Bi WL, Rapalino O, Liao W, Shen Q, Zhou H, Xiao B, Wang Y, Zhang PJ, Pinho MC, Wen PY, Batchelor TT, Boxerman JL, Arnaout O, Rosen BR, Gerstner ER, Yang L, Huang RY, Kalpathy-Cramer J. Automatic assessment of glioma burden: a deep learning algorithm for fully automated volumetric and bidimensional measurement. Neuro Oncol 2019; 21:1412-1422. [PMID: 31190077 PMCID: PMC6827825 DOI: 10.1093/neuonc/noz106] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Longitudinal measurement of glioma burden with MRI is the basis for treatment response assessment. In this study, we developed a deep learning algorithm that automatically segments abnormal fluid attenuated inversion recovery (FLAIR) hyperintensity and contrast-enhancing tumor, quantitating tumor volumes as well as the product of maximum bidimensional diameters according to the Response Assessment in Neuro-Oncology (RANO) criteria (AutoRANO). METHODS Two cohorts of patients were used for this study. One consisted of 843 preoperative MRIs from 843 patients with low- or high-grade gliomas from 4 institutions and the second consisted of 713 longitudinal postoperative MRI visits from 54 patients with newly diagnosed glioblastomas (each with 2 pretreatment "baseline" MRIs) from 1 institution. RESULTS The automatically generated FLAIR hyperintensity volume, contrast-enhancing tumor volume, and AutoRANO were highly repeatable for the double-baseline visits, with an intraclass correlation coefficient (ICC) of 0.986, 0.991, and 0.977, respectively, on the cohort of postoperative GBM patients. Furthermore, there was high agreement between manually and automatically measured tumor volumes, with ICC values of 0.915, 0.924, and 0.965 for preoperative FLAIR hyperintensity, postoperative FLAIR hyperintensity, and postoperative contrast-enhancing tumor volumes, respectively. Lastly, the ICCs for comparing manually and automatically derived longitudinal changes in tumor burden were 0.917, 0.966, and 0.850 for FLAIR hyperintensity volume, contrast-enhancing tumor volume, and RANO measures, respectively. CONCLUSIONS Our automated algorithm demonstrates potential utility for evaluating tumor burden in complex posttreatment settings, although further validation in multicenter clinical trials will be needed prior to widespread implementation.
Collapse
Affiliation(s)
- Ken Chang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Andrew L Beers
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Harrison X Bai
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James M Brown
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - K Ina Ly
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Joeky T Senders
- Computational Neuroscience Outcomes Center, Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Vasileios K Kavouridis
- Computational Neuroscience Outcomes Center, Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Alessandro Boaro
- Computational Neuroscience Outcomes Center, Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Chang Su
- Yale School of Medicine, New Haven, Connecticut, USA
| | - Wenya Linda Bi
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Otto Rapalino
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qin Shen
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Paul J Zhang
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marco C Pinho
- Department of Radiology and Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Tracy T Batchelor
- Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Jerrold L Boxerman
- Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Omar Arnaout
- Computational Neuroscience Outcomes Center, Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Elizabeth R Gerstner
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Li Yang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Jayashree Kalpathy-Cramer
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
213
|
Cagney DN, Sul J, Huang RY, Ligon KL, Wen PY, Alexander BM. The FDA NIH Biomarkers, EndpointS, and other Tools (BEST) resource in neuro-oncology. Neuro Oncol 2019; 20:1162-1172. [PMID: 29294069 DOI: 10.1093/neuonc/nox242] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In early 2016, the FDA and the National Institutes of Health (NIH) published the first version of the glossary included in the Biomarkers, EndpointS, and other Tools (BEST) resource.1 The BEST glossary was constructed to harmonize and clarify terms used in translational science and medical product development and to provide a common language used for communication by those agencies. It is considered a "living" document that will be updated in the future. This review will discuss the main biomarker and clinical outcome categories contained in the BEST glossary as they apply to neuro-oncology, as well as the overlapping and hierarchical relationships among them.
Collapse
Affiliation(s)
- Daniel N Cagney
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Joohee Sul
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Keith L Ligon
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Brian M Alexander
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
214
|
Glioma through the looking GLASS: molecular evolution of diffuse gliomas and the Glioma Longitudinal Analysis Consortium. Neuro Oncol 2019; 20:873-884. [PMID: 29432615 PMCID: PMC6280138 DOI: 10.1093/neuonc/noy020] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Adult diffuse gliomas are a diverse group of brain neoplasms that inflict a high emotional toll on patients and their families. The Cancer Genome Atlas and similar projects have provided a comprehensive understanding of the somatic alterations and molecular subtypes of glioma at diagnosis. However, gliomas undergo significant cellular and molecular evolution during disease progression. We review the current knowledge on the genomic and epigenetic abnormalities in primary tumors and after disease recurrence, highlight the gaps in the literature, and elaborate on the need for a new multi-institutional effort to bridge these knowledge gaps and how the Glioma Longitudinal Analysis Consortium (GLASS) aims to systemically catalog the longitudinal changes in gliomas. The GLASS initiative will provide essential insights into the evolution of glioma toward a lethal phenotype, with the potential to reveal targetable vulnerabilities and, ultimately, improved outcomes for a patient population in need.
Collapse
|
215
|
Boxerman JL, Zhang Z, Safriel Y, Rogg JM, Wolf RL, Mohan S, Marques H, Sorensen AG, Gilbert MR, Barboriak DP. Prognostic value of contrast enhancement and FLAIR for survival in newly diagnosed glioblastoma treated with and without bevacizumab: results from ACRIN 6686. Neuro Oncol 2019; 20:1400-1410. [PMID: 29590461 DOI: 10.1093/neuonc/noy049] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background ACRIN 6686/RTOG 0825 was a phase III trial of conventional chemoradiation plus adjuvant temozolomide with bevacizumab or without (placebo) in newly diagnosed glioblastoma. This study investigated whether changes in contrast-enhancing and fluid attenuated inversion recovery (FLAIR)-hyperintense tumor assessed by central reading prognosticate overall survival (OS). Methods Two hundred eighty-four patients (171 men; median age 57 y, range 19-79; 159 on bevacizumab) had MRI at post-op (baseline) and pre-cycle 4 of adjuvant temozolomide (22 wk post chemoradiation initiation). Four central readers measured bidimensional lesion enhancement (2D-T1) and FLAIR hyperintensity at both time points. Changes from baseline to pre-cycle 4 for both markers were dichotomized (increasing vs non-increasing). Cox proportional hazards model and Kaplan-Meier survival estimates were used for inference. Results Adjusting for treatment, increasing 2D-T1 (n = 262, hazard ratio [HR] = 2.07, 95% CI: 1.48-2.91, P < 0.0001) and FLAIR (n = 273, HR = 1.75, 95% CI: 1.26-2.41, P = 0.0008) significantly predicted worse OS. Median OS (days) was significantly shorter for patients with increasing versus non-increasing 2D-T1 for both bevacizumab (443 vs 535, P = 0.004) and placebo (526 vs 887, P = 0.001). Median OS was significantly shorter for patients with increasing versus non-increasing FLAIR for placebo (595 vs 872, P = 0.001), and trended similarly for bevacizumab (499 vs 535, P = 0.0935). Adjusting for 2D-T1 and treatment, increasing FLAIR represented significantly higher risk for death (HR = 1.59 [1.11-2.26], P = 0.01). Conclusion Increased 2D-T1 significantly predicts worse OS in both treatment groups, implying absence of a substantial proportion of pseudoprogression 22 weeks after initiation of standard therapy. FLAIR adds value beyond 2D-T1 in predicting OS, potentially addressing the pseudoresponse effect by substratifying bevacizumab-treated patients with non-increasing 2D-T1.
Collapse
Affiliation(s)
- Jerrold L Boxerman
- Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island
| | - Zheng Zhang
- Center for Statistical Sciences, Brown University, Providence, Rhode Island
| | - Yair Safriel
- Pharmascan Clinical Trials and Radiology Associates of Clearwater-University of South Florida, Clearwater, Florida
| | - Jeffrey M Rogg
- Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island
| | - Ronald L Wolf
- Department of Radiology, Neuroradiology Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Suyash Mohan
- Department of Radiology, Neuroradiology Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Helga Marques
- Center for Statistical Sciences, Brown University, Providence, Rhode Island
| | - A Gregory Sorensen
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts.,IMRIS, Deerfield Imaging, Inc, Minnetonka, Minnesota
| | - Mark R Gilbert
- Department of Neuro-oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Neuro-Oncology Branch of the National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Daniel P Barboriak
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
216
|
Gedye C, Sachchithananthan M, Leonard R, Jeffree RL, Buckland ME, Ziegler DS, Graeber MB, Day BW, McDonald KL, Lasocki A, Nowak AK. Driving innovation through collaboration: development of clinical annotation datasets for brain cancer biobanking. Neurooncol Pract 2019; 7:31-37. [PMID: 32257282 DOI: 10.1093/nop/npz036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background A key component of cancer research is the availability of clinical samples with appropriately annotated clinical data. Biobanks facilitate research by collecting/storing various types of clinical samples for research. Brain Cancer Biobanking Australia (BCBA) was established to facilitate the networking of brain cancer biobanking operations Australia-wide. Maximizing biospecimen utility in a networked biobanking environment requires the standardization of procedures and data across different sites. The aim of this research was to scope and develop a recommended clinical annotation dataset both for pediatric and adult brain cancer biobanks. Methods A multidisciplinary working group consisting of members from the BCBA Consortium was established to develop clinical dataset recommendations for brain cancer biobanks. A literature search was undertaken to identify any published clinical dataset recommendations for brain cancer biobanks. An audit of data items collected and stored by BCBA member biobanks was also conducted to survey current clinical data collection practices across the BCBA network. Results BCBA has developed a clinical annotation dataset recommendation for pediatric and adult brain cancer biobanks. The clinical dataset recommendation has 5 clinical data categories: demographic, clinical and radiological diagnosis and surgery, neuropathological diagnosis, patient treatment, and patient follow-up. The data fields have been categorized into 1 of 3 tiers; essential, preferred, and comprehensive. This enables biobanks and researchers to prioritize appropriately where resources are limited. Conclusion This dataset can be used to guide the integration of data from multiple existing biobanks for research studies and for planning prospective brain cancer biobanking activities.
Collapse
Affiliation(s)
- Craig Gedye
- Brain Cancer Biobanking Australia, National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, NSW, Australia.,Clinical Research Director, NSW Health Statewide Biobank, Camperdown NSW.,School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia.,Calvary Mater Newcastle, Waratah, NSW, Australia
| | - Mythily Sachchithananthan
- Brain Cancer Biobanking Australia, National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, NSW, Australia
| | - Robyn Leonard
- Brain Cancer Biobanking Australia, National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, NSW, Australia
| | - Rosalind L Jeffree
- Brain Cancer Biobanking Australia, National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, NSW, Australia.,Royal Brisbane and Women's Hospital, University of Queensland, Australia
| | - Michael E Buckland
- Brain Cancer Biobanking Australia, National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, NSW, Australia.,Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.,Discipline of Pathology, Brain & Mind Centre, University of Sydney, NSW, Australia
| | - David S Ziegler
- Brain Cancer Biobanking Australia, National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, NSW, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia.,School of Women's and Children's Health, University of New South Wales, Sydney, Australia.,Children's Cancer Institute, University of New South Wales, Sydney, Australia
| | - Manuel B Graeber
- Brain Cancer Biobanking Australia, National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, NSW, Australia.,Brain Tumor Research Laboratories, Brain and Mind Centre, The University of Sydney, NSW, Australia
| | - Bryan W Day
- Brain Cancer Biobanking Australia, National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, NSW, Australia.,QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Kerrie L McDonald
- Brain Cancer Biobanking Australia, National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, NSW, Australia.,Cure Brain Cancer Neuro-oncology Laboratory, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia.,Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Arian Lasocki
- Brain Cancer Biobanking Australia, National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, NSW, Australia.,Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Anna K Nowak
- Brain Cancer Biobanking Australia, National Health and Medical Research Council Clinical Trials Centre, The University of Sydney, NSW, Australia.,School of Medicine and Pharmacology, University of Western Australia, Crawley.,Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Australia
| |
Collapse
|
217
|
Kim D, Heo YJ, Jeong HW, Baek JW, Han JY, Lee JY, Jin SC, Baek HJ. Usefulness of the Delay Alternating with Nutation for Tailored Excitation Pulse with T1-Weighted Sampling Perfection with Application-Optimized Contrasts Using Different Flip Angle Evolution in the Detection of Cerebral Metastases: Comparison with MPRAGE Imaging. AJNR Am J Neuroradiol 2019; 40:1469-1475. [PMID: 31371358 DOI: 10.3174/ajnr.a6158] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 06/27/2019] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Contrast-enhanced T1-weighted sampling perfection with application-optimized contrasts by using different flip angle evolution (SPACE) with the delay alternating with nutation for tailored excitation (DANTE) pulse could suppress the blood flow signal and provide a higher contrast-to-noise ratio of enhancing lesion-to-brain parenchyma than the MPRAGE sequence. The purpose of our study was to evaluate the usefulness of SPACE with DANTE compared with MPRAGE for detecting brain metastases. MATERIALS AND METHODS Seventy-one patients who underwent contrast-enhanced SPACE with DANTE and MPRAGE sequences and who were suspected of having metastatic lesions were included. Two neuroradiologists determined the number of enhancing lesions, and diagnostic performance was evaluated using figure of merit, sensitivity, positive predictive value, interobserver agreement, and reading time. Contrast-to-noise ratiolesion/parenchyma and contrast-to-noise ratiowhite matter/gray matter were also assessed. RESULTS SPACE with DANTE (observer one, 328; observer two, 324) revealed significantly more small (<5 mm) enhancing lesions than MPRAGE (observer one, 175; observer two, 150) (P < 0.001 for observer 1, P ≤ .0001 for observer 2). Furthermore, SPACE with DANTE showed significantly higher figure of merit and sensitivity and shorter reading time than MPRAGE for both observers. The mean contrast-to-noise ratiolesion/parenchyma of SPACE with DANTE (52.3 ± 43.1) was significantly higher than that of MPRAGE (17.5 ± 19.3) (P ≤ .0001), but the mean contrast-to-noise ratiowhite matter/gray matter of SPACE with DANTE (-0.65 ± 1.39) was significantly lower than that of MPRAGE (3.08 ± 1.39) (P ≤ .0001). CONCLUSIONS Compared with MPRAGE, SPACE with DANTE significantly improves the detection of brain metastases.
Collapse
Affiliation(s)
- D Kim
- From the Departments of Radiology (D.K., Y.J.H., H.W.J., J.W.B., J.-Y.H.)
| | - Y J Heo
- From the Departments of Radiology (D.K., Y.J.H., H.W.J., J.W.B., J.-Y.H.)
| | - H W Jeong
- From the Departments of Radiology (D.K., Y.J.H., H.W.J., J.W.B., J.-Y.H.)
| | - J W Baek
- From the Departments of Radiology (D.K., Y.J.H., H.W.J., J.W.B., J.-Y.H.)
| | - J-Y Han
- From the Departments of Radiology (D.K., Y.J.H., H.W.J., J.W.B., J.-Y.H.)
| | - J Y Lee
- Internal Medicine (J.Y.L.), Inje University Busan Paik Hospital, Busan, Korea
| | - S-C Jin
- Department of Neurosurgery (S.-C.J.), Inje University Haeundae Paik Hospital, Busan, Republic of Korea
| | - H J Baek
- Department of Radiology (H.J.B.), Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon, Republic of Korea
| |
Collapse
|
218
|
Steidl E, Müller M, Müller A, Herrlinger U, Hattingen E. Longitudinal, leakage corrected and uncorrected rCBV during the first-line treatment of glioblastoma: a prospective study. J Neurooncol 2019; 144:409-417. [PMID: 31321614 DOI: 10.1007/s11060-019-03244-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE Dynamic susceptibility contrast (DSC) MR-perfusion is becoming a standard of care for the monitoring of glioblastoma. Yet, technical standards are lacking and measurements without leakage correction are still common. Also, data on leakage corrected measurements during stable disease is scarce. In this study we hypothesized that basic leakage correction would significantly enhance data quality during stable disease and improve progress detection. We furthermore investigated whether longitudinal data could increase diagnostic performance. METHODS Patients with histologically proven glioblastoma undergoing first-line therapy were prospectively recruited. We conducted DSC perfusion measurements without prebolus administration in 6-week intervals from the end of radiotherapy until progression. Maximum relative cerebral volume values (rCBVmax) with and without leakage correction were calculated using Philips IntelliSpace®. RESULTS We recruited 16 patients and conducted 82 MRI scans with a mean follow up of 7.2 month. During stable disease, corrected rCBVmax was significantly more stable than uncorrected rCBVmax. Detection of progression with a rCBVmax cutoff was better for corrected (specificity 86%) than for uncorrected rCBVmax (specificity 41%). Interestingly, the increase of corrected rCBVmax upon progression also had a good diagnostic performance with a combination of both cutoffs delivering the best result (sensitivity/specificity 89%/93%). CONCLUSION Corrected rCBVmax supports the imaging finding of a stable disease and large increases during longitudinal observation support the diagnosis of tumor progression. rCBV values without prebolus or leakage correction are not reliable to monitor glioblastomas. Further studies to investigate the value of longitudinal rCBV dynamics for the differentiation of real tumor progression from pseudoprogression are warranted.
Collapse
Affiliation(s)
- Eike Steidl
- Institute of Neuroradiology, University Hospital Frankfurt, Schleusenweg 2-16, 60528, Frankfurt, Germany.
- Department of Radiology, Neuroradiology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Mathias Müller
- Department of Radiology, Neuroradiology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
| | - Andreas Müller
- Department of Radiology, Neuroradiology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
| | - Ulrich Herrlinger
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
| | - Elke Hattingen
- Institute of Neuroradiology, University Hospital Frankfurt, Schleusenweg 2-16, 60528, Frankfurt, Germany
- Department of Radiology, Neuroradiology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
219
|
Kasten BB, Udayakumar N, Leavenworth JW, Wu AM, Lapi SE, McConathy JE, Sorace AG, Bag AK, Markert JM, Warram JM. Current and Future Imaging Methods for Evaluating Response to Immunotherapy in Neuro-Oncology. Theranostics 2019; 9:5085-5104. [PMID: 31410203 PMCID: PMC6691392 DOI: 10.7150/thno.34415] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/20/2019] [Indexed: 12/28/2022] Open
Abstract
Imaging plays a central role in evaluating responses to therapy in neuro-oncology patients. The advancing clinical use of immunotherapies has demonstrated that treatment-related inflammatory responses mimic tumor growth via conventional imaging, thus spurring the development of new imaging approaches to adequately distinguish between pseudoprogression and progressive disease. To this end, an increasing number of advanced imaging techniques are being evaluated in preclinical and clinical studies. These novel molecular imaging approaches will serve to complement conventional response assessments during immunotherapy. The goal of these techniques is to provide definitive metrics of tumor response at earlier time points to inform treatment decisions, which has the potential to improve patient outcomes. This review summarizes the available immunotherapy regimens, clinical response criteria, current state-of-the-art imaging approaches, and groundbreaking strategies for future implementation to evaluate the anti-tumor and immune responses to immunotherapy in neuro-oncology applications.
Collapse
Affiliation(s)
- Benjamin B. Kasten
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Neha Udayakumar
- School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianmei W. Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Anna M. Wu
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, United States
| | - Suzanne E. Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jonathan E. McConathy
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Anna G. Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Asim K. Bag
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - James M. Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jason M. Warram
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
220
|
Verburg N, Koopman T, Yaqub M, Hoekstra OS, Lammertsma AA, Schwarte LA, Barkhof F, Pouwels PJW, Heimans JJ, Reijneveld JC, Rozemuller AJM, Vandertop WP, Wesseling P, Boellaard R, de Witt Hamer PC. Direct comparison of [ 11C] choline and [ 18F] FET PET to detect glioma infiltration: a diagnostic accuracy study in eight patients. EJNMMI Res 2019; 9:57. [PMID: 31254208 PMCID: PMC6598977 DOI: 10.1186/s13550-019-0523-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023] Open
Abstract
Background Positron emission tomography (PET) is increasingly used to guide local treatment in glioma. The purpose of this study was a direct comparison of two potential tracers for detecting glioma infiltration, O-(2-[18F]-fluoroethyl)-l-tyrosine ([18F] FET) and [11C] choline. Methods Eight consecutive patients with newly diagnosed diffuse glioma underwent dynamic [11C] choline and [18F] FET PET scans. Preceding craniotomy, multiple stereotactic biopsies were obtained from regions inside and outside PET abnormalities. Biopsies were assessed independently for tumour presence by two neuropathologists. Imaging measurements were derived at the biopsy locations from 10 to 40 min [11C] choline and 20–40, 40–60 and 60–90 min [18F] FET intervals, as standardized uptake value (SUV) and tumour-to-brain ratio (TBR). Diagnostic accuracies of both tracers were compared using receiver operating characteristic analysis and generalized linear mixed modelling with consensus histopathological assessment as reference. Results Of the 74 biopsies, 54 (73%) contained tumour. [11C] choline SUV and [18F] FET SUV and TBR at all intervals were higher in tumour than in normal samples. For [18F] FET, the diagnostic accuracy of TBR was higher than that of SUV for intervals 40–60 min (area under the curve: 0.88 versus 0.81, p = 0.026) and 60–90 min (0.90 versus 0.81, p = 0.047). The diagnostic accuracy of [18F] FET TBR 60–90 min was higher than that of [11C] choline SUV 20–40 min (0.87 versus 0.67, p = 0.005). Conclusions [18F] FET was more accurate than [11C] choline for detecting glioma infiltration. Highest accuracy was found for [18F] FET TBR for the interval 60–90 min post-injection. Electronic supplementary material The online version of this article (10.1186/s13550-019-0523-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Niels Verburg
- Neurosurgical Center Amsterdam, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Thomas Koopman
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Otto S Hoekstra
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Lothar A Schwarte
- Department of Anaesthesiology, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,UCL institutes of Neurology & Healthcare Engineering, Gower St, Bloomsbury, London, WC1E 6BT, UK
| | - Petra J W Pouwels
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Jan J Heimans
- Department of Neurology, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Jaap C Reijneveld
- Department of Neurology, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - William P Vandertop
- Neurosurgical Center Amsterdam, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,Princess Máxima Center for Paediatric Oncology, and Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Philip C de Witt Hamer
- Neurosurgical Center Amsterdam, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
221
|
Schmainda KM, Prah MA, Zhang Z, Snyder BS, Rand SD, Jensen TR, Barboriak DP, Boxerman JL. Quantitative Delta T1 (dT1) as a Replacement for Adjudicated Central Reader Analysis of Contrast-Enhancing Tumor Burden: A Subanalysis of the American College of Radiology Imaging Network 6677/Radiation Therapy Oncology Group 0625 Multicenter Brain Tumor Trial. AJNR Am J Neuroradiol 2019; 40:1132-1139. [PMID: 31248863 DOI: 10.3174/ajnr.a6110] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/08/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND PURPOSE Brain tumor clinical trials requiring solid tumor assessment typically rely on the 2D manual delineation of enhancing tumors by ≥2 expert readers, a time-consuming step with poor interreader agreement. As a solution, we developed quantitative dT1 maps for the delineation of enhancing lesions. This retrospective analysis compares dT1 with 2D manual delineation of enhancing tumors acquired at 2 time points during the post therapeutic surveillance period of the American College of Radiology Imaging Network 6677/Radiation Therapy Oncology Group 0625 (ACRIN 6677/RTOG 0625) clinical trial. MATERIALS AND METHODS Patients enrolled in ACRIN 6677/RTOG 0625, a multicenter, randomized Phase II trial of bevacizumab in recurrent glioblastoma, underwent standard MR imaging before and after treatment initiation. For 123 patients from 23 institutions, both 2D manual delineation of enhancing tumors and dT1 datasets were evaluable at weeks 8 (n = 74) and 16 (n = 57). Using dT1, we assessed the radiologic response and progression at each time point. Percentage agreement with adjudicated 2D manual delineation of enhancing tumor reads and association between progression status and overall survival were determined. RESULTS For identification of progression, dT1 and adjudicated 2D manual delineation of enhancing tumor reads were in perfect agreement at week 8, with 73.7% agreement at week 16. Both methods showed significant differences in overall survival at each time point. When nonprogressors were further divided into responders versus nonresponders/nonprogressors, the agreement decreased to 70.3% and 52.6%, yet dT1 showed a significant difference in overall survival at week 8 (P = .01), suggesting that dT1 may provide greater sensitivity for stratifying subpopulations. CONCLUSIONS This study shows that dT1 can predict early progression comparable with the standard method but offers the potential for substantial time and cost savings for clinical trials.
Collapse
Affiliation(s)
- K M Schmainda
- From the Departments of Biophysics (K.M.S., M.A.P.) .,Radiology (K.M.S., S.D.R.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - M A Prah
- From the Departments of Biophysics (K.M.S., M.A.P.)
| | - Z Zhang
- Department of Biostatistics (Z.Z.).,Center for Statistical Sciences (Z.Z., B.S.S.), Brown University School of Public Health, Providence, Rhode Island
| | - B S Snyder
- Center for Statistical Sciences (Z.Z., B.S.S.), Brown University School of Public Health, Providence, Rhode Island
| | - S D Rand
- Radiology (K.M.S., S.D.R.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - T R Jensen
- Jensen Informatics LLC (T.R.J.), Brookfield, Wisconsin
| | - D P Barboriak
- Department of Radiology (D.P.B.), Duke University Medical Center, Durham, North Carolina.,Department of Diagnostic Imaging (D.P.B., J.L.B.), Rhode Island Hospital, Providence, Rhode Island
| | - J L Boxerman
- Department of Diagnostic Imaging (D.P.B., J.L.B.), Rhode Island Hospital, Providence, Rhode Island.,Warren Alpert Medical School of Brown University (J.L.B.), Providence, Rhode Island
| |
Collapse
|
222
|
Danieli L, Riccitelli GC, Distefano D, Prodi E, Ventura E, Cianfoni A, Kaelin-Lang A, Reinert M, Pravatà E. Brain Tumor-Enhancement Visualization and Morphometric Assessment: A Comparison of MPRAGE, SPACE, and VIBE MRI Techniques. AJNR Am J Neuroradiol 2019; 40:1140-1148. [PMID: 31221635 DOI: 10.3174/ajnr.a6096] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/08/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND PURPOSE Postgadolinium MR imaging is crucial for brain tumor diagnosis and morphometric assessment. We compared brain tumor enhancement visualization and the "target" object morphometry obtained with the most commonly used 3D MR imaging technique, MPRAGE, with 2 other routinely available techniques: sampling perfection with application-optimized contrasts by using different flip angle evolutions (SPACE) and volumetric interpolated brain examination (VIBE). MATERIALS AND METHODS Fifty-four contrast-enhancing tumors (38 gliomas and 16 metastases) were assessed using MPRAGE, VIBE, and SPACE techniques randomly acquired after gadolinium-based contrast agent administration on a 3T scanner. Enhancement conspicuity was assessed quantitatively by calculating the contrast rate and contrast-to-noise ratio, and qualitatively, by consensus visual comparative ratings. The total enhancing tumor volume and between-sequence discrepancy in the margin delineation were assessed on the corresponding 3D target objects contoured with a computer-assisted software for neuronavigation. The Wilcoxon signed rank and Pearson χ2 nonparametric tests were used to investigate between-sequence discrepancies in the contrast rate, contrast-to-noise ratio, visual conspicuity ratings, tumor volume, and margin delineation estimates. Differences were also tested for 1D (Response Evaluation Criteria in Solid Tumors) and 2D (Response Assessment in Neuro-Oncology) measurements. RESULTS Compared with MPRAGE, both SPACE and VIBE obtained higher contrast rate, contrast-to-noise ratio, and visual conspicuity ratings in both gliomas and metastases (P range, <.001-.001). The between-sequence 3D target object margin discrepancy ranged between 3% and 19.9% of lesion tumor volume. Larger tumor volumes, 1D and 2D measurements were obtained with SPACE (P range, <.01-.007). CONCLUSIONS Superior conspicuity for brain tumor enhancement can be achieved using SPACE and VIBE techniques, compared with MPRAGE. Discrepancies were also detected when assessing target object size and morphology, with SPACE providing more accurate estimates.
Collapse
Affiliation(s)
- L Danieli
- From the Departments of Neuroradiology (L.D., D.D., E.P., E.V., A.C., E.P.)
| | - G C Riccitelli
- Neurology (G.C.R., A.K.-L.).,Neuroimaging Research Unit (G.C.R.), Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - D Distefano
- From the Departments of Neuroradiology (L.D., D.D., E.P., E.V., A.C., E.P.)
| | - E Prodi
- From the Departments of Neuroradiology (L.D., D.D., E.P., E.V., A.C., E.P.)
| | - E Ventura
- From the Departments of Neuroradiology (L.D., D.D., E.P., E.V., A.C., E.P.)
| | - A Cianfoni
- From the Departments of Neuroradiology (L.D., D.D., E.P., E.V., A.C., E.P.).,Departments of Neuroradiology (A.C.)
| | - A Kaelin-Lang
- Neurology (G.C.R., A.K.-L.).,Neurology (A.K.-L.), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Faculty of Biomedical Sciences (A.K.-L., M.R.), Università della Svizzera Italiana, Lugano, Switzerland
| | - M Reinert
- Neurosurgery (M.R.), Neurocenter of Southern Switzerland, Lugano, Switzerland.,Faculty of Biomedical Sciences (A.K.-L., M.R.), Università della Svizzera Italiana, Lugano, Switzerland
| | - E Pravatà
- From the Departments of Neuroradiology (L.D., D.D., E.P., E.V., A.C., E.P.)
| |
Collapse
|
223
|
Romeo V, Stanzione A, Ugga L, Cuocolo R, Cocozza S, Ioannidou E, Brunetti A, Bisdas S. A Critical Appraisal of the Quality of Glioma Imaging Guidelines Using the AGREE II Tool: A EuroAIM Initiative. Front Oncol 2019; 9:472. [PMID: 31231610 PMCID: PMC6566105 DOI: 10.3389/fonc.2019.00472] [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] [Received: 03/14/2019] [Accepted: 05/16/2019] [Indexed: 12/24/2022] Open
Abstract
Background: Following the EuroAIM initiative to assess the quality of medical imaging guidelines by using the Appraisal of Guidelines for Research and Evaluation (AGREE) II instrument, we aimed to evaluate the quality of the current imaging guidelines in patients with gliomas. Methods: A literature search was conducted to identify eligible imaging guidelines considered in the management of adult patients with gliomas. The selected guidelines were evaluated using the AGREE II instrument by four independent appraisers. The agreement among the four appraisers was estimated using the intraclass correlation coefficient (ICC) analysis. Results: Seven guidelines were selected for the appraisal. Six out of the seven guidelines showed an average level of quality with only one showing a low quality. The highest scores were found in Domain 1 “Scope and purpose” (mean score = 81.2%) and Domain 4 “Clarity of presentation” (mean score = 77.6%). The remaining domains showed a low level of quality and, in particular, Domain 5 “Applicability” was the most critical with a mean score of 41.7%, mainly related to a minor attention to barriers and facilitators as well as costs and resources implications of applying the guidelines. The ICC analysis showed a very good agreement among the four appraisers with ICC values ranging from 0.907 to 0.993. Conclusions: The available guidelines on glioma imaging emerged as of average quality according to the AGREE II tool analysis. Based on these results, further efforts should be made in order to involve different professional bodies and stakeholders and increase patient and public involvement in any future guideline drafting as well as to improve the applicability of these guidelines into the clinical practice.
Collapse
Affiliation(s)
- Valeria Romeo
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Renato Cuocolo
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Sirio Cocozza
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Evangelia Ioannidou
- Medical School, University of Ioannina, Ioannina, Greece.,Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, London, United Kingdom
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Sotirios Bisdas
- Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, London, United Kingdom.,Department of Brain Repair and Rehabilitation, Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
224
|
Mills SJ, Radon MR, Baird RD, Hanemann CO, Keatley D, Lewis J, Pollock J, Sanghera P, Santarius T, Whitfield G, Zakaria R, Michael D. J. Utilization of volumetric magnetic resonance imaging for baseline and surveillance imaging in Neuro-oncology. Br J Radiol 2019; 92:20190059. [PMID: 30924680 PMCID: PMC6592091 DOI: 10.1259/bjr.20190059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/18/2019] [Accepted: 03/21/2019] [Indexed: 12/14/2022] Open
Abstract
The acquisition of volumetric post-contrast MRI has clear advantages in the interpretation of neuro-oncology studies but has yet to find its way into routine clinical practice beyond planning scans for surgery and radiotherapy. This commentary briefly highlights the benefits of these techniques whilst dispelling some of the perceived disadvantages.
Collapse
Affiliation(s)
| | - Mark R. Radon
- Department of Neuroradiology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| | | | | | - Debbie Keatley
- National Cancer Research Institute Metastases and Meningioma subgroup of the Brain Clinical Studies Group, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
225
|
Wang YL, Yao J, Chakhoyan A, Raymond C, Salamon N, Liau LM, Nghiemphu PL, Lai A, Pope WB, Nguyen N, Ji M, Cloughesy TF, Ellingson BM. Association between Tumor Acidity and Hypervascularity in Human Gliomas Using pH-Weighted Amine Chemical Exchange Saturation Transfer Echo-Planar Imaging and Dynamic Susceptibility Contrast Perfusion MRI at 3T. AJNR Am J Neuroradiol 2019; 40:979-986. [PMID: 31097430 DOI: 10.3174/ajnr.a6063] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 04/10/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND PURPOSE Acidification of the tumor microenvironment from abnormal metabolism along with angiogenesis to meet metabolic demands are both hallmarks of malignant brain tumors; however, the interdependency of tumor acidity and vascularity has not been explored. Therefore, our aim was to investigate the association between pH-sensitive amine chemical exchange saturation transfer echoplanar imaging (CEST-EPI) and relative cerebral blood volume (CBV) measurements obtained from dynamic susceptibility contrast (DSC) perfusion MRI in patients with gliomas. MATERIALS AND METHODS In this retrospective study, 90 patients with histologically confirmed gliomas were scanned between 2015 and 2018 (median age, 50.3 years; male/female ratio = 59:31). pH-weighting was obtained using chemical exchange saturation transfer echo-planar imaging estimation of the magnetization transfer ratio asymmetry at 3 ppm, and CBV was estimated using DSC-MR imaging. The voxelwise correlation and patient-wise median value correlation between the magnetization transfer ratio asymmetry at 3 ppm and CBV within T2-hyperintense lesions and contrast-enhancing lesions were evaluated using the Pearson correlation analysis. RESULTS General colocalization of elevated perfusion and high acidity was observed in tumors, with local intratumor heterogeneity. For patient-wise analysis, median CBV and magnetization transfer ratio asymmetry at 3 ppm within T2-hyperintense lesions were significantly correlated (R = 0.3180, P = .002), but not in areas of contrast enhancement (P = .52). The positive correlation in T2-hyperintense lesions remained within high-grade gliomas (R = 0.4128, P = .001) and in isocitrate dehydrogenase wild-type gliomas (R = 0.4300, P = .002), but not in World Health Organization II or in isocitrate dehydrogenase mutant tumors. Both magnetization transfer ratio asymmetry at 3 ppm and the voxelwise correlation between magnetization transfer ratio asymmetry and CBV were higher in high-grade gliomas compared with low-grade gliomas in T2-hyperintense tumors (magnetization transfer ratio asymmetry, P = .02; Pearson correlation, P = .01). The same trend held when comparing isocitrate dehydrogenase wild-type gliomas and isocitrate dehydrogenase mutant gliomas (magnetization transfer ratio asymmetry, P = .04; Pearson correlation, P = .01). CONCLUSIONS A positive linear correlation between CBV and acidity in areas of T2-hyperintense, nonenhancing tumor, but not enhancing tumor, was observed across patients. Local heterogeneity was observed within individual tumors.
Collapse
Affiliation(s)
- Y-L Wang
- From the UCLA Brain Tumor Imaging Laboratory (Y.-L.W., J.Y., A.C., C.R., B.M.E.).,Department of Radiology (Y.-L.W.), People's Liberation Army General Hospital, Beijing, China
| | - J Yao
- From the UCLA Brain Tumor Imaging Laboratory (Y.-L.W., J.Y., A.C., C.R., B.M.E.).,Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences (J.Y., A.C., C.R., N.S., W.B.P., B.M.E.).,Department of Bioengineering (J.Y., B.M.E.), Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, California
| | - A Chakhoyan
- From the UCLA Brain Tumor Imaging Laboratory (Y.-L.W., J.Y., A.C., C.R., B.M.E.).,Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences (J.Y., A.C., C.R., N.S., W.B.P., B.M.E.)
| | - C Raymond
- From the UCLA Brain Tumor Imaging Laboratory (Y.-L.W., J.Y., A.C., C.R., B.M.E.).,Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences (J.Y., A.C., C.R., N.S., W.B.P., B.M.E.)
| | - N Salamon
- Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences (J.Y., A.C., C.R., N.S., W.B.P., B.M.E.)
| | - L M Liau
- UCLA Brain Research Institute (L.M.L., A.L., B.M.E.).,Department of Neurosurgery (L.M.L.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - P L Nghiemphu
- Department of Neurology (P.L.N., A.L., N.N., M.J., T.F.C.)
| | - A Lai
- Department of Neurology (P.L.N., A.L., N.N., M.J., T.F.C.).,UCLA Brain Research Institute (L.M.L., A.L., B.M.E.)
| | - W B Pope
- Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences (J.Y., A.C., C.R., N.S., W.B.P., B.M.E.)
| | - N Nguyen
- Department of Neurology (P.L.N., A.L., N.N., M.J., T.F.C.)
| | - M Ji
- Department of Neurology (P.L.N., A.L., N.N., M.J., T.F.C.)
| | - T F Cloughesy
- Department of Neurology (P.L.N., A.L., N.N., M.J., T.F.C.)
| | - B M Ellingson
- From the UCLA Brain Tumor Imaging Laboratory (Y.-L.W., J.Y., A.C., C.R., B.M.E.) .,Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences (J.Y., A.C., C.R., N.S., W.B.P., B.M.E.).,Physics and Biology in Medicine (B.M.E.).,Department of Psychiatry and Biobehavioral Sciences (B.M.E.).,UCLA Brain Research Institute (L.M.L., A.L., B.M.E.).,Department of Bioengineering (J.Y., B.M.E.), Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
226
|
Arevalo OD, Soto C, Rabiei P, Kamali A, Ballester LY, Esquenazi Y, Zhu JJ, Riascos RF. Assessment of Glioblastoma Response in the Era of Bevacizumab: Longstanding and Emergent Challenges in the Imaging Evaluation of Pseudoresponse. Front Neurol 2019; 10:460. [PMID: 31133966 PMCID: PMC6514158 DOI: 10.3389/fneur.2019.00460] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 04/16/2019] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma is the deadliest primary malignant brain neoplasm, and despite the availability of many treatment options, its prognosis remains somber. Enhancement detected by magnetic resonance imaging (MRI) was considered the best imaging marker of tumor activity in glioblastoma for decades. However, its role as a surrogate marker of tumor viability has changed with the appearance of new treatment regimens and imaging modalities. The antiangiogenic therapy created an inflection point in the imaging assessment of glioblastoma response in clinical trials and clinical practice. Although BEV led to the improvement of enhancement, it did not necessarily mean tumor response. The decrease in the enhancement intensity represents a change in the permeability properties of the blood brain barrier, and presumably, the switch of the tumor growth pattern to an infiltrative non-enhancing phenotype. New imaging techniques for the assessment of cellularity, blood flow hemodynamics, and biochemistry have emerged to overcome this hurdle; nevertheless, designing tools to assess tumor response more accurately, and in so doing, improve the assessment of response to standard of care (SOC) therapies and to novel therapies, remains challenging.
Collapse
Affiliation(s)
- Octavio D Arevalo
- Department of Diagnostic and Interventional Radiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Carolina Soto
- Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Pejman Rabiei
- Department of Diagnostic and Interventional Radiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Arash Kamali
- Department of Diagnostic and Interventional Radiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Leomar Y Ballester
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jay-Jiguang Zhu
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Roy Francisco Riascos
- Department of Diagnostic and Interventional Radiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
227
|
Yun J, Park JE, Lee H, Ham S, Kim N, Kim HS. Radiomic features and multilayer perceptron network classifier: a robust MRI classification strategy for distinguishing glioblastoma from primary central nervous system lymphoma. Sci Rep 2019; 9:5746. [PMID: 30952930 PMCID: PMC6451024 DOI: 10.1038/s41598-019-42276-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 03/25/2019] [Indexed: 12/14/2022] Open
Abstract
We aimed to establish a high-performing and robust classification strategy, using magnetic resonance imaging (MRI), along with combinations of feature extraction and selection in human and machine learning using radiomics or deep features by employing a small dataset. Using diffusion and contrast-enhanced T1-weighted MR images obtained from patients with glioblastomas and primary central nervous system lymphomas, classification task was assigned to a combination of radiomic features and (1) supervised machine learning after feature selection or (2) multilayer perceptron (MLP) network; or MR image input without radiomic feature extraction to (3) two neuro-radiologists or (4) an end-to-end convolutional neural network (CNN). The results showed similar high performance in generalized linear model (GLM) classifier and MLP using radiomics features in the internal validation set, but MLP network remained robust in the external validation set obtained using different MRI protocols. CNN showed the lowest performance in both validation sets. Our results reveal that a combination of radiomic features and MLP network classifier serves a high-performing and generalizable model for classification task for a small dataset with heterogeneous MRI protocols.
Collapse
Affiliation(s)
- Jihye Yun
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-Ro 43-Gil Songpa-Gu, Seoul, 05505, Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil Songpa-Gu, Seoul, 05505, Korea.
| | - Hyunna Lee
- Health Innovation Big Data Center, Asan Institute for Life Science, 88 Olympic-Ro 43-Gil Songpa-Gu, Seoul, 05505, Korea
| | - Sungwon Ham
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-Ro 43-Gil Songpa-Gu, Seoul, 05505, Korea
| | - Namkug Kim
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-Ro 43-Gil Songpa-Gu, Seoul, 05505, Korea
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil Songpa-Gu, Seoul, 05505, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil Songpa-Gu, Seoul, 05505, Korea
| |
Collapse
|
228
|
Schmainda KM, Prah MA, Hu LS, Quarles CC, Semmineh N, Rand SD, Connelly JM, Anderies B, Zhou Y, Liu Y, Logan B, Stokes A, Baird G, Boxerman JL. Moving Toward a Consensus DSC-MRI Protocol: Validation of a Low-Flip Angle Single-Dose Option as a Reference Standard for Brain Tumors. AJNR Am J Neuroradiol 2019; 40:626-633. [PMID: 30923088 DOI: 10.3174/ajnr.a6015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/18/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE DSC-MR imaging using preload, intermediate (60°) flip angle and postprocessing leakage correction has gained traction as a standard methodology. Simulations suggest that DSC-MR imaging with flip angle = 30° and no preload yields relative CBV practically equivalent to the reference standard. This study tested this hypothesis in vivo. MATERIALS AND METHODS Eighty-four patients with brain lesions were enrolled in this 3-institution study. Forty-three patients satisfied the inclusion criteria. DSC-MR imaging (3T, single-dose gadobutrol, gradient recalled-echo-EPI, TE = 20-35 ms, TR = 1.2-1.63 seconds) was performed twice for each patient, with flip angle = 30°-35° and no preload (P-), which provided preload (P+) for the subsequent intermediate flip angle = 60°. Normalized relative CBV and standardized relative CBV maps were generated, including postprocessing with contrast agent leakage correction (C+) and without (C-) contrast agent leakage correction. Contrast-enhancing lesion volume, mean relative CBV, and contrast-to-noise ratio obtained with 30°/P-/C-, 30°/P-/C+, and 60°/P+/C- were compared with 60°/P+/C+ using the Lin concordance correlation coefficient and Bland-Altman analysis. Equivalence between the 30°/P-/C+ and 60°/P+/C+ protocols and the temporal SNR for the 30°/P- and 60°/P+ DSC-MR imaging data was also determined. RESULTS Compared with 60°/P+/C+, 30°/P-/C+ had closest mean standardized relative CBV (P = .61), highest Lin concordance correlation coefficient (0.96), and lowest Bland-Altman bias (μ = 1.89), compared with 30°/P-/C- (P = .02, Lin concordance correlation coefficient = 0.59, μ = 14.6) and 60°/P+/C- (P = .03, Lin concordance correlation coefficient = 0.88, μ = -10.1) with no statistical difference in contrast-to-noise ratios across protocols. The normalized relative CBV and standardized relative CBV were statistically equivalent at the 10% level using either the 30°/P-/C+ or 60°/P+/C+ protocols. Temporal SNR was not significantly different for 30°/P- and 60°/P+ (P = .06). CONCLUSIONS Tumor relative CBV derived from low-flip angle, no-preload DSC-MR imaging with leakage correction is an attractive single-dose alternative to the higher dose reference standard.
Collapse
Affiliation(s)
- K M Schmainda
- From the Departments of Biophysics (K.M.S., M.A.P.) .,Radiology (K.M.S., S.D.R.)
| | - M A Prah
- From the Departments of Biophysics (K.M.S., M.A.P.)
| | - L S Hu
- Departments of Radiology (L.S.H., Y.Z.)
| | - C C Quarles
- Division of Imaging Research (C.C.Q., N.S., A.S.), Barrow Neurological Institute, Phoenix, Arizona
| | - N Semmineh
- Division of Imaging Research (C.C.Q., N.S., A.S.), Barrow Neurological Institute, Phoenix, Arizona
| | | | | | - B Anderies
- Neurosurgery (B.A.), Mayo Clinic, Scottsdale, Arizona
| | - Y Zhou
- Departments of Radiology (L.S.H., Y.Z.)
| | - Y Liu
- Division of Biostatistics, Institute for Health and Society (Y.L., B.L.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - B Logan
- Division of Biostatistics, Institute for Health and Society (Y.L., B.L.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - A Stokes
- Division of Imaging Research (C.C.Q., N.S., A.S.), Barrow Neurological Institute, Phoenix, Arizona
| | - G Baird
- Department of Diagnostic Imaging (J.L.B., G.B.), Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - J L Boxerman
- Department of Diagnostic Imaging (J.L.B., G.B.), Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
229
|
Cloughesy TF, Mochizuki AY, Orpilla JR, Hugo W, Lee AH, Davidson TB, Wang AC, Ellingson BM, Rytlewski JA, Sanders CM, Kawaguchi ES, Du L, Li G, Yong WH, Gaffey SC, Cohen AL, Mellinghoff IK, Lee EQ, Reardon DA, O'Brien BJ, Butowski NA, Nghiemphu PL, Clarke JL, Arrillaga-Romany IC, Colman H, Kaley TJ, de Groot JF, Liau LM, Wen PY, Prins RM. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat Med 2019; 25:477-486. [PMID: 30742122 PMCID: PMC6408961 DOI: 10.1038/s41591-018-0337-7] [Citation(s) in RCA: 970] [Impact Index Per Article: 161.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/17/2018] [Indexed: 12/18/2022]
Abstract
Glioblastoma is the most common primary malignant brain tumor in adults and is associated with poor survival. The Ivy Foundation Early Phase Clinical Trials Consortium conducted a randomized, multi-institution clinical trial to evaluate immune responses and survival following neoadjuvant and/or adjuvant therapy with pembrolizumab in 35 patients with recurrent, surgically resectable glioblastoma. Patients who were randomized to receive neoadjuvant pembrolizumab, with continued adjuvant therapy following surgery, had significantly extended overall survival compared to patients that were randomized to receive adjuvant, post-surgical programmed cell death protein 1 (PD-1) blockade alone. Neoadjuvant PD-1 blockade was associated with upregulation of T cell- and interferon-γ-related gene expression, but downregulation of cell-cycle-related gene expression within the tumor, which was not seen in patients that received adjuvant therapy alone. Focal induction of programmed death-ligand 1 in the tumor microenvironment, enhanced clonal expansion of T cells, decreased PD-1 expression on peripheral blood T cells and a decreasing monocytic population was observed more frequently in the neoadjuvant group than in patients treated only in the adjuvant setting. These findings suggest that the neoadjuvant administration of PD-1 blockade enhances both the local and systemic antitumor immune response and may represent a more efficacious approach to the treatment of this uniformly lethal brain tumor.
Collapse
Affiliation(s)
- Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Medical and Molecular Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Aaron Y Mochizuki
- Division of Hematology/Oncology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Joey R Orpilla
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Willy Hugo
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alexander H Lee
- Department of Medical and Molecular Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tom B Davidson
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Division of Hematology/Oncology, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anthony C Wang
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Benjamin M Ellingson
- Jonsson Comprehensive Cancer Center, 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
| | | | | | - Eric S Kawaguchi
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lin Du
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gang Li
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
| | - William H Yong
- Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sarah C Gaffey
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Adam L Cohen
- Department of Neurosurgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Ingo K Mellinghoff
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eudocia Q Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Barbara J O'Brien
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas A Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Phioanh L Nghiemphu
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jennifer L Clarke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | | | - Howard Colman
- Department of Neurosurgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Thomas J Kaley
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John F de Groot
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linda M Liau
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Robert M Prins
- Department of Medical and Molecular Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| |
Collapse
|
230
|
Bi WL, Hosny A, Schabath MB, Giger ML, Birkbak NJ, Mehrtash A, Allison T, Arnaout O, Abbosh C, Dunn IF, Mak RH, Tamimi RM, Tempany CM, Swanton C, Hoffmann U, Schwartz LH, Gillies RJ, Huang RY, Aerts HJWL. Artificial intelligence in cancer imaging: Clinical challenges and applications. CA Cancer J Clin 2019; 69:127-157. [PMID: 30720861 PMCID: PMC6403009 DOI: 10.3322/caac.21552] [Citation(s) in RCA: 775] [Impact Index Per Article: 129.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Judgement, as one of the core tenets of medicine, relies upon the integration of multilayered data with nuanced decision making. Cancer offers a unique context for medical decisions given not only its variegated forms with evolution of disease but also the need to take into account the individual condition of patients, their ability to receive treatment, and their responses to treatment. Challenges remain in the accurate detection, characterization, and monitoring of cancers despite improved technologies. Radiographic assessment of disease most commonly relies upon visual evaluations, the interpretations of which may be augmented by advanced computational analyses. In particular, artificial intelligence (AI) promises to make great strides in the qualitative interpretation of cancer imaging by expert clinicians, including volumetric delineation of tumors over time, extrapolation of the tumor genotype and biological course from its radiographic phenotype, prediction of clinical outcome, and assessment of the impact of disease and treatment on adjacent organs. AI may automate processes in the initial interpretation of images and shift the clinical workflow of radiographic detection, management decisions on whether or not to administer an intervention, and subsequent observation to a yet to be envisioned paradigm. Here, the authors review the current state of AI as applied to medical imaging of cancer and describe advances in 4 tumor types (lung, brain, breast, and prostate) to illustrate how common clinical problems are being addressed. Although most studies evaluating AI applications in oncology to date have not been vigorously validated for reproducibility and generalizability, the results do highlight increasingly concerted efforts in pushing AI technology to clinical use and to impact future directions in cancer care.
Collapse
Affiliation(s)
- Wenya Linda Bi
- Assistant Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Ahmed Hosny
- Research Scientist, Department of Radiation Oncology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Matthew B. Schabath
- Associate Member, Department of Cancer EpidemiologyH. Lee Moffitt Cancer Center and Research InstituteTampaFL
| | - Maryellen L. Giger
- Professor of Radiology, Department of RadiologyUniversity of ChicagoChicagoIL
| | - Nicolai J. Birkbak
- Research Associate, The Francis Crick InstituteLondonUnited Kingdom
- Research Associate, University College London Cancer InstituteLondonUnited Kingdom
| | - Alireza Mehrtash
- Research Assistant, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
- Research Assistant, Department of Electrical and Computer EngineeringUniversity of British ColumbiaVancouverBCCanada
| | - Tavis Allison
- Research Assistant, Department of RadiologyColumbia University College of Physicians and SurgeonsNew YorkNY
- Research Assistant, Department of RadiologyNew York Presbyterian HospitalNew YorkNY
| | - Omar Arnaout
- Assistant Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Christopher Abbosh
- Research Fellow, The Francis Crick InstituteLondonUnited Kingdom
- Research Fellow, University College London Cancer InstituteLondonUnited Kingdom
| | - Ian F. Dunn
- Associate Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Raymond H. Mak
- Associate Professor, Department of Radiation Oncology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Rulla M. Tamimi
- Associate Professor, Department of MedicineBrigham and Women’s Hospital, Dana‐Farber Cancer Institute, Harvard Medical SchoolBostonMA
| | - Clare M. Tempany
- Professor of Radiology, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Charles Swanton
- Professor, The Francis Crick InstituteLondonUnited Kingdom
- Professor, University College London Cancer InstituteLondonUnited Kingdom
| | - Udo Hoffmann
- Professor of Radiology, Department of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Lawrence H. Schwartz
- Professor of Radiology, Department of RadiologyColumbia University College of Physicians and SurgeonsNew YorkNY
- Chair, Department of RadiologyNew York Presbyterian HospitalNew YorkNY
| | - Robert J. Gillies
- Professor of Radiology, Department of Cancer PhysiologyH. Lee Moffitt Cancer Center and Research InstituteTampaFL
| | - Raymond Y. Huang
- Assistant Professor, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Hugo J. W. L. Aerts
- Associate Professor, Departments of Radiation Oncology and Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
- Professor in AI in Medicine, Radiology and Nuclear Medicine, GROWMaastricht University Medical Centre (MUMC+)MaastrichtThe Netherlands
| |
Collapse
|
231
|
Rodriguez D, Chambers T, Warmuth-Metz M, Aliaga ES, Warren D, Calmon R, Hargrave D, Garcia J, Vassal G, Grill J, Zahlmann G, Morgan PS, Jaspan T. Evaluation of the Implementation of the Response Assessment in Neuro-Oncology Criteria in the HERBY Trial of Pediatric Patients with Newly Diagnosed High-Grade Gliomas. AJNR Am J Neuroradiol 2019; 40:568-575. [PMID: 30819765 DOI: 10.3174/ajnr.a5982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/31/2018] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE HERBY was a Phase II multicenter trial setup to establish the efficacy and safety of adding bevacizumab to radiation therapy and temozolomide in pediatric patients with newly diagnosed non-brain stem high-grade gliomas. This study evaluates the implementation of the radiologic aspects of HERBY. MATERIALS AND METHODS We analyzed multimodal imaging compliance rates and scan quality for participating sites, adjudication rates and reading times for the central review process, the influence of different Response Assessment in Neuro-Oncology criteria in the final response, the incidence of pseudoprogression, and the benefit of incorporating multimodal imaging into the decision process. RESULTS Multimodal imaging compliance rates were the following: diffusion, 82%; perfusion, 60%; and spectroscopy, 48%. Neuroradiologists' responses differed for 50% of scans, requiring adjudication, with a total average reading time per patient of approximately 3 hours. Pseudoprogression occurred in 10/116 (9%) cases, 8 in the radiation therapy/temozolomide arm and 2 in the bevacizumab arm (P < .01). Increased target enhancing lesion diameter was a reason for progression in 8/86 cases (9.3%) but never the only radiologic or clinical reason. Event-free survival was predicted earlier in 5/86 (5.8%) patients by multimodal imaging (diffusion, n = 4; perfusion, n = 1). CONCLUSIONS The addition of multimodal imaging to the response criteria modified the assessment in a small number of cases, determining progression earlier than structural imaging alone. Increased target lesion diameter, accounting for a large proportion of reading time, was never the only reason to designate disease progression.
Collapse
Affiliation(s)
- D Rodriguez
- Medical Physics and Clinical Engineering (D.R., P.S.M.)
| | - T Chambers
- Cardiff University, School of Medicine (T.C.), Cardiff, UK
| | - M Warmuth-Metz
- Würzburg University, Institute for Diagnostic and Interventional Neuroradiology (M.W.-M.), Würzburg, Germany
| | - E Sanchez Aliaga
- VU University Medical Center, Department of Radiology & Nuclear Medicine (E.S.A.), Amsterdam, the Netherlands
| | - D Warren
- Leeds Teaching Hospital, Department of Radiology (D.W.), Leeds, UK
| | - R Calmon
- Assistance Publique-Hôpitaux de Paris, Pediatric Radiology (R.C.), Paris, France
| | - D Hargrave
- Great Ormond Street Hospital, Haematology and Oncology Department (D.H.), London, UK
| | - J Garcia
- F. Hoffmann-La Roche (J.Garcia, G.Z.), Basel, Switzerland
| | - G Vassal
- Gustave Roussy and Paris-Sud University, Pediatric and Adolescent Oncology and Unite Mixte de Recherche (G.V., J.Grill), Villejuif, France
| | - J Grill
- Gustave Roussy and Paris-Sud University, Pediatric and Adolescent Oncology and Unite Mixte de Recherche (G.V., J.Grill), Villejuif, France
| | - G Zahlmann
- F. Hoffmann-La Roche (J.Garcia, G.Z.), Basel, Switzerland
| | - P S Morgan
- Medical Physics and Clinical Engineering (D.R., P.S.M.).,Nottingham Biomedical Research Centre of the UK National Institute of Health Research (P.S.M.), Nottingham, UK
| | - T Jaspan
- From Nottingham University Hospitals, Department of Radiology (T.J.)
| |
Collapse
|
232
|
pH-weighted amine chemical exchange saturation transfer echoplanar imaging (CEST-EPI) as a potential early biomarker for bevacizumab failure in recurrent glioblastoma. J Neurooncol 2019; 142:587-595. [PMID: 30806888 DOI: 10.1007/s11060-019-03132-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/21/2019] [Indexed: 11/27/2022]
Abstract
PURPOSE The objective of the current study was to explore the efficacy of using pH-weighted amine CEST-EPI as a potential non-invasive imaging biomarker for treatment response and/or failure in recurrent GBM patients treated with bevacizumab. METHOD A total of 11 patients with recurrent GBM treated with bevacizumab were included in this prospective study. CEST-EPI, perfusion MRI, and standardized anatomic MRI were obtained in patients before and after bevacizumab administration. CEST-EPI measures of magnetization transfer ratio asymmetry (MTRasym) at 3 ppm were used for pH-weighted imaging contrast. Multiple measures were examined for their association with progression-free survival (PFS). RESULT Tumor acidity, measured with MTRasym at 3 ppm, was significantly reduced in both contrast enhancing and non-enhancing tumor after bevacizumab (p = 0.0002 and p < 0.00001, respectively). The reduction in tumor acidity in both contrast enhancing and non-enhancing tumor was linearly correlated with PFS (p = 0.044 and p = 0.00026, respectively). In 9 of the 11 patients, areas of residual acidity were localized to areas of tumor recurrence, typically around 2 months prior to radiographic progression. Univariate (p = 0.006) and multivariate Cox regression controlling for age (p = 0.009) both indicated that change in tumor acidity (ΔMTRasym at 3 ppm) was a significant predictor of PFS. CONCLUSIONS This pilot study suggests pH-weighted amine CEST MRI may have value as a non-invasive, early imaging biomarker for bevacizumab treatment response and failure. Early decreases MTRasym at 3.0 ppm in recurrent GBM after bevacizumab may be associated with better PFS. Residual or emerging regions of acidity may colocalize to the site of tumor recurrence.
Collapse
|
233
|
Chakhoyan A, Yao J, Leu K, Pope WB, Salamon N, Yong W, Lai A, Nghiemphu PL, Everson RG, Prins RM, Liau LM, Nathanson DA, Cloughesy TF, Ellingson BM. Validation of vessel size imaging (VSI) in high-grade human gliomas using magnetic resonance imaging, image-guided biopsies, and quantitative immunohistochemistry. Sci Rep 2019; 9:2846. [PMID: 30808879 PMCID: PMC6391482 DOI: 10.1038/s41598-018-37564-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 12/04/2018] [Indexed: 01/19/2023] Open
Abstract
To evaluate the association between a vessel size index (VSIMRI) derived from dynamic susceptibility contrast (DSC) perfusion imaging using a custom spin-and-gradient echo echoplanar imaging (SAGE-EPI) sequence and quantitative estimates of vessel morphometry based on immunohistochemistry from image-guided biopsy samples. The current study evaluated both relative cerebral blood volume (rCBV) and VSIMRI in eleven patients with high-grade glioma (7 WHO grade III and 4 WHO grade IV). Following 26 MRI-guided glioma biopsies in these 11 patients, we evaluated tissue morphometry, including vessel density and average radius, using an automated procedure based on the endothelial cell marker CD31 to highlight tumor vasculature. Measures of rCBV and VSIMRI were then compared to histological measures. We demonstrate good agreement between VSI measured by MRI and histology; VSIMRI = 13.67 μm and VSIHistology = 12.60 μm, with slight overestimation of VSIMRI in grade III patients compared to histology. rCBV showed a moderate but significant correlation with vessel density (r = 0.42, p = 0.03), and a correlation was also observed between VSIMRI and VSIHistology (r = 0.49, p = 0.01). The current study supports the hypothesis that vessel size measures using MRI accurately reflect vessel caliber within high-grade gliomas, while traditional measures of rCBV are correlated with vessel density and not vessel caliber.
Collapse
Affiliation(s)
- Ararat Chakhoyan
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, 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, David Geffen School of Medicine, 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
| | - Kevin Leu
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, 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
| | - Whitney B Pope
- Department of Radiological Sciences, 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
| | - William Yong
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Albert Lai
- Department of Neurology, Ronald Reagan UCLA Medical Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Phioanh L Nghiemphu
- Department of Neurology, Ronald Reagan UCLA Medical Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Richard G Everson
- Department of Neurosurgery, Ronald Reagan UCLA Medical Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Robert M Prins
- Department of Neurosurgery, Ronald Reagan UCLA Medical Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Linda M Liau
- Department of Neurosurgery, Ronald Reagan UCLA Medical Center, University of California Los Angeles, Los Angeles, CA, USA
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, CA, USA
| | - Timothy F Cloughesy
- Department of Neurology, Ronald Reagan UCLA Medical Center, 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, David Geffen School of Medicine, 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, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
234
|
Radoul M, Najac C, Viswanath P, Mukherjee J, Kelly M, Gillespie AM, Chaumeil MM, Eriksson P, Santos RD, Pieper RO, Ronen SM. HDAC inhibition in glioblastoma monitored by hyperpolarized 13 C MRSI. NMR IN BIOMEDICINE 2019; 32:e4044. [PMID: 30561869 PMCID: PMC6545173 DOI: 10.1002/nbm.4044] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 10/11/2018] [Accepted: 10/31/2018] [Indexed: 05/20/2023]
Abstract
Vorinostat is a histone deacetylase (HDAC) inhibitor that inhibits cell proliferation and induces apoptosis in solid tumors, and is in clinical trials for the treatment of glioblastoma (GBM). The goal of this study was to assess whether hyperpolarized 13 C MRS and magnetic resonance spectroscopic imaging (MRSI) can detect HDAC inhibition in GBM models. First, we confirmed HDAC inhibition in U87 GBM cells and evaluated real-time dynamic metabolic changes using a bioreactor system with live vorinostat-treated or control cells. We found a significant 40% decrease in the 13 C MRS-detectable ratio of hyperpolarized [1-13 C]lactate to hyperpolarized [1-13 C]pyruvate, [1-13 C]Lac/Pyr, and a 37% decrease in the pseudo-rate constant, kPL , for hyperpolarized [1-13 C]lactate production, in vorinostat-treated cells compared with controls. To understand the underlying mechanism for this finding, we assessed the expression and activity of lactate dehydrogenase (LDH) (which catalyzes the pyruvate to lactate conversion), its associated cofactor nicotinamide adenine dinucleotide, the expression of monocarboxylate transporters (MCTs) MCT1 and MCT4 (which shuttle pyruvate and lactate in and out of the cell) and intracellular lactate levels. We found that the most likely explanation for our finding that hyperpolarized lactate is reduced in treated cells is a 30% reduction in intracellular lactate levels that occurs as a result of increased expression of both MCT1 and MCT4 in vorinostat-treated cells. In vivo 13 C MRSI studies of orthotopic tumors in mice also showed a significant 52% decrease in hyperpolarized [1-13 C]Lac/Pyr when comparing vorinostat-treated U87 GBM tumors with controls, and, as in the cell studies, this metabolic finding was associated with increased MCT1 and MCT4 expression in HDAC-inhibited tumors. Thus, the 13 C MRSI-detectable decrease in hyperpolarized [1-13 C]lactate production could serve as a biomarker of response to HDAC inhibitors.
Collapse
Affiliation(s)
- Marina Radoul
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California 94158, USA
| | - Chloé Najac
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California 94158, USA
| | - Pavithra Viswanath
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California 94158, USA
| | - Joydeep Mukherjee
- Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, USA
| | - Mark Kelly
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, USA
| | - Anne Marie Gillespie
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California 94158, USA
| | - Myriam M. Chaumeil
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California 94158, USA
- Department of Physical Therapy and Rehabilitation Science and Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California 94158, USA
| | - Pia Eriksson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California 94158, USA
| | - Romelyn Delos Santos
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California 94158, USA
| | - Russell O. Pieper
- Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, USA
| | - Sabrina M. Ronen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California 94158, USA
| |
Collapse
|
235
|
Warren KE, Vezina G, Poussaint TY, Warmuth-Metz M, Chamberlain MC, Packer RJ, Brandes AA, Reiss M, Goldman S, Fisher MJ, Pollack IF, Prados MD, Wen PY, Chang SM, Dufour C, Zurakowski D, Kortmann RD, Kieran MW. Response assessment in medulloblastoma and leptomeningeal seeding tumors: recommendations from the Response Assessment in Pediatric Neuro-Oncology committee. Neuro Oncol 2019; 20:13-23. [PMID: 28449033 DOI: 10.1093/neuonc/nox087] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Lack of standard response criteria in clinical trials for medulloblastoma and other seeding tumors complicates assessment of therapeutic efficacy and comparisons across studies. An international working group was established to develop consensus recommendations for response assessment. The aim is that these recommendations be prospectively evaluated in clinical trials, with the goal of achieving more reliable risk stratification and uniformity across clinical trials. Current practices and literature review were performed to identify major confounding issues and justify subsequently developed recommendations; in areas lacking scientific investigations, recommendations were based on experience of committee members and consensus was reached after discussion. Recommendations apply to both adult and pediatric patients with medulloblastoma and other seeding tumors. Response should be assessed using MR imaging (brain and spine), CSF cytology, and neurologic examination. Clinical imaging standards with minimum mandatory sequence acquisition that optimizes detection of leptomeningeal metastases are defined. We recommend central review prior to inclusion in treatment cohorts to ensure appropriate risk stratification and cohort inclusion. Consensus recommendations and response definitions for patients with medulloblastomas and other seeding tumors have been established; as with other Response Assessment in Neuro-Oncology recommendations, these need to now be prospectively validated in clinical trials.
Collapse
Affiliation(s)
- Katherine E Warren
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - Gilbert Vezina
- Department of Radiology, Children's National Medical Center, Washington, DC
| | - Tina Y Poussaint
- Department of Radiology, Boston Children's Hospital, Boston, Massachusetts
| | - Monika Warmuth-Metz
- Department of Neuroradiology, University Hospital Würzburg, Würzburg, Germany
| | - Marc C Chamberlain
- Department of Neurology, Seattle Cancer Care Alliance, Seattle, Washington
| | - Roger J Packer
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, DC
| | - Alba A Brandes
- Medical Oncology Department, AUSL-IRCCS Scienze Neurologiche, Bologna, Italy
| | - Moshe Reiss
- Division of Pediatric Neuro-Oncology, New York Medical College, Valhalla, New York
| | - Stewart Goldman
- Hematology-Oncology, Neuro-Oncology & Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Michael J Fisher
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ian F Pollack
- Department of Neurological Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Michael D Prados
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California.,Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Susan M Chang
- Department of Neurosurgery, University of California San Francisco, San Francisco, California
| | - Christelle Dufour
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Villejuif, France
| | - David Zurakowski
- Departments of Anesthesia & Surgery, Boston Children's Hospital, Boston, Massachusetts
| | - Rolf D Kortmann
- Department of Radiation Oncology, University of Leipzig, Leipzig, Germany
| | - Mark W Kieran
- Pediatric Neuro-Oncology, Dana Farber Boston Children's Cancer and Blood Disorder's Center, Boston, Massachusetts
| |
Collapse
|
236
|
Wiseman SJ, Meijboom R, Valdés Hernández MDC, Pernet C, Sakka E, Job D, Waldman AD, Wardlaw JM. Longitudinal multi-centre brain imaging studies: guidelines and practical tips for accurate and reproducible imaging endpoints and data sharing. Trials 2019; 20:21. [PMID: 30616680 PMCID: PMC6323670 DOI: 10.1186/s13063-018-3113-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 12/06/2018] [Indexed: 11/10/2022] Open
Abstract
Background Research involving brain imaging is important for understanding common brain diseases. Study endpoints can include features and measures derived from imaging modalities, providing a benchmark against which other phenotypical data can be assessed. In trials, imaging data provide objective evidence of beneficial and adverse outcomes. Multi-centre studies increase generalisability and statistical power. However, there is a lack of practical guidelines for the set-up and conduct of large neuroimaging studies. Methods We address this deficit by describing aspects of study design and other essential practical considerations that will help researchers avoid common pitfalls and data loss. Results The recommendations are grouped into seven categories: (1) planning, (2) defining the imaging endpoints, developing an imaging manual and managing the workflow, (3) performing a dummy run and testing the analysis methods, (4) acquiring the scans, (5) anonymising and transferring the data, (6) monitoring quality, and (7) using structured data and sharing data. Conclusions Implementing these steps will lead to valuable and usable data and help to avoid imaging data wastage.
Collapse
Affiliation(s)
- Stewart J Wiseman
- Edinburgh Imaging and Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK. .,UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK. .,CCBS, Chancellor's Building, Royal Infirmary of Edinburgh, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.
| | - Rozanna Meijboom
- Edinburgh Imaging and Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Maria Del C Valdés Hernández
- Edinburgh Imaging and Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Cyril Pernet
- Edinburgh Imaging and Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Eleni Sakka
- Edinburgh Imaging and Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Dominic Job
- Edinburgh Imaging and Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Adam D Waldman
- Edinburgh Imaging and Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Joanna M Wardlaw
- Edinburgh Imaging and Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
237
|
Radiation and Chemotherapy Induced Injury. Clin Neuroradiol 2019. [DOI: 10.1007/978-3-319-61423-6_68-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
238
|
Pizzini FB, Thust S, Jäger R. Clinical Presentations, Differential Diagnosis, and Imaging Work-Up of Cerebral Mass Lesions. Clin Neuroradiol 2019. [DOI: 10.1007/978-3-319-61423-6_56-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
239
|
van Dijken BR, van Laar PJ, Smits M, Dankbaar JW, Enting RH, van der Hoorn A. Perfusion MRI in treatment evaluation of glioblastomas: Clinical relevance of current and future techniques. J Magn Reson Imaging 2019; 49:11-22. [PMID: 30561164 PMCID: PMC6590309 DOI: 10.1002/jmri.26306] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 07/30/2018] [Indexed: 12/22/2022] Open
Abstract
Treatment evaluation of patients with glioblastomas is important to aid in clinical decisions. Conventional MRI with contrast is currently the standard method, but unable to differentiate tumor progression from treatment-related effects. Pseudoprogression appears as new enhancement, and thus mimics tumor progression on conventional MRI. Contrarily, a decrease in enhancement or edema on conventional MRI during antiangiogenic treatment can be due to pseudoresponse and is not necessarily reflective of a favorable outcome. Neovascularization is a hallmark of tumor progression but not for posttherapeutic effects. Perfusion-weighted MRI provides a plethora of additional parameters that can help to identify this neovascularization. This review shows that perfusion MRI aids to identify tumor progression, pseudoprogression, and pseudoresponse. The review provides an overview of the most applicable perfusion MRI methods and their limitations. Finally, future developments and remaining challenges of perfusion MRI in treatment evaluation in neuro-oncology are discussed. Level of Evidence: 3 Technical Efficacy: Stage 4 J. Magn. Reson. Imaging 2019;49:11-22.
Collapse
Affiliation(s)
- Bart R.J. van Dijken
- Department of Radiology, Medical Imaging Center (MIC)University Medical Center GroningenGroningenthe Netherlands
| | - Peter Jan van Laar
- Department of Radiology, Medical Imaging Center (MIC)University Medical Center GroningenGroningenthe Netherlands
| | - Marion Smits
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamthe Netherlands
| | - Jan Willem Dankbaar
- Department of RadiologyUniversity Medical Center UtrechtUtrechtthe Netherlands
| | - Roelien H. Enting
- Department of NeurologyUniversity Medical Center GroningenGroningenthe Netherlands
| | - Anouk van der Hoorn
- Department of Radiology, Medical Imaging Center (MIC)University Medical Center GroningenGroningenthe Netherlands
- Brain Tumour Imaging Group, Division of Neurosurgery, Department of Clinical NeurosciencesUniversity of Cambridge and Addenbrooke's HospitalCambridgeUK
| |
Collapse
|
240
|
Warren KE, Vezina G, Poussaint TY, Warmuth-Metz M, Packer RJ, Kieran MW. Response to Harreld re: "Response assessment in medulloblastoma and leptomeningeal seeding tumors: recommendations from the Response Assessment in Pediatric Neuro-Oncology Committee". Neuro Oncol 2018; 20:144-145. [PMID: 29329453 DOI: 10.1093/neuonc/nox219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Katherine E Warren
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Gilbert Vezina
- Department of Radiology, Children's National Medical Center, Rockville, Maryland, USA
| | - Tina Y Poussaint
- Department of Radiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Monika Warmuth-Metz
- Department of Neuroradiology, University Hospital Würzburg, Würzburg, Germany
| | - Roger J Packer
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Rockville, Maryland, USA
| | - Mark W Kieran
- Pediatric Neuro-Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| |
Collapse
|
241
|
Pérez-Beteta J, Martínez-González A, Pérez-García VM. A three-dimensional computational analysis of magnetic resonance images characterizes the biological aggressiveness in malignant brain tumours. J R Soc Interface 2018; 15:20180503. [PMID: 30958226 PMCID: PMC6303800 DOI: 10.1098/rsif.2018.0503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/05/2018] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma (GBM) is the most frequent and aggressive type of primary brain tumour. The development of image-based biomarkers from magnetic resonance images (MRIs) has been a topic of recent interest. GBMs on pre-treatment post-contrast T1-weighted (w) MRIs often appear as rim-shaped regions. In this research, we wanted to define rim-shape complexity (RSC) descriptors and study their value as indicators of the tumour's biological aggressiveness. We constructed a set of widths characterizing the rim-shaped contrast-enhancing areas in T1w MRIs, defined measures of the RSC and computed them for 311 GBM patients. Survival analysis, correlations and sensitivity studies were performed to assess the prognostic value of the measurements. All measures obtained from the histograms were found to depend on the class width to some extent. Several measures (FWHM and βR) had high prognostic value. Some histogram-independent measures were predictors of survival: maximum rim width, mean rim width and spherically averaged rim width. The later quantity allowed patients to be classified into subgroups with different rates of survival (mean difference 6.28 months, p = 0.006). In conclusion, some of the morphological quantifiers obtained from pre-treatment T1w MRIs provided information on the biological aggressiveness of GBMs. The results can be used to define prognostic measurements of clinical applicability.
Collapse
Affiliation(s)
- J. Pérez-Beteta
- Department of Mathematics, Mathematical Oncology Laboratory (MôLAB), University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | | | | |
Collapse
|
242
|
Harris RJ, Yao J, Chakhoyan A, Raymond C, Leu K, Liau LM, Nghiemphu PL, Lai A, Salamon N, Pope WB, Cloughesy TF, Ellingson BM. Simultaneous pH-sensitive and oxygen-sensitive MRI of human gliomas at 3 T using multi-echo amine proton chemical exchange saturation transfer spin-and-gradient echo echo-planar imaging (CEST-SAGE-EPI). Magn Reson Med 2018; 80:1962-1978. [PMID: 29626359 PMCID: PMC6107417 DOI: 10.1002/mrm.27204] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/05/2018] [Accepted: 03/11/2018] [Indexed: 01/09/2023]
Abstract
PURPOSE To introduce a new pH-sensitive and oxygen-sensitive MRI technique using amine proton CEST echo spin-and-gradient echo (SAGE) EPI (CEST-SAGE-EPI). METHODS pH-weighting was obtained using CEST estimations of magnetization transfer ratio asymmetry (MTRasym ) at 3 ppm, and oxygen-weighting was obtained using R2' measurements. Glutamine concentration, pH, and relaxation rates were varied in phantoms to validate simulations and estimate relaxation rates. The values of MTRasym and R2' in normal-appearing white matter, T2 hyperintensity, contrast enhancement, and macroscopic necrosis were measured in 47 gliomas. RESULTS Simulation and phantom results confirmed an increase in MTRasym with decreasing pH. The CEST-SAGE-EPI estimates of R2 , R2*, and R2' varied linearly with gadolinium diethylenetriamine penta-acetic acid concentration (R2 = 6.2 mM-1 ·sec-1 and R2* = 6.9 mM-1 ·sec-1 ). The CEST-SAGE-EPI and Carr-Purcell-Meiboom-Gill estimates of R2 (R2 = 0.9943) and multi-echo gradient-echo estimates of R2* (R2 = 0.9727) were highly correlated. T2 lesions had lower R2' and higher MTRasym compared with normal-appearing white matter, suggesting lower hypoxia and high acidity, whereas contrast-enhancement tumor regions had elevated R2' and MTRasym , indicating high hypoxia and acidity. CONCLUSION The CEST-SAGE-EPI technique provides simultaneous pH-sensitive and oxygen-sensitive image contrasts for evaluation of the brain tumor microenvironment. Advantages include fast whole-brain acquisition, in-line B0 correction, and simultaneous estimation of CEST effects, R2 , R2*, and R2' at 3 T.
Collapse
Affiliation(s)
- Robert J. Harris
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Physics and Biology in Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Dept. of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA
| | - Ararat Chakhoyan
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Kevin Leu
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Physics and Biology in Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Linda M. Liau
- UCLA Brain Research Institute (BRI), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Dept. of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Phioanh L. Nghiemphu
- Dept. of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Albert Lai
- Dept. of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- UCLA Brain Research Institute (BRI), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Noriko Salamon
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Whitney B. Pope
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Timothy F. Cloughesy
- Dept. of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Benjamin M. Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Physics and Biology in Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- Dept. of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA
- Dept. of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
- UCLA Brain Research Institute (BRI), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
243
|
Semmineh NB, Bell LC, Stokes AM, Hu LS, Boxerman JL, Quarles CC. Optimization of Acquisition and Analysis Methods for Clinical Dynamic Susceptibility Contrast MRI Using a Population-Based Digital Reference Object. AJNR Am J Neuroradiol 2018; 39:1981-1988. [PMID: 30309842 PMCID: PMC6239921 DOI: 10.3174/ajnr.a5827] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/08/2018] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE The accuracy of DSC-MR imaging CBV maps in glioblastoma depends on acquisition and analysis protocols. Multisite protocol heterogeneity has challenged standardization initiatives due to the difficulties of in vivo validation. This study sought to compare the accuracy of routinely used protocols using a digital reference object. MATERIALS AND METHODS The digital reference object consisted of approximately 10,000 simulated voxels recapitulating typical signal heterogeneity encountered in vivo. The influence of acquisition and postprocessing methods on CBV reliability was evaluated across 6912 parameter combinations, including contrast agent dosing schemes, pulse sequence parameters, field strengths, and postprocessing methods. Accuracy and precision were assessed using the concordance correlation coefficient and coefficient of variation. RESULTS Across all parameter space, the optimal protocol included full-dose contrast agent preload and bolus, intermediate (60°) flip angle, 30-ms TE, and postprocessing with a leakage-correction algorithm (concordance correlation coefficient = 0.97, coefficient of variation = 6.6%). Protocols with no preload or fractional dose preload and bolus using these acquisition parameters were generally less robust. However, a protocol with no preload, full-dose bolus, and low (30°) flip angle performed very well (concordance correlation coefficient = 0.93, coefficient of variation = 8.7% at 1.5T and concordance correlation coefficient = 0.92, coefficient of variation = 8.2% at 3T). CONCLUSIONS Schemes with full-dose preload and bolus maximize CBV accuracy and reduce variability, which could enable smaller sample sizes and more reliable detection of CBV changes in clinical trials. When a lower total contrast agent dose is desired, use of a low flip angle, no preload, and full-dose bolus protocol may provide an attractive alternative.
Collapse
Affiliation(s)
- N B Semmineh
- From the Department of Imaging Research (N.B.S., L.C.B., A.M.S., C.C.Q.), Barrow Neurological Institute, Phoenix, Arizona
| | - L C Bell
- From the Department of Imaging Research (N.B.S., L.C.B., A.M.S., C.C.Q.), Barrow Neurological Institute, Phoenix, Arizona
| | - A M Stokes
- From the Department of Imaging Research (N.B.S., L.C.B., A.M.S., C.C.Q.), Barrow Neurological Institute, Phoenix, Arizona
| | - L S Hu
- Department of Radiology (L.S.H.), Mayo Clinic Arizona, Phoenix, Arizona
| | - J L Boxerman
- Department of Diagnostic Imaging (J.L.B.), Rhode Island Hospital, Providence, Rhode Island
| | - C C Quarles
- From the Department of Imaging Research (N.B.S., L.C.B., A.M.S., C.C.Q.), Barrow Neurological Institute, Phoenix, Arizona
| |
Collapse
|
244
|
Freyschlag CF, Krieg SM, Kerschbaumer J, Pinggera D, Forster MT, Cordier D, Rossi M, Miceli G, Roux A, Reyes A, Sarubbo S, Smits A, Sierpowska J, Robe PA, Rutten GJ, Santarius T, Matys T, Zanello M, Almairac F, Mondot L, Jakola AS, Zetterling M, Rofes A, von Campe G, Guillevin R, Bagatto D, Lubrano V, Rapp M, Goodden J, De Witt Hamer PC, Pallud J, Bello L, Thomé C, Duffau H, Mandonnet E. Imaging practice in low-grade gliomas among European specialized centers and proposal for a minimum core of imaging. J Neurooncol 2018; 139:699-711. [PMID: 29992433 PMCID: PMC6132968 DOI: 10.1007/s11060-018-2916-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/29/2018] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Imaging studies in diffuse low-grade gliomas (DLGG) vary across centers. In order to establish a minimal core of imaging necessary for further investigations and clinical trials in the field of DLGG, we aimed to establish the status quo within specialized European centers. METHODS An online survey composed of 46 items was sent out to members of the European Low-Grade Glioma Network, the European Association of Neurosurgical Societies, the German Society of Neurosurgery and the Austrian Society of Neurosurgery. RESULTS A total of 128 fully completed surveys were received and analyzed. Most centers (n = 96, 75%) were academic and half of the centers (n = 64, 50%) adhered to a dedicated treatment program for DLGG. There were national differences regarding the sequences enclosed in MRI imaging and use of PET, however most included T1 (without and with contrast, 100%), T2 (100%) and TIRM or FLAIR (20, 98%). DWI is performed by 80% of centers and 61% of centers regularly performed PWI. CONCLUSION A minimal core of imaging composed of T1 (w/wo contrast), T2, TIRM/FLAIR, PWI and DWI could be identified. All morphologic images should be obtained in a slice thickness of ≤ 3 mm. No common standard could be obtained regarding advanced MRI protocols and PET. IMPORTANCE OF THE STUDY We believe that our study makes a significant contribution to the literature because we were able to determine similarities in numerous aspects of LGG imaging. Using the proposed "minimal core of imaging" in clinical routine will facilitate future cooperative studies.
Collapse
Affiliation(s)
- Christian F Freyschlag
- Department of Neurosurgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Sandro M Krieg
- Department of Neurosurgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Johannes Kerschbaumer
- Department of Neurosurgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Daniel Pinggera
- Department of Neurosurgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | | | - Dominik Cordier
- Department of Neurosurgery, Universitätsspital Basel, Basel, Switzerland
| | - Marco Rossi
- Neurosurgical Oncology Unit, Humanitas Research Hospital, IRCCS, Milan, Italy
| | - Gabriele Miceli
- Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - Alexandre Roux
- Department of Neurosurgery, Sainte-Anne Hospital, Paris Descartes University, Sorbonne Paris Cité, Paris, France
- Inserm U894, IMA-Brain, Centre de Psychiatrie et Neurosciences, Paris, France
| | - Andrés Reyes
- European Master's in Clinical Linguistics (EMCL), University of Groningen, Groningen, The Netherlands
- EMCL University of Potsdam, Potsdam, Germany
- Neuroscience Institute, and Laboratory of Experimental Psychology, Faculty of Psychology, El Bosque University, Bogotá, Colombia
| | - Silvio Sarubbo
- Division of Neurosurgery, Structural and Functional Connectivity Lab Project, "S. Chiara" Hospital, APSS, Trento, Italy
| | - Anja Smits
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden
| | - Joanna Sierpowska
- Cognition and Brain Plasticity Unit, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, Barcelona, Spain
- Department of Cognition, Development and Education Psychology, Barcelona, Spain
| | - Pierre A Robe
- Department of Neurology and Neurosurgery, Rudolf Magnus Brain Institute, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Geert-Jan Rutten
- Department of Neurosurgery, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands
| | - Thomas Santarius
- Department of Neurosurgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Tomasz Matys
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Marc Zanello
- Department of Neurosurgery, Sainte-Anne Hospital, Paris Descartes University, Sorbonne Paris Cité, Paris, France
- Inserm U894, IMA-Brain, Centre de Psychiatrie et Neurosciences, Paris, France
| | - Fabien Almairac
- Neurosurgery Department, Hôpital Pasteur 2, University Hospital of Nice, Nice, France
| | - Lydiane Mondot
- Radiology Department, Hôpital Pasteur 2, University Hospital of Nice, Nice, France
| | - Asgeir S Jakola
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg, Sweden
| | - Maria Zetterling
- Department of Neurosurgery, Institution of Neuroscience, Uppsala University Hospital, Uppsala, Sweden
| | - Adrià Rofes
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
- Department of Cognitive Science, Johns Hopkins University, Baltimore, USA
| | - Gord von Campe
- Department of Neurosurgery, Medical University Graz, Graz, Austria
| | - Remy Guillevin
- DACTIM, UMR CNRS 7348, Université de Poitiers et CHU de Poitiers, Poitiers, France
| | - Daniele Bagatto
- Neuroradiology Department, University Hospital Santa Maria della Misericordia, Udine, Italy
| | - Vincent Lubrano
- Department of Neurosurgery, CHU Toulouse, Toulouse, France
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Marion Rapp
- Department of Neurosurgery, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - John Goodden
- Department of Neurosurgery, The General Infirmary at Leeds, Leeds, West Yorkshire, UK
| | | | - Johan Pallud
- Department of Neurosurgery, Sainte-Anne Hospital, Paris Descartes University, Sorbonne Paris Cité, Paris, France
- Inserm U894, IMA-Brain, Centre de Psychiatrie et Neurosciences, Paris, France
| | - Lorenzo Bello
- Neurosurgical Oncology Unit, Humanitas Research Hospital, IRCCS, Milan, Italy
| | - Claudius Thomé
- Department of Neurosurgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Hugues Duffau
- Department of Neurosurgery, Hôpital Gui de Chauliac, Montpellier Medical University Center, Montpellier, France
- Institute of Neuroscience of Montpellier, INSERM U1051, University of Montpellier, Montpellier, France
| | - Emmanuel Mandonnet
- Department of Neurosurgery, Lariboisière Hospital, APHP, Paris, France
- University Paris 7, Paris, France
- IMNC, UMR 8165, Orsay, France
| |
Collapse
|
245
|
Stevens SP, Main C, Bailey S, Pizer B, English M, Phillips R, Peet A, Avula S, Wilne S, Wheatley K, Kearns PR, Wilson JS. The utility of routine surveillance screening with magnetic resonance imaging (MRI) to detect tumour recurrence in children with low-grade central nervous system (CNS) tumours: a systematic review. J Neurooncol 2018; 139:507-522. [PMID: 29948767 PMCID: PMC6132973 DOI: 10.1007/s11060-018-2901-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/12/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND Magnetic resonance imaging (MRI) is routinely used as a surveillance tool to detect early asymptomatic tumour recurrence with a view to improving patient outcomes. This systematic review aimed to assess its utility in children with low-grade CNS tumours. METHODS Using standard systematic review methods, twelve databases were searched up to January 2017. RESULTS Seven retrospective case series studies (n = 370 patients) were included, with average follow-up ranging from 5.6 to 7 years. No randomised controlled trials (RCTs) were identified. Due to study heterogeneity only a descriptive synthesis could be undertaken. Imaging was most frequent in the first year post-surgery (with 2-4 scans) reducing to around half this frequency in year two and annually thereafter for the duration of follow-up. Diagnostic yield ranged from 0.25 to 2%. Recurrence rates ranged from 5 to 41%, with most recurrences asymptomatic (range 65-100%). Collectively, 56% of recurrences had occurred within the first year post-treatment (46% in the first 6-months), 68% by year two and 90% by year five. Following recurrence, 90% of patients underwent treatment changes, mainly repeat surgery (72%). Five-year OS ranged from 96 to 100%, while five-year recurrence-free survival ranged from 67 to 100%. None of the studies reported quality of life measures. CONCLUSION This systematic review highlights the paucity of evidence currently available to assess the utility of MRI surveillance despite it being routine clinical practice and costly to patients, their families and healthcare systems. This needs to be evaluated within the context of an RCT.
Collapse
Affiliation(s)
- Simon P Stevens
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Caroline Main
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Simon Bailey
- Sir James Spence Institute of Child Health, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Barry Pizer
- Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Martin English
- Birmingham Women and Children's Hospital NHS Foundation Trust, Birmingham, UK
| | - Robert Phillips
- Centre for Reviews and Dissemination (CRD), University of York, York, UK
| | - Andrew Peet
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Shivaram Avula
- Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Sophie Wilne
- Queen's Medical Centre, Nottingham University Hospitals' NHS Trust, Nottingham, UK
| | - Keith Wheatley
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Pamela R Kearns
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Birmingham Women and Children's Hospital NHS Foundation Trust, Birmingham, UK
| | - Jayne S Wilson
- Cancer Research UK Clinical Trials Unit (CRCTU), Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
246
|
Yao J, Ruan D, Raymond C, Liau LM, Salamon N, Pope WB, Nghiemphu PL, Lai A, Cloughesy TF, Ellingson BM. Improving B 0 Correction for pH-Weighted Amine Proton Chemical Exchange Saturation Transfer (CEST) Imaging by Use of k-Means Clustering and Lorentzian Estimation. ACTA ACUST UNITED AC 2018; 4:123-137. [PMID: 30320212 PMCID: PMC6173788 DOI: 10.18383/j.tom.2018.00017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Amine chemical exchange saturation transfer (CEST) echoplanar imaging (EPI) provides unique pH and amino acid MRI contrast, enabling sensitive detection of altered microenvironment properties in various diseases. However, CEST contrast is sensitive to static magnetic field (B0) inhomogeneities. Here we propose 2 new B0 correction algorithms for use in correcting pH-weighted amine CEST EPI based on k-means clustering and Lorentzian fitting of CEST data: the iterative downsampling estimation using Lorentzian fitting and the 2-stage Lorentzian estimation with 4D polynomial fitting. Higher quality images of asymmetric magnetization transfer ratio (MTRasym) at 3.0 ppm could be obtained with the proposed algorithms than with the existing B0 correction methods. In particular, the proposed methods are shown to improve the intertissue consistency, interpatient consistency, and tumor region signal-to-noise ratio of MTRasym at 3.0 ppm images, with nonexcessive computation time.
Collapse
Affiliation(s)
- Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA
| | - Dan Ruan
- Departments of Radiation Oncology
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Linda M Liau
- Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Noriko Salamon
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Whitney B Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | | | | | | | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
247
|
Chakhoyan A, Woodworth DC, Harris RJ, Lai A, Nghiemphu PL, Liau LM, Pope WB, Cloughesy TF, Ellingson BM. Mono-exponential, diffusion kurtosis and stretched exponential diffusion MR imaging response to chemoradiation in newly diagnosed glioblastoma. J Neurooncol 2018; 139:651-659. [PMID: 29855771 PMCID: PMC6126989 DOI: 10.1007/s11060-018-2910-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/22/2018] [Indexed: 01/18/2023]
Abstract
PURPOSE To quantify changes and prognostic value of diffusion MRI measurements obtained using mono-exponential, diffusion kurtosis imaging (DKI) and stretched exponential (SE) models prior and after chemoradiation in newly diagnosed glioblastoma (GBM). METHODS Diffusion-weighted images (DWIs) were acquired in twenty-three patients following surgery, prior chemoradiation and within 7 days following completion of treatment, using b-values ranging from 0 to 5000s/mm2. Mono-exponential diffusion (apparent diffusion coefficient: ADC), isotropic (non-directional) DKI model with apparent diffusivity (Dapp) and kurtosis (Kapp) estimates as well as SE model with distributed-diffusion coefficient (DDC) and mean intra-voxel heterogeneity (α) were computed for all patients prior and after chemoradiation. Median values were calculated for normal appearing white matter (NAWM) and contrast-enhancing tumor (CET). The magnitudes of diffusion change prior and after chemoradiation were used to predict overall survival (OS). RESULTS Diffusivity in NAWM was consistent for all diffusion measures during chemoradiation, while diffusivity measurements (ADC, Dapp and DDC) within CET changed significantly. A strong positive correlation existed between ADC, Dapp, and DDC measurements prior to chemoradiation; however, this association was weak following chemoradiation, suggesting a more complex microstructural environment after cytotoxic therapy. When combined with baseline tumor volume and MGMT status, age and ADC changes added significant prognostic values, whereas more complex diffusion models did not show significant value in predicting OS. CONCLUSIONS Despite increased tissue complexity following chemoradiation, advanced diffusion models have longer acquisition times, provide largely comparable measures of diffusivity, and do not appear to provide additional prognostic value compared to mono-exponential ADC maps.
Collapse
Affiliation(s)
- Ararat Chakhoyan
- 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
| | - Davis C Woodworth
- 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 Biomedical Physics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert J Harris
- 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
| | - Albert Lai
- 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
| | - 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
| | - Whitney B Pope
- Department of Radiological Sciences, 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.
- UCLA Brain Tumor Imaging Laboratory (BTIL), Biomedical Physics, Psychiatry, and Bioengineering, Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.
| |
Collapse
|
248
|
Jang BS, Jeon SH, Kim IH, Kim IA. Prediction of Pseudoprogression versus Progression using Machine Learning Algorithm in Glioblastoma. Sci Rep 2018; 8:12516. [PMID: 30131513 PMCID: PMC6104063 DOI: 10.1038/s41598-018-31007-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/09/2018] [Indexed: 01/22/2023] Open
Abstract
We aimed to investigate the feasibility of machine learning (ML) algorithm to distinguish pseudoprogression (PsPD) from progression (PD) in patients with glioblastoma (GBM). We recruited the patients diagnosed as primary GBM who received gross total resection (GTR) and concurrent chemoradiotherapy in two institutions from April 2010 to April 2017 and presented suspicious contrast-enhanced lesion on brain magnetic resonance imaging (MRI) during follow-up. Patients from two institutions were allocated to training (N = 59) and testing (N = 19) datasets, respectively. We developed a convolutional neural network combined with a long short-term memory ML structure. MRI data, which was 9 axial post-contrast T1-weighted images in our study, and clinical features were incorporated (Model 1). In the testing set, the trained Model 1 resulted in AUC of 0.83, AUPRC of 0.87, and F1-score of 0.74 using optimal threshold. The performance was superior to that of Model 2 (CNN-LSTM model with MRI data alone) and Model 3 (random forest model with clinical feature alone). The developed algorithm involving MRI data and clinical features could help making decision during follow-up of patients with GBM treated with GTR and concurrent CCRT.
Collapse
Affiliation(s)
- Bum-Sup Jang
- Department of Radiation Oncology, Seoul National University Hospital, Seoul, Korea
| | - Seung Hyuck Jeon
- Department of Radiation Oncology, Seoul National University Hospital, Seoul, Korea
| | - Il Han Kim
- Department of Radiation Oncology, Seoul National University Hospital, Seoul, Korea
- Institute of Radiation Medicine, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - In Ah Kim
- Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnamsi, Korea.
- Institute of Radiation Medicine, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
249
|
Kong Z, Yan C, Zhu R, Wang J, Wang Y, Wang Y, Wang R, Feng F, Ma W. Imaging biomarkers guided anti-angiogenic therapy for malignant gliomas. NEUROIMAGE-CLINICAL 2018; 20:51-60. [PMID: 30069427 PMCID: PMC6067083 DOI: 10.1016/j.nicl.2018.07.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/02/2018] [Accepted: 07/03/2018] [Indexed: 12/24/2022]
Abstract
Antiangiogenic therapy is a universal approach to the treatment of malignant gliomas but fails to prolong the overall survival of newly diagnosed or recurrent glioblastoma patients. Imaging biomarkers are quantitative imaging parameters capable of objectively describing biological processes, pathological changes and treatment responses in some situations and have been utilized for outcome predictions of malignant gliomas in anti-angiogenic therapy. Advanced magnetic resonance imaging techniques (including perfusion-weighted imaging and diffusion-weighted imaging), positron emission computed tomography and magnetic resonance spectroscopy are imaging techniques that can be used to acquire imaging biomarkers, including the relative cerebral blood volume (rCBV), Ktrans, and the apparent diffusion coefficient (ADC). Imaging indicators for a better prognosis when treating malignant gliomas with antiangiogenic therapy include the following: a lower pre- or post-treatment rCBV, less change in rCBV during treatment, a lower pre-treatment Ktrans, a higher vascular normalization index during treatment, less change in arterio-venous overlap during treatment, lower pre-treatment ADC values for the lower peak, smaller ADC volume changes during treatment, and metabolic changes in glucose and phenylalanine. The investigation and utilization of these imaging markers may confront challenges, but may also promote further development of anti-angiogenic therapy. Despite considerable evidence, future prospective studies are critically needed to consolidate the current data and identify novel biomarkers. Anti-angiogenic therapy only benefits specific populations of glioma patients. Advanced imaging techniques can produce quantitative imaging biomarkers. Physiological and metabolic parameter can predict outcome for anti-angiogenic therapy. Larger prospective studies are needed to provide further evidence.
Collapse
Key Words
- 18F-FDOPA, 3,4-dihydroxy-6-[18F]-fluoro-l-phenylalanine
- 18F-FLT, [18F]-fluoro-3-deoxy-3-L-fluorothymidine
- ADC, apparent diffusion coefficient
- AVOL, arterio-venous overlap
- Anti-angiogenic
- BBB, blood brain barrier
- Biomarkers
- CBF, cerebral blood flow
- CBV, cerebral blood volume
- CNS, central nervous system
- CT, computed tomography
- D-2HG, D-2-hydroxypentanedioic acid
- DCE-MRI, dynamic contrast-enhanced magnetic resonance imaging
- DSC-MRI, dynamic susceptibility contrast magnetic resonance imaging
- DWI, diffusion-weighted imaging
- FDG, fluorodeoxyglucose
- FLAIR, fluid-attenuated inversion recovery
- FSE pcASL, fast spin echo pseudocontinuous artery spin labeling
- GBM, glioblastoma
- Glioma
- Imaging
- Ktrans, volume transfer constant between blood plasma and extravascular extracellular space
- MRI, magnetic resonance imaging
- MRS, magnetic resonance spectroscopy
- OS, overall survival
- PET, positron emission computed tomography
- PFS, progression-free survival
- PWI, perfusion-weighted imaging
- RANO, Response Assessment in Neuro-Oncology
- ROI, region of interest
- RSI, restriction spectrum imaging
- SUV, standardized uptake value
- TMZ, temozolomide
- Therapy
- VAI, vessel architectural imaging
- VEGF-A, vascular endothelial growth factor A
- VNI, vascular normalization index.
- fDMs, functional diffusion maps
- nGBM, newly diagnosed glioblastoma
- rCBF, relative cerebral blood flow
- rCBV, relative cerebral blood volume
- rGBM, recurrent glioblastoma
Collapse
Affiliation(s)
- Ziren Kong
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Chengrui Yan
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China; Department of Neurosurgery, Peking University International Hospital, Peking University, Beijing, China
| | - Ruizhe Zhu
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Jiaru Wang
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Yaning Wang
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China.
| | - Renzhi Wang
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China.
| | - Feng Feng
- Department of Radiology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China..
| | - Wenbin Ma
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
250
|
Regnery S, Adeberg S, Dreher C, Oberhollenzer J, Meissner JE, Goerke S, Windschuh J, Deike-Hofmann K, Bickelhaupt S, Zaiss M, Radbruch A, Bendszus M, Wick W, Unterberg A, Rieken S, Debus J, Bachert P, Ladd M, Schlemmer HP, Paech D. Chemical exchange saturation transfer MRI serves as predictor of early progression in glioblastoma patients. Oncotarget 2018; 9:28772-28783. [PMID: 29983895 PMCID: PMC6033360 DOI: 10.18632/oncotarget.25594] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/24/2018] [Indexed: 12/03/2022] Open
Abstract
PURPOSE To prospectively investigate chemical exchange saturation transfer (CEST) MRI in glioblastoma patients as predictor of early tumor progression after first-line treatment. EXPERIMENTAL DESIGN Twenty previously untreated glioblastoma patients underwent CEST MRI employing a 7T whole-body scanner. Nuclear Overhauser effect (NOE) as well as amide proton transfer (APT) CEST signals were isolated using Lorentzian difference (LD) analysis and relaxation compensated by the apparent exchange-dependent relaxation rate (AREX) evaluation. Additionally, NOE-weighted asymmetric magnetic transfer ratio (MTRasym) and downfield-NOE-suppressed APT (dns-APT) were calculated. Patient response to consecutive treatment was determined according to the RANO criteria. Mean signal intensities of each contrast in the whole tumor area were compared between early-progressive and stable disease. RESULTS Pre-treatment tumor signal intensity differed significantly regarding responsiveness to first-line therapy in NOE-LD (p = 0.0001), NOE-weighted MTRasym (p = 0.0186) and dns-APT (p = 0.0328) contrasts. Hence, significant prediction of early progression was possible employing NOE-LD (AUC = 0.98, p = 0.0005), NOE-weighted MTRasym (AUC = 0.83, p = 0.0166) and dns-APT (AUC = 0.80, p = 0.0318). The NOE-LD provided the highest sensitivity (91%) and specificity (100%). CONCLUSIONS CEST derived contrasts, particularly NOE-weighted imaging and dns-APT, yielded significant predictors of early progression after fist-line therapy in glioblastoma. Therefore, CEST MRI might be considered as non-invasive tool for customization of treatment in the future.
Collapse
Affiliation(s)
- Sebastian Regnery
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division of Radiology, Heidelberg, Germany
| | - Sebastian Adeberg
- German Cancer Research Center (DKFZ), HIRO (Heidelberg Institute for Radiation Oncology), Heidelberg, Germany
| | - Constantin Dreher
- German Cancer Research Center (DKFZ), Division of Radiology, Heidelberg, Germany
| | | | - Jan-Eric Meissner
- German Cancer Research Center (DKFZ), Division of Medical Physics in Radiology, Heidelberg, Germany
| | - Steffen Goerke
- German Cancer Research Center (DKFZ), Division of Medical Physics in Radiology, Heidelberg, Germany
| | - Johannes Windschuh
- German Cancer Research Center (DKFZ), Division of Medical Physics in Radiology, Heidelberg, Germany
| | | | | | | | - Alexander Radbruch
- German Cancer Research Center (DKFZ), Division of Radiology, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Rieken
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Bachert
- German Cancer Research Center (DKFZ), Division of Medical Physics in Radiology, Heidelberg, Germany
| | - Mark Ladd
- German Cancer Research Center (DKFZ), Division of Medical Physics in Radiology, Heidelberg, Germany
- Faculty of Physics and Astronomy, University of Heidelberg, Heidelberg, Germany
- Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | | | - Daniel Paech
- German Cancer Research Center (DKFZ), Division of Radiology, Heidelberg, Germany
| |
Collapse
|