1
|
Sanvito F, Kryukov I, Yao J, Teraishi A, Raymond C, Gao J, Miller C, Nghiemphu PL, Lai A, Liau LM, Patel K, Everson RG, Eldred BSC, Prins RM, Nathanson DA, Salamon N, Cloughesy TF, Ellingson BM. Advanced imaging characterization of post-chemoradiation glioblastoma stratified by diffusion MRI phenotypes known to predict favorable anti-VEGF response. J Neurooncol 2025; 173:583-594. [PMID: 40227555 DOI: 10.1007/s11060-025-05019-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE Recurrent glioblastomas showing a survival benefit from anti-VEGF agents are known to exhibit a distinct diffusion MRI phenotype. We aim to characterize advanced imaging features of this glioblastoma subset. METHODS MRI scans from 87 patients with IDH-wildtype glioblastoma were analyzed. All patients had completed standard chemoradiation and were anti-VEGF-naïve. Contrast-enhancing tumor segmentations were used to extract: the lowest peak of the double gaussian distribution of apparent diffusion coefficient values (ADCL) calculated from diffusion MRI, relative cerebral blood flow (rCBV) values from perfusion MRI, MTRasym @ 3ppm from pH-weighted amine CEST MRI, quantitative T2 and T2* relaxation times (qT2 and qT2*), T1w subtraction map values, and contrast-enhancing tumor volume. Lesions were categorized as high- or low-ADCL using a cutoff of 1240 µm2/s, according to previous studies. RESULTS High-ADCL lesions showed significantly lower rCBV (1.02 vs. 1.28, p = 0.0057), higher MTRasym @ 3ppm (2.36% vs. 2.10%, p = 0.0043), and higher qT2 (114.8 ms vs. 100.9 ms, p = 0.0094), compared to low-ADCL lesions. No group differences were seen in contrast-enhancing tumor volume, T1w subtraction map values, and qT2*, nor in clinical variables such as sex category, MGMT status, and EGFR status. Finally, no clear group-specific preferential locations were seen. CONCLUSION Post-chemoradiation glioblastomas with a diffusion MRI phenotype that is known to predict a favorable response to anti-VEGF (ADCL ≥1240 µm2/s) have distinct biological features, with different perfusion and metabolic characteristics, and T2 relaxation times.
Collapse
Affiliation(s)
- Francesco Sanvito
- 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
| | - Irina Kryukov
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ashley Teraishi
- 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
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - John Gao
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Cole Miller
- 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
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kunal Patel
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Richard G Everson
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Blaine S C Eldred
- 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
| | - Robert M Prins
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - David A Nathanson
- Department of Pharmacology, 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
| | - Timothy F Cloughesy
- 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 Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA, USA.
- Medical Scientist Training Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Anil A, Stokes AM, Karis JP, Bell LC, Eschbacher J, Jennings K, Prah MA, Hu LS, Boxerman JL, Schmainda KM, Quarles CC. Identification of a Single-Dose, Low-Flip-Angle-Based CBV Threshold for Fractional Tumor Burden Mapping in Recurrent Glioblastoma. AJNR Am J Neuroradiol 2024; 45:1545-1551. [PMID: 38782593 PMCID: PMC11448978 DOI: 10.3174/ajnr.a8357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/21/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND AND PURPOSE DSC-MR imaging can be used to generate fractional tumor burden (FTB) maps via application of relative CBV thresholds to spatially differentiate glioblastoma recurrence from posttreatment radiation effects (PTRE). Image-localized histopathology was previously used to validate FTB maps derived from a reference DSC-MR imaging protocol by using preload, a moderate flip angle (MFA, 60°), and postprocessing leakage correction. Recently, a DSC-MR imaging protocol with a low flip angle (LFA, 30°) with no preload was shown to provide leakage-corrected relative CBV (rCBV) equivalent to the reference protocol. This study aimed to identify the rCBV thresholds for the LFA protocol that generate the most accurate FTB maps, concordant with those obtained from the reference MFA protocol. MATERIALS AND METHODS Fifty-two patients with grade-IV glioblastoma who had prior surgical resection and received chemotherapy and radiation therapy were included in the study. Two sets of DSC-MR imaging data were collected sequentially first by using LFA protocol with no preload, which served as the preload for the subsequent MFA protocol. Standardized relative CBV maps (sRCBV) were obtained for each patient and coregistered with the anatomic postcontrast T1-weighted images. The reference MFA-based FTB maps were computed by using previously published sRCBV thresholds (1.0 and 1.56). A receiver operating characteristics (ROC) analysis was conducted to identify the optimal, voxelwise LFA sRCBV thresholds, and the sensitivity, specificity, and accuracy of the LFA-based FTB maps were computed with respect to the MFA-based reference. RESULTS The mean sRCBV values of tumors across patients exhibited strong agreement (concordance correlation coefficient = 0.99) between the 2 protocols. Using the ROC analysis, the optimal lower LFA threshold that accurately distinguishes PTRE from tumor recurrence was found to be 1.0 (sensitivity: 87.77%; specificity: 90.22%), equivalent to the ground truth. To identify aggressive tumor regions, the ROC analysis identified an upper LFA threshold of 1.37 (sensitivity: 90.87%; specificity: 91.10%) for the reference MFA threshold of 1.56. CONCLUSIONS For LFA-based FTB maps, an sRCBV threshold of 1.0 and 1.37 can differentiate PTRE from recurrent tumors. FTB maps aid in surgical planning, guiding pathologic diagnosis and treatment strategies in the recurrent setting. This study further confirms the reliability of single-dose LFA-based DSC-MR imaging.
Collapse
Affiliation(s)
- Aliya Anil
- From the Cancer System Imaging (A.A., C.C.Q.), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ashley M Stokes
- Division of Neuroimaging Research and Barrow Neuroimaging Innovation Center (A.M.S.), Barrow Neurological Institute, Phoenix, Arizona
| | - John P Karis
- Department of Neuroradiology (J.P.K.), Barrow Neurological Institute, Phoenix, Arizona
| | - Laura C Bell
- Clinical Imaging Group (L.C.B.), Genentech Inc., San Francisco, California
| | - Jennifer Eschbacher
- Department of Neuropathology (J.E.), Barrow Neurological Institute, Phoenix, Arizona
| | - Kristofer Jennings
- Department of Biostatistics (K.J.), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Melissa A Prah
- Department of Biophysics (M.A.P., K.M.S.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Leland S Hu
- Department of Radiology (L.S.H.), Division of Neuroradiology, Mayo Clinic Arizona, Phoenix, Arizona
| | - Jerrold L Boxerman
- Department of Diagnostic Imaging (J.L.B.), Rhode Island Hospital, Providence, Rhode Island
| | - Kathleen M Schmainda
- Department of Biophysics (M.A.P., K.M.S.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - C Chad Quarles
- From the Cancer System Imaging (A.A., C.C.Q.), The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
3
|
Vu C, Shen J, Gonzalez Zacarias C, Xu B, Baas K, Choi S, Nederveen A, Wood JC. Contrast-free dynamic susceptibility contrast using sinusoidal and bolus oxygenation challenges. NMR IN BIOMEDICINE 2024; 37:e5111. [PMID: 38297919 PMCID: PMC10987281 DOI: 10.1002/nbm.5111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/10/2023] [Accepted: 01/08/2024] [Indexed: 02/02/2024]
Abstract
Deoxygenation-based dynamic susceptibility contrast (dDSC) MRI uses respiratory challenges as a source of endogenous contrast as an alternative to gadolinium injection. These gas challenges induce T2*-weighted MRI signal losses, after which tracer kinetics modeling was applied to calculate cerebral perfusion. This work compares three gas challenges, desaturation (transient hypoxia), resaturation (transient normoxia), and SineO2 (sinusoidal modulation of end-tidal oxygen pressures) in a cohort of 10 healthy volunteers (age 37 ± 11 years; 60% female). Perfusion estimates consisted of cerebral blood flow (CBF), cerebral blood volume (CBV), and mean transit time (MTT). Calculations were computed using a traditional tracer kinetics model in the time domain for desaturation and resaturation and in the frequency domain for SineO2. High correlations and limits of agreement were observed among the three deoxygenation-based paradigms for CBV, although MTT and CBF estimates varied with the hypoxic stimulus. Cross-modality correlation with gadolinium DSC was lower, particularly for MTT, but on a par with agreement between the other perfusion references. Overall, this work demonstrated the feasibility and reliability of oxygen respiratory challenges to measure brain perfusion. Additional work is needed to assess the utility of dDSC in the diagnostic evaluation of various pathologies such as ischemic strokes, brain tumors, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Chau Vu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Jian Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Clio Gonzalez Zacarias
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA
| | - Botian Xu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Koen Baas
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location AMC, Amsterdam, Netherlands
| | - Soyoung Choi
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA
| | - Aart Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location AMC, Amsterdam, Netherlands
| | - John C. Wood
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
- Division of Cardiology, Children’s Hospital Los Angeles, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
4
|
Prah MA, Schmainda KM. Practical guidance to identify and troubleshoot suboptimal DSC-MRI results. FRONTIERS IN RADIOLOGY 2024; 4:1307586. [PMID: 38445104 PMCID: PMC10913595 DOI: 10.3389/fradi.2024.1307586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/02/2024] [Indexed: 03/07/2024]
Abstract
Relative cerebral blood volume (rCBV) derived from dynamic susceptibility contrast (DSC) perfusion MR imaging (pMRI) has been shown to be a robust marker of neuroradiological tumor burden. Recent consensus recommendations in pMRI acquisition strategies have provided a pathway for pMRI inclusion in diverse patient care centers, regardless of size or experience. However, even with proper implementation and execution of the DSC-MRI protocol, issues will arise that many centers may not easily recognize or be aware of. Furthermore, missed pMRI issues are not always apparent in the resulting rCBV images, potentiating inaccurate or missed radiological diagnoses. Therefore, we gathered from our database of DSC-MRI datasets, true-to-life examples showcasing the breakdowns in acquisition, postprocessing, and interpretation, along with appropriate mitigation strategies when possible. The pMRI issues addressed include those related to image acquisition and postprocessing with a focus on contrast agent administration, timing, and rate, signal-to-noise quality, and susceptibility artifact. The goal of this work is to provide guidance to minimize and recognize pMRI issues to ensure that only quality data is interpreted.
Collapse
Affiliation(s)
- Melissa A. Prah
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kathleen M. Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI,United States
| |
Collapse
|
5
|
Sabeghi P, Zarand P, Zargham S, Golestany B, Shariat A, Chang M, Yang E, Rajagopalan P, Phung DC, Gholamrezanezhad A. Advances in Neuro-Oncological Imaging: An Update on Diagnostic Approach to Brain Tumors. Cancers (Basel) 2024; 16:576. [PMID: 38339327 PMCID: PMC10854543 DOI: 10.3390/cancers16030576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
This study delineates the pivotal role of imaging within the field of neurology, emphasizing its significance in the diagnosis, prognostication, and evaluation of treatment responses for central nervous system (CNS) tumors. A comprehensive understanding of both the capabilities and limitations inherent in emerging imaging technologies is imperative for delivering a heightened level of personalized care to individuals with neuro-oncological conditions. Ongoing research in neuro-oncological imaging endeavors to rectify some limitations of radiological modalities, aiming to augment accuracy and efficacy in the management of brain tumors. This review is dedicated to the comparison and critical examination of the latest advancements in diverse imaging modalities employed in neuro-oncology. The objective is to investigate their respective impacts on diagnosis, cancer staging, prognosis, and post-treatment monitoring. By providing a comprehensive analysis of these modalities, this review aims to contribute to the collective knowledge in the field, fostering an informed approach to neuro-oncological care. In conclusion, the outlook for neuro-oncological imaging appears promising, and sustained exploration in this domain is anticipated to yield further breakthroughs, ultimately enhancing outcomes for individuals grappling with CNS tumors.
Collapse
Affiliation(s)
- Paniz Sabeghi
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Paniz Zarand
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717411, Iran;
| | - Sina Zargham
- Department of Basic Science, California Northstate University College of Medicine, 9700 West Taron Drive, Elk Grove, CA 95757, USA;
| | - Batis Golestany
- Division of Biomedical Sciences, Riverside School of Medicine, University of California, 900 University Ave., Riverside, CA 92521, USA;
| | - Arya Shariat
- Kaiser Permanente Los Angeles Medical Center, 4867 W Sunset Blvd, Los Angeles, CA 90027, USA;
| | - Myles Chang
- Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, CA 90089, USA;
| | - Evan Yang
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Priya Rajagopalan
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Daniel Chang Phung
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Ali Gholamrezanezhad
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| |
Collapse
|
6
|
Al-Gizawiy MM, Wujek RT, Alhajala HS, Cobb JM, Prah MA, Doan NB, Connelly JM, Chitambar CR, Schmainda KM. Potent in vivo efficacy of oral gallium maltolate in treatment-resistant glioblastoma. Front Oncol 2024; 13:1278157. [PMID: 38288102 PMCID: PMC10822938 DOI: 10.3389/fonc.2023.1278157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/26/2023] [Indexed: 01/31/2024] Open
Abstract
Background Treatment-resistant glioblastoma (trGBM) is an aggressive brain tumor with a dismal prognosis, underscoring the need for better treatment options. Emerging data indicate that trGBM iron metabolism is an attractive therapeutic target. The novel iron mimetic, gallium maltolate (GaM), inhibits mitochondrial function via iron-dependent and -independent pathways. Methods In vitro irradiated adult GBM U-87 MG cells were tested for cell viability and allowed to reach confluence prior to stereotactic implantation into the right striatum of male and female athymic rats. Advanced MRI at 9.4T was carried out weekly starting two weeks after implantation. Daily oral GaM (50mg/kg) or vehicle were provided on tumor confirmation. Longitudinal MRI parameters were processed for enhancing tumor ROIs in OsiriX 8.5.1 (lite) with Imaging Biometrics Software (Imaging Biometrics LLC). Statistical analyses included Cox proportional hazards regression models, Kaplan-Meier survival plots, linear mixed model comparisons, and t-statistic for slopes comparison as indicator of tumor growth rate. Results In this study we demonstrate non-invasively, using longitudinal MRI surveillance, the potent antineoplastic effects of GaM in a novel rat xenograft model of trGBM, as evidenced by extended suppression of tumor growth (23.56 mm3/week untreated, 5.76 mm3/week treated, P < 0.001), a blunting of tumor perfusion, and a significant survival benefit (median overall survival: 30 days untreated, 56 days treated; P < 0.001). The therapeutic effect was confirmed histologically by the presence of abundant cytotoxic cellular swelling, a significant reduction in proliferation markers (P < 0.01), and vessel normalization characterized by prominent vessel pruning, loss of branching, and uniformity of vessel lumina. Xenograft tumors in the treatment group were further characterized by an absence of an invasive edge and a significant reduction in both, MIB-1% and mitotic index (P < 0.01 each). Transferrin receptor and ferroportin expression in GaM-treated tumors illustrated cellular iron deprivation. Additionally, treatment with GaM decreased the expression of pro-angiogenic markers (von Willebrand Factor and VEGF) and increased the expression of anti-angiogenic markers, such as Angiopoietin-2. Conclusion Monotherapy with the iron-mimetic GaM profoundly inhibits trGBM growth and significantly extends disease-specific survival in vivo.
Collapse
Affiliation(s)
- Mona M. Al-Gizawiy
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Robert T. Wujek
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Hisham S. Alhajala
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jonathan M. Cobb
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Melissa A. Prah
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ninh B. Doan
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jennifer M. Connelly
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Christopher R. Chitambar
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kathleen M. Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
7
|
Yadav VK, Mohan S, Agarwal S, de Godoy LL, Rajan A, Nasrallah MP, Bagley SJ, Brem S, Loevner LA, Poptani H, Singh A, Chawla S. Distinction of pseudoprogression from true progression in glioblastomas using machine learning based on multiparametric magnetic resonance imaging and O 6-methylguanine-methyltransferase promoter methylation status. Neurooncol Adv 2024; 6:vdae159. [PMID: 39502470 PMCID: PMC11535496 DOI: 10.1093/noajnl/vdae159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Background It is imperative to differentiate true progression (TP) from pseudoprogression (PsP) in glioblastomas (GBMs). We sought to investigate the potential of physiologically sensitive quantitative parameters derived from diffusion and perfusion magnetic resonance imaging (MRI), and molecular signature combined with machine learning in distinguishing TP from PsP in GBMs in the present study. Methods GBM patients (n = 93) exhibiting contrast-enhancing lesions within 6 months after completion of standard treatment underwent 3T MRI. Final data analyses were performed on 75 patients as O6-methylguanine-DNA-methyltransferase (MGMT) status was available only from these patients. Subsequently, patients were classified as TP (n = 55) or PsP (n = 20) based on histological features or mRANO criteria. Quantitative parameters were computed from contrast-enhancing regions of neoplasms. PsP datasets were artificially augmented to achieve balanced class distribution in 2 groups (TP and PsP). A random forest algorithm was applied to select the optimized features. The data were randomly split into training and testing subsets in an 8:2 ratio. To develop a robust prediction model in distinguishing TP from PsP, several machine-learning classifiers were employed. The cross-validation and receiver operating characteristic (ROC) curve analyses were performed to determine the diagnostic performance. Results The quadratic support vector machine was found to be the best classifier in distinguishing TP from PsP with a training accuracy of 91%, cross-validation accuracy of 86%, and testing accuracy of 85%. Additionally, ROC analysis revealed an accuracy of 85%, sensitivity of 70%, and specificity of 100%. Conclusions Machine learning using quantitative multiparametric MRI may be a promising approach to distinguishing TP from PsP in GBMs.
Collapse
Affiliation(s)
- Virendra Kumar Yadav
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sumeet Agarwal
- Yardi School of Artificial Intelligence, Indian Institute of Technology Delhi, New Delhi, India
- Department of Electical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Laiz Laura de Godoy
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Archith Rajan
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - MacLean P Nasrallah
- Department of Clinical Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephen J Bagley
- Department of Medicine, Division of Hematology-Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laurie A Loevner
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Harish Poptani
- Department of Molecular and Clinical Cancer Medicine, Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Anup Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
- Yardi School of Artificial Intelligence, Indian Institute of Technology Delhi, New Delhi, India
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
8
|
Martucci M, Ferranti AM, Schimperna F, Infante A, Magnani F, Olivi A, D'Alessandris QG, Gessi M, Chiesa S, Mazzarella C, Russo R, Giordano C, Gaudino S. Magnetic resonance imaging-derived parameters to predict response to regorafenib in recurrent glioblastoma. Neuroradiology 2023; 65:1439-1445. [PMID: 37247021 DOI: 10.1007/s00234-023-03169-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/21/2023] [Indexed: 05/30/2023]
Abstract
PURPOSE Regorafenib is a multikinase inhibitor, approved as a preferred regimen for recurrent glioblastoma (rGB). Although its effects on prolonging survival could seem modest, it is still unclear whether a subset of patients, potentially identifiable by imaging biomarkers, might experience a more substantial positive effect. Our aim was to evaluate the potential value of magnetic resonance imaging-derived parameters as non-invasive biomarkers to predict response to regorafenib in patients with rGB. METHODS 20 patients with rGB underwent conventional and advanced MRI at diagnosis (before surgery), at recurrence and at first follow-up (3 months) during regorafenib. Maximum relative cerebral blood volume (rCBVmax) value, intra-tumoral susceptibility signals (ITSS), apparent diffusion coefficient (ADC) values, and contrast-enhancing tumor volumes were tested for correlation with response to treatment, progression-free survival (PFS), and overall survival (OS). Response at first follow-up was assessed according to Response Assessment in Neuro-Oncology (RANO) criteria. RESULTS 8/20 patients showed stable disease at first follow-up. rCBVmax values of the primary glioblastoma (before surgery) significantly correlated to treatment response; specifically, patients with stable disease displayed higher rCBVmax compared to progressive disease (p = 0.04, 2-group t test). Moreover, patients with stable disease showed longer PFS (p = 0.02, 2-group t test) and OS (p = 0.04, 2-group t test). ITSS, ADC values, and contrast-enhancing tumor volumes showed no correlation with treatment response, PFS nor OS. CONCLUSION Our results suggest that rCBVmax of the glioblastoma at diagnosis could serve as a non-invasive biomarker of treatment response to regorafenib in patients with rGB.
Collapse
Affiliation(s)
- Matia Martucci
- UOSD Neuroradiologia Diagnostica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy.
| | - Andrea Maurizio Ferranti
- Istituto di Radiologia, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Francesco Schimperna
- Istituto di Radiologia, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Amato Infante
- UOC Radiologia d'Urgenza, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Francesca Magnani
- Istituto di Radiologia, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Alessandro Olivi
- UOC Neurochirurgia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Quintino Giorgio D'Alessandris
- UOC Neurochirurgia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Marco Gessi
- UOS Neuropatologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Silvia Chiesa
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Ciro Mazzarella
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Rosellina Russo
- UOSD Neuroradiologia Diagnostica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Carolina Giordano
- UOSD Neuroradiologia Diagnostica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Simona Gaudino
- UOSD Neuroradiologia Diagnostica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| |
Collapse
|
9
|
Breda-Yepes M, Rodríguez-Hernández LA, Gómez-Figueroa E, Mondragón-Soto MG, Arellano-Flores G, Hernández-Hernández A, Rodríguez-Rubio HA, Martínez P, Reyes-Moreno I, Álvaro-Heredia JA, Gutiérrez Aceves GA, Villanueva-Castro E, Sangrador-Deitos MV, Alonso-Vanegas M, Guerrero-Juárez V, González-Aguilar A. Relative cerebral blood volume as response predictor in the treatment of recurrent glioblastoma with anti-angiogenic therapy. Clin Neurol Neurosurg 2023; 233:107904. [PMID: 37499302 DOI: 10.1016/j.clineuro.2023.107904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/08/2023] [Accepted: 07/16/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Glioblastoma is one of the most common brain tumors in adult populations, usually carrying a poor prognosis. While several studies have researched the impact of anti-angiogenic therapies, especially anti-VEFG treatments in glioblastoma, few have attempted to assess its progress using imaging studies. PURPOSE We attempted to analyze whether relative cerebral blood volume (rCBV) from dynamic susceptibility-weighted contrast-enhanced MRI (DSC-MRI) could predict response in patients with glioblastoma undergoing Bevacizumab (BVZ) treatment. METHODS We performed a retrospective study evaluating patients with recurrent glioblastoma receiving anti-angiogenic therapy with BVZ between 2012 and 2017 in our institution. Patients were scheduled for routine MRIs at baseline and first-month follow-up visits. Studies were processed for DSC-MRI, cT1, and FLAIR images, from which relative cerebral blood volume measurements were obtained. We assessed patient response using the Response Assessment in Neuro-Oncology (RANO) working group criteria and overall survival. RESULTS 40 patients were included in the study and were classified as Bevacizumab responders and non-responders. The average rCBV before treatment was 4.5 for both groups, and average rCBV was 2.5 for responders and 5.4 for non-responders. ROC curve set a cutoff point of 3.7 for rCBV predictive of response to BVZ. Cox Multivariate analysis only showed rCBV as a predictive factor of OS. CONCLUSION A statistically significant difference was found in rCBV between patients who responded and those who did not respond to BVZ treatment. rCBV may be a low-cost and effective marker to assess response to Bevacizumab treatment in GBM.
Collapse
Affiliation(s)
- Michele Breda-Yepes
- Department of Neurosurgery, National Institute of Neurology and Neurosurgery, Mexico
| | | | | | | | | | | | | | - Pablo Martínez
- Department of Neurosurgery, National Institute of Neurology and Neurosurgery, Mexico
| | | | - Juan A Álvaro-Heredia
- Department of Neurosurgery, National Institute of Neurology and Neurosurgery, Mexico
| | | | | | | | - Mario Alonso-Vanegas
- Department of Neurosurgery, National Institute of Neurology and Neurosurgery, Mexico
| | | | - Alberto González-Aguilar
- The American British Cowdray (ABC) Medical Center, Mexico City, Mexico; Department of Neuro-Oncology, National Institute of Neurology and Neurosurgery, Mexico; Emergency Department, National Institute of Neurology and Neurosurgery, Mexico.
| |
Collapse
|
10
|
Shiroishi MS, Weinert D, Cen SY, Varghese B, Dondlinger T, Prah M, Mendoza J, Nazemi S, Ameli N, Amini N, Shohas S, Chen S, Bigjahan B, Zada G, Chen T, Neman-Ebrahim J, Chang EL, Chow FE, Fan Z, Yang W, Attenello FJ, Ye J, Kim PE, Patel VN, Lerner A, Acharya J, Hu LS, Quarles CC, Boxerman JL, Wu O, Schmainda KM. A cross-sectional study to test equivalence of low- versus intermediate-flip angle dynamic susceptibility contrast MRI measures of relative cerebral blood volume in patients with high-grade gliomas at 1.5 Tesla field strength. Front Oncol 2023; 13:1156843. [PMID: 37799462 PMCID: PMC10548232 DOI: 10.3389/fonc.2023.1156843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 08/21/2023] [Indexed: 10/07/2023] Open
Abstract
Introduction 1.5 Tesla (1.5T) remain a significant field strength for brain imaging worldwide. Recent computer simulations and clinical studies at 3T MRI have suggested that dynamic susceptibility contrast (DSC) MRI using a 30° flip angle ("low-FA") with model-based leakage correction and no gadolinium-based contrast agent (GBCA) preload provides equivalent relative cerebral blood volume (rCBV) measurements to the reference-standard acquisition using a single-dose GBCA preload with a 60° flip angle ("intermediate-FA") and model-based leakage correction. However, it remains unclear whether this holds true at 1.5T. The purpose of this study was to test this at 1.5T in human high-grade glioma (HGG) patients. Methods This was a single-institution cross-sectional study of patients who had undergone 1.5T MRI for HGG. DSC-MRI consisted of gradient-echo echo-planar imaging (GRE-EPI) with a low-FA without preload (30°/P-); this then subsequently served as a preload for the standard intermediate-FA acquisition (60°/P+). Both normalized (nrCBV) and standardized relative cerebral blood volumes (srCBV) were calculated using model-based leakage correction (C+) with IBNeuro™ software. Whole-enhancing lesion mean and median nrCBV and srCBV from the low- and intermediate-FA methods were compared using the Pearson's, Spearman's and intraclass correlation coefficients (ICC). Results Twenty-three HGG patients composing a total of 31 scans were analyzed. The Pearson and Spearman correlations and ICCs between the 30°/P-/C+ and 60°/P+/C+ acquisitions demonstrated high correlations for both mean and median nrCBV and srCBV. Conclusion Our study provides preliminary evidence that for HGG patients at 1.5T MRI, a low FA, no preload DSC-MRI acquisition can be an appealing alternative to the reference standard higher FA acquisition that utilizes a preload.
Collapse
Affiliation(s)
- Mark S. Shiroishi
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Marina del Rey, CA, United States
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Dane Weinert
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Steven Y. Cen
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Bino Varghese
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | | | - Melissa Prah
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jesse Mendoza
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Sina Nazemi
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Nima Ameli
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Negin Amini
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Salman Shohas
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Shannon Chen
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Bavrina Bigjahan
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Gabriel Zada
- Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Thomas Chen
- Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Josh Neman-Ebrahim
- Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Eric L. Chang
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Frances E. Chow
- Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Zhaoyang Fan
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Wensha Yang
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Frank J. Attenello
- Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Jason Ye
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Paul E. Kim
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Vishal N. Patel
- Department of Radiology, Mayo Clinic, Jacksonville, FL, United States
| | - Alexander Lerner
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Jay Acharya
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Leland S. Hu
- Department of Radiology, Mayo Clinic, Phoenix, AZ, United States
| | - C. Chad Quarles
- Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jerrold L. Boxerman
- Department of Diagnostic Imaging, The Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Ona Wu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Kathleen M. Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
11
|
Laudicella R, Mantarro C, Catalfamo B, Alongi P, Gaeta M, Minutoli F, Baldari S, Bisdas S. PET Imaging in Gliomas. RADIOLOGY‐NUCLEAR MEDICINE DIAGNOSTIC IMAGING 2023:194-218. [DOI: 10.1002/9781119603627.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Boxerman JL, Snyder BS, Barboriak DP, Schmainda KM. Early post-bevacizumab change in rCBV from DSC-MRI identifies pseudoresponse in recurrent glioblastoma: Results from ACRIN 6677/RTOG 0625. Front Oncol 2023; 13:1061502. [PMID: 36776298 PMCID: PMC9909012 DOI: 10.3389/fonc.2023.1061502] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/02/2023] [Indexed: 01/27/2023] Open
Abstract
Background Progressive enhancement predicted poor survival in ACRIN 6677/RTOG 0625, a multi-center trial of bevacizumab with irinotecan or temozolomide in recurrent glioblastoma, but pseudoresponse likely limited enhancement-based survival prognostication in T1 non-progressors. We aimed to determine whether early change in cerebral blood volume from baseline (ΔCBV) could further stratify the T1 non-progressors according to overall (OS) and progression-free (PFS) survival. Methods 37/123 enrolled patients had DSC-MRI, including 13, 15, and 8 patients without 2D-T1 progression at 2, 8, and 16 weeks post-treatment initiation, respectively. Mean CBV normalized to white matter (nRCBV) and mean standardized CBV (sRCBV) were extracted from enhancing tumor. ROC curves were derived for ΔCBV using six-month PFS and one-year OS as reference standards. Kaplan-Meier survival estimates and log-rank test compared PFS and OS for both ΔCBV (increase vs. decrease) and T1 response status (stable vs. decreasing enhancement). Results PFS and OS were significantly worse for increasing CBV at 2 weeks (p=0.003 and p=0.002 for nRCBV, and p=0.03 and p=0.03 for sRCBV, respectively), but not for 2D-T1 patients with stable vs. decreasing enhancement (p=0.44 and p=0.86, respectively). ΔCBV at week 2 was also a good prognostic marker for OS-1 and PFS-6 using ROC analysis. By contrast, 2D-T1 response status at weeks 2, 8, and 16 was not associated with PFS-6. ΔCBV at 16 weeks (p=0.008 for sRCBV) but not 8 weeks (p=0.74 for nRCBV and p=0.56 for sRCBV) was associated with significant difference in median survival, but no difference in survival was observed for 2D-T1 patients with stable vs. decreasing enhancement at 8 weeks (p=0.69) or 16 weeks (p=0.21). At 16 weeks, OS did not differ significantly between 2D-T1 progressors and 2D-T1 non-progressors with increasing CBV (median survival 3.3 months post week 16 scan vs. 9.2 months, respectively; p=0.13), suggesting that 2D-T1 non-progressors with increasing CBV may have a prognosis like that of 2D-T1 progressors. Conclusion After 2 weeks of anti-angiogenic therapy, ΔCBV in 2D-T1 non-progressors significantly prognosticated PFS and OS, whereas 2D-T1 response status did not, identifying a subpopulation that benefits from bevacizumab. Combining 2D-T1 progression and ΔCBV may yield a response assessment paradigm with 3-tiered OS stratification.
Collapse
Affiliation(s)
- Jerrold L. Boxerman
- Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, United States
| | - Bradley S. Snyder
- Center for Statistical Sciences, Brown University School of Public Health, Providence, RI, United States
| | - Daniel P. Barboriak
- Department of Radiology, Duke University Medical Center, Durham, NC, United States
| | - Kathleen M. Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
13
|
Fu S, Li L, Li X, Wu Q, Wang X, Huang Y, Hu H, Cao D. Case report: Long-term partial response of apatinib plus paclitaxel as second-line therapy in a patient with metastatic gastric cancer. Front Pharmacol 2022; 13:888106. [PMID: 36034835 PMCID: PMC9400896 DOI: 10.3389/fphar.2022.888106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/28/2022] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer is the second most prevalent cancer and the second leading cause of cancer-related death in China. The prognosis of metastatic gastric cancer is poor with a median overall survival of 8–10 months. Apatinib, an oral small-molecule, selective vascular endothelial growth factor receptor-2 tyrosine kinase inhibitor, is approved as third-line or subsequent therapy for gastric cancer in China. Several recent small-scale studies and case reports showed that it may be great help in improvement of prognosis as second-line treatment in patients with advanced or metastatic gastric cancer. Here, we present a case of advanced gastric adenocarcinoma with multiple hepatic metastases who was treated with apatinib plus paclitaxel as second-line therapy, realized a long progression-free survival of 37 months. Until 29 January 2022, the disease remains an efficacy of partial response. We believe that the good outcome of this case is not an accident, because of the typically hyper-vascular of his liver metastases, the treatment toxicities of hypertension and proteinuria, all may be potential predictive biomarkers for anti-angiogenic treatments.
Collapse
Affiliation(s)
- Shengya Fu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, SC, China
- Second Department of Oncology, Sichuan Friendship Hospital, Chengdu, SC, China
| | - Linjuan Li
- Department of Abdominal Oncology, West China Hospital, Sichuan University/ West China School of Nursing, Sichuan University, Chengdu, SC, China
| | - Xiaofen Li
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Qiang Wu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Xiaohui Wang
- Second Department of Oncology, Sichuan Friendship Hospital, Chengdu, SC, China
| | - Yan Huang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Haoyue Hu
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Medicine School of University of Electronic Science and Technology, Chengdu, SC, China
| | - Dan Cao
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, SC, China
- *Correspondence: Dan Cao,
| |
Collapse
|
14
|
Withey SB, MacPherson L, Oates A, Powell S, Novak J, Abernethy L, Pizer B, Grundy R, Morgan PS, Bailey S, Mitra D, Arvanitis TN, Auer DP, Avula S, Peet AC. Dynamic susceptibility-contrast magnetic resonance imaging with contrast agent leakage correction aids in predicting grade in pediatric brain tumours: a multicenter study. Pediatr Radiol 2022; 52:1134-1149. [PMID: 35290489 PMCID: PMC9107460 DOI: 10.1007/s00247-021-05266-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 08/31/2021] [Accepted: 12/11/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Relative cerebral blood volume (rCBV) measured using dynamic susceptibility-contrast MRI can differentiate between low- and high-grade pediatric brain tumors. Multicenter studies are required for translation into clinical practice. OBJECTIVE We compared leakage-corrected dynamic susceptibility-contrast MRI perfusion parameters acquired at multiple centers in low- and high-grade pediatric brain tumors. MATERIALS AND METHODS Eighty-five pediatric patients underwent pre-treatment dynamic susceptibility-contrast MRI scans at four centers. MRI protocols were variable. We analyzed data using the Boxerman leakage-correction method producing pixel-by-pixel estimates of leakage-uncorrected (rCBVuncorr) and corrected (rCBVcorr) relative cerebral blood volume, and the leakage parameter, K2. Histological diagnoses were obtained. Tumors were classified by high-grade tumor. We compared whole-tumor median perfusion parameters between low- and high-grade tumors and across tumor types. RESULTS Forty tumors were classified as low grade, 45 as high grade. Mean whole-tumor median rCBVuncorr was higher in high-grade tumors than low-grade tumors (mean ± standard deviation [SD] = 2.37±2.61 vs. -0.14±5.55; P<0.01). Average median rCBV increased following leakage correction (2.54±1.63 vs. 1.68±1.36; P=0.010), remaining higher in high-grade tumors than low grade-tumors. Low-grade tumors, particularly pilocytic astrocytomas, showed T1-dominant leakage effects; high-grade tumors showed T2*-dominance (mean K2=0.017±0.049 vs. 0.002±0.017). Parameters varied with tumor type but not center. Median rCBVuncorr was higher (mean = 1.49 vs. 0.49; P=0.015) and K2 lower (mean = 0.005 vs. 0.016; P=0.013) in children who received a pre-bolus of contrast agent compared to those who did not. Leakage correction removed the difference. CONCLUSION Dynamic susceptibility-contrast MRI acquired at multiple centers helped distinguish between children's brain tumors. Relative cerebral blood volume was significantly higher in high-grade compared to low-grade tumors and differed among common tumor types. Vessel leakage correction is required to provide accurate rCBV, particularly in low-grade enhancing tumors.
Collapse
Affiliation(s)
- Stephanie B Withey
- RRPPS, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Oncology, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Lesley MacPherson
- Radiology, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Adam Oates
- Radiology, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Stephen Powell
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Jan Novak
- Oncology, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Department of Psychology, Aston Brain Centre, School of Life and Health Sciences, Aston University, Birmingham, UK
| | | | - Barry Pizer
- Oncology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Richard Grundy
- The Children's Brain Tumour Research Centre, University of Nottingham, Nottingham, UK
| | - Paul S Morgan
- The Children's Brain Tumour Research Centre, University of Nottingham, Nottingham, UK
- Medical Physics, Nottingham University Hospitals, Nottingham, UK
- Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK
| | - Simon Bailey
- Sir James Spence Institute of Child Health, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Dipayan Mitra
- Neuroradiology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Theodoros N Arvanitis
- Oncology, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Institute of Digital Healthcare, WMG, University of Warwick, Coventry, UK
| | - Dorothee P Auer
- Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK
- Neuroradiology, Nottingham University Hospitals Trust, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham, UK
| | - Shivaram Avula
- Radiology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Andrew C Peet
- Oncology, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK.
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK.
- Children's Brain Tumour Research Team, 4th Floor Institute of Child Health, Birmingham Women's and Children's Hospital NHS Foundation Trust, Steelhouse Lane, Birmingham, B4 6NH, UK.
| |
Collapse
|
15
|
Malik DG, Rath TJ, Urcuyo Acevedo JC, Canoll PD, Swanson KR, Boxerman JL, Quarles CC, Schmainda KM, Burns TC, Hu LS. Advanced MRI Protocols to Discriminate Glioma From Treatment Effects: State of the Art and Future Directions. FRONTIERS IN RADIOLOGY 2022; 2:809373. [PMID: 37492687 PMCID: PMC10365126 DOI: 10.3389/fradi.2022.809373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/01/2022] [Indexed: 07/27/2023]
Abstract
In the follow-up treatment of high-grade gliomas (HGGs), differentiating true tumor progression from treatment-related effects, such as pseudoprogression and radiation necrosis, presents an ongoing clinical challenge. Conventional MRI with and without intravenous contrast serves as the clinical benchmark for the posttreatment surveillance imaging of HGG. However, many advanced imaging techniques have shown promise in helping better delineate the findings in indeterminate scenarios, as posttreatment effects can often mimic true tumor progression on conventional imaging. These challenges are further confounded by the histologic admixture that can commonly occur between tumor growth and treatment-related effects within the posttreatment bed. This review discusses the current practices in the surveillance imaging of HGG and the role of advanced imaging techniques, including perfusion MRI and metabolic MRI.
Collapse
Affiliation(s)
- Dania G. Malik
- Department of Radiology, Mayo Clinic, Phoenix, AZ, United States
| | - Tanya J. Rath
- Department of Radiology, Mayo Clinic, Phoenix, AZ, United States
| | - Javier C. Urcuyo Acevedo
- Mathematical Neurooncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, United States
| | - Peter D. Canoll
- Departments of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Kristin R. Swanson
- Mathematical Neurooncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, United States
| | - Jerrold L. Boxerman
- Department of Diagnostic Imaging, Brown University, Providence, RI, United States
| | - C. Chad Quarles
- Department of Neuroimaging Research & Barrow Neuroimaging Innovation Center, Barrow Neurologic Institute, Phoenix, AZ, United States
| | - Kathleen M. Schmainda
- Department of Biophysics & Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Terry C. Burns
- Departments of Neurologic Surgery and Neuroscience, Mayo Clinic, Rochester, MN, United States
| | - Leland S. Hu
- Department of Radiology, Mayo Clinic, Phoenix, AZ, United States
- Mathematical Neurooncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, United States
| |
Collapse
|
16
|
Luo X, Zhu Y, Zhang Y, Zhang Q, Wang X, Deng X. Parameters of MR perfusion-weighted imaging predict the response and prognosis to high-dose methotrexate-based chemotherapy in immunocompetent patients with primary central nervous system lymphoma. J Clin Neurosci 2022; 95:151-158. [DOI: 10.1016/j.jocn.2021.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 11/26/2021] [Accepted: 12/07/2021] [Indexed: 01/04/2023]
|
17
|
Stokes AM, Bergamino M, Alhilali L, Hu LS, Karis JP, Baxter LC, Bell LC, Quarles CC. Evaluation of single bolus, dual-echo dynamic susceptibility contrast MRI protocols in brain tumor patients. J Cereb Blood Flow Metab 2021; 41:3378-3390. [PMID: 34415211 PMCID: PMC8669280 DOI: 10.1177/0271678x211039597] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Relative cerebral blood volume (rCBV) obtained from dynamic susceptibility contrast (DSC) MRI is adversely impacted by contrast agent leakage in brain tumors. Using simulations, we previously demonstrated that multi-echo DSC-MRI protocols provide improvements in contrast agent dosing, pulse sequence flexibility, and rCBV accuracy. The purpose of this study is to assess the in-vivo performance of dual-echo acquisitions in patients with brain tumors (n = 59). To verify pulse sequence flexibility, four single-dose dual-echo acquisitions were tested with variations in contrast agent dose, flip angle, and repetition time, and the resulting dual-echo rCBV was compared to standard single-echo rCBV obtained with preload (double-dose). Dual-echo rCBV was comparable to standard double-dose single-echo protocols (mean (standard deviation) tumor rCBV 2.17 (1.28) vs. 2.06 (1.20), respectively). High rCBV similarity was observed (CCC = 0.96), which was maintained across both flip angle (CCC = 0.98) and repetition time (CCC = 0.96) permutations, demonstrating that dual-echo acquisitions provide flexibility in acquisition parameters. Furthermore, a single dual-echo acquisition was shown to enable quantification of both perfusion and permeability metrics. In conclusion, single-dose dual-echo acquisitions provide similar rCBV to standard double-dose single-echo acquisitions, suggesting contrast agent dose can be reduced while providing significant pulse sequence flexibility and complementary tumor perfusion and permeability metrics.
Collapse
Affiliation(s)
- Ashley M Stokes
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Maurizio Bergamino
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Lea Alhilali
- Neuroradiology, Southwest Neuroimaging at Barrow Neurological Institute, Phoenix, AZ, USA
| | - Leland S Hu
- Department of Radiology, Division of Neuroradiology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - John P Karis
- Neuroradiology, Southwest Neuroimaging at Barrow Neurological Institute, Phoenix, AZ, USA
| | - Leslie C Baxter
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, USA.,Department of Radiology, Division of Neuroradiology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Laura C Bell
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, USA
| | - C Chad Quarles
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW This review aims to cover current MRI techniques for assessing treatment response in brain tumors, with a focus on radio-induced lesions. RECENT FINDINGS Pseudoprogression and radionecrosis are common radiological entities after brain tumor irradiation and are difficult to distinguish from real progression, with major consequences on daily patient care. To date, shortcomings of conventional MRI have been largely recognized but morphological sequences are still used in official response assessment criteria. Several complementary advanced techniques have been proposed but none of them have been validated, hampering their clinical use. Among advanced MRI, brain perfusion measures increase diagnostic accuracy, especially when added with spectroscopy and susceptibility-weighted imaging. However, lack of reproducibility, because of several hard-to-control variables, is still a major limitation for their standardization in routine protocols. Amide Proton Transfer is an emerging molecular imaging technique that promises to offer new metrics by indirectly quantifying intracellular mobile proteins and peptide concentration. Preliminary studies suggest that this noncontrast sequence may add key biomarkers in tumor evaluation, especially in posttherapeutic settings. SUMMARY Benefits and pitfalls of conventional and advanced imaging on posttreatment assessment are discussed and the potential added value of APT in this clinicoradiological evolving scenario is introduced.
Collapse
Affiliation(s)
- Lucia Nichelli
- Department of Neuroradiology, Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière-Charles Foix
- Sorbonne Université, INSERM, CNRS, Assistance Publique-Hôpitaux de Paris, Institut du Cerveau et de la Moelle épinière, boulevard de l’Hôpital, Paris
| | - Stefano Casagranda
- Department of Research & Innovation, Olea Medical, avenue des Sorbiers, La Ciotat, France
| |
Collapse
|
19
|
Wang Y, Chen W, Shi Y, Yan C, Kong Z, Wang Y, Wang Y, Ma W. Imposing Phase II and Phase III Clinical Trials of Targeted Drugs for Glioblastoma: Current Status and Progress. Front Oncol 2021; 11:719623. [PMID: 34568049 PMCID: PMC8458950 DOI: 10.3389/fonc.2021.719623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/24/2021] [Indexed: 12/21/2022] Open
Abstract
The most common primary intracranial tumor is glioma, among which glioblastoma (GBM) has the worst prognosis. Because of the high degree of malignancy of GBM and frequent recurrence after surgery, postoperative therapy, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy, is particularly important. A wide variety of targeted drugs have undergone phase III clinical trials for patients with GBM, but these drugs do not work for all patients, and few patients in these trials have prolonged overall survival. In this review, some imposing phase III clinical trials of targeted drugs for glioma are introduced, and some prospective phase II clinical trials that have been completed or are in progress are summarized. In addition, the mechanisms of these drugs are briefly introduced, and deficiencies of these clinical trials are analyzed. This review aims to provide a comprehensive overview of current research on targeted drugs for glioma to clarify future research directions.
Collapse
Affiliation(s)
- Yaning Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqi Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yixin Shi
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chengrui Yan
- Department of Neurosurgery, Peking University International Hospital, Beijing, China
| | - Ziren Kong
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuekun Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
20
|
Mohan S, Wang S, Chawla S, Abdullah K, Desai A, Maloney E, Brem S. Multiparametric MRI assessment of response to convection-enhanced intratumoral delivery of MDNA55, an interleukin-4 receptor targeted immunotherapy, for recurrent glioblastoma. Surg Neurol Int 2021; 12:337. [PMID: 34345478 PMCID: PMC8326072 DOI: 10.25259/sni_353_2021] [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] [Received: 04/11/2021] [Accepted: 06/09/2021] [Indexed: 11/04/2022] Open
Abstract
Background Glioblastoma (GBM) is the most common malignant brain tumor and carries a dismal prognosis. Attempts to develop biologically targeted therapies are challenging as the blood-brain barrier can limit drugs from reaching their target when administered through conventional (intravenous or oral) routes. Furthermore, systemic toxicity of drugs often limits their therapeutic potential. To circumvent these problems, convection-enhanced delivery (CED) provides direct, targeted, intralesional therapy with a secondary objective to alter the tumor microenvironment from an immunologically "cold" (nonresponsive) to an "inflamed" (immunoresponsive) tumor. Case Description We report a patient with right occipital recurrent GBM harboring poor prognostic genotypes who was treated with MRI-guided CED of a fusion protein MDNA55 (a targeted toxin directed toward the interleukin-4 receptor). The patient underwent serial anatomical, diffusion, and perfusion MRI scans before initiation of targeted therapy and at 1, 3-month posttherapy. Increased mean diffusivity along with decreased fractional anisotropy and maximum relative cerebral blood volume was noted at follow-up periods relative to baseline. Conclusion Our findings suggest that diffusion and perfusion MRI techniques may be useful in evaluating early response to CED of MDNA55 in recurrent GBM patients.
Collapse
Affiliation(s)
- Suyash Mohan
- Department of Radiology, Division of Neuroradiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Sumei Wang
- Department of Radiology, Division of Neuroradiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Sanjeev Chawla
- Department of Radiology, Division of Neuroradiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Kalil Abdullah
- Department of Neurosurgery, University of Texas-Southwestern Medical Center, Dallas, Texas, United States
| | - Arati Desai
- Department of Medicine Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Eileen Maloney
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
21
|
Durgin JS, Henderson F, Nasrallah MP, Mohan S, Wang S, Lacey SF, Melenhorst JJ, Desai AS, Lee JYK, Maus MV, June CH, Brem S, O'Connor RS, Binder Z, O'Rourke DM. Case Report: Prolonged Survival Following EGFRvIII CAR T Cell Treatment for Recurrent Glioblastoma. Front Oncol 2021; 11:669071. [PMID: 34026647 PMCID: PMC8138201 DOI: 10.3389/fonc.2021.669071] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/07/2021] [Indexed: 12/01/2022] Open
Abstract
Autologous chimeric antigen receptor (CAR) T cells targeted to epidermal growth factor receptor variant III (CAR T-EGFRvIII) have been developed and administered experimentally to treat patients with IDH1 wildtype recurrent glioblastoma (rGBM) (NCT02209376). We report the case of a 59-year-old patient who received a single peripheral infusion of CAR T-EGFRvIII cells and survived 36 months after disease recurrence, exceeding expected survival for recurrent glioblastoma. Post-infusion histopathologic analysis of tissue obtained during a second stage surgical resection revealed immunosuppressive adaptive changes in the tumor tissue as well as reduced EGFRvIII expression. Serial brain imaging demonstrated a significant reduction in relative cerebral blood volume (rCBV), a measure strongly associated with tumor proliferative activity, at early time points following CAR T treatment. Notably, CAR T-EGFRvIII cells persisted in her peripheral circulation during 29 months of follow-up, the longest period of CAR T persistence reported in GBM trials to date. These findings in a long-term survivor show that peripherally administered CAR T-EGFRvIII cells can persist for years in the circulation and suggest that this cell therapy approach could be optimized to achieve broader efficacy in recurrent GBM patients.
Collapse
Affiliation(s)
- Joseph S Durgin
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.,Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pathology & Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Fraser Henderson
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, United States
| | - MacLean P Nasrallah
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.,Department of Pathology & Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Suyash Mohan
- Department of Radiology, Division of Neuroradiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Sumei Wang
- Department of Radiology, Division of Neuroradiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Simon F Lacey
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, United States
| | - Jan Joseph Melenhorst
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.,Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pathology & Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Arati S Desai
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.,Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - John Y K Lee
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, United States
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pathology & Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Steven Brem
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.,Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Roddy S O'Connor
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pathology & Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Zev Binder
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.,Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Donald M O'Rourke
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.,Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
22
|
Schmainda KM, Prah MA, Marques H, Kim E, Barboriak DP, Boxerman JL. Value of dynamic contrast perfusion MRI to predict early response to bevacizumab in newly diagnosed glioblastoma: results from ACRIN 6686 multicenter trial. Neuro Oncol 2021; 23:314-323. [PMID: 32678438 PMCID: PMC7906067 DOI: 10.1093/neuonc/noaa167] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In Radiation Therapy Oncology Group (RTOG) 0825, a phase III trial of standard therapy with bevacizumab or without (placebo) in newly diagnosed glioblastoma, 44 patients underwent dynamic contrast enhanced (DCE) and/or dynamic susceptibility contrast (DSC) MRI in the American College of Radiology Imaging Network (ACRIN) trial 6686. The association between early changes in relative cerebral blood volume (rCBV) and volume transfer constant (Ktrans) with overall survival (OS) was evaluated. METHODS MRI was performed at postop baseline (S0), immediately before (S1), 1 day after (S2), and 7 weeks after (S3) bevacizumab or placebo initiation. Mean normalized and standardized rCBV (nRCBV, sRCBV) and Ktrans were measured within contrast-enhancing lesion. Wilcoxon rank sum tests compared parameter changes from S1-S2 and S1-S3. Association with OS and progression-free survival (PFS) were determined using Kaplan-Meier and log-rank tests. Treatment response for groups stratified by pretreatment nRCBV (S0, S1) was explored. The intraclass correlation coefficient and repeatability coefficient for the placebo arm (S1-S2) were used to assess repeatability. RESULTS Evaluable were 27-36 datasets per time point. Significant differences between treatment arms were found for changes in nRCBV and sRCBV from S1-S2 and S1-S3, and in Ktrans for S1-S3. Improved PFS (P = 0.05) but not OS (P = 0.46) was observed. High pretreatment rCBV predicted improved OS for bevacizumab-treated patients. Based on the intraclass correlation coefficient, sRCBV (0.92) was more repeatable than nRCBV (0.71) and Ktrans (0.75), consistent with repeatability coefficient values. CONCLUSIONS Bevacizumab significantly changes rCBV but not Ktrans as early as 1 day posttreatment in newly diagnosed glioblastoma unrelated to outcomes. Improvements in clinical trial design to maximize rCBV benefit are indicated.
Collapse
Affiliation(s)
- Kathleen M Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Melissa A Prah
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Helga Marques
- Department of Biostatistics and Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
| | - Eunhee Kim
- Merck Research Laboratories, Kenilworth, New Jersey
| | | | - Jerrold L Boxerman
- Department of Diagnostic Imaging, Rhode Island Hospital, and Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
23
|
Song J, Kadaba P, Kravitz A, Hormigo A, Friedman J, Belani P, Hadjipanayis C, Ellingson BM, Nael K. Multiparametric MRI for early identification of therapeutic response in recurrent glioblastoma treated with immune checkpoint inhibitors. Neuro Oncol 2021; 22:1658-1666. [PMID: 32193547 DOI: 10.1093/neuonc/noaa066] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Physiologic changes quantified by diffusion and perfusion MRI have shown utility in predicting treatment response in glioblastoma (GBM) patients treated with cytotoxic therapies. We aimed to investigate whether quantitative changes in diffusion and perfusion after treatment by immune checkpoint inhibitors (ICIs) would determine 6-month progression-free survival (PFS6) in patients with recurrent GBM. METHODS Inclusion criteria for this retrospective study were: (i) diagnosis of recurrent GBM treated with ICIs and (ii) availability of diffusion and perfusion in pre and post ICI MRI (iii) at ≥6 months follow-up from treatment. After co-registration, mean values of the relative apparent diffusion coefficient (rADC), Ktrans (volume transfer constant), Ve (extravascular extracellular space volume) and Vp (plasma volume), and relative cerebral blood volume (rCBV) were calculated from a volume-of-interest of the enhancing tumor. Final assignment of stable/improved versus progressive disease was determined on 6-month follow-up using modified Response Assessment in Neuro-Oncology criteria. RESULTS Out of 19 patients who met inclusion criteria and follow-up (mean ± SD: 7.8 ± 1.4 mo), 12 were determined to have tumor progression, while 7 had treatment response after 6 months of ICI treatment. Only interval change of rADC was suggestive of treatment response. Patients with treatment response (6/7: 86%) had interval increased rADC, while 11/12 (92%) with tumor progression had decreased rADC (P = 0.001). Interval change in rCBV, Ktrans, Vp, and Ve were not indicative of treatment response within 6 months. CONCLUSIONS In patients with recurrent GBM, interval change in rADC is promising in assessing treatment response versus progression within the first 6 months following ICI treatment. KEY POINTS • In recurrent GBM treated with ICIs, interval change in rADC suggests early treatment response.• Interval change in rADC can be used as an imaging biomarker to determine PFS6.• Interval change in MR perfusion and permeability measures do not suggest ICI treatment response.
Collapse
Affiliation(s)
- Joseph Song
- Icahn School of Medicine at Mount Sinai, Department of Radiology (Neuroimaging Advanced and Exploratory Lab), New York, New York
| | - Priyanka Kadaba
- Icahn School of Medicine at Mount Sinai, Department of Radiology (Neuroimaging Advanced and Exploratory Lab), New York, New York
| | - Amanda Kravitz
- Icahn School of Medicine at Mount Sinai, Department of Radiology (Neuroimaging Advanced and Exploratory Lab), New York, New York
| | - Adilia Hormigo
- Department of Neurology, Medicine (Div Hem Onc), The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joshua Friedman
- Department of Neurology, Medicine (Div Hem Onc), The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Puneet Belani
- Icahn School of Medicine at Mount Sinai, Department of Radiology (Neuroimaging Advanced and Exploratory Lab), New York, New York
| | | | - 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
| | - Kambiz Nael
- 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
|
24
|
Al Feghali KA, Randall JW, Liu DD, Wefel JS, Brown PD, Grosshans DR, McAvoy SA, Farhat MA, Li J, McGovern SL, McAleer MF, Ghia AJ, Paulino AC, Sulman EP, Penas-Prado M, Wang J, de Groot J, Heimberger AB, Armstrong TS, Gilbert MR, Mahajan A, Guha-Thakurta N, Chung C. Phase II trial of proton therapy versus photon IMRT for GBM: secondary analysis comparison of progression-free survival between RANO versus clinical assessment. Neurooncol Adv 2021; 3:vdab073. [PMID: 34337411 PMCID: PMC8320688 DOI: 10.1093/noajnl/vdab073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND This secondary image analysis of a randomized trial of proton radiotherapy (PT) versus photon intensity-modulated radiotherapy (IMRT) compares tumor progression based on clinical radiological assessment versus Response Assessment in Neuro-Oncology (RANO). METHODS Eligible patients were enrolled in the randomized trial and had MR imaging at baseline and follow-up beyond 12 weeks from completion of radiotherapy. "Clinical progression" was based on a clinical radiology report of progression and/or change in treatment for progression. RESULTS Of 90 enrolled patients, 66 were evaluable. Median clinical progression-free survival (PFS) was 10.8 (range: 9.4-14.7) months; 10.8 months IMRT versus 11.2 months PT (P = .14). Median RANO-PFS was 8.2 (range: 6.9, 12): 8.9 months IMRT versus 6.6 months PT (P = .24). RANO-PFS was significantly shorter than clinical PFS overall (P = .001) and for both the IMRT (P = .01) and PT (P = .04) groups. There were 31 (46.3%) discrepant cases of which 17 had RANO progression more than a month prior to clinical progression, and 14 had progression by RANO but not clinical criteria. CONCLUSIONS Based on this secondary analysis of a trial of PT versus IMRT for glioblastoma, while no difference in PFS was noted relative to treatment technique, RANO criteria identified progression more often and earlier than clinical assessment. This highlights the disconnect between measures of tumor response in clinical trials versus clinical practice. With growing efforts to utilize real-world data and personalized treatment with timely adaptation, there is a growing need to improve the consistency of determining tumor progression within clinical trials and clinical practice.
Collapse
Affiliation(s)
- Karine A Al Feghali
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - James W Randall
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Diane D Liu
- Department of Biostatistics, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey S Wefel
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - David R Grosshans
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sarah A McAvoy
- Department of Radiation Oncology, University of Maryland, Baltimore, Maryland, USA
| | - Maguy A Farhat
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jing Li
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Susan L McGovern
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Mary F McAleer
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Amol J Ghia
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Arnold C Paulino
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Erik P Sulman
- Department of Radiation Oncology, NYU Langone, New York, New York, USA
| | - Marta Penas-Prado
- Department of Neuro-Oncology, National Institutes of Health, Bethesda, Maryland, USA
| | - Jihong Wang
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - John de Groot
- Department of Neuro-Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Amy B Heimberger
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Terri S Armstrong
- Department of Neuro-Oncology, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark R Gilbert
- Department of Neuro-Oncology, National Institutes of Health, Bethesda, Maryland, USA
| | - Anita Mahajan
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Caroline Chung
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
25
|
Boxerman JL, Quarles CC, Hu LS, Erickson BJ, Gerstner ER, Smits M, Kaufmann TJ, Barboriak DP, Huang RH, Wick W, Weller M, Galanis E, Kalpathy-Cramer J, Shankar L, Jacobs P, Chung C, van den Bent MJ, Chang S, Al Yung WK, Cloughesy TF, Wen PY, Gilbert MR, Rosen BR, Ellingson BM, Schmainda KM. Consensus recommendations for a dynamic susceptibility contrast MRI protocol for use in high-grade gliomas. Neuro Oncol 2020; 22:1262-1275. [PMID: 32516388 PMCID: PMC7523451 DOI: 10.1093/neuonc/noaa141] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Despite the widespread clinical use of dynamic susceptibility contrast (DSC) MRI, DSC-MRI methodology has not been standardized, hindering its utilization for response assessment in multicenter trials. Recently, the DSC-MRI Standardization Subcommittee of the Jumpstarting Brain Tumor Drug Development Coalition issued an updated consensus DSC-MRI protocol compatible with the standardized brain tumor imaging protocol (BTIP) for high-grade gliomas that is increasingly used in the clinical setting and is the default MRI protocol for the National Clinical Trials Network. After reviewing the basis for controversy over DSC-MRI protocols, this paper provides evidence-based best practices for clinical DSC-MRI as determined by the Committee, including pulse sequence (gradient echo vs spin echo), BTIP-compliant contrast agent dosing (preload and bolus), flip angle (FA), echo time (TE), and post-processing leakage correction. In summary, full-dose preload, full-dose bolus dosing using intermediate (60°) FA and field strength-dependent TE (40-50 ms at 1.5 T, 20-35 ms at 3 T) provides overall best accuracy and precision for cerebral blood volume estimates. When single-dose contrast agent usage is desired, no-preload, full-dose bolus dosing using low FA (30°) and field strength-dependent TE provides excellent performance, with reduced contrast agent usage and elimination of potential systematic errors introduced by variations in preload dose and incubation time.
Collapse
Affiliation(s)
- Jerrold L Boxerman
- Department of Diagnostic Imaging, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Representative of the Eastern Cooperative Oncology Group–American College of Radiology Imaging Network (ECOG-ACRIN) Cancer Research Group
- Representative of the American Society of Neuroradiology (ASNR)
- Representative of the American Society of Functional Neuroradiology (ASFNR)
| | - Chad C Quarles
- Department of Neuroimaging Research and Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Leland S Hu
- Department of Radiology, Mayo Clinic, Phoenix, Arizona, USA
- Representative of the Alliance for Clinical Trials in Oncology
- Representative of the American Society of Neuroradiology (ASNR)
| | - Bradley J Erickson
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Representative of the Alliance for Clinical Trials in Oncology
- Representative of the RSNA Quantitative Imaging Biomarker Alliance (QIBA)
- Representative of the American Society of Neuroradiology (ASNR)
| | - Elizabeth R Gerstner
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Representative of the Adult Brain Tumor Consortium (ABTC)
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC–University Medical Center Rotterdam, Rotterdam, Netherlands
- Representative of the European Organisation for Research and Treatment of Cancer (EORTC)
| | - Timothy J Kaufmann
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Representative of the Alliance for Clinical Trials in Oncology
| | - Daniel P Barboriak
- Department of Radiology, Duke University School of Medicine, Durham, North Carolina, USA
- Representative of the Eastern Cooperative Oncology Group–American College of Radiology Imaging Network (ECOG-ACRIN) Cancer Research Group
- Representative of the RSNA Quantitative Imaging Biomarker Alliance (QIBA)
- Representative of the American Society of Neuroradiology (ASNR)
| | - Raymond H Huang
- Department of Radiology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Wolfgang Wick
- Department of Neurooncology, National Center of Tumor Disease, University Clinic Heidelberg, Heidelberg, Germany
- Representative of the European Organisation for Research and Treatment of Cancer (EORTC)
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
- Representative of the European Organisation for Research and Treatment of Cancer (EORTC)
| | - Evanthia Galanis
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
- Representative of the Alliance for Clinical Trials in Oncology
| | - Jayashree Kalpathy-Cramer
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lalitha Shankar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), Bethesda, Maryland, USA
| | - Paula Jacobs
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), Bethesda, Maryland, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Representative of the Alliance for Clinical Trials in Oncology
| | - Martin J van den Bent
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
- Representative of the European Organisation for Research and Treatment of Cancer (EORTC)
| | - Susan Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - W K Al Yung
- Department of Neuro-Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program and UCLA Brain Tumor Imaging Laboratory (BTIL), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
- Representative of the Adult Brain Tumor Consortium (ABTC)
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute (NCI), Bethesda, Maryland, USA
- Representative of the Radiation Therapy Oncology Group (RTOG)
| | - Bruce R Rosen
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin M Ellingson
- UCLA Neuro-Oncology Program and UCLA Brain Tumor Imaging Laboratory (BTIL), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Departments of Radiological Sciences, Psychiatry, and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Representative of the Adult Brain Tumor Consortium (ABTC)
- Representative of the Ivy Consortium for Early Phase Clinical Trials
- Representative of the Eastern Cooperative Oncology Group–American College of Radiology Imaging Network (ECOG-ACRIN) Cancer Research Group
- Representative of the RSNA Quantitative Imaging Biomarker Alliance (QIBA)
- Representative of the American Society of Neuroradiology (ASNR)
| | - Kathleen M Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Representative of the Eastern Cooperative Oncology Group–American College of Radiology Imaging Network (ECOG-ACRIN) Cancer Research Group
| |
Collapse
|
26
|
Magnetic resonance imaging evaluation of brain glioma before postoperative radiotherapy. Clin Transl Oncol 2020; 23:820-826. [PMID: 32857338 DOI: 10.1007/s12094-020-02474-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE To investigate the magnetic resonance imaging (MRI) images of brain glioma before postoperative radiotherapy, and to provide reference for the delineation of postoperative radiotherapy target area. METHODS Retrospective analysis was performed on 106 cases of brain glioma confirmed by surgery and pathology in our hospital, including 70 cases of high-grade glioma (HGG) and 36 cases of low-grade glioma (LGG). The MRI images of the lesions within 1 month before and after surgery were analyzed, the apparent diffusion coefficient (ADC) values in the near and far tumor areas were measured, respectively, and the corresponding rADC values were calculated. RESULTS The incidence of residual tumors of postoperative HGG and LGG was 0, 15.7% (0/36, 11/70), respectively. The incidence of postoperative reactive enhancement was 11.0% and 52.9% (4/36 and 37/70), respectively. About 30.6% and 81.4% (11/36 and 57/70) of patients with adjacent meningeal enhancement were found in the operative area. CONCLUSIONS The MRI images of HGG and LGG before postoperative radiotherapy had certain characteristics, providing a favorable guidance for the delineation of the target area of radiotherapy and the formulation of treatment plan.
Collapse
|
27
|
Kickingereder P, Brugnara G, Hansen MB, Nowosielski M, Pflüger I, Schell M, Isensee F, Foltyn M, Neuberger U, Kessler T, Sahm F, Wick A, Heiland S, Weller M, Platten M, von Deimling A, Maier-Hein KH, Østergaard L, van den Bent MJ, Gorlia T, Wick W, Bendszus M. Noninvasive Characterization of Tumor Angiogenesis and Oxygenation in Bevacizumab-treated Recurrent Glioblastoma by Using Dynamic Susceptibility MRI: Secondary Analysis of the European Organization for Research and Treatment of Cancer 26101 Trial. Radiology 2020; 297:164-175. [PMID: 32720870 DOI: 10.1148/radiol.2020200978] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Relevance of antiangiogenic treatment with bevacizumab in patients with glioblastoma is controversial because progression-free survival benefit did not translate into an overall survival (OS) benefit in randomized phase III trials. Purpose To perform longitudinal characterization of intratumoral angiogenesis and oxygenation by using dynamic susceptibility contrast agent-enhanced (DSC) MRI and evaluate its potential for predicting outcome from administration of bevacizumab. Materials and Methods In this secondary analysis of the prospective randomized phase II/III European Organization for Research and Treatment of Cancer 26101 trial conducted between October 2011 and December 2015 in 596 patients with first recurrence of glioblastoma, the subset of patients with availability of anatomic MRI and DSC MRI at baseline and first follow-up was analyzed. Patients were allocated into those administered bevacizumab (hereafter, the BEV group; either bevacizumab monotherapy or bevacizumab with lomustine) and those not administered bevacizumab (hereafter, the non-BEV group with lomustine monotherapy). Contrast-enhanced tumor volume, noncontrast-enhanced T2 fluid-attenuated inversion recovery (FLAIR) signal abnormality volume, Gaussian-normalized relative cerebral blood volume (nrCBV), Gaussian-normalized relative blood flow (nrCBF), and tumor metabolic rate of oxygen (nTMRO2) was quantified. The predictive ability of these imaging parameters was assessed with multivariable Cox regression and formal interaction testing. Results A total of 254 of 596 patients were evaluated (mean age, 57 years ± 11; 155 men; 161 in the BEV group and 93 in non-BEV group). Progression-free survival was longer in the BEV group (3.7 months; 95% confidence interval [CI]: 3.0, 4.2) compared with the non-BEV group (2.5 months; 95% CI: 1.5, 2.9; P = .01), whereas OS was not different (P = .15). The nrCBV decreased for the BEV group (-16.3%; interquartile range [IQR], -39.5% to 12.0%; P = .01), but not for the non-BEV group (1.2%; IQR, -17.9% to 23.3%; P = .19) between baseline and first follow-up. An identical pattern was observed for both nrCBF and nTMRO2 values. Contrast-enhanced tumor and noncontrast-enhanced T2 FLAIR signal abnormality volumes decreased for the BEV group (-66% [IQR, -83% to -35%] and -33% [IQR, -71% to -5%], respectively; P < .001 for both), whereas they increased for the non-BEV group (30% [IQR, -17% to 98%], P = .001; and 10% [IQR, -13% to 82%], P = .02, respectively) between baseline and first follow-up. None of the assessed MRI parameters were predictive for OS in the BEV group. Conclusion Bevacizumab treatment decreased tumor volumes, angiogenesis, and oxygenation, thereby reflecting its effectiveness for extending progression-free survival; however, these parameters were not predictive of overall survival (OS), which highlighted the challenges of identifying patients that derive an OS benefit from bevacizumab. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Dillon in this issue.
Collapse
Affiliation(s)
- Philipp Kickingereder
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Gianluca Brugnara
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Mikkel Bo Hansen
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Martha Nowosielski
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Irada Pflüger
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Marianne Schell
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Fabian Isensee
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Martha Foltyn
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Ulf Neuberger
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Tobias Kessler
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Felix Sahm
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Antje Wick
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Sabine Heiland
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Michael Weller
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Michael Platten
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Andreas von Deimling
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Klaus H Maier-Hein
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Leif Østergaard
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Martin J van den Bent
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Thierry Gorlia
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Wolfgang Wick
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| | - Martin Bendszus
- From the Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany (P.K., G.B., I.P., M.S., M.F., U.N., S.H., M.B.); Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University Hospital, Aarhus, Denmark (M.B.H., L.Ø.); Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany (M.N., T.K., A.W., W.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (M.N.); Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.I., K.H.M.H.); Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (T.K., W.W.); Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany (F.S., A.v.D.); Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany (F.S., A.v.D.); Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (M.W.); Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany (M.P.); Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany (K.H.M.H.); Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark (L.Ø.); Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands (M.J.v.d.B.); and European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium (T.G.)
| |
Collapse
|
28
|
Park JE, Kim HS, Park SY, Jung SC, Kim JH, Heo HY. Identification of Early Response to Anti-Angiogenic Therapy in Recurrent Glioblastoma: Amide Proton Transfer–weighted and Perfusion-weighted MRI compared with Diffusion-weighted MRI. Radiology 2020; 295:397-406. [DOI: 10.1148/radiol.2020191376] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Ji Eun Park
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| | - Ho Sung Kim
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| | - Seo Young Park
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| | - Seung Chai Jung
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| | - Jeong Hoon Kim
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| | - Hye-Young Heo
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., S.C.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea; and Department of Radiology, Johns Hopkins University, Baltimore, Md (H.Y.H.)
| |
Collapse
|
29
|
Park JE, Kim HS. [Current Applications and Future Perspectives of Brain Tumor Imaging]. TAEHAN YONGSANG UIHAKHOE CHI 2020; 81:467-487. [PMID: 36238631 PMCID: PMC9431910 DOI: 10.3348/jksr.2020.81.3.467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 11/29/2022]
Abstract
뇌종양의 진단 및 치료 반응 평가의 기본이 되는 영상기법은 해부학적 영상이다. 현재 임상에서 사용 가능한 영상기법들 중 확산 강조 영상 및 관류 영상이 추가적인 정보를 제공하고 있다. 최근에는 종양의 유전체 변이와 이질성 평가가 중요해지면서 라디오믹스와 딥러닝을 이용한 영상분석기법의 임상 응용이 기대되고 있다. 본 종설에서는 뇌종양 영상 임상 적용에서 여전히 중요한 해부학적 영상을 중심으로 한 자기공명영상 촬영 권고안, 최신 영상기법 중 확산 강조 영상 및 관류 영상의 기본 원리, 병태생리학적 배경 및 임상응용, 마지막으로 최근 컴퓨터 기술의 발전으로 많이 연구되고 있는 라디오믹스와 딥러닝의 뇌종양에서의 향후 활용가치에 대해 기술하고자 한다.
Collapse
|
30
|
Stokes AM, Semmineh NB, Nespodzany A, Bell LC, Quarles CC. Systematic assessment of multi-echo dynamic susceptibility contrast MRI using a digital reference object. Magn Reson Med 2019; 83:109-123. [PMID: 31400035 DOI: 10.1002/mrm.27914] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/14/2019] [Accepted: 07/02/2019] [Indexed: 11/07/2022]
Abstract
PURPOSE Brain tumor dynamic susceptibility contrast (DSC) MRI is adversely impacted by T1 and T 2 ∗ contrast agent leakage effects that result in inaccurate hemodynamic metrics. While multi-echo acquisitions remove T1 leakage effects, there is no consensus on the optimal set of acquisition parameters. Using a computational approach, we systematically evaluated a wide range of acquisition strategies to determine the optimal multi-echo DSC-MRI perfusion protocol. METHODS Using a population-based DSC-MRI digital reference object (DRO), we assessed the influence of preload dosing (no preload and full dose preload), field strength (1.5 and 3T), pulse sequence parameters (echo time, repetition time, and flip angle), and leakage correction on relative cerebral blood volume (rCBV) and flow (rCBF) accuracy. We also compared multi-echo DSC-MRI protocols with standard single-echo protocols. RESULTS Multi-echo DSC-MRI is highly consistent across all protocols, and multi-echo rCBV (with or without use of a preload dose) had higher accuracy than single-echo rCBV. Regression analysis showed that choice of repetition time and flip angle had minimal impact on multi-echo rCBV and rCBV, indicating the potential for significant flexibility in acquisition parameters. The echo time combination had minimal impact on rCBV, though longer echo times should be avoided, particularly at higher field strengths. Leakage correction improved rCBV accuracy in all cases. Multi-echo rCBF was less biased than single-echo rCBF, although rCBF accuracy was reduced overall relative to rCBV. CONCLUSIONS Multi-echo acquisitions were more robust than single-echo, essentially decoupling both repetition time and flip angle from rCBV accuracy. Multi-echo acquisitions obviate the need for preload dosing, although leakage correction to remove residual T 2 ∗ leakage effects remains compulsory for high rCBV accuracy.
Collapse
Affiliation(s)
- Ashley M Stokes
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona
| | - Natenael B Semmineh
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona
| | - Ashley Nespodzany
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona
| | - Laura C Bell
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona
| | - C Chad Quarles
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona
| |
Collapse
|
31
|
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
|
32
|
Correlation of radiological and immunochemical parameters with clinical outcome in patients with recurrent glioblastoma treated with Bevacizumab. Clin Transl Oncol 2019; 21:1413-1423. [PMID: 30877636 DOI: 10.1007/s12094-019-02070-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 02/23/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Some phase 2 trials had reported encouraging progression-free survival with Bevacizumab in monotherapy or combined with chemotherapy in glioblastoma. However, phase 3 trials showed a significant improvement in progression free survival without a benefit in overall survival. To date, there are no predictive biomarker of response for Bevacizumab in glioblastoma. METHODS We used Immunochemical analysis on tumor samples and pretreatment and post-treatment perfusion-MRI to try to identify possible predictive angiogenesis-related biomarkers of response and survival in patients with glioblastoma treated with bevacizumab in the first recurrence. We analyzed histological parameters: vascular proliferation, mitotic number and Ki-67 index; molecular factors: MGMT promoter methylation, EGFR amplification and EGFR variant III; immunohistochemical: MET, Midkine, HIF1, VEGFA, VEGF-R2, CD44, Olig2, microvascular area and microvascular density; and radiological: rCBV. RESULTS In the statistical analysis, no significant correlation of any histological, molecular, microvascular or radiological parameters could be demonstrated with the response rate, PFS or OS with bevacizumab treatment. CONCLUSION Unfortunately, in this histopathological, molecular, immunohistochemical and neuroradiological study we did not find any predictive biomarker of response or survival benefit for Bevacizumab in glioblastoma.
Collapse
|
33
|
Comparisons Between PET With 11C-Methyl-L-Methionine and Arterial Spin Labeling Perfusion Imaging in Recurrent Glioblastomas Treated With Bevacizumab. Clin Nucl Med 2019; 44:186-193. [PMID: 30562194 DOI: 10.1097/rlu.0000000000002417] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of this study was to clarify whether arterial spin labeling (ASL) perfusion imaging can assess biological effects from bevacizumab (BEV) therapy as reliably as PET with C-methyl-L-methionine (C-met-PET). MATERIALS AND METHODS Twenty-four patients with recurrent glioblastoma were examined using both ASL and C-met-PET before and 4 and 8 weeks after starting BEV treatment. Tumor-to-normal brain (T/N) ratios, fluctuations in T/N ratio, and tumor volumes were compared between ASL and C-met-PET. Accuracy of predicting patient with long progression-free survival (PFS) was assessed for T/N ratios and fluctuations for ASL and C-met-PET in each phase and in each period using receiver operating characteristic curves. Between 2 groups of patients assigned by cutoff values from receiver operating characteristic curves, PFS was compared in each phase or in each period. RESULTS T/N ratios, fluctuations in ratio, and tumor volumes correlated significantly between ASL and C-met-PET at all time points and all periods. Arterial spin labeling was eligible as a predictor for long PFS only in assessment of fluctuations in T/N ratio. However, the most accurate predictors for long PFS were T/N ratio from C-met-PET at 8 weeks and the fluctuation from baseline to 4 weeks in T/N ratio from C-met-PET. CONCLUSIONS Blood flows on ASL correlated with accumulations of C-met on PET in recurrent glioblastoma under BEV treatment. Although C-met-PET offered superior accuracy for predicting patients with long PFS from time points, ASL offered reliable prediction of long PFS, provided that fluctuations in T/N ratio between consecutive scans are assessed.
Collapse
|
34
|
Kasenene A, Baidya A, Shams S, Xu HB. Evaluation of tumor response to antiangiogenic therapy in patients with recurrent gliomas using contrast-enhanced perfusion-weighted magnetic resonance imaging techniques: A meta-analysis. World J Meta-Anal 2019; 7:51-65. [DOI: 10.13105/wjma.v7.i2.51] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/07/2019] [Accepted: 02/14/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND It is of vital importance to find radiologic biomarkers that can accurately predict treatment response. Usually, the initiation of antiangiogenic therapy causes a rapid decrease in the contrast enhancing tumor. However, the treatment response is observed only in a fraction of patients due to the partial radiological response secondary to stabilization of abnormal vessels which does not essentially indicate a true antitumor effect. Perfusion-weighted magnetic resonance imaging (PW-MRI) techniques have shown implicitness as a strong imaging biomarker for gliomas since they give hemodynamic information of blood vessels. Hence, there is a rapid expansion of PW-MRI related studies and clinical applications.
AIM To determine the diagnostic performance of PW-MRI techniques including: (A) dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI); and (B) dynamic susceptibility contrast magnetic resonance imaging (DSC-MRI) for evaluating response to antiangiogenic therapy in patients with recurrent gliomas.
METHODS Databases such as PubMed (MEDLINE included), EMBASE, and Google Scholar were searched for relevant original articles. The included studies were assessed for methodological quality with the Quality Assessment of Diagnostic Accuracy Studies 2 tool. Medical imaging follow-up or histopathological analysis was used as the reference standard. The data were extracted by two reviewers independently, and then the sensitivity, specificity, summary receiver operating characteristic curve, area under the curve (AUC), and heterogeneity were calculated using Meta-Disc 1.4 software.
RESULTS This study analyzed a total of six articles. The overall sensitivity for DCE-MRI and DSC-MRI was 0.69 [95% confidence interval (CI): 0.53-0.82], and the specificity was 0.99 (95%CI: 0.93-1) by a random effects model (DerSimonianee-Laird model). The likelihood ratio (LR) +, LR-, and diagnostic odds ratio (DOR) were 12.84 (4.54-36.28), 0.35 (0.22-0.53), and 24.44 (7.19-83.06), respectively. The AUC (± SE) was 0.9921 (± 0.0120), and the Q* index (± SE) was 0.9640 (± 0.0323). For DSC-MRI, the sensitivity was 0.73, the specificity was 0.98, the LR+ was 7.82, the LR- was 0.32, the DOR was 31.65, the AUC (± SE) was 0.9925 (± 0.0132), and the Q* index was 0.9649 (± 0.0363). For DCE-MRI, the sensitivity was 0.41, the specificity was 0.97, the LR+ was 5.34, the LR- was 0.71, the DOR was 8.76, the AUC (± SE) was 0.9922 (± 0.2218), and the Q* index was 0.8935 (± 0.3037).
CONCLUSION This meta-analysis demonstrated a beneficial value of PW-MRI (DSC-MRI and DCE-MRI) in monitoring the response of recurrent gliomas to antiangiogenic therapy, with reasonable sensitivity, specificity, +LR, and -LR.
Collapse
Affiliation(s)
- Akanganyira Kasenene
- Department of Radiology and Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Aju Baidya
- Department of Radiology and Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Salman Shams
- Department of Radiology and Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Hai-Bo Xu
- Department of Radiology and Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
35
|
Baseline multicentric tumors, distant recurrences and leptomeningeal dissemination predict poor survival in patients with recurrent glioblastomas receiving bevacizumab. J Neurooncol 2018; 142:149-159. [DOI: 10.1007/s11060-018-03075-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/06/2018] [Indexed: 12/14/2022]
|
36
|
Probing tumor microenvironment in patients with newly diagnosed glioblastoma during chemoradiation and adjuvant temozolomide with functional MRI. Sci Rep 2018; 8:17062. [PMID: 30459364 PMCID: PMC6244161 DOI: 10.1038/s41598-018-34820-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 10/24/2018] [Indexed: 12/18/2022] Open
Abstract
Functional MRI may identify critical windows of opportunity for drug delivery and distinguish between early treatment responders and non-responders. Using diffusion-weighted, dynamic contrast-enhanced, and dynamic susceptibility contrast MRI, as well as pro-angiogenic and pro-inflammatory blood markers, we prospectively studied the physiologic tumor-related changes in fourteen newly diagnosed glioblastoma patients during standard therapy. 153 MRI scans and blood collection were performed before chemoradiation (baseline), weekly during chemoradiation (week 1–6), monthly before each cycle of adjuvant temozolomide (pre-C1-C6), and after cycle 6. The apparent diffusion coefficient, volume transfer coefficient (Ktrans), and relative cerebral blood volume (rCBV) and flow (rCBF) were calculated within the tumor and edema regions and compared to baseline. Cox regression analysis was used to assess the effect of clinical variables, imaging, and blood markers on progression-free (PFS) and overall survival (OS). After controlling for additional covariates, high baseline rCBV and rCBF within the edema region were associated with worse PFS (microvessel rCBF: HR = 7.849, p = 0.044; panvessel rCBV: HR = 3.763, p = 0.032; panvessel rCBF: HR = 3.984; p = 0.049). The same applied to high week 5 and pre-C1 Ktrans within the tumor region (week 5 Ktrans: HR = 1.038, p = 0.003; pre-C1 Ktrans: HR = 1.029, p = 0.004). Elevated week 6 VEGF levels were associated with worse OS (HR = 1.034; p = 0.004). Our findings suggest a role for rCBV and rCBF at baseline and Ktrans and VEGF levels during treatment as markers of response. Functional imaging changes can differ substantially between tumor and edema regions, highlighting the variable biologic and vascular state of tumor microenvironment during therapy.
Collapse
|
37
|
Miyoshi F, Shinohara Y, Kambe A, Kuya K, Murakami A, Kurosaki M, Ogawa T. Utility of intravoxel incoherent motion magnetic resonance imaging and arterial spin labeling for recurrent glioma after bevacizumab treatment. Acta Radiol 2018; 59:1372-1379. [PMID: 29471670 DOI: 10.1177/0284185118759707] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background Detecting recurrence of glioma on magnetic resonance imaging (MRI) is getting more and more important, especially after administration of new anti-tumor agent. However, it is still hard to identify. Purpose To examine the utility of intravoxel incoherent motion (IVIM) MRI and arterial spin labeling-cerebral blood flow (ASL-CBF) for recurrent glioma after initiation of bevacizumab (BEV) treatment. Material and Methods Thirteen patients (7 men, 6 women; age range = 41-82 years) with glioma (high grade, n = 11; low grade, n = 2) were enrolled in the study. IVIM parameters including apparent diffusion coefficient (ADC), true diffusion coefficient (D), and perfusion fraction (f) were obtained with 14 different b-values. We identified tumor progression during BEV therapy by MRI monitoring consisting of diffusion-weighted imaging (DWI), fluid-attenuated inversion recovery (FLAIR) imaging, and contrast-enhanced T1-weighted (CE-T1W) imaging by measuring tumor area. We also measured each parameter of IVIM and ASL-CBF, and calculated relative ADC (rADC), relative D (rD), relative f (rf), and relative CBF (rCBF) by obtaining the ratio between each area and the contralateral cerebral white matter. We calculated the rate of change (Δ) by subtracting values from those from the preceding MRI study, and obtained Spearman's rank correlation coefficient (rs). Results Tumor progression was identified in nine patients (high grade, n = 7; low grade, n = 2). Negative correlations were identified between ΔrD and ΔDWI area (rs = -0.583), and between ΔrD and ΔCE-T1W imaging area (rs = -0.605). Conclusion Tumor progression after BEV treatment can be identified by decreasing rD.
Collapse
Affiliation(s)
- Fuminori Miyoshi
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Yuki Shinohara
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Atsushi Kambe
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Keita Kuya
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Atsushi Murakami
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Masamichi Kurosaki
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Toshihide Ogawa
- Division of Radiology, Department of Pathophysiological Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
38
|
Grossmann P, Narayan V, Chang K, Rahman R, Abrey L, Reardon DA, Schwartz LH, Wen PY, Alexander BM, Huang R, Aerts HJWL. Quantitative imaging biomarkers for risk stratification of patients with recurrent glioblastoma treated with bevacizumab. Neuro Oncol 2018; 19:1688-1697. [PMID: 28499022 DOI: 10.1093/neuonc/nox092] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Anti-angiogenic therapy with bevacizumab is the most widely used treatment option for recurrent glioblastoma, but therapeutic response varies substantially and effective biomarkers for patient selection are not available. To this end, we determine whether novel quantitative radiomic strategies on the basis of MRI have the potential to noninvasively stratify survival and progression in this patient population. Methods In an initial cohort of 126 patients, we identified a distinct set of features representative of the radiographic phenotype on baseline (pretreatment) MRI. These selected features were evaluated on a second cohort of 165 patients from the multicenter BRAIN trial with prospectively acquired clinical and imaging data. Features were evaluated in terms of prognostic value for overall survival (OS), progression-free survival (PFS), and progression within 3, 6, and 9 months using baseline imaging and first follow-up imaging at 6 weeks posttreatment initiation. Results Multivariable analysis of features derived at baseline imaging resulted in significant stratification of OS (hazard ratio [HR] = 2.5; log-rank P = 0.001) and PFS (HR = 4.5; log-rank P = 2.1 × 10-5) in validation data. These stratifications were stronger compared with clinical or volumetric covariates (permutation test false discovery rate [FDR] <0.05). Univariable analysis of a prognostic textural heterogeneity feature (information correlation) derived from postcontrast T1-weighted imaging revealed significantly higher scores for patients who progressed within 3 months (Wilcoxon test P = 8.8 × 10-8). Generally, features derived from postcontrast T1-weighted imaging yielded higher prognostic power compared with precontrast enhancing T2-weighted imaging. Conclusion Radiomics provides prognostic value for survival and progression in patients with recurrent glioblastoma receiving bevacizumab treatment. These results could lead to the development of quantitative pretreatment biomarkers to predict benefit from bevacizumab using standard of care imaging.
Collapse
Affiliation(s)
- Patrick Grossmann
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Product Development, Pharma Division, F. Hoffmann-La Roche Ltd., Basel, Switzerland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York, USA
| | - Vivek Narayan
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Product Development, Pharma Division, F. Hoffmann-La Roche Ltd., Basel, Switzerland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York, USA
| | - Ken Chang
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Product Development, Pharma Division, F. Hoffmann-La Roche Ltd., Basel, Switzerland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York, USA
| | - Rifaquat Rahman
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Product Development, Pharma Division, F. Hoffmann-La Roche Ltd., Basel, Switzerland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York, USA
| | - Lauren Abrey
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Product Development, Pharma Division, F. Hoffmann-La Roche Ltd., Basel, Switzerland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York, USA
| | - David A Reardon
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Product Development, Pharma Division, F. Hoffmann-La Roche Ltd., Basel, Switzerland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York, USA
| | - Lawrence H Schwartz
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Product Development, Pharma Division, F. Hoffmann-La Roche Ltd., Basel, Switzerland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York, USA
| | - Patrick Y Wen
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Product Development, Pharma Division, F. Hoffmann-La Roche Ltd., Basel, Switzerland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York, USA
| | - Brian M Alexander
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Product Development, Pharma Division, F. Hoffmann-La Roche Ltd., Basel, Switzerland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York, USA
| | - Raymond Huang
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Product Development, Pharma Division, F. Hoffmann-La Roche Ltd., Basel, Switzerland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York, USA
| | - Hugo J W L Aerts
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Product Development, Pharma Division, F. Hoffmann-La Roche Ltd., Basel, Switzerland; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
39
|
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
|
40
|
Ly KI, Gerstner ER. The Role of Advanced Brain Tumor Imaging in the Care of Patients with Central Nervous System Malignancies. Curr Treat Options Oncol 2018; 19:40. [PMID: 29931476 DOI: 10.1007/s11864-018-0558-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OPINION STATEMENT T1-weighted post-contrast and T2-weighted fluid-attenuated inversion recovery (FLAIR) magnetic resonance imaging (MRI) constitute the gold standard for diagnosis and response assessment in neuro-oncologic patients but are limited in their ability to accurately reflect tumor biology and metabolism, particularly over the course of a patient's treatment. Advanced MR imaging methods are sensitized to different biophysical processes in tissue, including blood perfusion, tumor metabolism, and chemical composition of tissue, and provide more specific information on tissue physiology than standard MRI. This review provides an overview of the most common and emerging advanced imaging modalities in the field of brain tumor imaging and their applications in the care of neuro-oncologic patients.
Collapse
Affiliation(s)
- K Ina Ly
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9E, Boston, MA, 02114, USA
| | - Elizabeth R Gerstner
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9E, Boston, MA, 02114, USA.
| |
Collapse
|
41
|
Schmainda KM, Prah MA, Rand SD, Liu Y, Logan B, Muzi M, Rane SD, Da X, Yen YF, Kalpathy-Cramer J, Chenevert TL, Hoff B, Ross B, Cao Y, Aryal MP, Erickson B, Korfiatis P, Dondlinger T, Bell L, Hu L, Kinahan PE, Quarles CC. Multisite Concordance of DSC-MRI Analysis for Brain Tumors: Results of a National Cancer Institute Quantitative Imaging Network Collaborative Project. AJNR Am J Neuroradiol 2018; 39:1008-1016. [PMID: 29794239 DOI: 10.3174/ajnr.a5675] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/07/2018] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Standard assessment criteria for brain tumors that only include anatomic imaging continue to be insufficient. While numerous studies have demonstrated the value of DSC-MR imaging perfusion metrics for this purpose, they have not been incorporated due to a lack of confidence in the consistency of DSC-MR imaging metrics across sites and platforms. This study addresses this limitation with a comparison of multisite/multiplatform analyses of shared DSC-MR imaging datasets of patients with brain tumors. MATERIALS AND METHODS DSC-MR imaging data were collected after a preload and during a bolus injection of gadolinium contrast agent using a gradient recalled-echo-EPI sequence (TE/TR = 30/1200 ms; flip angle = 72°). Forty-nine low-grade (n = 13) and high-grade (n = 36) glioma datasets were uploaded to The Cancer Imaging Archive. Datasets included a predetermined arterial input function, enhancing tumor ROIs, and ROIs necessary to create normalized relative CBV and CBF maps. Seven sites computed 20 different perfusion metrics. Pair-wise agreement among sites was assessed with the Lin concordance correlation coefficient. Distinction of low- from high-grade tumors was evaluated with the Wilcoxon rank sum test followed by receiver operating characteristic analysis to identify the optimal thresholds based on sensitivity and specificity. RESULTS For normalized relative CBV and normalized CBF, 93% and 94% of entries showed good or excellent cross-site agreement (0.8 ≤ Lin concordance correlation coefficient ≤ 1.0). All metrics could distinguish low- from high-grade tumors. Optimum thresholds were determined for pooled data (normalized relative CBV = 1.4, sensitivity/specificity = 90%:77%; normalized CBF = 1.58, sensitivity/specificity = 86%:77%). CONCLUSIONS By means of DSC-MR imaging data obtained after a preload of contrast agent, substantial consistency resulted across sites for brain tumor perfusion metrics with a common threshold discoverable for distinguishing low- from high-grade tumors.
Collapse
Affiliation(s)
- K M Schmainda
- From the Department of Radiology (K.M.S., M.A.P., S.D.R.)
| | - M A Prah
- From the Department of Radiology (K.M.S., M.A.P., S.D.R.)
| | - S D Rand
- From the Department of Radiology (K.M.S., M.A.P., S.D.R.).,Department of Radiology (M.M., S.D.R., P.E.K.), University of Washington, Seattle, Washington
| | - Y Liu
- Division of Biostatistics (Y.L., B.L.), Institute for Health and Society, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - B Logan
- Division of Biostatistics (Y.L., B.L.), Institute for Health and Society, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - M Muzi
- Department of Radiology (M.M., S.D.R., P.E.K.), University of Washington, Seattle, Washington
| | - S D Rane
- From the Department of Radiology (K.M.S., M.A.P., S.D.R.)
| | - X Da
- Department of Radiology (X.D.), Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts
| | - Y-F Yen
- Athinoula A. Martinos Center for Biomedical Imaging (Y.-F.Y., J.K.-C.), Department of Radiology, Harvard Medical School/Massachusetts General Hospital, Charlestown, Massachusetts
| | - J Kalpathy-Cramer
- Athinoula A. Martinos Center for Biomedical Imaging (Y.-F.Y., J.K.-C.), Department of Radiology, Harvard Medical School/Massachusetts General Hospital, Charlestown, Massachusetts
| | | | - B Hoff
- Department of Radiology (T.L.C., B.H., B.R.)
| | - B Ross
- Department of Radiology (T.L.C., B.H., B.R.)
| | - Y Cao
- Departments of Radiation Oncology, Radiology, and Biomedical Engineering (Y.C., M.P.A.), University of Michigan, Ann Arbor, Michigan
| | - M P Aryal
- Departments of Radiation Oncology, Radiology, and Biomedical Engineering (Y.C., M.P.A.), University of Michigan, Ann Arbor, Michigan
| | - B Erickson
- Department of Radiology (B.E., P.K.), Mayo Clinic, Rochester, Minnesota
| | - P Korfiatis
- Department of Radiology (B.E., P.K.), Mayo Clinic, Rochester, Minnesota
| | - T Dondlinger
- Imaging Biometrics LLC (T.D.), Elm Grove, Wisconsin
| | - L Bell
- Division of Imaging Research (L.B., C.C.Q.), Barrow Neurological Institute, Phoenix, Arizona
| | - L Hu
- Department of Radiology (L.H.), Mayo Clinic, Scottsdale, Arizona
| | - P E Kinahan
- Department of Radiology (M.M., S.D.R., P.E.K.), University of Washington, Seattle, Washington
| | - C C Quarles
- Division of Imaging Research (L.B., C.C.Q.), Barrow Neurological Institute, Phoenix, Arizona
| |
Collapse
|
42
|
Pope WB, Brandal G. Conventional and advanced magnetic resonance imaging in patients with high-grade glioma. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2018; 62:239-253. [PMID: 29696946 DOI: 10.23736/s1824-4785.18.03086-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Magnetic resonance imaging is integral to the care of patients with high-grade gliomas. Anatomic detail can be acquired with conventional structural imaging, but newer approaches also add capabilities to interrogate image-derived physiologic and molecular characteristics of central nervous system neoplasms. These advanced imaging techniques are increasingly employed to generate biomarkers that better reflect tumor burden and therapy response. The following is an overview of current strategies based on advanced magnetic resonance imaging that are used in the assessment of high-grade glioma patients with an emphasis on how novel imaging biomarkers can potentially advance patient care.
Collapse
Affiliation(s)
- Whitney B Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA, USA -
| | - Garth Brandal
- Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
43
|
Goyal P, Tenenbaum M, Gupta S, Kochar PS, Bhatt AA, Mangla M, Kumar Y, Mangla R. Survival prediction based on qualitative MRI diffusion signature in patients with recurrent high grade glioma treated with bevacizumab. Quant Imaging Med Surg 2018; 8:268-279. [PMID: 29774180 DOI: 10.21037/qims.2018.04.05] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Bevacizumab was approved by the FDA for the treatment of recurrent or progressive glioblastoma (GBM). Imaging responses are typically assessed by gadolinium-enhanced MRI. We sought to determine the significance of qualitative diffusion signature (manifest as variable degree of dark signal) on ADC maps in recurrent gliomas after treatment with bevacizumab. Methods We performed an institutional review board (IRB) approved retrospective study on patients who underwent MRI of the brain after 8 weeks of receiving bevacizumab for recurrent glioma. Patients were divided into three groups based on qualitative diffusion signature: (I) lesion not bright on diffusion weighted imaging (DWI) suggestive of no restricted diffusion (FDR0); (II) lesion bright on DWI with corresponding homogenous dark signal on apparent diffusion coefficient (ADC) maps suggestive of focal restricted diffusion likely due to bevacizumab induced necrosis (FDRn); and (III) lesion bright on DWI with corresponding homogenous faint dark signal on ADC maps suggestive of focal restricted diffusion likely due to viable tumor or heterogeneous spectrum of dark and faint dark signals on ADC maps suggestive of focal restricted diffusion likely due to viable tumor surrounding the bevacizumab induced necrosis (FDRt). Results Based on the qualitative signal on diffusion weighted sequences after bevacizumab therapy, total number of patients in group (I) were 14 (36%), in group (II) were 17 (44%); and in group (III) were 8 (20%). The median overall survival (OS) from the time of recurrence in patients belonging to group (II) was 364 days vs. 183 days for those with group (I) vs. 298 days for group (III). On simultaneous comparison of survival differences in all three groups by Kaplan-Meier analysis, group (II) was significant in predicting survival with P values for the log-rank tests <0.033. Conclusions In patients with recurrent glioma treated with bevacizumab, the presence of homogenous dark signal (FDRn) on ADC maps at 8 weeks follow-up MRI correlated with a longer survival. Thus, use of this qualitative diffusion signature in adjunct to contrast enhanced MRI may have the widest potential impact on routine clinical care for patients with recurrent high-grade gliomas. However, prospective studies analysing its predictive value are warranted.
Collapse
Affiliation(s)
- Pradeep Goyal
- Department of Radiology, St. Vincent's Medical Center, Bridgeport, CT, USA
| | - Mary Tenenbaum
- Department of Radiology, UMMS-Baystate Regional Campus, Springfield, MA, USA.,Department of Radiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Sonali Gupta
- Department of Medicine, St. Vincent's Medical Center, Bridgeport, CT, USA
| | - Puneet S Kochar
- Department of Radiology, Yale New Haven Health Bridgeport Hospital, Bridgeport, CT, USA
| | - Alok A Bhatt
- Department of Radiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Manisha Mangla
- Department of Public Health, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yogesh Kumar
- Department of Radiology, Columbia University at Bassett Healthcare, Cooperstown, NY, USA
| | - Rajiv Mangla
- Department of Radiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Radiology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
44
|
Nandu H, Wen PY, Huang RY. Imaging in neuro-oncology. Ther Adv Neurol Disord 2018; 11:1756286418759865. [PMID: 29511385 PMCID: PMC5833173 DOI: 10.1177/1756286418759865] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/18/2018] [Indexed: 12/11/2022] Open
Abstract
Imaging plays several key roles in managing brain tumors, including diagnosis, prognosis, and treatment response assessment. Ongoing challenges remain as new therapies emerge and there are urgent needs to find accurate and clinically feasible methods to noninvasively evaluate brain tumors before and after treatment. This review aims to provide an overview of several advanced imaging modalities including magnetic resonance imaging and positron emission tomography (PET), including advances in new PET agents, and summarize several key areas of their applications, including improving the accuracy of diagnosis and addressing the challenging clinical problems such as evaluation of pseudoprogression and anti-angiogenic therapy, and rising challenges of imaging with immunotherapy.
Collapse
Affiliation(s)
- Hari Nandu
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02445, USA
| |
Collapse
|
45
|
Mullen KM, Huang RY. An Update on the Approach to the Imaging of Brain Tumors. Curr Neurol Neurosci Rep 2017; 17:53. [PMID: 28516376 DOI: 10.1007/s11910-017-0760-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Neuroimaging plays a critical role in diagnosis of brain tumors and in assessment of response to therapy. However, challenges remain, including accurately and reproducibly assessing response to therapy, defining endpoints for neuro-oncology trials, providing prognostic information, and differentiating progressive disease from post-therapeutic changes particularly in the setting of antiangiogenic and other novel therapies. RECENT FINDINGS Recent advances in the imaging of brain tumors include application of advanced MRI imaging techniques to assess tumor response to therapy and analysis of imaging features correlating to molecular markers, grade, and prognosis. This review aims to summarize recent advances in imaging as applied to current diagnostic and therapeutic neuro-oncologic challenges.
Collapse
Affiliation(s)
- Katherine M Mullen
- Department of Radiology, Brigham and Women's Hospital, 75 Francis St, Boston, MA, 02115, USA
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, 75 Francis St, Boston, MA, 02115, USA.
| |
Collapse
|
46
|
Margiewicz S, Cordova C, Chi AS, Jain R. State of the Art Treatment and Surveillance Imaging of Glioblastomas. Semin Roentgenol 2017; 53:23-36. [PMID: 29405952 DOI: 10.1053/j.ro.2017.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
| | - Christine Cordova
- Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY
| | - Andrew S Chi
- Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY
| | - Rajan Jain
- Department of Radiology, NYU School of Medicine, New York, NY; Department of Neurosurgery, NYU School of Medicine, New York, NY.
| |
Collapse
|
47
|
Comparison of 18F-FET PET and perfusion-weighted MRI for glioma grading: a hybrid PET/MR study. Eur J Nucl Med Mol Imaging 2017; 44:2257-2265. [PMID: 28831534 DOI: 10.1007/s00259-017-3812-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022]
Abstract
PURPOSE Both perfusion-weighted MR imaging (PWI) and O-(2-18F-fluoroethyl)-L-tyrosine PET (18F-FET) provide grading information in cerebral gliomas. The aim of this study was to compare the diagnostic value of 18F-FET PET and PWI for tumor grading in a series of patients with newly diagnosed, untreated gliomas using an integrated PET/MR scanner. METHODS Seventy-two patients with untreated gliomas [22 low-grade gliomas (LGG), and 50 high-grade gliomas (HGG)] were investigated with 18F-FET PET and PWI using a hybrid PET/MR scanner. After visual inspection of PET and PWI maps (rCBV, rCBF, MTT), volumes of interest (VOIs) with a diameter of 16 mm were centered upon the maximum of abnormality in the tumor area in each modality and the contralateral unaffected hemisphere. Mean and maximum tumor-to-brain ratios (TBRmean, TBRmax) were calculated. In addition, Time-to-Peak (TTP) and slopes of time-activity curves were calculated for 18F-FET PET. Diagnostic accuracies of 18F-FET PET and PWI for differentiating low-grade glioma (LGG) from high-grade glioma (HGG) were evaluated by receiver operating characteristic analyses (area under the curve; AUC). RESULTS The diagnostic accuracy of 18F-FET PET and PWI to discriminate LGG from HGG was similar with highest AUC values for TBRmean and TBRmax of 18F-FET PET uptake (0.80, 0.83) and for TBRmean and TBRmax of rCBV (0.80, 0.81). In case of increased signal in the tumor area with both methods (n = 32), local hot-spots were incongruent in 25 patients (78%) with a mean distance of 10.6 ± 9.5 mm. Dynamic FET PET and combination of different parameters did not further improve diagnostic accuracy. CONCLUSIONS Both 18F-FET PET and PWI discriminate LGG from HGG with similar diagnostic performance. Regional abnormalities in the tumor area are usually not congruent indicating that tumor grading by 18F-FET PET and PWI is based on different pathophysiological phenomena.
Collapse
|
48
|
Abstract
Despite the fact that MRI has evolved to become the standard method for diagnosis and monitoring of patients with brain tumours, conventional MRI sequences have two key limitations: the inability to show the full extent of the tumour and the inability to differentiate neoplastic tissue from nonspecific, treatment-related changes after surgery, radiotherapy, chemotherapy or immunotherapy. In the past decade, PET involving the use of radiolabelled amino acids has developed into an important diagnostic tool to overcome some of the shortcomings of conventional MRI. The Response Assessment in Neuro-Oncology working group - an international effort to develop new standardized response criteria for clinical trials in brain tumours - has recommended the additional use of amino acid PET imaging for brain tumour management. Concurrently, a number of advanced MRI techniques such as magnetic resonance spectroscopic imaging and perfusion weighted imaging are under clinical evaluation to target the same diagnostic problems. This Review summarizes the clinical role of amino acid PET in relation to advanced MRI techniques for differential diagnosis of brain tumours; delineation of tumour extent for treatment planning and biopsy guidance; post-treatment differentiation between tumour progression or recurrence versus treatment-related changes; and monitoring response to therapy. An outlook for future developments in PET and MRI techniques is also presented.
Collapse
Affiliation(s)
- Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4) Forschungszentrum Jülich, Wilhelm-Johnen-Strasse, D-52425 Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4) Forschungszentrum Jülich, Wilhelm-Johnen-Strasse, D-52425 Jülich, Germany.,Department of Neurology, University of Cologne, Kerpener Strasse 62, D-50937 Cologne, Germany.,Center for Integrated Oncology, Josef-Stelzmann-Strasse 9, D-50937 Cologne, Germany
| | - Elke Hattingen
- Department of Neuroradiology and Center for Integrated Oncology, University of Bonn, Sigmund-Freud-Strasse 25, D-53127 Bonn, Germany
| | - Nadim Jon Shah
- Institute of Neuroscience and Medicine (INM-3, INM-4) Forschungszentrum Jülich, Wilhelm-Johnen-Strasse, D-52425 Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Pauwelsstrasse 30, D-52074 Aachen, Germany.,Monash Institute of Medical Engineering, Department of Electrical and Computer Systems Engineering, and Monash Biomedical Imaging, School of Psychological Sciences, Monash University, 18 Innovation Walk, Clayton Campus, Wellington Road, Melbourne, Victoria 3800, Australia
| |
Collapse
|
49
|
Lucas JT, Knapp BJ, Uh J, Hua CH, Merchant TE, Hwang SN, Patay Z, Broniscer A. Posttreatment DSC-MRI is Predictive of Early Treatment Failure in Children with Supratentorial High-Grade Glioma Treated with Erlotinib. Clin Neuroradiol 2017; 28:393-400. [PMID: 28382379 DOI: 10.1007/s00062-017-0580-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/15/2017] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND PURPOSE The role of perfusion imaging in the management of pediatric high grade glioma is unclear. We evaluated the ability of dynamic susceptibility contrast magnetic resonance imaging (DSC-MRI) to determine grade, evaluate post-treatment response and predict treatment failure. MATERIAL AND METHODS In this study 22 patients with high-grade glioma underwent biopsy and were treated with concurrent and sequential radiotherapy and erlotinib as part of a phase I/II clinical trial (NCT00124657). Preradiotherapy, immediate postradiotherapy, 6‑month and treatment failure DSC MR images were reviewed, registered, and processed for the ratio of cerebral blood flow (CBF) and cerebral blood volume (CBV). Processed, derived perfusion, and T1-weighted images (T1WI), T2WI, and fluid attenuation inversion recovery (FLAIR) MRI sequences were used for segmentation and extraction of tumor perfusion parameters at all time points. Patient, tumor, treatment, and outcome data were summarized and related to perfusion data. RESULTS Regional CBF in tumors increased from diagnosis to postradiotherapy, while they decreased to levels below those at diagnosis from postradiotherapy to 6‑month follow-up. At 6 months, the median regional CBF was higher in tumors that progressed (median 1.16) than in those that did not (median, 0.95; P < 0.05). Patients with regional CBF ratios above 1.4 at diagnosis had shorter survival times than did those with regional CBF ratios below 1.4 (P = 0.77). Tumors with a regional CBV above 1.15 at the postradiotherapy (1-3 months) follow-up scan were associated with an earlier time to death than that of tumors with a regional CBV below 1.15 (P < 0.05). CONCLUSION Posttreatment perfusion characteristics are prognostic and may help predict survival. Overall, perfusion MRI is useful for managing pediatric high-grade glioma and should be incorporated into future clinical trials.
Collapse
Affiliation(s)
- John T Lucas
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 210, 38105-3678, Memphis, TN, USA.
| | - Brendan J Knapp
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jinsoo Uh
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 210, 38105-3678, Memphis, TN, USA
| | - Chia-Ho Hua
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 210, 38105-3678, Memphis, TN, USA
| | - Thomas E Merchant
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 210, 38105-3678, Memphis, TN, USA
| | - Scott N Hwang
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zoltan Patay
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Alberto Broniscer
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
50
|
Zhang J, Liu H, Tong H, Wang S, Yang Y, Liu G, Zhang W. Clinical Applications of Contrast-Enhanced Perfusion MRI Techniques in Gliomas: Recent Advances and Current Challenges. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:7064120. [PMID: 29097933 PMCID: PMC5612612 DOI: 10.1155/2017/7064120] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/23/2017] [Indexed: 01/12/2023]
Abstract
Gliomas possess complex and heterogeneous vasculatures with abnormal hemodynamics. Despite considerable advances in diagnostic and therapeutic techniques for improving tumor management and patient care in recent years, the prognosis of malignant gliomas remains dismal. Perfusion-weighted magnetic resonance imaging techniques that could noninvasively provide superior information on vascular functionality have attracted much attention for evaluating brain tumors. However, nonconsensus imaging protocols and postprocessing analysis among different institutions impede their integration into standard-of-care imaging in clinic. And there have been very few studies providing a comprehensive evidence-based and systematic summary. This review first outlines the status of glioma theranostics and tumor-associated vascular pathology and then presents an overview of the principles of dynamic contrast-enhanced MRI (DCE-MRI) and dynamic susceptibility contrast-MRI (DSC-MRI), with emphasis on their recent clinical applications in gliomas including tumor grading, identification of molecular characteristics, differentiation of glioma from other brain tumors, treatment response assessment, and predicting prognosis. Current challenges and future perspectives are also highlighted.
Collapse
Affiliation(s)
- Junfeng Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Heng Liu
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Haipeng Tong
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Sumei Wang
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Yizeng Yang
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Weiguo Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing 400042, China
| |
Collapse
|