1
|
Lin J, Su CQ, Tang WT, Xia ZW, Lu SS, Hong XN. Radiomic features on multiparametric MRI for differentiating pseudoprogression from recurrence in high-grade gliomas. Acta Radiol 2024:2841851241283781. [PMID: 39380365 DOI: 10.1177/02841851241283781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
BACKGROUND Distinguishing between tumor recurrence and pseudoprogression (PsP) in high-grade glioma postoperatively is challenging. This study aims to enhance this differentiation using a combination of intratumoral and peritumoral radiomics. PURPOSE To assess the effectiveness of intratumoral and peritumoral radiomics in improving the differentiation between high-grade glioma recurrence and pseudoprogression after surgery. MATERIAL AND METHODS A total of 109 cases were randomly divided into training and validation sets, with 1316 features extracted from intratumoral and peritumoral volumes of interest (VOIs) on conventional magnetic resonance imaging (MRI) and apparent diffusion coefficient (ADC) maps. Feature selection was performed using the mRMR algorithm, resulting in intratumoral (100 features), peritumoral (100 features), and combined (200 features) subsets. Optimal features were then selected using PCC and RFE algorithms and modeled using LR, SVM, and LDA classifiers. Diagnostic performance was compared using area under the receiver operating characteristic curve (AUC), evaluated in the validation set. A nomogram was established using radscores from intratumoral, peritumoral, and combined models. RESULTS The combined model, utilizing 14 optimal features (8 peritumoral, 6 intratumoral) and LR as the best classifier, outperformed the single intratumoral and peritumoral models. In the training set, the AUC values for the combined model, intratumoral model, and peritumoral model were 0.938, 0.921, and 0.847, respectively; in the validation set, the AUC values were 0.841, 0.755, and 0.705. The nomogram model demonstrated AUCs of 0.960 (training set) and 0.850 (validation set). CONCLUSION The combination of intratumoral and peritumoral radiomics is effective in distinguishing high-grade glioma recurrence from pseudoprogression after surgery.
Collapse
Affiliation(s)
- Jie Lin
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Chun-Qiu Su
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Wen-Tian Tang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Zhi-Wei Xia
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Shan-Shan Lu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Xun-Ning Hong
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| |
Collapse
|
2
|
Śledzińska-Bebyn P, Furtak J, Bebyn M, Serafin Z. Beyond conventional imaging: Advancements in MRI for glioma malignancy prediction and molecular profiling. Magn Reson Imaging 2024; 112:63-81. [PMID: 38914147 DOI: 10.1016/j.mri.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/20/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
This review examines the advancements in magnetic resonance imaging (MRI) techniques and their pivotal role in diagnosing and managing gliomas, the most prevalent primary brain tumors. The paper underscores the importance of integrating modern MRI modalities, such as diffusion-weighted imaging and perfusion MRI, which are essential for assessing glioma malignancy and predicting tumor behavior. Special attention is given to the 2021 WHO Classification of Tumors of the Central Nervous System, emphasizing the integration of molecular diagnostics in glioma classification, significantly impacting treatment decisions. The review also explores radiogenomics, which correlates imaging features with molecular markers to tailor personalized treatment strategies. Despite technological progress, MRI protocol standardization and result interpretation challenges persist, affecting diagnostic consistency across different settings. Furthermore, the review addresses MRI's capacity to distinguish between tumor recurrence and pseudoprogression, which is vital for patient management. The necessity for greater standardization and collaborative research to harness MRI's full potential in glioma diagnosis and personalized therapy is highlighted, advocating for an enhanced understanding of glioma biology and more effective treatment approaches.
Collapse
Affiliation(s)
- Paulina Śledzińska-Bebyn
- Department of Radiology, 10th Military Research Hospital and Polyclinic, 85-681 Bydgoszcz, Poland.
| | - Jacek Furtak
- Department of Clinical Medicine, Faculty of Medicine, University of Science and Technology, Bydgoszcz, Poland; Department of Neurosurgery, 10th Military Research Hospital and Polyclinic, 85-681 Bydgoszcz, Poland
| | - Marek Bebyn
- Department of Internal Diseases, 10th Military Clinical Hospital and Polyclinic, 85-681 Bydgoszcz, Poland
| | - Zbigniew Serafin
- Department of Radiology and Diagnostic Imaging, Nicolaus Copernicus University, Collegium Medicum, Bydgoszcz, Poland
| |
Collapse
|
3
|
Li K, Zhu Q, Yang J, Zheng Y, Du S, Song M, Peng Q, Yang R, Liu Y, Qi L. Imaging and Liquid Biopsy for Distinguishing True Progression From Pseudoprogression in Gliomas, Current Advances and Challenges. Acad Radiol 2024; 31:3366-3383. [PMID: 38614827 DOI: 10.1016/j.acra.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/14/2024] [Accepted: 03/18/2024] [Indexed: 04/15/2024]
Abstract
RATIONALE AND OBJECTIVES Gliomas are aggressive brain tumors with a poor prognosis. Assessing treatment response is challenging because magnetic resonance imaging (MRI) may not distinguish true progression (TP) from pseudoprogression (PsP). This review aims to discuss imaging techniques and liquid biopsies used to distinguish TP from PsP. MATERIALS AND METHODS This review synthesizes existing literature to examine advances in imaging techniques, such as magnetic resonance diffusion imaging (MRDI), perfusion-weighted imaging (PWI) MRI, and liquid biopsies, for identifying TP or PsP through tumor markers and tissue characteristics. RESULTS Advanced imaging techniques, including MRDI and PWI MRI, have proven effective in delineating tumor tissue properties, offering valuable insights into glioma behavior. Similarly, liquid biopsy has emerged as a potent tool for identifying tumor-derived markers in biofluids, offering a non-invasive glimpse into tumor evolution. Despite their promise, these methodologies grapple with significant challenges. Their sensitivity remains inconsistent, complicating the accurate differentiation between TP and PSP. Furthermore, the absence of standardized protocols across platforms impedes the reliability of comparisons, while inherent biological variability adds complexity to data interpretation. CONCLUSION Their potential applications have been highlighted, but gaps remain before routine clinical use. Further research is needed to develop and validate these promising methods for distinguishing TP from PsP in gliomas.
Collapse
Affiliation(s)
- Kaishu Li
- Department of Neurosurgery, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China; Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), 1# Jiazi Road, Foshan, Guangdong 528300, China.; Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qihui Zhu
- Department of Neurosurgery, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Junyi Yang
- Department of Neurosurgery, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Yin Zheng
- Department of Neurosurgery, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Siyuan Du
- Institute of Digestive Disease of Guangzhou Medical University, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Meihui Song
- Institute of Digestive Disease of Guangzhou Medical University, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Qian Peng
- Institute of Digestive Disease of Guangzhou Medical University, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Runwei Yang
- Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), 1# Jiazi Road, Foshan, Guangdong 528300, China
| | - Yawei Liu
- Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), 1# Jiazi Road, Foshan, Guangdong 528300, China
| | - Ling Qi
- Institute of Digestive Disease of Guangzhou Medical University, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China.
| |
Collapse
|
4
|
Sanvito F, Raymond C, Cho NS, Yao J, Hagiwara A, Orpilla J, Liau LM, Everson RG, Nghiemphu PL, Lai A, Prins R, Salamon N, Cloughesy TF, Ellingson BM. Simultaneous quantification of perfusion, permeability, and leakage effects in brain gliomas using dynamic spin-and-gradient-echo echoplanar imaging MRI. Eur Radiol 2024; 34:3087-3101. [PMID: 37882836 PMCID: PMC11045669 DOI: 10.1007/s00330-023-10215-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/05/2023] [Accepted: 07/27/2023] [Indexed: 10/27/2023]
Abstract
OBJECTIVE To determine the feasibility and biologic correlations of dynamic susceptibility contrast (DSC), dynamic contrast enhanced (DCE), and quantitative maps derived from contrast leakage effects obtained simultaneously in gliomas using dynamic spin-and-gradient-echo echoplanar imaging (dynamic SAGE-EPI) during a single contrast injection. MATERIALS AND METHODS Thirty-eight patients with enhancing brain gliomas were prospectively imaged with dynamic SAGE-EPI, which was processed to compute traditional DSC metrics (normalized relative cerebral blood flow [nrCBV], percentage of signal recovery [PSR]), DCE metrics (volume transfer constant [Ktrans], extravascular compartment [ve]), and leakage effect metrics: ΔR2,ss* (reflecting T2*-leakage effects), ΔR1,ss (reflecting T1-leakage effects), and the transverse relaxivity at tracer equilibrium (TRATE, reflecting the balance between ΔR2,ss* and ΔR1,ss). These metrics were compared between patient subgroups (treatment-naïve [TN] vs recurrent [R]) and biological features (IDH status, Ki67 expression). RESULTS In IDH wild-type gliomas (IDHwt-i.e., glioblastomas), previous exposure to treatment determined lower TRATE (p = 0.002), as well as higher PSR (p = 0.006), Ktrans (p = 0.17), ΔR1,ss (p = 0.035), ve (p = 0.006), and ADC (p = 0.016). In IDH-mutant gliomas (IDHm), previous treatment determined higher Ktrans and ΔR1,ss (p = 0.026). In TN-gliomas, dynamic SAGE-EPI metrics tended to be influenced by IDH status (p ranging 0.09-0.14). TRATE values above 142 mM-1s-1 were exclusively seen in TN-IDHwt, and, in TN-gliomas, this cutoff had 89% sensitivity and 80% specificity as a predictor of Ki67 > 10%. CONCLUSIONS Dynamic SAGE-EPI enables simultaneous quantification of brain tumor perfusion and permeability, as well as mapping of novel metrics related to cytoarchitecture (TRATE) and blood-brain barrier disruption (ΔR1,ss), with a single contrast injection. CLINICAL RELEVANCE STATEMENT Simultaneous DSC and DCE analysis with dynamic SAGE-EPI reduces scanning time and contrast dose, respectively alleviating concerns about imaging protocol length and gadolinium adverse effects and accumulation, while providing novel leakage effect metrics reflecting blood-brain barrier disruption and tumor tissue cytoarchitecture. KEY POINTS • Traditionally, perfusion and permeability imaging for brain tumors requires two separate contrast injections and acquisitions. • Dynamic spin-and-gradient-echo echoplanar imaging enables simultaneous perfusion and permeability imaging. • Dynamic spin-and-gradient-echo echoplanar imaging provides new image contrasts reflecting blood-brain barrier disruption and cytoarchitecture characteristics.
Collapse
Affiliation(s)
- Francesco Sanvito
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, 924 Westwood Blvd, Los Angeles, CA, 90024, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
- Unit of Radiology, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Viale Camillo Golgi 19, 27100, Pavia, Italy
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, 924 Westwood Blvd, Los Angeles, CA, 90024, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
| | - Nicholas S Cho
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, 924 Westwood Blvd, Los Angeles, CA, 90024, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
- Medical Scientist Training Program, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, 7400 Boelter Hall, Los Angeles, CA, 90095, USA
| | - Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, 924 Westwood Blvd, Los Angeles, CA, 90024, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
| | - Akifumi Hagiwara
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, 924 Westwood Blvd, Los Angeles, CA, 90024, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
- Department of Radiology, Juntendo University School of Medicine, Bunkyo City, 2-Chōme-1-1 Hongō, Tokyo, 113-8421, Japan
| | - Joey Orpilla
- Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
| | - Richard G Everson
- Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
| | - Phioanh L Nghiemphu
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
| | - Albert Lai
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
| | - Robert Prins
- Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
| | - Noriko Salamon
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, 924 Westwood Blvd, Los Angeles, CA, 90024, USA.
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA.
- Medical Scientist Training Program, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, 7400 Boelter Hall, Los Angeles, CA, 90095, USA.
- Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA.
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, 885 Tiverton Dr, Los Angeles, CA, 90095, USA.
| |
Collapse
|
5
|
Hoggarth AR, Muthukumar S, Thomas SM, Crowley J, Kiser J, Witcher MR. Clinical Theranostics in Recurrent Gliomas: A Review. Cancers (Basel) 2024; 16:1715. [PMID: 38730666 PMCID: PMC11083317 DOI: 10.3390/cancers16091715] [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: 03/30/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Gliomas represent the most commonly occurring tumors in the central nervous system and account for approximately 80% of all malignant primary brain tumors. With a high malignancy and recurrence risk, the prognosis of high-grade gliomas is poor, with a mean survival time of 12-18 months. While contrast-enhanced MRI serves as the standard diagnostic imaging modality for gliomas, it faces limitations in the evaluation of recurrent gliomas, failing to distinguish between treatment-related changes and tumor progression, and offers no direct therapeutic options. Recent advances in imaging modalities have attempted to address some of these limitations, including positron emission tomography (PET), which has demonstrated success in delineating tumor margins and guiding the treatment of recurrent gliomas. Additionally, with the advent of theranostics in nuclear medicine, PET tracers, when combined with therapeutic agents, have also evolved beyond a purely diagnostic modality, serving both diagnostic and therapeutic roles. This review will discuss the growing involvement of theranostics in diagnosing and treating recurrent gliomas and address the associated impact on quality of life and functional recovery.
Collapse
Affiliation(s)
- Austin R. Hoggarth
- Department of Neurosurgery, Carilion Clinic, 1906 Belleview Avenue, Roanoke, VA 24014, USA;
- Virginia Tech Carilion School of Medicine, 2 Riverside Circle, Roanoke, VA 24016, USA; (S.M.); (S.M.T.)
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Sankar Muthukumar
- Virginia Tech Carilion School of Medicine, 2 Riverside Circle, Roanoke, VA 24016, USA; (S.M.); (S.M.T.)
| | - Steven M. Thomas
- Virginia Tech Carilion School of Medicine, 2 Riverside Circle, Roanoke, VA 24016, USA; (S.M.); (S.M.T.)
| | - James Crowley
- Carilion Clinic Radiology, Roanoke, VA 24016, USA; (J.C.); (J.K.)
| | - Jackson Kiser
- Carilion Clinic Radiology, Roanoke, VA 24016, USA; (J.C.); (J.K.)
| | - Mark R. Witcher
- Department of Neurosurgery, Carilion Clinic, 1906 Belleview Avenue, Roanoke, VA 24014, USA;
- Virginia Tech Carilion School of Medicine, 2 Riverside Circle, Roanoke, VA 24016, USA; (S.M.); (S.M.T.)
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
6
|
Valerius AR, Webb MJ, Hammad N, Sener U, Malani R. Cerebrospinal Fluid Liquid Biopsies in the Evaluation of Adult Gliomas. Curr Oncol Rep 2024; 26:377-390. [PMID: 38488990 DOI: 10.1007/s11912-024-01517-6] [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] [Accepted: 03/05/2024] [Indexed: 03/17/2024]
Abstract
PURPOSE OF REVIEW This review aims to discuss recent research regarding the biomolecules explored in liquid biopsies and their potential clinical uses for adult-type diffuse gliomas. RECENT FINDINGS Evaluation of tumor biomolecules via cerebrospinal fluid (CSF) is an emerging technology in neuro-oncology. Studies to date have already identified various circulating tumor DNA, extracellular vesicle, micro-messenger RNA and protein biomarkers of interest. These biomarkers show potential to assist in multiple avenues of central nervous system (CNS) tumor evaluation, including tumor differentiation and diagnosis, treatment selection, response assessment, detection of tumor progression, and prognosis. In addition, CSF liquid biopsies have the potential to better characterize tumor heterogeneity compared to conventional tissue collection and CNS imaging. Current imaging modalities are not sufficient to establish a definitive glioma diagnosis and repeated tissue sampling via conventional biopsy is risky, therefore, there is a great need to improve non-invasive and minimally invasive sampling methods. CSF liquid biopsies represent a promising, minimally invasive adjunct to current approaches which can provide diagnostic and prognostic information as well as aid in response assessment.
Collapse
Affiliation(s)
| | - Mason J Webb
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Nouran Hammad
- Jordan University of Science and Technology School of Medicine, Irbid, Jordan
| | - Ugur Sener
- Department of Neurology, Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Rachna Malani
- University of UT - Huntsman Cancer Institute (Department of Neurosurgery), Salt Lake City, UT, USA
| |
Collapse
|
7
|
Fan H, Luo Y, Gu F, Tian B, Xiong Y, Wu G, Nie X, Yu J, Tong J, Liao X. Artificial intelligence-based MRI radiomics and radiogenomics in glioma. Cancer Imaging 2024; 24:36. [PMID: 38486342 PMCID: PMC10938723 DOI: 10.1186/s40644-024-00682-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/03/2024] [Indexed: 03/18/2024] Open
Abstract
The specific genetic subtypes that gliomas exhibit result in variable clinical courses and the need to involve multidisciplinary teams of neurologists, epileptologists, neurooncologists and neurosurgeons. Currently, the diagnosis of gliomas pivots mainly around the preliminary radiological findings and the subsequent definitive surgical diagnosis (via surgical sampling). Radiomics and radiogenomics present a potential to precisely diagnose and predict survival and treatment responses, via morphological, textural, and functional features derived from MRI data, as well as genomic data. In spite of their advantages, it is still lacking standardized processes of feature extraction and analysis methodology among different research groups, which have made external validations infeasible. Radiomics and radiogenomics can be used to better understand the genomic basis of gliomas, such as tumor spatial heterogeneity, treatment response, molecular classifications and tumor microenvironment immune infiltration. These novel techniques have also been used to predict histological features, grade or even overall survival in gliomas. In this review, workflows of radiomics and radiogenomics are elucidated, with recent research on machine learning or artificial intelligence in glioma.
Collapse
Affiliation(s)
- Haiqing Fan
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Yilin Luo
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Fang Gu
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Bin Tian
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Yongqin Xiong
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Guipeng Wu
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Xin Nie
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Jing Yu
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Juan Tong
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Xin Liao
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China.
| |
Collapse
|
8
|
Hilario A, Salvador E, Cardenas A, Romero J, Lechuga C, Chen Z, Martinez de Aragon A, Perez-Nuñez A, Hernandez-Lain A, Sepulveda J, Lagares A, Toldos O, Rodriguez-Gonzalez V, Ramos A. Low rCBV values in glioblastoma tumor progression under chemoradiotherapy. Neuroradiology 2024; 66:317-323. [PMID: 38183424 DOI: 10.1007/s00234-023-03279-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 12/26/2023] [Indexed: 01/08/2024]
Abstract
PURPOSE After standard treatment for glioblastoma, perfusion MRI remains challenging for differentiating tumor progression from post-treatment changes. Our objectives were (1) to correlate rCBV values at diagnosis and at first tumor progression and (2) to analyze the relationship of rCBV values at tumor recurrence with enhancing volume, localization of tumor progression, and time elapsed since the end of radiotherapy in tumor recurrence. METHODS Inclusion criteria were (1) age > 18 years, (2) histologically confirmed glioblastoma treated with STUPP regimen, and (3) tumor progression according to RANO criteria > 12 weeks after radiotherapy. Co-registration of segmented enhancing tumor VOIs with dynamic susceptibility contrast perfusion MRI was performed using Olea Sphere software. For tumor recurrence, we correlated rCBV values with enhancing tumor volume, with recurrence localization, and with time elapsed from the end of radiotherapy to progression. Analyses were performed with SPSS software. RESULTS Sixty-four patients with glioblastoma were included in the study. Changes in rCBV values between diagnosis and first tumor progression were significant (p < 0.001), with a mean and median decreases of 32% and 46%, respectively. Mean rCBV values were also different (p < 0.01) when tumors progressed distally (radiation field rCBV values of 1.679 versus 3.409 distally). However, changes and, therefore, low rCBV values after radiotherapy in tumor recurrence were independent of time. CONCLUSION Chemoradiation alters tumor perfusion and rCBV values may be decreased in the setting of tumor progression. Changes in rCBV values with respect to diagnosis, with low rCBV in tumor progression, are independent of time but related to the site of recurrence.
Collapse
Affiliation(s)
- A Hilario
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain.
| | - E Salvador
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - A Cardenas
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - J Romero
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - C Lechuga
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - Z Chen
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - A Martinez de Aragon
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - A Perez-Nuñez
- Department of Neurosurgery, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - A Hernandez-Lain
- Department of Neuropathology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - J Sepulveda
- Department of Medical Oncology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - A Lagares
- Department of Neurosurgery, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - O Toldos
- Department of Neuropathology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - V Rodriguez-Gonzalez
- Department of Radiation Oncology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| | - A Ramos
- Department of Radiology, Hospital 12 de Octubre, Avenida de Cordoba s/n, 28041, Madrid, Spain
| |
Collapse
|
9
|
Bhattacharya K, Rastogi S, Mahajan A. Post-treatment imaging of gliomas: challenging the existing dogmas. Clin Radiol 2024; 79:e376-e392. [PMID: 38123395 DOI: 10.1016/j.crad.2023.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 10/23/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Gliomas are the commonest malignant central nervous system tumours in adults and imaging is the cornerstone of diagnosis, treatment, and post-treatment follow-up of these patients. With the ever-evolving treatment strategies post-treatment imaging and interpretation in glioma remains challenging, more so with the advent of anti-angiogenic drugs and immunotherapy, which can significantly alter the appearance in this setting, thus making interpretation of routine imaging findings such as contrast enhancement, oedema, and mass effect difficult to interpret. This review details the various methods of management of glioma including the upcoming novel therapies and their impact on imaging findings, with a comprehensive description of the imaging findings in conventional and advanced imaging techniques. A systematic appraisal for the existing and emerging techniques of imaging in these settings and their clinical application including various response assessment guidelines and artificial intelligence based response assessment will also be discussed.
Collapse
Affiliation(s)
- K Bhattacharya
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - S Rastogi
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - A Mahajan
- Department of imaging, The Clatterbridge Cancer Centre, NHS Foundation Trust, Pembroke Place, Liverpool L7 8YA, UK; University of Liverpool, Liverpool L69 3BX, UK.
| |
Collapse
|
10
|
Douri K, Iorio-Morin C, Mercure-Cyr R, Figueiredo G, Touchette CJ, Masson-Côté L, Mathieu D. Response Assessment in Brain Metastases Managed by Stereotactic Radiosurgery: A Reappraisal of the RANO-BM Criteria. Curr Oncol 2023; 30:9382-9391. [PMID: 37999099 PMCID: PMC10670467 DOI: 10.3390/curroncol30110679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Brain metastases (BM) are increasingly being treated using stereotactic radiosurgery (SRS). Standardized response criteria are necessary to improve research and treatment protocols. This study's goal was to validate the RANO-BM criteria thresholds for tumor progression in a cohort of patients with brain metastases managed using SRS. METHODS We performed a retrospective analysis of patients treated at least twice with SRS for brain metastases. Local progression, as defined by RANO-BM criteria, was compared to our multidisciplinary tumor board's treatment recommendation. A ROC curve was generated using varying diameter thresholds to assess the sensitivity and specificity of current RANO-BM criteria. RESULTS 249 metastases in 67 patients were included in the analysis. RANO-BM criteria current progression thresholds yielded a sensitivity of 38%, a specificity of 95%, a positive predictive value of 71%, and a negative predictive value of 84% relative to our tumor board's treatment recommendation. Modified RANO-BM criteria using absolute diameter differences of 2.5 mm yielded a sensitivity of 83%, a specificity of 87%, a positive predictive value of 67% and a negative predictive value of 94%. CONCLUSIONS Current RANO-BM criteria unreliably identifies clinically relevant tumor progression. The use of absolute diameter differences thresholds appears superior in our BM cohort.
Collapse
Affiliation(s)
- Keiss Douri
- Division of Neurosurgery, Department of Surgery, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada; (K.D.); (C.I.-M.); (C.J.T.)
| | - Christian Iorio-Morin
- Division of Neurosurgery, Department of Surgery, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada; (K.D.); (C.I.-M.); (C.J.T.)
| | - Rosalie Mercure-Cyr
- Division of Neurosurgery, Department of Surgery, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada; (K.D.); (C.I.-M.); (C.J.T.)
| | - Gabrielle Figueiredo
- Department of Radiology, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada;
| | - Charles Jean Touchette
- Division of Neurosurgery, Department of Surgery, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada; (K.D.); (C.I.-M.); (C.J.T.)
| | - Laurence Masson-Côté
- Division of Radiation Oncology, Department of Medicine, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada;
| | - David Mathieu
- Division of Neurosurgery, Department of Surgery, Université de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5H3, Canada; (K.D.); (C.I.-M.); (C.J.T.)
| |
Collapse
|
11
|
Liao D, Liu YC, Liu JY, Wang D, Liu XF. Differentiating tumour progression from pseudoprogression in glioblastoma patients: a monoexponential, biexponential, and stretched-exponential model-based DWI study. BMC Med Imaging 2023; 23:119. [PMID: 37697237 PMCID: PMC10494379 DOI: 10.1186/s12880-023-01082-7] [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: 12/10/2022] [Accepted: 08/19/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND To investigate the diagnostic performance of parameters derived from monoexponential, biexponential, and stretched-exponential diffusion-weighted imaging models in differentiating tumour progression from pseudoprogression in glioblastoma patients. METHODS Forty patients with pathologically confirmed glioblastoma exhibiting enhancing lesions after completion of chemoradiation therapy were enrolled in the study, which were then classified as tumour progression and pseudoprogression. All patients underwent conventional and multi-b diffusion-weighted MRI. The apparent diffusion coefficient (ADC) from a monoexponential model, the true diffusion coefficient (D), pseudodiffusion coefficient (D*) and perfusion fraction (f) from a biexponential model, and the distributed diffusion coefficient (DDC) and intravoxel heterogeneity index (α) from a stretched-exponential model were compared between tumour progression and pseudoprogression groups. Receiver operating characteristic curves (ROC) analysis was used to investigate the diagnostic performance of different DWI parameters. Interclass correlation coefficient (ICC) was used to evaluate the consistency of measurements. RESULTS The values of ADC, D, DDC, and α values were lower in tumour progression patients than that in pseudoprogression patients (p < 0.05). The values of D* and f were higher in tumour progression patients than that in pseudoprogression patients (p < 0.05). Diagnostic accuracy for differentiating tumour progression from pseudoprogression was highest for α(AUC = 0.94) than that for ADC (AUC = 0.91), D (AUC = 0.92), D* (AUC = 0.81), f (AUC = 0.75), and DDC (AUC = 0.88). CONCLUSIONS Multi-b DWI is a promising method for differentiating tumour progression from pseudoprogression with high diagnostic accuracy. In addition, the α derived from stretched-exponential model is the most promising DWI parameter for the prediction of tumour progression in glioblastoma patients.
Collapse
Affiliation(s)
- Dan Liao
- Department of Radiology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002 China
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010 China
| | - Yuan-Cheng Liu
- Department of Radiology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002 China
| | - Jiang-Yong Liu
- Department of Radiology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002 China
| | - Di Wang
- Department of Radiology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002 China
| | - Xin-Feng Liu
- Department of Radiology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002 China
| |
Collapse
|
12
|
Xiang Y, Tang W, Wang J, Wang Z, Bi N. Pseudoprogression of thoracic tumor after radiotherapy in the era of immunotherapy: a case series. Front Oncol 2023; 13:1021253. [PMID: 37576884 PMCID: PMC10419187 DOI: 10.3389/fonc.2023.1021253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 07/18/2023] [Indexed: 08/15/2023] Open
Abstract
Pseudoprogression is rarely mentioned after radiotherapy except for central nervous system tumors. With the widespread of immunotherapy, the incidence of pseudoprogression of thoracic tumor after radiotherapy is increasing. This study summarized the clinical features of pseudoprogression in 4 patients who had underwent thoracic radiotherapy after and/or followed by immunotherapy. All of them had received chemotherapy and immunotherapy before thoracic radiotherapy. After radiotherapy, pseudoprogression occurred within 3 months after initiation of immune consolidation/rechallenge therapy. At least a 20% increase in the sum of the longest diameter of target lesions were measured on their chest image. During this period, patients' ECOG PS scores remained stable, specific serum tumor markers did not increase significantly. Treatment strategies did not change after pseudoprogression. The causes of radiographic pseudoprogression in this case series may be attributed to disturbances such as pneumonitis, atelectasis, mucus blockages and infection. In the era of immunotherapy, pseudoprogression of thoracic tumors after chest radiotherapy might become a common phenomenon. It is important for us to identify pseudoprogression based on patient's general status, radiological changes, and laboratory tests.
Collapse
Affiliation(s)
- Yongbo Xiang
- Department of Radiation Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Tang
- Center for National Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianyang Wang
- Department of Radiation Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhijie Wang
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Bi
- Department of Radiation Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Mut M, Adiguzel Z, Cakir-Aktas C, Hanalioğlu Ş, Gungor-Topcu G, Kiyga E, Isikay I, Sarac A, Soylemezoglu F, Strobel T, Ampudia-Mesias E, Cameron C, Aslan T, Tekirdas E, Hayran M, Oguz KK, Henzler C, Saydam N, Saydam O. Extracellular-Vesicle-Based Cancer Panels Diagnose Glioblastomas with High Sensitivity and Specificity. Cancers (Basel) 2023; 15:3782. [PMID: 37568598 PMCID: PMC10417317 DOI: 10.3390/cancers15153782] [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: 05/31/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Glioblastoma is one of the most devastating neoplasms of the central nervous system. This study focused on the development of serum extracellular vesicle (EV)-based glioblastoma tumor marker panels that can be used in a clinic to diagnose glioblastomas and to monitor tumor burden, progression, and regression in response to treatment. RNA sequencing studies were performed using RNA isolated from serum EVs from both patients (n = 85) and control donors (n = 31). RNA sequencing results for preoperative glioblastoma EVs compared to control EVs revealed 569 differentially expressed genes (DEGs, 2XFC, FDR < 0.05). By using these DEGs, we developed serum-EV-based biomarker panels for the following glioblastomas: wild-type IDH1 (96% sensitivity/80% specificity), MGMT promoter methylation (91% sensitivity/73% specificity), p53 gene mutation (100% sensitivity/89% specificity), and TERT promoter mutation (89% sensitivity/100% specificity). This is the first study showing that serum-EV-based biomarker panels can be used to diagnose glioblastomas with a high sensitivity and specificity.
Collapse
Affiliation(s)
- Melike Mut
- Department of Neurosurgery, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey; (Ş.H.); (I.I.); (T.A.); (E.T.)
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara 06230, Turkey;
| | - Zelal Adiguzel
- TUBİTAK, GEBI, Gebze, Kocaeli 41470, Turkey; (Z.A.); (G.G.-T.); (E.K.); (A.S.)
- Faculty of Medicine KUTTAM, Koç University, Davutpaşa Street No. 4 Topkapi, Istanbul 34010, Turkey
| | - Canan Cakir-Aktas
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara 06230, Turkey;
| | - Şahin Hanalioğlu
- Department of Neurosurgery, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey; (Ş.H.); (I.I.); (T.A.); (E.T.)
| | - Gamze Gungor-Topcu
- TUBİTAK, GEBI, Gebze, Kocaeli 41470, Turkey; (Z.A.); (G.G.-T.); (E.K.); (A.S.)
| | - Ezgi Kiyga
- TUBİTAK, GEBI, Gebze, Kocaeli 41470, Turkey; (Z.A.); (G.G.-T.); (E.K.); (A.S.)
| | - Ilkay Isikay
- Department of Neurosurgery, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey; (Ş.H.); (I.I.); (T.A.); (E.T.)
| | - Aydan Sarac
- TUBİTAK, GEBI, Gebze, Kocaeli 41470, Turkey; (Z.A.); (G.G.-T.); (E.K.); (A.S.)
| | - Figen Soylemezoglu
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey;
| | - Thomas Strobel
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Elisabet Ampudia-Mesias
- Division of Hematology and Oncology, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN 55455, USA; (E.A.-M.); (C.C.)
| | - Charles Cameron
- Division of Hematology and Oncology, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN 55455, USA; (E.A.-M.); (C.C.)
| | - Tulay Aslan
- Department of Neurosurgery, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey; (Ş.H.); (I.I.); (T.A.); (E.T.)
| | - Eray Tekirdas
- Department of Neurosurgery, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey; (Ş.H.); (I.I.); (T.A.); (E.T.)
| | - Mutlu Hayran
- Department of Preventive Oncology, Hacettepe University Cancer Institute, Ankara 06230, Turkey;
| | - Kader Karli Oguz
- Department of Radiology, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey;
| | - Christine Henzler
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA;
| | | | - Okay Saydam
- Division of Hematology and Oncology, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN 55455, USA; (E.A.-M.); (C.C.)
| |
Collapse
|
14
|
Anzalone N, Politi LS, Caulo M. Editorial: Untangling post-treatment follow up of brain tumors: the role of neuroimaging. FRONTIERS IN RADIOLOGY 2023; 3:1204517. [PMID: 37492383 PMCID: PMC10364976 DOI: 10.3389/fradi.2023.1204517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/18/2023] [Indexed: 07/27/2023]
Affiliation(s)
- Nicoletta Anzalone
- Vita e Salute University, San Raffaele Hospital (IRCCS), Milan, Italy
- Neuroradiology Department, San Raffaele Hospital and Vita e Salute University, Milan, Italy
| | - Letterio S. Politi
- Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Massimo Caulo
- University of Studies G. d'Annunzio Chieti and Pescara, Chieti, Italy
- Radiology Department, D'Annunzio University, Chieti, Italy
| |
Collapse
|
15
|
Wang Y, Liu R, Zhang Q, Dong M, Wang D, Chen J, Ou Y, Luo H, Yang K, Wang X. Charged particle therapy for high-grade gliomas in adults: a systematic review. Radiat Oncol 2023; 18:29. [PMID: 36755321 PMCID: PMC9906872 DOI: 10.1186/s13014-022-02187-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/20/2022] [Indexed: 02/10/2023] Open
Abstract
High-grade gliomas are the most common intracranial malignancies, and their current prognosis remains poor despite standard aggressive therapy. Charged particle beams have unique physical and biological properties, especially high relative biological effectiveness (RBE) of carbon ion beam might improve the clinical treatment outcomes of malignant gliomas. We systematically reviewed the safety, efficacy, and dosimetry of carbon-ion or proton radiotherapy to treat high-grade gliomas. The protocol is detailed in the online PROSPERO database, registration No. CRD42021258495. PubMed, EMBASE, Web of Science, and The Cochrane Library databases were collected for data analysis on charged particle radiotherapy for high-grade gliomas. Until July 2022, two independent reviewers extracted data based on inclusion and exclusion criteria. Eleven articles were eligible for further analysis. Overall survival rates were marginally higher in patients with the current standard of care than those receiving concurrent intensity-modulated radiotherapy plus temozolomide. The most common side effects of carbon-ion-related therapy were grade 1-2 (such as dermatitis, headache, and alopecia). Long-term toxicities (more than three to six months) usually present as radiation necrosis; however, toxicities higher than grade 3 were not observed. Similarly, dermatitis, headache, and alopecia are among the most common acute side effects of proton therapy treatment. Despite improvement in survival rates, the method of dose-escalation using proton boost is associated with severe brain necrosis which should not be clinically underestimated. Regarding dosimetry, two studies compared proton therapy and intensity-modulated radiation therapy plans. Proton therapy plans aimed to minimize dose exposure to non-target tissues while maintaining target coverage. The use of charged-particle radiotherapy seems to be effective with acceptable adverse effects when used either alone or as a boost. The tendency of survival outcome shows that carbon ion boost is seemingly superior to proton boost. The proton beam could provide good target coverage, and it seems to reduce dose exposure to contralateral organs at risk significantly. This can potentially reduce the treatment-related dose- and volume-related side effects in long-term survivors, such as neurocognitive impairment. High-quality randomized control trials should be conducted in the future. Moreover, Systemic therapeutic options that can be paired with charged particles are necessary.
Collapse
Affiliation(s)
- Yuhang Wang
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Ruifeng Liu
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China ,grid.410726.60000 0004 1797 8419Department of Postgraduate, University of Chinese Academy of Sciences, Beijing, China ,Heavy Ion Therapy Center, Lanzhou Heavy Ions Hospital, Lanzhou, China
| | - Qiuning Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China. .,Department of Postgraduate, University of Chinese Academy of Sciences, Beijing, China. .,Heavy Ion Therapy Center, Lanzhou Heavy Ions Hospital, Lanzhou, China.
| | - Meng Dong
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Dandan Wang
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Junru Chen
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yuhong Ou
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Hongtao Luo
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China ,grid.410726.60000 0004 1797 8419Department of Postgraduate, University of Chinese Academy of Sciences, Beijing, China ,Heavy Ion Therapy Center, Lanzhou Heavy Ions Hospital, Lanzhou, China
| | - Kehu Yang
- grid.32566.340000 0000 8571 0482Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaohu Wang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China. .,The First School of Clinical Medicine, Lanzhou University, Lanzhou, China. .,Department of Postgraduate, University of Chinese Academy of Sciences, Beijing, China. .,Heavy Ion Therapy Center, Lanzhou Heavy Ions Hospital, Lanzhou, China.
| |
Collapse
|
16
|
Muthukumar S, Darden J, Crowley J, Witcher M, Kiser J. A Comparison of PET Tracers in Recurrent High-Grade Gliomas: A Systematic Review. Int J Mol Sci 2022; 24:ijms24010408. [PMID: 36613852 PMCID: PMC9820099 DOI: 10.3390/ijms24010408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
Humans with high-grade gliomas have a poor prognosis, with a mean survival time of just 12-18 months for patients who undergo standard-of-care tumor resection and adjuvant therapy. Currently, surgery and chemoradiotherapy serve as standard treatments for this condition, yet these can be complicated by the tumor location, growth rate and recurrence. Currently, gadolinium-based, contrast-enhanced magnetic resonance imaging (CE-MRI) serves as the predominant imaging modality for recurrent high-grade gliomas, but it faces several drawbacks, including its inability to distinguish tumor recurrence from treatment-related changes and its failure to reveal the entirety of tumor burden (de novo or recurrent) due to limitations inherent to gadolinium contrast. As such, alternative imaging modalities that can address these limitations, including positron emission tomography (PET), are worth pursuing. To this end, the identification of PET-based markers for use in imaging of recurrent high-grade gliomas is paramount. This review will highlight several PET radiotracers that have been implemented in clinical practice and provide a comparison between them to assess the efficacy of these tracers.
Collapse
Affiliation(s)
| | - Jordan Darden
- Carilion Clinic Neurosurgery, Roanoke, VA 24016, USA
| | | | - Mark Witcher
- Carilion Clinic Neurosurgery, Roanoke, VA 24016, USA
| | - Jackson Kiser
- Carilion Clinic Radiology, Roanoke, VA 24016, USA
- Correspondence:
| |
Collapse
|
17
|
Yao Y, Tan X, Yin W, Kou Y, Wang X, Jiang X, Chen S, Liu Y, Dang J, Yin J, Cheng Z. Performance of 18 F-FAPI PET/CT in assessing glioblastoma before radiotherapy: a pilot study. BMC Med Imaging 2022; 22:226. [PMID: 36566187 PMCID: PMC9789562 DOI: 10.1186/s12880-022-00952-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND We aimed to determine the performance of 18 F-FAPI PET/CT used for preprocedural assessment of glioblastoma before radiotherapy. METHODS Twelve glioblastoma patients having undergone incomplete surgical resection or biopsy were examined with 18 F-FAPI PET/CT and MRI scanning before radiotherapy. All patients had confirmed tumor residues according to findings of histopathological and/or long-term clinical and radiological follow-ups. Lesion characterization data, including SUVmax and tumor-to-background ratio (TBR) on PET/CT were attained. PET/CT and MRI findings were compared in terms of number of lesions. The correlation between immunohistochemistry, molecular expression, and PET/CT parameters was also evaluated. RESULTS 18 F-FAPI PET/CT detected 16 FAPI-avid out of 23 lesions in 12 patients described on MRI. MRI was statistically different from 18 F-FAPI PET/CT for lesion detection according to the exact McNemar statistical test (P = 0.0156). The SUVmax and TBR of the glioblastomas was 7.08 ± 3.55 and 19.95 ± 13.22, respectively. The sensitivity and positive predictive value (PPV) of 18 F-FAPI PET were 69.6% and 100%, respectively. Neither the Ki-67 index nor the molecular expression was correlated with the FAPI-PET/CT parameters. CONCLUSION 18 F-FAPI PET/CT detects glioblastomas at a lower rate than MRI. However, the 100% PPV of the examination may make it useful for differentiating controversial lesions detected on MRI. The 18 F-FAPI-avid lesions are displayed more clearly probably due to a higher TBR. 18 F-FAPI PET/CT imaging might find application in glioblastoma biopsy and radiotherapy planning.
Collapse
Affiliation(s)
- Yutang Yao
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Xiaofei Tan
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Wenya Yin
- grid.54549.390000 0004 0369 4060Department of Radiation Oncology, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, 610041 Chengdu, China
| | - Ying Kou
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Xiaoxiong Wang
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Xiao Jiang
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China ,grid.410655.30000 0001 0157 8259Institute of Isotope, China Institute of Atomic Energy, 102413 Beijing, China
| | - Shirong Chen
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Yongli Liu
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Jun Dang
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| | - Jun Yin
- grid.54549.390000 0004 0369 4060Department of Radiation Oncology, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, 610041 Chengdu, China
| | - Zhuzhong Cheng
- grid.54549.390000 0004 0369 4060Department of Nuclear Medicine, Sichuan Cancer Hospital&Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, No.55, Section 4, South People’s Road, Sichuan 610041 Chengdu, China
| |
Collapse
|
18
|
Zhao M, Ma H, Cheng P, Yang H, Zhao Y, Han Q. Apatinib combined with temozolomide treatment for pseudoprogression in glioblastoma: A case report. Medicine (Baltimore) 2022; 101:e32156. [PMID: 36626518 PMCID: PMC9750629 DOI: 10.1097/md.0000000000032156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
RATIONALE Glioblastoma is the most common malignant tumor of the central nervous system, which originates from glial cells and corresponding precursors. Due to its strong invasion and rapid growth, the prognosis of patients after treatment is very poor and easy to relapse. PATIENT CONCERNS In August 2015, a 48 years old man with a relapse of glioblastoma. DIAGNOSES The patient was diagnosed by computed tomography, magnetic resonance imaging, and pathological biopsy in this case report. INTERVENTIONS The patient underwent 2 surgeries, radiotherapy, and multiple regular chemotherapy sessions over the next 6 years. Apatinib, an inhibitor of vascular endothelial growth factor receptor 2 was given to treat recurrent glioma. OUTCOMES It was found that radiotherapy combined with temozolomide administration often increased the size of the original lesion or produced a new glioblastoma lesion. The lesion development was similar to tumor progression, which was called pseudoprogression. And it significantly prolonged the survival of this patient. LESSONS Surgery, radiotherapy and chemotherapy with apatinib and temozolomide are effective to treat the patients with pseudoprogression in glioblastoma.
Collapse
Affiliation(s)
- Mingming Zhao
- First Ward of Cancer Center, People’s Hospital of Henan University, Zhengzhou, China
| | - Haodong Ma
- First Ward of Cancer Center, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Peng Cheng
- First Ward of Cancer Center, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Hongjie Yang
- First Ward of Cancer Center, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yang Zhao
- First Ward of Cancer Center, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Qian Han
- First Ward of Cancer Center, Henan Provincial People’s Hospital, Zhengzhou, China
- * Correspondence: Qian Han, First Ward of Cancer Center, Henan Provincial People’s Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China (e-mail: )
| |
Collapse
|
19
|
Jing H, Yang F, Peng K, Qin D, He Y, Yang G, Zhang H. Multimodal MRI-Based Radiomic Nomogram for the Early Differentiation of Recurrence and Pseudoprogression of High-Grade Glioma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4667117. [PMID: 36246986 PMCID: PMC9553483 DOI: 10.1155/2022/4667117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/18/2022]
Abstract
Objective To evaluate the diagnostic value of multimodal MRI radiomics based on T2-weighted fluid attenuated inversion recovery imaging (T2WI-FLAIR) combined with T1-weighted contrast enhanced imaging (T1WI-CE) in the early differentiation of high-grade glioma recurrence from pseudoprogression. Methods A total of one hundred eighteen patients with brain gliomas who were diagnosed from March 2014 to April 2020 were retrospectively analyzed. According to the clinical characteristics, the patients were randomly split into a training group (n = 83) and a test group (n = 35) at a 7 : 3 ratio. The region of interest (ROI) was delineated, and 2632 radiomic features were extracted. We used multiple logistic regression to establish a classification model, including the T1 model, T2 model, and T1 + T2 model, to differentiate recurrence from pseudoprogression. The diagnostic efficiency of the model was evaluated by calculating the area under the receiver operating characteristic curve (AUC) and accuracy (ACC) and by analyzing the calibration curve of the nomogram and decision curve. Results There were 75 cases of recurrence and 43 cases of pseudoprogression. The diagnostic efficacies of the multimodal MRI-based radiomic model were relatively high. The AUC values and ACC of the training group were 0.831 and 77.11%, respectively, and the AUC values and ACC of the test group were 0.829 and 88.57%, respectively. The calibration curve of the nomogram showed that the discrimination probability was consistent with the actual occurrence in the training group, and the discrimination probability was roughly the same as the actual occurrence in the test group. In the decision curve analysis, the T1 + T2 model showed greater overall net efficiency. Conclusion The multimodal MRI radiomic model has relatively high efficiency in the early differentiation of recurrence from pseudoprogression, and it could be helpful for clinicians in devising correct treatment plans so that patients can be treated in a timely and accurate manner.
Collapse
Affiliation(s)
- Hui Jing
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Radiology, The Sixth Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Fan Yang
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Kun Peng
- Department of Radiology, The Sixth Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Danlei Qin
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yexin He
- Department of Radiology, Shanxi Provincial People's Hospital, Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
| | - Guoqiang Yang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hui Zhang
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi Province, China
- Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
20
|
The Role of Apparent Diffusion Coefficient Values in Glioblastoma: Differentiating Tumor Progression Versus Treatment-Related Changes. J Comput Assist Tomogr 2022; 46:923-928. [PMID: 36112011 DOI: 10.1097/rct.0000000000001373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Glioblastoma represents the most common primary brain malignancy with a median survival of 15 months. Follow-up examinations are crucial to establish the presence of tumor recurrence, as well as treatment-associated changes such as ischemic infarction and radiation effects. Even though magnetic resonance imaging is a valuable tool, a histopathological diagnosis is often required because of imaging overlap between tumor recurrence and treatment associated changes. We set out to measure the apparent diffusion coefficient (ADC) values of the lesions in magnetic resonance imaging scans of treated glioblastoma patients to investigate if ADC values could accurately differentiate between tumor progression, radiation-related changes, and ischemic infarctions. METHODS We evaluated ADC values among 3 groups, patients with tumor progression, radiation necrosis, and ischemic infarctions. The regions of interest were placed in the areas of greatest hypointensity among solid lesions using the ADC maps, excluding areas with necrotic, cystic, or hemorrhagic changes. The ADC values of the contralateral normal appearing white matter were also measured as the reference value for each patient. The relative ADC (rADC) values were measured for all 3 groups. Comparison between lesions and normal white matter was evaluated by Wilcoxon signed test. RESULTS A total of 157 patients were included in the study; 49 patients classified as tumor progression, 58 patients as radiation necrosis, and 50 patients as ischemic infarctions. The mean ± SD ADC value was 752.8 ± 132.5 for tumor progression, 479.0 ± 105.2 for radiation-related changes, and 250.1 ± 57.2 for ischemic infarctions. The mean ± SD rADC value was 1.07 ± 0.22 for tumor progression, 0.66 ± 0.14 for radiation necrosis, and 0.34 ± 0.08 for ischemic infarctions. The mean rADC values were significantly higher in tumor progression, compared with both radiation necrosis and ischemic changes (P < 0.001). CONCLUSIONS The present study demonstrates that ADC values are a helpful tool to differentiate between tumor progression, radiation necrosis, and posttreatment ischemic changes.
Collapse
|
21
|
Kim S, Hoch MJ, Peng L, Somasundaram A, Chen Z, Weinberg BD. A brain tumor reporting and data system to optimize imaging surveillance and prognostication in high-grade gliomas. J Neuroimaging 2022; 32:1185-1192. [PMID: 36045502 DOI: 10.1111/jon.13044] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE High-grade glioma (HGG), including glioblastoma, is the most common primary brain neoplasm and has a dismal prognosis. After initial treatment, follow-up decisions are guided by longitudinal MRI performed at routine intervals. The Brain Tumor Reporting and Data System (BT-RADS) is a proposed structured reporting system for posttreatment brain MRIs. The purpose of this study is to determine the relationship between BT-RADS scores and overall survival in HGG patients. METHODS Chart review of grade 4 glioma patients who had an MRI at a single institution from November 2018 to November 2019 was performed. BT-RADS scores, tumor characteristics, and overall survival were recorded. Likelihood of improvement, stability, or worsening on the subsequent study was calculated for each score. Survival analysis was performed using Kaplan-Meier method, log-rank test, and a time-dependent cox model. Significance level of .05 was used. RESULTS The study identified 91 HGG patients who underwent a total of 538 MRIs. Mean age of patients was 57 years old. Score with the highest likelihood for worsening on the next follow-up was 3b. The risk of death was 53% higher with each incremental increase in BT-RADS scores (hazard ratio, 1.53; 95% confidence interval [CI], 1.07-2.19; p = .019). The risk of death was 167% higher in O-6-methylguanine-DNA-methyltransferase unmethylated tumors (hazard ratio, 2.67; 95% CI, 1.34-5.33; p = .005). CONCLUSIONS BT-RADS scores can be used as a reference guide to anticipate whether patients' subsequent MRI will be improved, stable, or worsened. The scoring system can also be used to predict clinical outcomes and prognosis.
Collapse
Affiliation(s)
- Sera Kim
- Department of Radiology, University of California, San Francisco, San Francisco, California, USA
| | - Michael J Hoch
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lingyi Peng
- Department of Statistics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aravind Somasundaram
- Department of Radiology and Imaging Sciences, Emory University Hospital, Atlanta, Georgia, USA
| | - Zhengjia Chen
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Brent D Weinberg
- Department of Radiology and Imaging Sciences, Emory University Hospital, Atlanta, Georgia, USA
| |
Collapse
|
22
|
Eichkorn T, Lischalk JW, Sandrini E, Meixner E, Regnery S, Held T, Bauer J, Bahn E, Harrabi S, Hörner-Rieber J, Herfarth K, Debus J, König L. Iatrogenic Influence on Prognosis of Radiation-Induced Contrast Enhancements in Patients with Glioma WHO 1-3 following Photon and Proton Radiotherapy. Radiother Oncol 2022; 175:133-143. [PMID: 36041565 DOI: 10.1016/j.radonc.2022.08.025] [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: 03/17/2022] [Revised: 07/20/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Radiation-induced contrast enhancement (RICE) is a common side effect following radiotherapy for glioma, but both diagnosis and handling are challenging. Due to the potential risks associated with RICE and its challenges in differentiating RICE from tumor progression, it is critical to better understand how RICE prognosis depends on iatrogenic influence. MATERIALS AND METHODS We identified 99 patients diagnosed with RICE who were previously treated with either photon or proton therapy for World Health Organization (WHO) grade 1-3 primary gliomas. Post-treatment brain MRI-based volumetric analysis and clinical data collection was performed at multiple time points. RESULTS The most common histologic subtypes were astrocytoma (50%) and oligodendroglioma (46%). In 67%, it was graded WHO grade 2 and in 86% an IDH mutation was present. RICE first occurred after 16 months (range: 1 - 160) in median. At initial RICE occurrence, 39% were misinterpreted as tumor progression. A tumor-specific therapy including chemotherapy or re-irradiation led to a RICE size progression in 86% and 92% of cases, respectively and RICE symptom progression in 57% and 65% of cases, respectively. A RICE-specific therapy such as corticosteroids or Bevacizumab for larger or symptomatic RICE led to a RICE size regression in 81% of cases with symptom stability or regression in 62% of cases. CONCLUSIONS While with chemotherapy and re-irradiation a RICE progression was frequently observed, anti-edematous or anti-VEGF treatment frequently went along with a RICE regression. For RICE, correct diagnosis and treatment decisions are challenging and critical and should be made interdisciplinarily.
Collapse
Affiliation(s)
- Tanja Eichkorn
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany; National Center for Tumor diseases (NCT), Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Jonathan W Lischalk
- Department of Radiation Oncology, Perlmutter Cancer Center at New York University Langone Health at Long Island, New York, NY, USA.
| | - Elisabetta Sandrini
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany; National Center for Tumor diseases (NCT), Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Eva Meixner
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany; National Center for Tumor diseases (NCT), Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Sebastian Regnery
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany; National Center for Tumor diseases (NCT), Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Thomas Held
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany; National Center for Tumor diseases (NCT), Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Julia Bauer
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Emanuel Bahn
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Semi Harrabi
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany; National Center for Tumor diseases (NCT), Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Juliane Hörner-Rieber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany; National Center for Tumor diseases (NCT), Heidelberg, Germany; Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; German Cancer Consortium (DKTK), partner site Heidelberg, Germany.
| | - Klaus Herfarth
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany; National Center for Tumor diseases (NCT), Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany; National Center for Tumor diseases (NCT), Heidelberg, Germany; Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; German Cancer Consortium (DKTK), partner site Heidelberg, Germany.
| | - Laila König
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany; National Center for Tumor diseases (NCT), Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
23
|
Mammadov O, Akkurt BH, Musigmann M, Ari AP, Blömer DA, Kasap DN, Henssen DJ, Nacul NG, Sartoretti E, Sartoretti T, Backhaus P, Thomas C, Stummer W, Heindel W, Mannil M. Radiomics for pseudoprogression prediction in high grade gliomas: added value of MR contrast agent. Heliyon 2022; 8:e10023. [PMID: 35965975 PMCID: PMC9364026 DOI: 10.1016/j.heliyon.2022.e10023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/04/2022] [Accepted: 07/18/2022] [Indexed: 10/31/2022] Open
Abstract
Objective Material & methods Results Conclusion Radiomics allows for prediction of pseudoprogression in high-grade gliomas. Use of contrast media boosts the performance of the Radiomics prediction model.
Collapse
|
24
|
The Value of Dynamic Contrast-Enhanced Magnetic Resonance Imaging (DCE-MRI) in the Differentiation of Pseudoprogression and Recurrence of Intracranial Gliomas. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:5680522. [PMID: 35935318 PMCID: PMC9337951 DOI: 10.1155/2022/5680522] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022]
Abstract
Objective The objective of this study was to determine the value of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) in assessing postoperative changes in intracranial gliomas. Method A total of fifty-one patients who had new enhanced lesions after surgical resection followed by standard radiotherapy and chemotherapy were collected retrospectively from October 2014 to June 2021. The patients were divided into a pseudoprogression group (15 cases) and a recurrence group (36 cases) according to the pathological results of the second operation or a follow-up of more than six months. The follow-up data of all patients were complete, and DCE-MRI was performed. The images were processed to obtain the quantitative parameters Ktrans, Ve, and Kep and the semiquantitative parameter iAUC, which were analysed with relevant statistical software. Results First, the difference in Ktrans and iAUC values between the two groups was statistically significant (P < 0.05), and the difference in Ve and Kep values was not statistically significant (P > 0.05). Second, by comparing the area under the curve, threshold, sensitivity and specificity of Ktrans, and iAUC, it was found that the iAUC threshold value was slightly higher than that of Ktrans, and the specificity of Ktrans was equal to that of iAUC, while the area under the curve and sensitivity of Ktrans were higher than those of iAUC. Third, Ktrans and iAUC had high accuracy in diagnosing recurrence and pseudoprogression, and Ktrans had higher accuracy than iAUC. Conclusion In this study, DCE-MRI has a certain diagnostic value in the early differentiation of recurrence and pseudoprogression, offering a new method for the diagnosis and assessment of gliomas after surgery.
Collapse
|
25
|
Krishnakumar HN, Son C. Delayed cerebral pseudoaneurysm following surgical and combined-modality therapy for glioblastoma multiforme: illustrative case. JOURNAL OF NEUROSURGERY: CASE LESSONS 2022; 4:CASE22129. [PMID: 35855012 PMCID: PMC9274294 DOI: 10.3171/case22129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/18/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND
Post–radiation therapy and chemotherapy cerebral pseudoaneurysms are rare entities. Within previous tumor treatment areas on nonvascular imaging, they are potentially confused as recurrent tumor.
OBSERVATIONS
A 61-year-old man was a long-term survivor of glioblastoma multiforme whose treatment consisted of open biopsy followed by radiotherapy to 60 Gy and systemic carmustine. On surveillance imaging, enlargement of a posttreatment cyst and new enhancing lateral “mural nodule” was first noticed approximately 16 years after initial treatment. Over 12 months, both continued to enlarge. Initially referred to as recurrence, subsequent angiography showed the mural nodule to be an unruptured distal middle cerebral artery pseudoaneurysm within the previous tumor bed. The patient underwent repeat craniotomy for clipping of the aneurysm and biopsy of the cyst wall, which was negative for malignancy.
LESSONS
Delayed pseudoaneurysms following radiation therapy and chemotherapy for malignant brain tumors are rare but have been previously reported. Their appearance on cross-sectional imaging can mimic recurrence, and they should be kept in the differential of new, circumscribed enhancement within such treatment areas.
Collapse
Affiliation(s)
- Hari N. Krishnakumar
- Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Colin Son
- Neurosurgical Associates of San Antonio, San Antonio, Texas; and
- School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, Texas
| |
Collapse
|
26
|
Greuter L, Frank N, Guzman R, Soleman J. The Clinical Applications of Liquid Biopsies in Pediatric Brain Tumors: A Systematic Literature Review. Cancers (Basel) 2022; 14:cancers14112683. [PMID: 35681663 PMCID: PMC9179879 DOI: 10.3390/cancers14112683] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/03/2022] [Accepted: 05/25/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Brain tumors are the most common solid cancer in children and are traditionally diagnosed via a tissue biopsy or resection. Liquid biopsy offers the possibility to characterize brain tumors based on their circulating DNA in blood, cerebrospinal fluid or even urine. Moreover, disease progress can be monitored accurately and sometimes even detected before radiographic progression. More trials are needed to standardize the use of liquid biopsy in pediatric brain tumors. Abstract Background: Pediatric brain tumors are the most common solid tumor in children. Traditionally, tumor diagnosis and molecular analysis were carried out on tumor tissue harvested either via biopsy or resection. However, liquid biopsy allows analysis of circulating tumor DNA in corporeal fluids such as cerebrospinal fluid or blood. Methods: We performed a systematic review in Pubmed and Embase regarding the role of liquid biopsy in pediatric brain tumors. Results: Nine studies with a total of 570 patients were included. The preferred corporeal fluid for analysis with a relatively high yield of ct-DNA was cerebrospinal fluid (CSF). For high-grade glioma, liquid biopsy can successfully characterize H3K27mutations and predict tumor progression before it is radiographically detected. Moreover, liquid biopsy has the potential to distinguish between pseudo-progression and actual progression. In medulloblastoma, ct-DNA in the CSF can be used as a surrogate marker of measurable residual disease and correlates with response to therapy and progression of the tumor up to three months before radiographic detection. Conclusion: Liquid biopsy is primarily useful in high-grade pediatric brain tumors such as diffuse midline glioma or medulloblastoma. Disease detection and monitoring is feasible for both tumor entities. More trials to standardize its use for pediatric brain tumors are necessary.
Collapse
Affiliation(s)
- Ladina Greuter
- Department of Neurosurgery, University Hospital of Basel, 4031 Basel, Switzerland; (N.F.); (R.G.); (J.S.)
- Correspondence:
| | - Nicole Frank
- Department of Neurosurgery, University Hospital of Basel, 4031 Basel, Switzerland; (N.F.); (R.G.); (J.S.)
| | - Raphael Guzman
- Department of Neurosurgery, University Hospital of Basel, 4031 Basel, Switzerland; (N.F.); (R.G.); (J.S.)
- Department of Neurosurgery and Pediatric Neurosurgery, University Hospital of Basel and Children’s Hospital, 4056 Basel, Switzerland
- Faculty of Medicine, University of Basel, 4056 Basel, Switzerland
| | - Jehuda Soleman
- Department of Neurosurgery, University Hospital of Basel, 4031 Basel, Switzerland; (N.F.); (R.G.); (J.S.)
- Department of Neurosurgery and Pediatric Neurosurgery, University Hospital of Basel and Children’s Hospital, 4056 Basel, Switzerland
- Faculty of Medicine, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
27
|
Sun P, Wu Z, Xiao Y, Wu H, Di Q, Zhao X, Quan J, Tang H, Wang Q, Chen W. TfR-T12 short peptide and pH sensitive cell transmembrane peptide modified nano-composite micelles for glioma treatment via remodeling tumor microenvironment. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 41:102516. [PMID: 35131469 DOI: 10.1016/j.nano.2022.102516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 11/18/2021] [Accepted: 01/07/2022] [Indexed: 10/19/2022]
Abstract
Two kinds of amphiphilic block copolymers of TfR-T12-PEG-PLGA and TATH7-PEG-PLGA were synthesized to self-assembly nano-composite micelles for encapsulating paclitaxel and imiquimod synchronously. TfR-T12 peptide modified nano-composite micelles can pass through BBB in a TfR-mediated way to achieve targeted delivery of chemotherapeutic drugs, and pH sensitive TATH7 peptide modified nano-composite micelles enhanced uptake efficiency more significantly under pH 5.5 medium than pH 7.4 medium. The results of pharmacodynamic evaluation in vivo showed that the nano-composite micelles had achieved good anti-tumor effect in subcutaneous and normotopia glioma models, and effectively prolonged the life cycle of tumor-bearing mice. The nano-composite micelles regulated the immunosuppression phenomenon of tumor microenvironment significantly, and promoted the M1 polarization of TAMs, then enhanced the proliferation and activation of CD8+ T cells in tumor microenvironment. It comes to conclusion that the nano-composite micelle achieves the purpose of effective treatment of glioma by chemotherapy combined with immunotherapy.
Collapse
Affiliation(s)
- Ping Sun
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China; Institute of Pharmaceutics, School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Zherui Wu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Yue Xiao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Han Wu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Qianqian Di
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Xibao Zhao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Jiazheng Quan
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Haimei Tang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weilin Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China.
| |
Collapse
|
28
|
Gonçalves SI, Simões RV, Shemesh N. Short TE downfield magnetic resonance spectroscopy in a mouse model of brain glioma. Magn Reson Med 2022; 88:524-536. [PMID: 35315536 DOI: 10.1002/mrm.29243] [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: 07/27/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 11/11/2022]
Abstract
PURPOSE Enhanced cell proliferation in tumors can be associated with altered metabolic profiles and dramatic microenvironmental changes. Downfield magnetic resonance spectroscopy (MRS) has received increasing attention due to its ability to report on labile resonances of molecules not easily detected in upfield 1 H MRS. Image-selected-in-vivo-spectroscopy-relaxation enhanced MRS (iRE-MRS) was recently introduced for acquiring short echo-time (TE) spectra. Here, iRE-MRS was used to investigate in-vivo downfield spectra in glioma-bearing mice. METHODS Experiments were performed in vivo in an immunocompetent glioma mouse model at 9.4 T using a cryogenic coil. iRE-MRS spectra were acquired in N = 6 glioma-bearing mice (voxel size = 2.23 mm3 ) and N = 6 control mice. Spectra were modeled by a sum of Lorentzian peaks simulating known downfield resonances, and differences between controls and tumors were quantified using relative peak areas. RESULTS Short TE tumor spectra exhibited large qualitative differences compared to control spectra. Most peaks appeared modulated, with strong attenuation of NAA (∼7.82, 7.86 ppm) and changes in relative peak areas between 6.75 and 8.49 ppm. Peak areas tended to be smaller for DF6.83 , DF7.60 , DF8.18 and NAA; and larger for DF7.95 and DF8.24 . Differences were also detected in signals resonating above 8.5 ppm, assumed to arise from NAD+. CONCLUSIONS In-vivo downfield 1 H iRE-MRS of mouse glioma revealed differences between controls and tumor bearing mice, including in metabolites which are not easily detectable in the more commonly investigated upfield spectrum. These findings motivate future downfield MRS investigations exploring pH and exchange contributions to these differences.
Collapse
Affiliation(s)
| | - Rui V Simões
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Noam Shemesh
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
29
|
Hemodynamic Imaging in Cerebral Diffuse Glioma-Part A: Concept, Differential Diagnosis and Tumor Grading. Cancers (Basel) 2022; 14:cancers14061432. [PMID: 35326580 PMCID: PMC8946242 DOI: 10.3390/cancers14061432] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 11/17/2022] Open
Abstract
Diffuse gliomas are the most common primary malignant intracranial neoplasms. Aside from the challenges pertaining to their treatment-glioblastomas, in particular, have a dismal prognosis and are currently incurable-their pre-operative assessment using standard neuroimaging has several drawbacks, including broad differentials diagnosis, imprecise characterization of tumor subtype and definition of its infiltration in the surrounding brain parenchyma for accurate resection planning. As the pathophysiological alterations of tumor tissue are tightly linked to an aberrant vascularization, advanced hemodynamic imaging, in addition to other innovative approaches, has attracted considerable interest as a means to improve diffuse glioma characterization. In the present part A of our two-review series, the fundamental concepts, techniques and parameters of hemodynamic imaging are discussed in conjunction with their potential role in the differential diagnosis and grading of diffuse gliomas. In particular, recent evidence on dynamic susceptibility contrast, dynamic contrast-enhanced and arterial spin labeling magnetic resonance imaging are reviewed together with perfusion-computed tomography. While these techniques have provided encouraging results in terms of their sensitivity and specificity, the limitations deriving from a lack of standardized acquisition and processing have prevented their widespread clinical adoption, with current efforts aimed at overcoming the existing barriers.
Collapse
|
30
|
Stumpo V, Guida L, Bellomo J, Van Niftrik CHB, Sebök M, Berhouma M, Bink A, Weller M, Kulcsar Z, Regli L, Fierstra J. Hemodynamic Imaging in Cerebral Diffuse Glioma-Part B: Molecular Correlates, Treatment Effect Monitoring, Prognosis, and Future Directions. Cancers (Basel) 2022; 14:1342. [PMID: 35267650 PMCID: PMC8909110 DOI: 10.3390/cancers14051342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/05/2023] Open
Abstract
Gliomas, and glioblastoma in particular, exhibit an extensive intra- and inter-tumoral molecular heterogeneity which represents complex biological features correlating to the efficacy of treatment response and survival. From a neuroimaging point of view, these specific molecular and histopathological features may be used to yield imaging biomarkers as surrogates for distinct tumor genotypes and phenotypes. The development of comprehensive glioma imaging markers has potential for improved glioma characterization that would assist in the clinical work-up of preoperative treatment planning and treatment effect monitoring. In particular, the differentiation of tumor recurrence or true progression from pseudoprogression, pseudoresponse, and radiation-induced necrosis can still not reliably be made through standard neuroimaging only. Given the abundant vascular and hemodynamic alterations present in diffuse glioma, advanced hemodynamic imaging approaches constitute an attractive area of clinical imaging development. In this context, the inclusion of objective measurable glioma imaging features may have the potential to enhance the individualized care of diffuse glioma patients, better informing of standard-of-care treatment efficacy and of novel therapies, such as the immunotherapies that are currently increasingly investigated. In Part B of this two-review series, we assess the available evidence pertaining to hemodynamic imaging for molecular feature prediction, in particular focusing on isocitrate dehydrogenase (IDH) mutation status, MGMT promoter methylation, 1p19q codeletion, and EGFR alterations. The results for the differentiation of tumor progression/recurrence from treatment effects have also been the focus of active research and are presented together with the prognostic correlations identified by advanced hemodynamic imaging studies. Finally, the state-of-the-art concepts and advancements of hemodynamic imaging modalities are reviewed together with the advantages derived from the implementation of radiomics and machine learning analyses pipelines.
Collapse
Affiliation(s)
- Vittorio Stumpo
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Lelio Guida
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Jacopo Bellomo
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Christiaan Hendrik Bas Van Niftrik
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Martina Sebök
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Moncef Berhouma
- Department of Neurosurgical Oncology and Vascular Neurosurgery, Pierre Wertheimer Neurological and Neurosurgical Hospital, Hospices Civils de Lyon, 69500 Lyon, France;
| | - Andrea Bink
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
- Department of Neuroradiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Michael Weller
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
- Department of Neurology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Zsolt Kulcsar
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
- Department of Neuroradiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| | - Jorn Fierstra
- Department of Neurosurgery, University Hospital Zurich, 8091 Zurich, Switzerland; (L.G.); (J.B.); (C.H.B.V.N.); (M.S.); (L.R.); (J.F.)
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, 8057 Zurich, Switzerland; (A.B.); (M.W.); (Z.K.)
| |
Collapse
|
31
|
Carrete LR, Young JS, Cha S. Advanced Imaging Techniques for Newly Diagnosed and Recurrent Gliomas. Front Neurosci 2022; 16:787755. [PMID: 35281485 PMCID: PMC8904563 DOI: 10.3389/fnins.2022.787755] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Management of gliomas following initial diagnosis requires thoughtful presurgical planning followed by regular imaging to monitor treatment response and survey for new tumor growth. Traditional MR imaging modalities such as T1 post-contrast and T2-weighted sequences have long been a staple of tumor diagnosis, surgical planning, and post-treatment surveillance. While these sequences remain integral in the management of gliomas, advances in imaging techniques have allowed for a more detailed characterization of tumor characteristics. Advanced MR sequences such as perfusion, diffusion, and susceptibility weighted imaging, as well as PET scans have emerged as valuable tools to inform clinical decision making and provide a non-invasive way to help distinguish between tumor recurrence and pseudoprogression. Furthermore, these advances in imaging have extended to the operating room and assist in making surgical resections safer. Nevertheless, surgery, chemotherapy, and radiation treatment continue to make the interpretation of MR changes difficult for glioma patients. As analytics and machine learning techniques improve, radiomics offers the potential to be more quantitative and personalized in the interpretation of imaging data for gliomas. In this review, we describe the role of these newer imaging modalities during the different stages of management for patients with gliomas, focusing on the pre-operative, post-operative, and surveillance periods. Finally, we discuss radiomics as a means of promoting personalized patient care in the future.
Collapse
Affiliation(s)
- Luis R. Carrete
- University of California San Francisco School of Medicine, San Francisco, CA, United States
| | - Jacob S. Young
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Jacob S. Young,
| | - Soonmee Cha
- Department of Radiology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
32
|
Stumpo V, Sebök M, van Niftrik CHB, Seystahl K, Hainc N, Kulcsar Z, Weller M, Regli L, Fierstra J. Feasibility of glioblastoma tissue response mapping with physiologic BOLD imaging using precise oxygen and carbon dioxide challenge. MAGMA (NEW YORK, N.Y.) 2022; 35:29-44. [PMID: 34874499 DOI: 10.1007/s10334-021-00980-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Innovative physiologic MRI development focuses on depiction of heterogenous vascular and metabolic features in glioblastoma. For this feasibility study, we employed blood oxygenation level-dependent (BOLD) MRI with standardized and precise carbon dioxide (CO2) and oxygen (O2) modulation to investigate specific tumor tissue response patterns in patients with newly diagnosed glioblastoma. MATERIALS AND METHODS Seven newly diagnosed untreated patients with suspected glioblastoma were prospectively included to undergo a BOLD study with combined CO2 and O2 standardized protocol. %BOLD signal change/mmHg during hypercapnic, hypoxic, and hyperoxic stimulus was calculated in the whole brain, tumor lesion and segmented volumes of interest (VOI) [contrast-enhancing (CE) - tumor, necrosis and edema] to analyze their tissue response patterns. RESULTS Quantification of BOLD signal change after gas challenges can be used to identify specific responses to standardized stimuli in glioblastoma patients. Integration of this approach with automatic VOI segmentation grants improved characterization of tumor subzones and edema. Magnitude of BOLD signal change during the 3 stimuli can be visualized at voxel precision through color-coded maps overlayed onto whole brain and identified VOIs. CONCLUSIONS Our preliminary investigation shows good feasibility of BOLD with standardized and precise CO2 and O2 modulation as an emerging physiologic imaging technique to detail specific glioblastoma characteristics. The unique tissue response patterns generated can be further investigated to better detail glioblastoma lesions and gauge treatment response.
Collapse
Affiliation(s)
- Vittorio Stumpo
- Department of Neurosurgery, University Hospital Zurich, Frauenklinikstrasse 10, 8091, Zurich, Switzerland. .,Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Martina Sebök
- Department of Neurosurgery, University Hospital Zurich, Frauenklinikstrasse 10, 8091, Zurich, Switzerland.,Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christiaan Hendrik Bas van Niftrik
- Department of Neurosurgery, University Hospital Zurich, Frauenklinikstrasse 10, 8091, Zurich, Switzerland.,Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Katharina Seystahl
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Nicolin Hainc
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland
| | - Zsolt Kulcsar
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Weller
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, University Hospital Zurich, Frauenklinikstrasse 10, 8091, Zurich, Switzerland.,Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jorn Fierstra
- Department of Neurosurgery, University Hospital Zurich, Frauenklinikstrasse 10, 8091, Zurich, Switzerland.,Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
33
|
Eisenhut F, Engelhorn T, Arinrad S, Brandner S, Coras R, Putz F, Fietkau R, Doerfler A, Schmidt MA. A Comparison of Single- and Multiparametric MRI Models for Differentiation of Recurrent Glioblastoma from Treatment-Related Change. Diagnostics (Basel) 2021; 11:diagnostics11122281. [PMID: 34943518 PMCID: PMC8700236 DOI: 10.3390/diagnostics11122281] [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: 10/27/2021] [Revised: 11/24/2021] [Accepted: 12/03/2021] [Indexed: 12/02/2022] Open
Abstract
To evaluate single- and multiparametric MRI models to differentiate recurrent glioblastoma (GBM) and treatment-related changes (TRC) in clinical routine imaging. Selective and unselective apparent diffusion coefficient (ADC) and minimum, mean, and maximum cerebral blood volume (CBV) measurements in the lesion were performed. Minimum, mean, and maximum ratiosCBV (CBVlesion to CBVhealthy white matter) were computed. All data were tested for lesion discrimination. A multiparametric model was compiled via multiple logistic regression using data demonstrating significant difference between GBM and TRC and tested for its diagnostic strength in an independent patient cohort. A total of 34 patients (17 patients with recurrent GBM and 17 patients with TRC) were included. ADC measurements showed no significant difference between both entities. All CBV and ratiosCBV measurements were significantly higher in patients with recurrent GBM than TRC. A minimum CBV of 8.5, mean CBV of 116.5, maximum CBV of 327 and ratioCBV minimum of 0.17, ratioCBV mean of 2.26 and ratioCBV maximum of 3.82 were computed as optimal cut-off values. By integrating these parameters in a multiparametric model and testing it in an independent patient cohort, 9 of 10 patients, i.e., 90%, were classified correctly. The multiparametric model further improves radiological discrimination of GBM from TRC in comparison to single-parameter approaches and enables reliable identification of recurrent tumors.
Collapse
Affiliation(s)
- Felix Eisenhut
- Department of Neuroradiology, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany; (T.E.); (A.D.); (M.A.S.)
- Correspondence: ; Tel.: +49-913185-44838
| | - Tobias Engelhorn
- Department of Neuroradiology, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany; (T.E.); (A.D.); (M.A.S.)
| | - Soheil Arinrad
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany; (S.A.); (S.B.)
| | - Sebastian Brandner
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany; (S.A.); (S.B.)
| | - Roland Coras
- Department of Neuropathology, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany;
| | - Florian Putz
- Department of Radiation Oncology, University Hospital Erlangen, Universitaetsstrasse 27, 91054 Erlangen, Germany; (F.P.); (R.F.)
| | - Rainer Fietkau
- Department of Radiation Oncology, University Hospital Erlangen, Universitaetsstrasse 27, 91054 Erlangen, Germany; (F.P.); (R.F.)
| | - Arnd Doerfler
- Department of Neuroradiology, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany; (T.E.); (A.D.); (M.A.S.)
| | - Manuel A. Schmidt
- Department of Neuroradiology, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany; (T.E.); (A.D.); (M.A.S.)
| |
Collapse
|
34
|
Olson JJ. Congress of Neurological Surgeons systematic review and evidence-based guidelines for the treatment of adults with progressive glioblastoma update: introduction and methods. J Neurooncol 2021; 158:133-137. [PMID: 34694566 DOI: 10.1007/s11060-021-03850-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 09/17/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The management of glioblastoma at the time of progression is an important facet of all physicians involved in neuro-oncology. This is an update of the evidence-based guidelines for management of progressive glioblastoma published by the Congress of Neurological Surgeons and American Association of Neurological Surgeons in 2014. METHODS The medical literature from July 1, 2012 through March 31, 2019 was searched in MEDLINE® and Embase® and the Cochrane Database of Systematic Reviews, Cochrane Controlled Trials Registry, and Cochrane Database of Abstracts of Reviews of Effects to determine if information was available to update, modify or create new recommendations related to imaging, cytoreductive surgery, neuropathology, radiotherapy, cytotoxic chemotherapy, targeted therapy, and immunotherapy. RESULTS The writing group utilized the information from the updated literature search to formulate recommendations that were exclusively evidence based and not founded on potentially biased consensus or expert opinion. CONCLUSION The series of guideline documents provides an update of the information and recommendations that could be derived in the 2014 version. It sets a benchmark as to what we really know about the management of this difficult disease. It also provides clues to key investigations that are necessary to move us toward truly effective disease control.
Collapse
Affiliation(s)
- Jeffrey J Olson
- Department of Neurosurgery, School of Medicine, Emory University, 1365 Clifton Road Northeast, Bldg. B, Fl 2, Ste 2200, Atlanta, GA, 30322, USA.
| |
Collapse
|
35
|
Galldiks N, Niyazi M, Grosu AL, Kocher M, Langen KJ, Law I, Minniti G, Kim MM, Tsien C, Dhermain F, Soffietti R, Mehta MP, Weller M, Tonn JC. Contribution of PET imaging to radiotherapy planning and monitoring in glioma patients - a report of the PET/RANO group. Neuro Oncol 2021; 23:881-893. [PMID: 33538838 DOI: 10.1093/neuonc/noab013] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The management of patients with glioma usually requires multimodality treatment including surgery, radiotherapy, and systemic therapy. Accurate neuroimaging plays a central role for radiotherapy planning and follow-up after radiotherapy completion. In order to maximize the radiation dose to the tumor and to minimize toxic effects on the surrounding brain parenchyma, reliable identification of tumor extent and target volume delineation is crucial. The use of positron emission tomography (PET) for radiotherapy planning and monitoring in gliomas has gained considerable interest over the last several years, but Class I data are not yet available. Furthermore, PET has been used after radiotherapy for response assessment and to distinguish tumor progression from pseudoprogression or radiation necrosis. Here, the Response Assessment in Neuro-Oncology (RANO) working group provides a summary of the literature and recommendations for the use of PET imaging for radiotherapy of patients with glioma based on published studies, constituting levels 1-3 evidence according to the Oxford Centre for Evidence-based Medicine.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3,-4), Research Center Juelich, Juelich, Germany.,Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Düsseldorf, Cologne and Aachen, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Anca L Grosu
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg, Germany
| | - Martin Kocher
- Institute of Neuroscience and Medicine (INM-3,-4), Research Center Juelich, Juelich, Germany.,Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3,-4), Research Center Juelich, Juelich, Germany.,Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Düsseldorf, Cologne and Aachen, Germany.,Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, University Hospital Copenhagen, Copenhagen, Denmark
| | - Giuseppe Minniti
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.,IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Christina Tsien
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Frederic Dhermain
- Department of Radiation Therapy, Institut de Cancerologie Gustave Roussy, Villejuif, France
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA.,Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Michael Weller
- Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Jörg-Christian Tonn
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
36
|
Hong CS, Beckta JM, Kundishora AJ, Elsamadicy AA, Chiang VL. Laser interstitial thermal therapy for treatment of cerebral radiation necrosis. Int J Hyperthermia 2021; 37:68-76. [PMID: 32672119 DOI: 10.1080/02656736.2020.1760362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Radiation necrosis is a well described complication after radiosurgical treatment of intracranial pathologies - best recognized after the treatment of patients with arteriovenous malformations and brain metastases but possibly also affecting patients treated with radiosurgery for meningioma. The pathophysiology of radiation necrosis is still not well understood but is most likely a secondary local tissue inflammatory response to brain tissue injured by radiation. Radiation necrosis in brain metastases patients may present radiographically and behave clinically like recurrent tumor. Differentiation between radiation necrosis and recurrent tumor has been difficult based on radiographic changes alone. Biopsy or craniotomy therefore remains the gold standard method of diagnosis. For symptomatic patients, corticosteroids are first-line therapy, but patients may fail medical management due to intolerance of chronic steroids or persistence of symptoms. In these cases, open surgical resection has been shown to be successful in management of surgically amenable lesions but may be suboptimal in patients with deep-seated lesions or extensive prior cranial surgical history, both carrying high risk for peri-operative morbidity. Laser interstitial thermal therapy has emerged as a viable, alternative surgical option. In addition to allowing access to tissue for diagnosis, thermal treatment of the lesion can also be delivered precisely and accurately under real-time imaging guidance. This review highlights the pertinent studies that have shaped the impetus for use of laser interstitial thermal therapy in the treatment of radiation necrosis, reviewing indications, outcomes, and nuances toward successful application of this technology in patients with suspected radiation necrosis.
Collapse
Affiliation(s)
- Christopher S Hong
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Jason M Beckta
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Adam J Kundishora
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Aladine A Elsamadicy
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Veronica L Chiang
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA.,Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
37
|
Bapuraj JR, Perni K, Gomez-Hassan D, Srinivasan A. Imaging Surveillance of Gliomas: Role of Basic and Advanced Imaging Techniques. Radiol Clin North Am 2021; 59:395-407. [PMID: 33926685 DOI: 10.1016/j.rcl.2021.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
It is essential to be aware of widely accepted criteria for grading of treatment response in both high-grade and low-grade gliomas. These criteria primarily take into account responses of measurable and nonmeasurable lesions on T2-weighted, fluid-attenuated inversion recovery, and postcontrast images to determine a final category of response for the patient. The additional role that other advanced imaging techniques, such as diffusion and perfusion imaging, can play in the surveillance of these tumors is discussed in this article.
Collapse
Affiliation(s)
- Jayapalli Rajiv Bapuraj
- Division of Neuroradiology, Department of Radiology, University of Michigan Medical School, Michigan Medicine, 1500 East Medical Center Drive, UMHS, B2-A209, Ann Arbor, MI 48109, USA
| | - Krishna Perni
- Division of Neuroradiology, Department of Radiology, University of Michigan Medical School, Michigan Medicine, 1500 East Medical Center Drive, UMHS, B2-A209, Ann Arbor, MI 48109, USA
| | - Diana Gomez-Hassan
- Division of Neuroradiology, Department of Radiology, University of Michigan Medical School, Michigan Medicine, 4260 Plymouth Road, Ann Arbor, MI 48105, USA
| | - Ashok Srinivasan
- Division of Neuroradiology, Department of Radiology, University of Michigan Medical School, Michigan Medicine, 1500 East Medical Center Drive, UMHS, B2-A209, Ann Arbor, MI 48109, USA.
| |
Collapse
|
38
|
Patel M, Zhan J, Natarajan K, Flintham R, Davies N, Sanghera P, Grist J, Duddalwar V, Peet A, Sawlani V. Machine learning-based radiomic evaluation of treatment response prediction in glioblastoma. Clin Radiol 2021; 76:628.e17-628.e27. [PMID: 33941364 DOI: 10.1016/j.crad.2021.03.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/29/2021] [Indexed: 11/16/2022]
Abstract
AIM To investigate machine learning based models combining clinical, radiomic, and molecular information to distinguish between early true progression (tPD) and pseudoprogression (psPD) in patients with glioblastoma. MATERIALS AND METHODS A retrospective analysis was undertaken of 76 patients (46 tPD, 30 psPD) with early enhancing disease following chemoradiotherapy for glioblastoma. Outcome was determined on follow-up until 6 months post-chemoradiotherapy. Models comprised clinical characteristics, O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status, and 307 quantitative imaging features extracted from enhancing disease and perilesional oedema masks on early post-chemoradiotherapy contrast-enhanced T1-weighted imaging, T2-weighted imaging (T2WI), and apparent diffusion coefficient (ADC) maps. Feature selection was performed within bootstrapped cross-validated recursive feature elimination with a random forest algorithm. Naive Bayes five-fold cross-validation was used to validate the final model. RESULTS Top selected features included age, MGMT promoter methylation status, two shape-based features from the enhancing disease mask, three radiomic features from the enhancing disease mask on ADC, and one radiomic feature from the perilesional oedema mask on T2WI. The final model had an area under the receiver operating characteristics curve (AUC) of 0.80, sensitivity 78.2%, specificity 66.7%, and accuracy of 73.7%. CONCLUSION Incorporating a machine learning-based approach using quantitative radiomic features from standard-of-care magnetic resonance imaging (MRI), in combination with clinical characteristics and MGMT promoter methylation status has a complementary effect and improves model performance for early prediction of glioblastoma treatment response.
Collapse
Affiliation(s)
- M Patel
- University of Birmingham, Birmingham, UK; Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - J Zhan
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; The Affiliated Hospital of Qingdao University, Qingdao Shi, Shandong Sheng, China
| | - K Natarajan
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - R Flintham
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - N Davies
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - P Sanghera
- University of Birmingham, Birmingham, UK; Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - J Grist
- University of Birmingham, Birmingham, UK
| | - V Duddalwar
- Departments of Radiology, Urology and Biomedical Engineering, University of Southern California, USA
| | - A Peet
- University of Birmingham, Birmingham, UK; Birmingham Children's Hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - V Sawlani
- University of Birmingham, Birmingham, UK; Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| |
Collapse
|
39
|
Multimodal assessment of disease activity in glioblastoma : A single center experience. Wien Klin Wochenschr 2021; 133:1148-1154. [PMID: 33877437 DOI: 10.1007/s00508-021-01859-2] [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: 02/11/2021] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Assessment of disease activity in glioblastoma (GBM) can be challenging due to several clinical and radiological pitfalls. Besides MRI, FET-PET and neurocognitive assessment (NA) are used in several neuro-oncological centers in order to improve the specificity of response assessment. We performed a retrospective study to investigate whether the assessment by RANO (Response Assessment in NeuroOncology) corresponds to FET-PET imaging and NA results. Moreover, the concordance of RANO with a final recommendation of an interdisciplinary neuro-oncological tumor board recommendation (TBR) was analyzed. METHODS We enrolled 25 consecutive patients with newly diagnosed histologically confirmed GBM in a pilot study, accounting for 81 multimodal test results. All patients were selected after undergoing consecutive follow-up comprising MRI, FET-PET, and NA with a subsequent TBR. Results were analyzed for correlations between RANO, FET-PET and NA. An additional consistency analysis was performed to elucidate the impact of RANO on decision making. RESULTS A highly statistically significant correlation was found between RANO and FET-PET and NA results (all P < 0.01); however, 26% of follow-up tests exhibited inconsistent results in multimodal assessment, among which RANO was only 48% in accordance with the final TBR. The concordance of NA and FET-PET with the final TBR was 67% and 86%, respectively. CONCLUSION The RANO proved its value in the context of multimodal assessment of disease activity in GBM; however, because the implementation of multimodal assessment showed a considerably high percentage of inconsistent results, further studies are required to investigate the relationship between different assessment techniques, in addition to their overall significance to response rating.
Collapse
|
40
|
Freiburg Neuropathology Case Conference : Contrast-enhancing Brain Lesion 6 Months after Resection and Combined Radiotherapy and Chemotherapy of an Unmethylated but IDH R132H-mutated Glioblastoma Multiforme. Clin Neuroradiol 2021; 31:283-288. [PMID: 33625551 PMCID: PMC7943525 DOI: 10.1007/s00062-021-01006-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 10/28/2022]
|
41
|
Zheng L, Zhou ZR, Yu Q, Shi M, Yang Y, Zhou X, Li C, Wei Q. The Definition and Delineation of the Target Area of Radiotherapy Based on the Recurrence Pattern of Glioblastoma After Temozolomide Chemoradiotherapy. Front Oncol 2021; 10:615368. [PMID: 33692942 PMCID: PMC7937883 DOI: 10.3389/fonc.2020.615368] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/30/2020] [Indexed: 11/13/2022] Open
Abstract
Radiotherapy is an important treatment for glioblastoma (GBM), but there is no consensus on the target delineation for GBM radiotherapy. The Radiation Therapy Oncology Group (RTOG) and European Organisation for Research and Treatment of Cancer (EORTC) each have their own rules. Our center adopted a target volume delineation plan based on our previous studies. This study focuses on the recurrence pattern of GBM patients whose target delineations did not intentionally include the T2/fluid-attenuated inversion recovery (FLAIR) hyperintensity area outside of the gross tumor volume (GTV). We prospectively collected 162 GBM cases and retrospectively analysed the clinical data and continuous dynamic magnetic resonance images (MRI) of 55 patients with recurrent GBM. All patients received concurrent radiotherapy and chemotherapy with temozolomide (TMZ). The GTV that we defined includes the postoperative T1-weighted MRI enhancement area and resection cavity. Clinical target volume 1 (CTV1) and CTV2 were defined as GTVs with 1 and 2 cm margins, respectively. Planning target volume 1 (PTV1) and PTV2 were defined as CTV1 and CTV2 plus a 3 mm margin with prescribed doses of 60 and 54 Gy, respectively. The first recurrent contrast-enhanced T1-weighted MRI was introduced into the Varian Eclipse radiotherapy planning system and fused with the original planning computed tomography (CT) images to determine the recurrence pattern. The median follow-up time was 15.8 months. The median overall survival (OS) and progression-free survival (PFS) were 17.7 and 7.0 months, respectively. Among the patients, 44 had central recurrences, two had in-field recurrences, one had marginal recurrence occurred, 11 had distant recurrences, and three had subependymal recurrences. Five patients had multiple recurrence patterns. Compared to the EORTC protocol, target delineation that excludes the adjacent T2/FLAIR hyperintensity area reduces the brain volume exposed to high-dose radiation (P = 0.000) without an increased risk of marginal recurrence. Therefore, it is worthwhile to conduct a clinical trial investigating the feasibility of intentionally not including the T2/FLAIR hyperintensity region outside of the GTV.
Collapse
Affiliation(s)
- Lin Zheng
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Radiation Oncology, Taizhou Cancer Hospital, Wenling, China
| | - Zhi-Rui Zhou
- Radiation Oncology Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - QianQian Yu
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minghan Shi
- Département de l'éducation aux adultes, Cégep Saint-Jean-sur-Richelieu, Brossard, QC, Canada
| | - Yang Yang
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaofeng Zhou
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Li
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qichun Wei
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
42
|
Oronsky B, Reid TR, Oronsky A, Sandhu N, Knox SJ. A Review of Newly Diagnosed Glioblastoma. Front Oncol 2021; 10:574012. [PMID: 33614476 PMCID: PMC7892469 DOI: 10.3389/fonc.2020.574012] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/28/2020] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma is an aggressive and inevitably recurrent primary intra-axial brain tumor with a dismal prognosis. The current mainstay of treatment involves maximally safe surgical resection followed by radiotherapy over a 6-week period with concomitant temozolomide chemotherapy followed by temozolomide maintenance. This review provides a summary of the epidemiological, clinical, histologic and genetic characteristics of newly diagnosed disease as well as the current standard of care and potential future therapeutic prospects.
Collapse
Affiliation(s)
- Bryan Oronsky
- Department of Clinical Research, EpicentRx, San Diego, CA, United States
| | - Tony R. Reid
- Department of Medical Oncology, UC San Diego School of Medicine, San Diego, CA, United States
| | - Arnold Oronsky
- Department of Clinical Research, InterWest Partners, Menlo Park, CA, United States
| | - Navjot Sandhu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
| | - Susan J. Knox
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
43
|
Sanvito F, Castellano A, Falini A. Advancements in Neuroimaging to Unravel Biological and Molecular Features of Brain Tumors. Cancers (Basel) 2021; 13:cancers13030424. [PMID: 33498680 PMCID: PMC7865835 DOI: 10.3390/cancers13030424] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Advanced neuroimaging is gaining increasing relevance for the characterization and the molecular profiling of brain tumor tissue. On one hand, for some tumor types, the most widespread advanced techniques, investigating diffusion and perfusion features, have been proven clinically feasible and rather robust for diagnosis and prognosis stratification. In addition, 2-hydroxyglutarate spectroscopy, for the first time, offers the possibility to directly measure a crucial molecular marker. On the other hand, numerous innovative approaches have been explored for a refined evaluation of tumor microenvironments, particularly assessing microstructural and microvascular properties, and the potential applications of these techniques are vast and still to be fully explored. Abstract In recent years, the clinical assessment of primary brain tumors has been increasingly dependent on advanced magnetic resonance imaging (MRI) techniques in order to infer tumor pathophysiological characteristics, such as hemodynamics, metabolism, and microstructure. Quantitative radiomic data extracted from advanced MRI have risen as potential in vivo noninvasive biomarkers for predicting tumor grades and molecular subtypes, opening the era of “molecular imaging” and radiogenomics. This review presents the most relevant advancements in quantitative neuroimaging of advanced MRI techniques, by means of radiomics analysis, applied to primary brain tumors, including lower-grade glioma and glioblastoma, with a special focus on peculiar oncologic entities of current interest. Novel findings from diffusion MRI (dMRI), perfusion-weighted imaging (PWI), and MR spectroscopy (MRS) are hereby sifted in order to evaluate the role of quantitative imaging in neuro-oncology as a tool for predicting molecular profiles, stratifying prognosis, and characterizing tumor tissue microenvironments. Furthermore, innovative technological approaches are briefly addressed, including artificial intelligence contributions and ultra-high-field imaging new techniques. Lastly, after providing an overview of the advancements, we illustrate current clinical applications and future perspectives.
Collapse
Affiliation(s)
- Francesco Sanvito
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.S.); (A.F.)
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Unit of Radiology, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Antonella Castellano
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.S.); (A.F.)
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Correspondence: ; Tel.: +39-02-2643-3015
| | - Andrea Falini
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.S.); (A.F.)
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
44
|
Le Fèvre C, Lhermitte B, Ahle G, Chambrelant I, Cebula H, Antoni D, Keller A, Schott R, Thiery A, Constans JM, Noël G. Pseudoprogression versus true progression in glioblastoma patients: A multiapproach literature review: Part 1 - Molecular, morphological and clinical features. Crit Rev Oncol Hematol 2020; 157:103188. [PMID: 33307200 DOI: 10.1016/j.critrevonc.2020.103188] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/12/2020] [Accepted: 11/23/2020] [Indexed: 01/04/2023] Open
Abstract
With new therapeutic protocols, more patients treated for glioblastoma have experienced a suspicious radiologic image of progression (pseudoprogression) during follow-up. Pseudoprogression should be differentiated from true progression because the disease management is completely different. In the case of pseudoprogression, the follow-up continues, and the patient is considered stable. In the case of true progression, a treatment adjustment is necessary. Presently, a pseudoprogression diagnosis certainly needs to be pathologically confirmed. Some important efforts in the radiological, histopathological, and genomic fields have been made to differentiate pseudoprogression from true progression, and the assessment of response criteria exists but remains limited. The aim of this paper is to highlight clinical and pathological markers to differentiate pseudoprogression from true progression through a literature review.
Collapse
Affiliation(s)
- Clara Le Fèvre
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Benoît Lhermitte
- Département of Pathology, Hautepierre University Hospital, 1, Avenue Molière, 67200, Strasbourg, France
| | - Guido Ahle
- Departement of Neurology, Hôpitaux Civils de Colmar, 39 Avenue de la Liberté, 68024, Colmar, France
| | - Isabelle Chambrelant
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Hélène Cebula
- Departement of Neurosurgery, Hautepierre University Hospital, 1, Avenue Molière, 67200, Strasbourg, France
| | - Delphine Antoni
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Audrey Keller
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Roland Schott
- Departement of Medical Oncology, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Alicia Thiery
- Department of Public Health, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France
| | - Jean-Marc Constans
- Department of Radiology, Amiens-Pïcardie University Hospital, 1 rond point du Professeur Christian Cabrol, 80054 Amiens Cedex 1, France
| | - Georges Noël
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200, Strasbourg Cedex, France.
| |
Collapse
|
45
|
Wu XF, Liang X, Wang XC, Qin JB, Zhang L, Tan Y, Zhang H. Differentiating high-grade glioma recurrence from pseudoprogression: Comparing diffusion kurtosis imaging and diffusion tensor imaging. Eur J Radiol 2020; 135:109445. [PMID: 33341429 DOI: 10.1016/j.ejrad.2020.109445] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/15/2020] [Accepted: 11/25/2020] [Indexed: 01/11/2023]
Abstract
PURPOSE To compare the diagnostic value of DKI and DTI in differentiation of high-grade glioma recurrence and pseudoprogression (PsP). METHOD Forty patients with high-grade gliomas who exhibited new enhancing lesions (24 high-grade glioma recurrence and 16 PsP) within 6 months after surgery followed by completion of chemoradiation therapy. All patients underwent repeat surgery or biopsy after routine MRI and DKI (including DTI). They were histologically classified into high-grade glioma recurrence and PsP groups. DKI (mean kurtosis [MK], axial kurtosis [Ka], and radial kurtosis [Kr]) and DTI (mean diffusivity [MD] and fractional anisotropy [FA]) parameters in the enhancing lesions and in the perilesional edema were measured. Inter-group differences between high-grade glioma recurrence and PsP were compared using the Mann-Whitney U test The receiver operating characteristic (ROC) curve was used to assess differential diagnostic efficacy of each parameter, and Z-scores were used to compare the value between DKI and DTI. RESULTS Relative MK (rMK) was significantly higher and relative MD (rMD) was significantly lower in the enhancing lesions of high-grade glioma recurrence compared to PsP (P < 0.001, P = 0.006, respectively). The AUC was 0.914 for rMK and 0.760 for rMD, and this difference was significant (P = 0.030). In the perilesional edema, rMK values were significantly higher and rMD values were significantly lower in high-grade glioma recurrence compared to PsP (P < 0.001, P = 0.005). CONCLUSIONS DKI had superior performance in differentiating high-grade glioma recurrence from PsP, and rMK appeared to be the best independent predictor.
Collapse
Affiliation(s)
- Xiao-Feng Wu
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China; College of Medical Imaging, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Xiao Liang
- Shanxi Provincial People's Hospital, Taiyuan 030001, Shanxi Province, China
| | - Xiao-Chun Wang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China; College of Medical Imaging, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Jiang-Bo Qin
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China
| | - Lei Zhang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China
| | - Yan Tan
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China; College of Medical Imaging, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China.
| | - Hui Zhang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China; College of Medical Imaging, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China.
| |
Collapse
|
46
|
Advanced magnetic resonance imaging to support clinical drug development for malignant glioma. Drug Discov Today 2020; 26:429-441. [PMID: 33249294 DOI: 10.1016/j.drudis.2020.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/23/2020] [Accepted: 11/18/2020] [Indexed: 11/22/2022]
Abstract
Even though the treatment options and survival of patients with glioblastoma multiforme (GBM), the most common type of malignant glioma, have improved over the past decade, there is still a high unmet medical need to develop novel therapies. Complexity in pathology and therapy require biomarkers to characterize tumors, to define malignant and active areas, to assess disease prognosis, and to quantify and monitor therapy response. While conventional magnetic resonance imaging (MRI) techniques have improved these assessments, limitations remain. In this review, we evaluate the role of various non-invasive biomarkers based on advanced structural and functional MRI techniques in the context of GBM drug development over the past 5 years.
Collapse
|
47
|
Sun P, Xiao Y, Di Q, Ma W, Ma X, Wang Q, Chen W. Transferrin Receptor-Targeted PEG-PLA Polymeric Micelles for Chemotherapy Against Glioblastoma Multiforme. Int J Nanomedicine 2020; 15:6673-6688. [PMID: 32982226 PMCID: PMC7494234 DOI: 10.2147/ijn.s257459] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background The safe and efficient delivery of chemotherapeutic agents is critical to glioma therapy. However, chemotherapy for glioma is extremely challenging because the blood–brain barrier (BBB) rigorously prevents drugs from reaching the tumor region. Materials and Methods TfR-T12 peptide-modified PEG-PLA polymer was synthesized to deliver paclitaxel (PTX) for glioma therapy. TfR was significantly expressed on brain capillary endothelial cells and glioma cells; therefore, TfR-T12 peptide-modified micelles can cross the BBB system and target glioma cells. Results TfR-T12-PEG-PLA/PTX polymeric micelles (TfR-T12-PMs) could be absorbed rapidly by tumor cells, and traversed effectively the BBB monolayers. TfR-T12-PMs can effectively inhibit the proliferation of U87MG cells in vitro, and TfR-T12-PMs loaded with paclitaxel presented better antiglioma effect with prolonged median survival of nude mice-bearing glioma than the unmodified PMs. Conclusion The TfR-T12-PMs could effectively overcome the BBB barrier and accomplish glioma-targeted drug delivery, thus validating its potential in improving the therapeutic outcome in multiforme.
Collapse
Affiliation(s)
- Ping Sun
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen 518060, People's Republic of China.,Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, People's Republic of China
| | - Yue Xiao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen 518060, People's Republic of China
| | - Qianqian Di
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen 518060, People's Republic of China
| | - Wenjing Ma
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen 518060, People's Republic of China
| | - Xingyu Ma
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen 518060, People's Republic of China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University of Medicine School, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Weilin Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen 518060, People's Republic of China
| |
Collapse
|
48
|
Wen PY, Weller M, Lee EQ, Alexander BM, Barnholtz-Sloan JS, Barthel FP, Batchelor TT, Bindra RS, Chang SM, Chiocca EA, Cloughesy TF, DeGroot JF, Galanis E, Gilbert MR, Hegi ME, Horbinski C, Huang RY, Lassman AB, Le Rhun E, Lim M, Mehta MP, Mellinghoff IK, Minniti G, Nathanson D, Platten M, Preusser M, Roth P, Sanson M, Schiff D, Short SC, Taphoorn MJB, Tonn JC, Tsang J, Verhaak RGW, von Deimling A, Wick W, Zadeh G, Reardon DA, Aldape KD, van den Bent MJ. Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol 2020; 22:1073-1113. [PMID: 32328653 PMCID: PMC7594557 DOI: 10.1093/neuonc/noaa106] [Citation(s) in RCA: 598] [Impact Index Per Article: 149.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastomas are the most common form of malignant primary brain tumor and an important cause of morbidity and mortality. In recent years there have been important advances in understanding the molecular pathogenesis and biology of these tumors, but this has not translated into significantly improved outcomes for patients. In this consensus review from the Society for Neuro-Oncology (SNO) and the European Association of Neuro-Oncology (EANO), the current management of isocitrate dehydrogenase wildtype (IDHwt) glioblastomas will be discussed. In addition, novel therapies such as targeted molecular therapies, agents targeting DNA damage response and metabolism, immunotherapies, and viral therapies will be reviewed, as well as the current challenges and future directions for research.
Collapse
Affiliation(s)
- Patrick Y Wen
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Weller
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Eudocia Quant Lee
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Brian M Alexander
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jill S Barnholtz-Sloan
- Case Western Reserve University School of Medicine and University Hospitals of Cleveland, Cleveland, Ohio, USA
| | - Floris P Barthel
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Tracy T Batchelor
- Department of Neurology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Susan M Chang
- University of California San Francisco, San Francisco, California, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Timothy F Cloughesy
- David Geffen School of Medicine, Department of Neurology, University of California Los Angeles, Los Angeles, California, USA
| | - John F DeGroot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Monika E Hegi
- Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Raymond Y Huang
- Division of Neuroradiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew B Lassman
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, NewYork-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, USA
| | - Emilie Le Rhun
- University of Lille, Inserm, Neuro-oncology, General and Stereotaxic Neurosurgery service, University Hospital of Lille, Lille, France; Breast Cancer Department, Oscar Lambret Center, Lille, France and Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Ingo K Mellinghoff
- Department of Neurology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Giuseppe Minniti
- Radiation Oncology Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - David Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Heidelberg, Germany
| | - Matthias Preusser
- Division of Oncology, Department of Medicine, Medical University of Vienna, Vienna, Austria
| | - Patrick Roth
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Marc Sanson
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière – Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - David Schiff
- University of Virginia School of Medicine, Division of Neuro-Oncology, Department of Neurology, University of Virginia, Charlottesville, Virginia, USA
| | - Susan C Short
- Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, UK
| | - Martin J B Taphoorn
- Department of Neurology, Medical Center Haaglanden, The Hague and Department of Neurology, Leiden University Medical Center, the Netherlands
| | | | - Jonathan Tsang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Roel G W Verhaak
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Andreas von Deimling
- Neuropathology and Clinical Cooperation Unit Neuropathology, University Heidelberg and German Cancer Center, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology and Neuro-oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Gelareh Zadeh
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, Canada
| | - David A Reardon
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kenneth D Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | | |
Collapse
|
49
|
Neurologic Complications of Cranial Radiation Therapy and Strategies to Prevent or Reduce Radiation Toxicity. Curr Neurol Neurosci Rep 2020; 20:34. [DOI: 10.1007/s11910-020-01051-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
Gesundheit B, Ben-David E, Posen Y, Ellis R, Wollmann G, Schneider EM, Aigner K, Brauns L, Nesselhut T, Ackva I, Weisslein C, Thaller A. Effective Treatment of Glioblastoma Multiforme With Oncolytic Virotherapy: A Case-Series. Front Oncol 2020; 10:702. [PMID: 32477944 PMCID: PMC7241257 DOI: 10.3389/fonc.2020.00702] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/15/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma multiforme (GBM) remains an incurable condition, associated with a median survival time of 15 months with best standard of care and 5-year survival rate of <10%. We report on four GBM patients on combination treatment regimens that included oncolytic virus (OV) immunotherapy, who achieved clinical and radiological responses with long-term survival, thus far, of up to 14 years, and good quality of life. We discuss the radiological findings that provide new insights into this treatment, the scientific rationale of this innovative and promising therapy, and considerations for future research.
Collapse
Affiliation(s)
| | - Eliel Ben-David
- Department of Radiology, Shaare Zedek Medical Center, Jerusalem, Israel
| | | | | | - Guido Wollmann
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Innsbruck, Austria
| | - E Marion Schneider
- Division of Experimental Anesthesiology, University Hospital Ulm, Ulm, Germany
| | | | | | | | - Ingrid Ackva
- Praxisklinik fuer Allgemeinmedizin, Markt Berolzheim, Germany
| | | | - Arno Thaller
- Praxisklinik fuer Allgemeinmedizin, Markt Berolzheim, Germany
| |
Collapse
|