1
|
Vollmuth P, Karschnia P, Sahm F, Park YW, Ahn SS, Jain R. A Radiologist's Guide to IDH-Wildtype Glioblastoma for Efficient Communication With Clinicians: Part II-Essential Information on Post-Treatment Imaging. Korean J Radiol 2025; 26:368-389. [PMID: 40015559 PMCID: PMC11955384 DOI: 10.3348/kjr.2024.0983] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/08/2024] [Accepted: 11/30/2024] [Indexed: 03/01/2025] Open
Abstract
Owing to recent advancements in various postoperative treatment modalities, such as radiation, chemotherapy, antiangiogenic treatment, and immunotherapy, the radiological and clinical assessment of patients with isocitrate dehydrogenase-wildtype glioblastoma using post-treatment imaging has become increasingly challenging. This review highlights the challenges in differentiating treatment-related changes such as pseudoprogression, radiation necrosis, and pseudoresponse from true tumor progression and aims to serve as a guideline for efficient communication with clinicians for optimal management of patients with post-treatment imaging.
Collapse
Affiliation(s)
- Philipp Vollmuth
- Division for Computational Radiology & Clinical AI (CCIBonn.ai), Clinic for Neuroradiology, University Hospital Bonn, Bonn, Germany
- Medical Faculty Bonn, University of Bonn, Bonn, Germany
- Division of Medical Image Computing, German Cancer Research Center, Heidelberg, Germany
| | - Philipp Karschnia
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany
- Department of Neurosurgery, Friedrich-Alexander-University University, Erlangen-Nuremberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yae Won Park
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Sung Soo Ahn
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Rajan Jain
- Department of Radiology, New York University Grossman School of Medicine, New York, USA
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, USA
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Henssen D, Meijer F, Verburg FA, Smits M. Challenges and opportunities for advanced neuroimaging of glioblastoma. Br J Radiol 2023; 96:20211232. [PMID: 36062962 PMCID: PMC10997013 DOI: 10.1259/bjr.20211232] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 08/10/2022] [Accepted: 08/25/2022] [Indexed: 11/05/2022] Open
Abstract
Glioblastoma is the most aggressive of glial tumours in adults. On conventional magnetic resonance (MR) imaging, these tumours are observed as irregular enhancing lesions with areas of infiltrating tumour and cortical expansion. More advanced imaging techniques including diffusion-weighted MRI, perfusion-weighted MRI, MR spectroscopy and positron emission tomography (PET) imaging have found widespread application to diagnostic challenges in the setting of first diagnosis, treatment planning and follow-up. This review aims to educate readers with regard to the strengths and weaknesses of the clinical application of these imaging techniques. For example, this review shows that the (semi)quantitative analysis of the mentioned advanced imaging tools was found useful for assessing tumour aggressiveness and tumour extent, and aids in the differentiation of tumour progression from treatment-related effects. Although these techniques may aid in the diagnostic work-up and (post-)treatment phase of glioblastoma, so far no unequivocal imaging strategy is available. Furthermore, the use and further development of artificial intelligence (AI)-based tools could greatly enhance neuroradiological practice by automating labour-intensive tasks such as tumour measurements, and by providing additional diagnostic information such as prediction of tumour genotype. Nevertheless, due to the fact that advanced imaging and AI-diagnostics is not part of response assessment criteria, there is no harmonised guidance on their use, while at the same time the lack of standardisation severely hampers the definition of uniform guidelines.
Collapse
Affiliation(s)
- Dylan Henssen
- Department of Medical Imaging, Radboud university medical
center, Nijmegen, The Netherlands
| | - Frederick Meijer
- Department of Medical Imaging, Radboud university medical
center, Nijmegen, The Netherlands
| | - Frederik A. Verburg
- Department of Medical Imaging, Radboud university medical
center, Nijmegen, The Netherlands
| | - Marion Smits
- Department of Medical Imaging, Radboud university medical
center, Nijmegen, The Netherlands
| |
Collapse
|
5
|
Diffusion-weighted imaging and arterial spin labeling radiomics features may improve differentiation between radiation-induced brain injury and glioma recurrence. Eur Radiol 2022; 33:3332-3342. [PMID: 36576544 DOI: 10.1007/s00330-022-09365-3] [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] [Received: 08/24/2022] [Revised: 11/16/2022] [Accepted: 12/05/2022] [Indexed: 12/29/2022]
Abstract
OBJECTIVES To determine whether radiomics features derived from diffusion-weighted imaging (DWI) and arterial spin labeling (ASL) can improve the differentiation between radiation-induced brain injury (RIBI) and tumor recurrence (TR) in glioma patients. METHODS A total of 4199 radiomics features were extracted from conventional MRI, apparent diffusion coefficient (ADC), and cerebral blood flow (CBF) maps, obtained from 96 pathologically confirmed WHO grade 2~4 gliomas with enhancement after standard treatment. The intraclass correlation coefficient (ICC) was used to test segmentation stability between two doctors. Radiomics features were selected using the Mann-Whitney U test, LASSO regression, and RFE algorithms. Four machine learning classifiers were adopted to establish radiomics models. The diagnostic performance of multiparameter, conventional, and single-parameter MRI radiomics models was compared using the area under the curve (AUC). The models were evaluated in the subsequent independent validation set (n = 30). RESULTS Eight important radiomics features (3 from conventional MRI, 1 from ADC, and 4 from CBF) were selected. Support vector machine (SVM) was chosen as the optimal classifier. The diagnostic performance of the multiparameter MRI radiomics model (AUC 0.96) was higher than that of the conventional MRI (AUC 0.88), ADC (AUC 0.91), and CBF (AUC 0.95) radiomics models. For subgroup analysis, the multiparameter MRI radiomics model showed similar performance, with AUCs of 0.98 in WHO grade 2~3 and 0.96 in WHO grade 4. CONCLUSION The incorporation of noninvasive DWI and ASL into the MRI radiomics model improved the diagnostic performance in differentiating RIBI from TR; ASL, especially, played a significant role. KEY POINTS • The multiparameter MRI radiomics model was superior to the conventional MRI radiomics model in differentiating glioma recurrence from radiation-induced brain injury. • Diffusion and perfusion MRI could improve the ability of the radiomics model in predicting the progression in patients with glioma. • Arterial spin labeling played an important role in predicting glioma progression using radiomics models.
Collapse
|
6
|
Li W, Le NN, Onishi N, Newitt DC, Wilmes LJ, Gibbs JE, Carmona-Bozo J, Liang J, Partridge SC, Price ER, Joe BN, Kornak J, Magbanua MJM, Nanda R, LeStage B, Esserman LJ, I-Spy Imaging Working Group, I-Spy Investigator Network, Van't Veer LJ, Hylton NM. Diffusion-Weighted MRI for Predicting Pathologic Complete Response in Neoadjuvant Immunotherapy. Cancers (Basel) 2022; 14:4436. [PMID: 36139594 PMCID: PMC9497087 DOI: 10.3390/cancers14184436] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 11/22/2022] Open
Abstract
This study tested the hypothesis that a change in the apparent diffusion coefficient (ADC) measured in diffusion-weighted MRI (DWI) is an independent imaging marker, and ADC performs better than functional tumor volume (FTV) for assessing treatment response in patients with locally advanced breast cancer receiving neoadjuvant immunotherapy. A total of 249 patients were randomized to standard neoadjuvant chemotherapy with pembrolizumab (pembro) or without pembrolizumab (control). DCE-MRI and DWI, performed prior to and 3 weeks after the start of treatment, were analyzed. Percent changes of tumor ADC metrics (mean, 5th to 95th percentiles of ADC histogram) and FTV were evaluated for the prediction of pathologic complete response (pCR) using a logistic regression model. The area under the ROC curve (AUC) estimated for the percent change in mean ADC was higher in the pembro cohort (0.73, 95% confidence interval [CI]: 0.52 to 0.93) than in the control cohort (0.63, 95% CI: 0.43 to 0.83). In the control cohort, the percent change of the 95th percentile ADC achieved the highest AUC, 0.69 (95% CI: 0.52 to 0.85). In the pembro cohort, the percent change of the 25th percentile ADC achieved the highest AUC, 0.75 (95% CI: 0.55 to 0.95). AUCs estimated for percent change of FTV were 0.61 (95% CI: 0.39 to 0.83) and 0.66 (95% CI: 0.47 to 0.85) for the pembro and control cohorts, respectively. Tumor ADC may perform better than FTV to predict pCR at an early treatment time-point during neoadjuvant immunotherapy.
Collapse
Affiliation(s)
- Wen Li
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Nu N Le
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Natsuko Onishi
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - David C Newitt
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Lisa J Wilmes
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Jessica E Gibbs
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Julia Carmona-Bozo
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Jiachao Liang
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Savannah C Partridge
- Department of Radiology, University of Washington, 1100 Fairview Ave N, Seattle, Washington, DC 98109, USA
| | - Elissa R Price
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Bonnie N Joe
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - John Kornak
- Department of Epidemiology and Biostatistics, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | - Mark Jesus M Magbanua
- Department of Laboratory Medicine, University of California, 2340 Sutter Street, San Francisco, CA 94115, USA
| | - Rita Nanda
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Barbara LeStage
- I-SPY 2 Advocacy Group, 499 Illinois Street, San Francisco, CA 94158, USA
| | - Laura J Esserman
- Department of Surgery, University of California, 550 16th Street, San Francisco, CA 94158, USA
| | | | - I-Spy Investigator Network
- Department of Radiology, University of Washington, 1100 Fairview Ave N, Seattle, Washington, DC 98109, USA
| | - Laura J Van't Veer
- Department of Laboratory Medicine, University of California, 2340 Sutter Street, San Francisco, CA 94115, USA
| | - Nola M Hylton
- Department of Radiology and Biomedical Imaging, University of California, 550 16th Street, San Francisco, CA 94158, USA
| |
Collapse
|
7
|
Qin D, Yang G, Jing H, Tan Y, Zhao B, Zhang H. Tumor Progression and Treatment-Related Changes: Radiological Diagnosis Challenges for the Evaluation of Post Treated Glioma. Cancers (Basel) 2022; 14:cancers14153771. [PMID: 35954435 PMCID: PMC9367286 DOI: 10.3390/cancers14153771] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Glioma is the most common primary malignant tumor of the adult central nervous system. Despite aggressive multimodal treatment, its prognosis remains poor. During follow-up, it remains challenging to distinguish treatment-related changes from tumor progression in treated patients with gliomas due to both share clinical symptoms and morphological imaging characteristics (with new and/or increasing enhancing mass lesions). The early effective identification of tumor progression and treatment-related changes is of great significance for the prognosis and treatment of gliomas. We believe that advanced neuroimaging techniques can provide additional information for distinguishing both at an early stage. In this article, we focus on the research of magnetic resonance imaging technology and artificial intelligence in tumor progression and treatment-related changes. Finally, it provides new ideas and insights for clinical diagnosis. Abstract As the most common neuro-epithelial tumors of the central nervous system in adults, gliomas are highly malignant and easy to recurrence, with a dismal prognosis. Imaging studies are indispensable for tracking tumor progression (TP) or treatment-related changes (TRCs). During follow-up, distinguishing TRCs from TP in treated patients with gliomas remains challenging as both share similar clinical symptoms and morphological imaging characteristics (with new and/or increasing enhancing mass lesions) and fulfill criteria for progression. Thus, the early identification of TP and TRCs is of great significance for determining the prognosis and treatment. Histopathological biopsy is currently the gold standard for TP and TRC diagnosis. However, the invasive nature of this technique limits its clinical application. Advanced imaging methods (e.g., diffusion magnetic resonance imaging (MRI), perfusion MRI, magnetic resonance spectroscopy (MRS), positron emission tomography (PET), amide proton transfer (APT) and artificial intelligence (AI)) provide a non-invasive and feasible technical means for identifying of TP and TRCs at an early stage, which have recently become research hotspots. This paper reviews the current research on using the abovementioned advanced imaging methods to identify TP and TRCs of gliomas. First, the review focuses on the pathological changes of the two entities to establish a theoretical basis for imaging identification. Then, it elaborates on the application of different imaging techniques and AI in identifying the two entities. Finally, the current challenges and future prospects of these techniques and methods are discussed.
Collapse
Affiliation(s)
- Danlei Qin
- College of Medical Imaging, Shanxi Medical University, Taiyuan 030001, China;
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School, Hospital of Stomatology, Taiyuan 030001, China
| | - Guoqiang Yang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan 030001, China; (G.Y.); (Y.T.)
| | - Hui Jing
- Department of MRI, The Six Hospital, Shanxi Medical University, Taiyuan 030008, China;
| | - Yan Tan
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan 030001, China; (G.Y.); (Y.T.)
| | - Bin Zhao
- College of Medical Imaging, Shanxi Medical University, Taiyuan 030001, China;
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School, Hospital of Stomatology, Taiyuan 030001, China
- Correspondence: (B.Z.); (H.Z.)
| | - Hui Zhang
- College of Medical Imaging, Shanxi Medical University, Taiyuan 030001, China;
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan 030001, China; (G.Y.); (Y.T.)
- Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, Taiyuan 030001, China
- Correspondence: (B.Z.); (H.Z.)
| |
Collapse
|
8
|
Taylor C, Ekert JO, Sefcikova V, Fersht N, Samandouras G. Discriminators of pseudoprogression and true progression in high-grade gliomas: A systematic review and meta-analysis. Sci Rep 2022; 12:13258. [PMID: 35918373 PMCID: PMC9345984 DOI: 10.1038/s41598-022-16726-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 07/14/2022] [Indexed: 11/09/2022] Open
Abstract
High-grade gliomas remain the most common primary brain tumour with limited treatments options and early recurrence rates following adjuvant treatments. However, differentiating true tumour progression (TTP) from treatment-related effects or pseudoprogression (PsP), may critically influence subsequent management options. Structural MRI is routinely employed to evaluate treatment responses, but misdiagnosis of TTP or PsP may lead to continuation of ineffective or premature cessation of effective treatments, respectively. A systematic review and meta-analysis were conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses method. Embase, MEDLINE, Web of Science and Google Scholar were searched for methods applied to differentiate PsP and TTP, and studies were selected using pre-specified eligibility criteria. The sensitivity and specificity of included studies were summarised. Three of the identified methods were compared in a separate subgroup meta-analysis. Thirty studies assessing seven distinct neuroimaging methods in 1372 patients were included in the systematic review. The highest performing methods in the subgroup analysis were DWI (AUC = 0.93 [0.91-0.95]) and DSC-MRI (AUC = 0.93 [0.90-0.95]), compared to DCE-MRI (AUC = 0.90 [0.87-0.93]). 18F-fluoroethyltyrosine PET (18F-FET PET) and amide proton transfer-weighted MRI (APTw-MRI) also showed high diagnostic accuracy, but results were based on few low-powered studies. Both DWI and DSC-MRI performed with high sensitivity and specificity for differentiating PsP from TTP. Considering the technical parameters and feasibility of each identified method, the authors suggested that, at present, DSC-MRI technique holds the most clinical potential.
Collapse
Affiliation(s)
- Chris Taylor
- UCL Queen Square Institute of Neurology, University College London, Gower St., Bloomsbury, Queen Square, London, WC1E 6BT, UK.
| | - Justyna O Ekert
- Wellcome Centre for Human Neuroimaging, University College London, 12 Queen Square, London, UK
| | - Viktoria Sefcikova
- UCL Queen Square Institute of Neurology, University College London, Gower St., Bloomsbury, Queen Square, London, WC1E 6BT, UK
| | - Naomi Fersht
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - George Samandouras
- Wellcome Centre for Human Neuroimaging, University College London, 12 Queen Square, London, UK
- Victor Horsley Department of Neurosurgery, The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| |
Collapse
|
9
|
Starck L, Skeie BS, Moen G, Grüner R. Dynamic Susceptibility Contrast MRI May Contribute in Prediction of Stereotactic Radiosurgery Outcome in Brain Metastases. Neurooncol Adv 2022; 4:vdac070. [PMID: 35673606 PMCID: PMC9167634 DOI: 10.1093/noajnl/vdac070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Following stereotactic radiosurgery (SRS), predicting treatment response is not possible at an early stage using structural imaging alone. Hence, the current study aims at investigating whether dynamic susceptibility contrast (DSC)-MRI estimated prior to SRS can provide predictive biomarkers in response to SRS treatment and characterize vascular characteristics of pseudo-progression. Methods In this retrospective study, perfusion-weighted DSC-MRI image data acquired with a temporal resolution of 1.45 seconds were collected from 41 patients suffering from brain metastases. Outcome was defined based on lesion volume changes in time (determined on structural images) or death. Motion correction and manual lesion delineation were performed prior to semi-automated, voxel-wise perfusion analysis. Statistical testing was performed using linear regression and a significance threshold at P = .05. Age, sex, primary cancers (pulmonary cancer and melanoma), lesion volume, and dichotomized survival time were added as covariates in the linear regression models (ANOVA). Results Relative cerebral blood volume (rCBV) and relative cerebral blood flow (rCBF) were found to be significantly lower prior to SRS treatment in patients with increasing lesion volume or early death post-SRS (P ≤ .01). Conclusion Unfavorable treatment outcome may be linked to low perfusion prior to SRS. Pseudo-progression may be preceded by a transient rCBF increase post-SRS. However, results should be verified in different or larger patient material.
Collapse
Affiliation(s)
- Lea Starck
- Department of Physics and Technology, University of Bergen, Norway
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | | | - Gunnar Moen
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Renate Grüner
- Department of Physics and Technology, University of Bergen, Norway
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
10
|
Li Y, Ma Y, Wu Z, Xie R, Zeng F, Cai H, Lui S, Song B, Chen L, Wu M. Advanced Imaging Techniques for Differentiating Pseudoprogression and Tumor Recurrence After Immunotherapy for Glioblastoma. Front Immunol 2021; 12:790674. [PMID: 34899760 PMCID: PMC8656432 DOI: 10.3389/fimmu.2021.790674] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/08/2021] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant tumor of the central nervous system with poor prognosis. Although the field of immunotherapy in glioma is developing rapidly, glioblastoma is still prone to recurrence under strong immune intervention. The major challenges in the process of immunotherapy are evaluating the curative effect, accurately distinguishing between treatment-related reactions and tumor recurrence, and providing guidance for clinical decision-making. Since the conventional magnetic resonance imaging (MRI) is usually difficult to distinguish between pseudoprogression and the true tumor progression, many studies have used various advanced imaging techniques to evaluate treatment-related responses. Meanwhile, criteria for efficacy evaluation of immunotherapy are constantly updated and improved. A standard imaging scheme to evaluate immunotherapeutic response will benefit patients finally. This review mainly summarizes the application status and future trend of several advanced imaging techniques in evaluating the efficacy of GBM immunotherapy.
Collapse
Affiliation(s)
- Yan Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Yiqi Ma
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Zijun Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Ruoxi Xie
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Fanxin Zeng
- Department of Clinic Medical Center, Dazhou Central Hospital, Dazhou, China
| | - Huawei Cai
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Su Lui
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Song
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China.,Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, China
| | - Min Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.,Department of Clinic Medical Center, Dazhou Central Hospital, Dazhou, China
| |
Collapse
|
11
|
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
|
12
|
Radiomics-based neural network predicts recurrence patterns in glioblastoma using dynamic susceptibility contrast-enhanced MRI. Sci Rep 2021; 11:9974. [PMID: 33976264 PMCID: PMC8113258 DOI: 10.1038/s41598-021-89218-z] [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: 12/10/2020] [Accepted: 04/22/2021] [Indexed: 02/03/2023] Open
Abstract
Glioblastoma remains the most devastating brain tumor despite optimal treatment, because of the high rate of recurrence. Distant recurrence has distinct genomic alterations compared to local recurrence, which requires different treatment planning both in clinical practice and trials. To date, perfusion-weighted MRI has revealed that perfusional characteristics of tumor are associated with prognosis. However, not much research has focused on recurrence patterns in glioblastoma: namely, local and distant recurrence. Here, we propose two different neural network models to predict the recurrence patterns in glioblastoma that utilizes high-dimensional radiomic profiles based on perfusion MRI: area under the curve (AUC) (95% confidence interval), 0.969 (0.903-1.000) for local recurrence; 0.864 (0.726-0.976) for distant recurrence for each patient in the validation set. This creates an opportunity to provide personalized medicine in contrast to studies investigating only group differences. Moreover, interpretable deep learning identified that salient radiomic features for each recurrence pattern are related to perfusional intratumoral heterogeneity. We also demonstrated that the combined salient radiomic features, or "radiomic risk score", increased risk of recurrence/progression (hazard ratio, 1.61; p = 0.03) in multivariate Cox regression on progression-free survival.
Collapse
|
13
|
Sun YZ, Yan LF, Han Y, Nan HY, Xiao G, Tian Q, Pu WH, Li ZY, Wei XC, Wang W, Cui GB. Differentiation of Pseudoprogression from True Progressionin Glioblastoma Patients after Standard Treatment: A Machine Learning Strategy Combinedwith Radiomics Features from T 1-weighted Contrast-enhanced Imaging. BMC Med Imaging 2021; 21:17. [PMID: 33535988 PMCID: PMC7860032 DOI: 10.1186/s12880-020-00545-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/28/2020] [Indexed: 12/29/2022] Open
Abstract
Background Based on conventional MRI images, it is difficult to differentiatepseudoprogression from true progressionin GBM patients after standard treatment, which isa critical issue associated with survival. The aim of this study was to evaluate the diagnostic performance of machine learning using radiomics modelfrom T1-weighted contrast enhanced imaging(T1CE) in differentiating pseudoprogression from true progression after standard treatment for GBM. Methods Seventy-sevenGBM patients, including 51 with true progression and 26 with pseudoprogression,who underwent standard treatment and T1CE, were retrospectively enrolled.Clinical information, including sex, age, KPS score, resection extent, neurological deficit and mean radiation dose, were also recorded collected for each patient. The whole tumor enhancementwas manually drawn on the T1CE image, and a total of texture 9675 features were extracted and fed to a two-step feature selection scheme. A random forest (RF) classifier was trained to separate the patients by their outcomes.The diagnostic efficacies of the radiomics modeland radiologist assessment were further compared by using theaccuracy (ACC), sensitivity and specificity. Results No clinical features showed statistically significant differences between true progression and pseudoprogression.The radiomic classifier demonstrated ACC, sensitivity, and specificity of 72.78%(95% confidence interval [CI]: 0.45,0.91), 78.36%(95%CI: 0.56,1.00) and 61.33%(95%CI: 0.20,0.82).The accuracy, sensitivity and specificity of three radiologists’ assessment were66.23%(95% CI: 0.55,0.76), 61.50%(95% CI: 0.43,0.78) and 68.62%(95% CI: 0.55,0.80); 55.84%(95% CI: 0.45,0.66),69.25%(95% CI: 0.50,0.84) and 49.13%(95% CI: 0.36,0.62); 55.84%(95% CI: 0.45,0.66), 69.23%(95% CI: 0.50,0.84) and 47.06%(95% CI: 0.34,0.61), respectively. Conclusion T1CE–based radiomics showed better classification performance compared with radiologists’ assessment.The radiomics modelwas promising in differentiating pseudoprogression from true progression.
Collapse
Affiliation(s)
- Ying-Zhi Sun
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Lin-Feng Yan
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Yu Han
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Hai-Yan Nan
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Gang Xiao
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Qiang Tian
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Wen-Hui Pu
- Student Brigade, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Ze-Yang Li
- Student Brigade, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | | | - Wen Wang
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| | - Guang-Bin Cui
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
14
|
Strauss SB, Meng A, Ebani EJ, Chiang GC. Imaging Glioblastoma Posttreatment: Progression, Pseudoprogression, Pseudoresponse, Radiation Necrosis. Neuroimaging Clin N Am 2021; 31:103-120. [PMID: 33220823 DOI: 10.1016/j.nic.2020.09.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Radiographic monitoring of posttreatment glioblastoma is important for clinical trials and determining next steps in management. Evaluation for tumor progression is confounded by the presence of treatment-related radiographic changes, making a definitive determination less straight-forward. The purpose of this article was to describe imaging tools available for assessing treatment response in glioblastoma, as well as to highlight the definitions, pathophysiology, and imaging features typical of true progression, pseudoprogression, pseudoresponse, and radiation necrosis.
Collapse
Affiliation(s)
- Sara B Strauss
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Alicia Meng
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Edward J Ebani
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Gloria C Chiang
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA.
| |
Collapse
|
15
|
Le Fèvre C, Constans JM, Chambrelant I, Antoni D, Bund C, Leroy-Freschini B, Schott R, Cebula H, Noël G. Pseudoprogression versus true progression in glioblastoma patients: A multiapproach literature review. Part 2 - Radiological features and metric markers. Crit Rev Oncol Hematol 2021; 159:103230. [PMID: 33515701 DOI: 10.1016/j.critrevonc.2021.103230] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/10/2021] [Accepted: 01/16/2021] [Indexed: 12/28/2022] Open
Abstract
After chemoradiotherapy for glioblastoma, pseudoprogression can occur and must be distinguished from true progression to correctly manage glioblastoma treatment and follow-up. Conventional treatment response assessment is evaluated via conventional MRI (contrast-enhanced T1-weighted and T2/FLAIR), which is unreliable. The emergence of advanced MRI techniques, MR spectroscopy, and PET tracers has improved pseudoprogression diagnostic accuracy. This review presents a literature review of the different imaging techniques and potential imaging biomarkers to differentiate pseudoprogression from true progression.
Collapse
Affiliation(s)
- Clara Le Fèvre
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Jean-Marc Constans
- Department of Radiology, Amiens-Picardie University Hospital, 1 rond-point du Professeur Christian Cabrol, 80054, Amiens Cedex 1, France.
| | - Isabelle Chambrelant
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Delphine Antoni
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Caroline Bund
- Department of Nuclear Medicine, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Benjamin Leroy-Freschini
- Department of Nuclear Medicine, 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.
| | - Hélène Cebula
- Departement of Neurosurgery, Hautepierre University Hospital, 1, avenue Molière, 67200, Strasbourg, France.
| | - Georges Noël
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| |
Collapse
|
16
|
Wang L, Wei L, Wang J, Li N, Gao Y, Ma H, Qu X, Zhang M. Evaluation of perfusion MRI value for tumor progression assessment after glioma radiotherapy: A systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e23766. [PMID: 33350761 PMCID: PMC7769293 DOI: 10.1097/md.0000000000023766] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/22/2020] [Accepted: 11/15/2020] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVES This study aimed to evaluate the diagnostic performance of magnetic resonance perfusion-weighted imaging (PWI) as a noninvasive method to assess post-treatment radiation effect and tumor progression in patients with glioma. METHODS A systematic literature search was performed in the PubMed, Cochrane Library, and Embase databases up to March 2020. The quality of the included studies was assessed by the quality assessment of diagnostic accuracy studies 2. Data were extracted to calculate sensitivity, specificity, and diagnostic odds ratio (DOR), 95% Confidence interval (CI) and analyze the heterogeneity of the studies (Spearman correlation coefficient, I2 test). We performed meta-regression and subgroup analyses to identify the impact of study heterogeneity. RESULTS Twenty studies were included, with available data for analysis on 939 patients and 968 lesions. All included studies used dynamic susceptibility contrast (DSC) PWI, four also used dynamic contrast-enhanced PWI, and three also used arterial spin marker imaging PWI. When DSC was considered, the pooled sensitivity and specificity were 0.83 (95% CI, 0.79 to 0.86) and 0.83 (95% CI, 0.78 to 0.87), respectively; pooled DOR, 21.31 (95% CI, 13.07 to 34.73); area under the curve (AUC), 0.887; Q∗, 0.8176. In studies using dynamic contrast-enhanced, the pooled sensitivity and specificity were 0.73 (95% CI, 0.66 to 0.80) and 0.80 (95% CI, 0.69 to 0.88), respectively; pooled DOR, 10.83 (95% CI, 2.01 to 58.43); AUC, 0.9416; Q∗, 0.8795. In studies using arterial spin labeling, the pooled sensitivity and specificity were 0.79 (95% CI, 0.69 to 0.87) and 0.78 (95% CI, 0.67 to 0.87), respectively; pooled DOR, 15.63 (95% CI, 4.61 to 53.02); AUC, 0.8786; Q∗, 0.809. CONCLUSIONS Perfusion magnetic resonance imaging displays moderate overall accuracy in identifying post-treatment radiation effect and tumor progression in patients with glioma. Based on the current evidence, DSC-PWI is a relatively reliable option for assessing tumor progression after glioma radiotherapy.
Collapse
Affiliation(s)
| | - Lizhou Wei
- Department of neurosurgery, Xijing hospital, Fourth military medical university
| | | | - Na Li
- Department of radiology, Ninth Hospital of Xi’an
| | - Yanzhong Gao
- Department of radiology, Ninth Hospital of Xi’an
| | - Hongge Ma
- Department of radiology, Ninth Hospital of Xi’an
| | - Xinran Qu
- Department of radiology, Ninth Hospital of Xi’an
| | - Ming Zhang
- Department of Radiology, the First Affiliated Hospital of Xi ’an Jiao tong University, Shaanxi Province, China
| |
Collapse
|
17
|
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: 23] [Impact Index Per Article: 4.6] [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
|
18
|
Tsakiris C, Siempis T, Alexiou GA, Zikou A, Sioka C, Voulgaris S, Argyropoulou MI. Differentiation Between True Tumor Progression of Glioblastoma and Pseudoprogression Using Diffusion-Weighted Imaging and Perfusion-Weighted Imaging: Systematic Review and Meta-analysis. World Neurosurg 2020; 144:e100-e109. [DOI: 10.1016/j.wneu.2020.07.218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 01/08/2023]
|
19
|
Sawlani V, Patel MD, Davies N, Flintham R, Wesolowski R, Ughratdar I, Pohl U, Nagaraju S, Petrik V, Kay A, Jacob S, Sanghera P, Wykes V, Watts C, Poptani H. Multiparametric MRI: practical approach and pictorial review of a useful tool in the evaluation of brain tumours and tumour-like lesions. Insights Imaging 2020; 11:84. [PMID: 32681296 PMCID: PMC7367972 DOI: 10.1186/s13244-020-00888-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/24/2020] [Indexed: 12/17/2022] Open
Abstract
MRI has a vital role in the assessment of intracranial lesions. Conventional MRI has limited specificity and multiparametric MRI using diffusion-weighted imaging, perfusion-weighted imaging and magnetic resonance spectroscopy allows more accurate assessment of the tissue microenvironment. The purpose of this educational pictorial review is to demonstrate the role of multiparametric MRI for diagnosis, treatment planning and for assessing treatment response, as well as providing a practical approach for performing and interpreting multiparametric MRI in the clinical setting. A variety of cases are presented to demonstrate how multiparametric MRI can help differentiate neoplastic from non-neoplastic lesions compared to conventional MRI alone.
Collapse
Affiliation(s)
- Vijay Sawlani
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK.
- University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Markand Dipankumar Patel
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
- University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Nigel Davies
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
| | - Robert Flintham
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
| | - Roman Wesolowski
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
| | - Ismail Ughratdar
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
| | - Ute Pohl
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
| | - Santhosh Nagaraju
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
| | - Vladimir Petrik
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
| | - Andrew Kay
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
| | - Saiju Jacob
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
- University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Paul Sanghera
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
| | - Victoria Wykes
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
- University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Colin Watts
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
- University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Harish Poptani
- Centre for Pre-Clinical Imaging, Department of Cellular and Molecular Physiology, University of Liverpool, Crown Street, Liverpool, L69 3BX, UK
| |
Collapse
|
20
|
|
21
|
Yu Y, Ma Y, Sun M, Jiang W, Yuan T, Tong D. Meta-analysis of the diagnostic performance of diffusion magnetic resonance imaging with apparent diffusion coefficient measurements for differentiating glioma recurrence from pseudoprogression. Medicine (Baltimore) 2020; 99:e20270. [PMID: 32501974 PMCID: PMC7306328 DOI: 10.1097/md.0000000000020270] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/13/2020] [Accepted: 04/15/2020] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE The accurate differentiation of glioma recurrence from pseudoprogression (PSP) after therapy remains a considerable clinical challenge. Several studies have shown that diffusion magnetic resonance imaging (MRI) has potential value in distinguishing these 2 outcomes. The current meta-analysis examined the diagnostic accuracy of diffusion MRI with the apparent diffusion coefficient (ADC) in the differentiation of glioma recurrence from PSP. METHOD PubMed, Embase, Cochrane Library, and Chinese Biomedical databases were reviewed to identify studies that fulfilled our inclusion/exclusion criteria and were published on or before May 5, 2019. Threshold effects; heterogeneity; pooled sensitivity (SENS), specificity, positive likelihood ratio, and negative likelihood ratio; and diagnostic odds ratio were calculated. The overall diagnostic usefulness of diffusion MRI-derived ADC values was assessed by calculating the area under the curve (AUC) following summary receiver operating characteristic (SROC) analysis. RESULTS Six eligible studies examined a total of 214 patients. Calculation of pooled values indicated the SENS was 0.95 (95% confidence interval [CI] = 0.89-0.98), specificity was 0.83 (95% CI = 0.72-0.91), positive likelihood ratio was 4.82 (95% CI = 2.93-7.93), negative likelihood ratio was 0.08 (95% CI = 0.04-0.17), and diagnostic odds ratio was 59.63 (95% CI = 22.63-157.37). The SROC AUC was 0.9322. Publication bias was not significant, and SENS analysis indicated the results were relatively stable. CONCLUSIONS Our meta-analysis indicated that diffusion MRI with quantitative ADC is an effective approach for differentiation of glioma recurrence from PSP, and can be used as an auxiliary tool to diagnose glioma progression.
Collapse
Affiliation(s)
| | | | - Mengyao Sun
- Department of Internal Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
| | | | | | | |
Collapse
|
22
|
Advanced multimodal imaging in differentiating glioma recurrence from post-radiotherapy changes. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 151:281-297. [PMID: 32448612 DOI: 10.1016/bs.irn.2020.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gliomas are the most common malignant primary brain tumor, and their prognosis is extremely poor. Radiotherapy is an important treatment for glioma patients, but the changes caused by radiotherapy have brought difficulties in clinical image evaluation because differentiating glioma recurrence from post-radiotherapy changes including pseudo-progression (PD) and radiation necrosis (RN) remains a challenge. Therefore, accurate and reliable imaging evaluation is very important for making clinical decisions. In recent years, advanced multimodal imaging techniques have been applied to achieve the goal of better differentiating glioma recurrence from post-radiotherapy changes for minimizing errors associated with interpretation of treatment effects. In this review, we discuss the recent applications of advanced multimodal imaging such as diffusion MRI sequences, amide proton transfer MRI sequences, perfusion MRI sequences, MR spectroscopy and multinuclides PET/CT in the evaluation of post-radiotherapy treatment response in glioma patients and highlight their potential role in differentiating post-radiotherapy changes from glioma recurrence.
Collapse
|
23
|
Vajapeyam S, Brown D, Billups C, Patay Z, Vezina G, Shiroishi MS, Law M, Baxter P, Onar-Thomas A, Fangusaro JR, Dunkel IJ, Poussaint TY. Advanced ADC Histogram, Perfusion, and Permeability Metrics Show an Association with Survival and Pseudoprogression in Newly Diagnosed Diffuse Intrinsic Pontine Glioma: A Report from the Pediatric Brain Tumor Consortium. AJNR Am J Neuroradiol 2020; 41:718-724. [PMID: 32241771 DOI: 10.3174/ajnr.a6499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/10/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE Diffuse intrinsic pontine glioma is a lethal childhood brain cancer with dismal prognosis and MR imaging is the primary methodology used for diagnosis and monitoring. Our aim was to determine whether advanced diffusion, perfusion, and permeability MR imaging metrics predict survival and pseudoprogression in children with newly diagnosed diffuse intrinsic pontine glioma. MATERIALS AND METHODS A clinical trial using the poly (adenosine diphosphate ribose) polymerase (PARP) inhibitor veliparib concurrently with radiation therapy, followed by maintenance therapy with veliparib + temozolomide, in children with diffuse intrinsic pontine glioma was conducted by the Pediatric Brain Tumor Consortium. Standard MR imaging, DWI, dynamic contrast-enhanced perfusion, and DSC perfusion were performed at baseline and approximately every 2 months throughout treatment. ADC histogram metrics of T2-weighted FLAIR and enhancing tumor volume, dynamic contrast-enhanced permeability metrics for enhancing tumors, and tumor relative CBV from DSC perfusion MR imaging were calculated. Baseline values, post-radiation therapy changes, and longitudinal trends for all metrics were evaluated for associations with survival and pseudoprogression. RESULTS Fifty children were evaluable for survival analyses. Higher baseline relative CBV was associated with shorter progression-free survival (P = .02, Q = 0.089) and overall survival (P = .006, Q = 0.055). Associations of higher baseline mean transfer constant from the blood plasma into the extravascular extracellular space with shorter progression-free survival (P = .03, Q = 0.105) and overall survival (P = .03, Q = 0.102) trended toward significance. An increase in relative CBV with time was associated with shorter progression-free survival (P < .001, Q < 0.001) and overall survival (P = .004, Q = 0.043). Associations of longitudinal mean extravascular extracellular volume fraction with progression-free survival (P = .03, Q = 0.104) and overall survival (P = .03, Q = 0.105) and maximum transfer constant from the blood plasma into the extravascular extracellular space with progression-free survival (P = .03, Q = 0.102) trended toward significance. Greater increases with time were associated with worse outcomes. True radiologic progression showed greater post-radiation therapy decreases in mode_ADC_FLAIR compared with pseudoprogression (means, -268.15 versus -26.11, P = .01.) CONCLUSIONS: ADC histogram, perfusion, and permeability MR imaging metrics in diffuse intrinsic pontine glioma are useful in predicting survival and pseudoprogression.
Collapse
Affiliation(s)
- S Vajapeyam
- From the Radiology (S.V., T.Y.P.), Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - D Brown
- DF/HCC Tumor Imaging Metrics Core (D.B.), Massachusetts General Hospital, Boston, Massachusetts
| | | | - Z Patay
- Diagnostic Imaging (Z.P.), St. Jude Children's Research Hospital, Memphis, Tennessee
| | - G Vezina
- Radiology (G.V.), Children's National Medical Center, Washington, DC
| | - M S Shiroishi
- Radiology (M.S.S.), Keck Medical Center of USC, Los Angeles, California
| | - M Law
- Neuroscience (M.L.), Monash University, Melbourne, Australia
| | - P Baxter
- Cancer and Hematology Center (P.B.), Texas Children's Hospital, Houston, Texas
| | | | - J R Fangusaro
- Aflac Cancer and Blood Disorders Center (J.R.F.), Children's Healthcare of Atlanta, Atlanta, Georgia
| | - I J Dunkel
- Pediatrics (I.J.D.), Memorial Sloan Kettering Cancer Center, New York, New York
| | - T Y Poussaint
- From the Radiology (S.V., T.Y.P.), Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
24
|
Gharzeddine K, Hatzoglou V, Holodny AI, Young RJ. MR Perfusion and MR Spectroscopy of Brain Neoplasms. Radiol Clin North Am 2019; 57:1177-1188. [PMID: 31582043 DOI: 10.1016/j.rcl.2019.07.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Advances in imaging techniques, such as MR perfusion and spectroscopy, are increasingly indispensable in the management and treatment plans of brain neoplasms: from diagnosing, molecular/genetic typing and grading neoplasms, augmenting biopsy results and improving accuracy, to ultimately directing and monitoring treatment and response. New developments in treatment methods have resulted in new diagnostic challenges for conventional MR imaging, such as pseudoprogression, where MR perfusion has the widest current application. MR spectroscopy is showing increasing promise in noninvasively determining genetic subtypes and, potentially, susceptibility to molecular targeted therapies.
Collapse
Affiliation(s)
- Karem Gharzeddine
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Vaios Hatzoglou
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, 1275 York Avenue, New York, NY 10065, USA
| | - Andrei I Holodny
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, Weill Cornell Graduate School of Medical Sciences, 1275 York Avenue, New York, NY 10065, USA.
| | - Robert J Young
- Brain Imaging, Neuroradiology Research, Neuroradiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
25
|
Manhard MK, Bilgic B, Liao C, Han S, Witzel T, Yen YF, Setsompop K. Accelerated whole-brain perfusion imaging using a simultaneous multislice spin-echo and gradient-echo sequence with joint virtual coil reconstruction. Magn Reson Med 2019; 82:973-983. [PMID: 31069861 PMCID: PMC6692914 DOI: 10.1002/mrm.27784] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Dynamic susceptibility contrast imaging requires high temporal sampling, which poses limits on achievable spatial coverage and resolution. Additionally, more encoding-intensive multi-echo acquisitions for quantitative imaging are desired to mitigate contrast leakage effects, which further limits spatial encoding. We present an accelerated sequence that provides whole-brain coverage at an improved spatio-temporal resolution, to allow for dynamic quantitative R2 and R2 * mapping during contrast-enhanced imaging. METHODS A multi-echo spin and gradient-echo sequence was implemented with simultaneous multislice acquisition. Complementary k-space sampling between repetitions and joint virtual coil reconstruction were used along with a dynamic phase-matching technique to achieve high-quality reconstruction at 9-fold acceleration, which enabled 2 × 2 × 5 mm whole-brain imaging at TR of 1.5 to 1.7 seconds. The multi-echo images from this sequence were fit to achieve quantitative R2 and R2 * maps for each repetition, and subsequently used to find perfusion measures including cerebral blood flow and cerebral blood volume. RESULTS Images reconstructed using joint virtual coil show improved image quality and g-factor compared with conventional reconstruction methods, resulting in improved quantitative maps with a 9-fold acceleration factor and whole-brain coverage during the dynamic perfusion acquisition. CONCLUSION The method presented shows the advantage of using a joint virtual coil-GRAPPA reconstruction to allow for high acceleration factors while maintaining reliable image quality for quantitative perfusion mapping, with the potential to improve tumor diagnostics and monitoring.
Collapse
Affiliation(s)
- Mary Kate Manhard
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Berkin Bilgic
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Congyu Liao
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - SoHyun Han
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Thomas Witzel
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Yi-Fen Yen
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Kawin Setsompop
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Health Sciences and Technology, MIT, Cambridge, MA, USA
| |
Collapse
|
26
|
Strauss SB, Meng A, Ebani EJ, Chiang GC. Imaging Glioblastoma Posttreatment: Progression, Pseudoprogression, Pseudoresponse, Radiation Necrosis. Radiol Clin North Am 2019; 57:1199-1216. [PMID: 31582045 DOI: 10.1016/j.rcl.2019.07.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Radiographic monitoring of posttreatment glioblastoma is important for clinical trials and determining next steps in management. Evaluation for tumor progression is confounded by the presence of treatment-related radiographic changes, making a definitive determination less straight-forward. The purpose of this article was to describe imaging tools available for assessing treatment response in glioblastoma, as well as to highlight the definitions, pathophysiology, and imaging features typical of true progression, pseudoprogression, pseudoresponse, and radiation necrosis.
Collapse
Affiliation(s)
- Sara B Strauss
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Alicia Meng
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Edward J Ebani
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Gloria C Chiang
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA.
| |
Collapse
|
27
|
Carceller F, Jerome NP, Fowkes LA, Khabra K, Mackinnon A, Bautista F, Marshall LV, Vaidya S, Mandeville H, Morgan V, Leach MO, Koh DM. Post-radiotherapy apparent diffusion coefficient (ADC) in children and young adults with high-grade gliomas and diffuse intrinsic pontine gliomas. Pediatr Hematol Oncol 2019; 36:103-112. [PMID: 30978130 DOI: 10.1080/08880018.2019.1592267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 03/05/2019] [Indexed: 01/14/2023]
Abstract
Objectives: Diffusion-weighted magnetic resonance imaging (DW-MRI) offers potential to monitor response and predict survival in high-grade gliomas (HGG) and diffuse intrinsic pontine gliomas (DIPG). We hypothesized that post-radiotherapy DW-MRI may provide prognostic imaging biomarkers in children and young adults with these tumors. Methods: Patients aged ≤21 years diagnosed between 2005 and 2012 were eligible. The tumor median apparent diffusion coefficient (ADC) and its 5th percentile (C5-ADC) were determined at the first post-radiotherapy scan and at the time of radiological progression. DW-MRI parameters were correlated with survival endpoints, temozolomide use and pseudoprogression, when it occurred. Results: Out of 40 patients (20 HGG, 20 DIPG), 23 had evaluable DW-MRI post-radiotherapy and 25 at radiological progression. There were 6 episodes of pseudoprogression. Hazard ratios (95%CI) for progression-free survival were 0.998 (0.993-1.003) for median ADC and 1.003 (0.996-1.010) for C5-ADC. Hazard ratios (95%CI) for overall survival were 1.0009 (0.996-1.006) for median ADC and 0.998 (0.992-1.004) for C5-ADC. Post-radiotherapy median and C5-ADC values were not significantly different between patients treated with radiotherapy alone versus radiotherapy/temozolomide. The median and C5-ADC values were not significantly different at the time of pseudoprogression compared to those at tumor progression. Conclusions: Post-radiotherapy median ADC and C5-ADC were not prognostic, nor able to differentiate radiosensitization with temozolomide or occurrence of pseudoprogression in this cohort of HGG and DIPG patients. Further exploration of alternative DW parameters, study timepoints or data modeling may contribute to the development of prognostic/predictive imaging biomarkers for children and young adults with HGG or DIPG.
Collapse
Affiliation(s)
- Fernando Carceller
- a Paediatric Neuro-Oncology and Drug Development Teams, Children & Young People's Unit , The Royal Marsden NHS Foundation Trust , London , UK
- b Division of Clinical Studies and Cancer Therapeutics , The Institute of Cancer Research , London , UK
| | - Neil P Jerome
- c Cancer Research UK Cancer Imaging Centre , The Institute of Cancer Research , London , UK
- d Department of Circulation and Medical Imaging , NTNU - Norwegian University of Science and Technology , Trondheim , Norway
| | - Lucy A Fowkes
- e Department of Radiology , The Royal Marsden NHS Foundation Trust , London , UK
| | - Komel Khabra
- f The Royal Marsden NHS Foundation Trust , Research Data Management and Statistics Unit , London , UK
- g MRC Clinical Trials Unit, University College London , London , UK
| | - Andrew Mackinnon
- e Department of Radiology , The Royal Marsden NHS Foundation Trust , London , UK
| | | | - Lynley V Marshall
- a Paediatric Neuro-Oncology and Drug Development Teams, Children & Young People's Unit , The Royal Marsden NHS Foundation Trust , London , UK
- b Division of Clinical Studies and Cancer Therapeutics , The Institute of Cancer Research , London , UK
| | - Sucheta Vaidya
- a Paediatric Neuro-Oncology and Drug Development Teams, Children & Young People's Unit , The Royal Marsden NHS Foundation Trust , London , UK
- b Division of Clinical Studies and Cancer Therapeutics , The Institute of Cancer Research , London , UK
| | - Henry Mandeville
- i Department of Radiotherapy , The Royal Marsden NHS Foundation Trust , London , UK
| | - Veronica Morgan
- e Department of Radiology , The Royal Marsden NHS Foundation Trust , London , UK
| | - Martin O Leach
- c Cancer Research UK Cancer Imaging Centre , The Institute of Cancer Research , London , UK
| | - Dow-Mu Koh
- e Department of Radiology , The Royal Marsden NHS Foundation Trust , London , UK
| |
Collapse
|
28
|
Du S, Sun H, Gao S, Xin J, Lu Z, Chen Z, Pan S, Guo Q. Relationship between 18F-FDG PET metabolic parameters and MRI intravoxel incoherent motion (IVIM) histogram parameters and their correlations with clinicopathological features of cervical cancer: evidence from integrated PET/MRI. Clin Radiol 2019; 74:178-186. [DOI: 10.1016/j.crad.2018.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 11/09/2018] [Indexed: 12/14/2022]
|
29
|
Integrated PET-MRI for Glioma Surveillance: Perfusion-Metabolism Discordance Rate and Association With Molecular Profiling. AJR Am J Roentgenol 2019; 212:883-891. [PMID: 30779663 DOI: 10.2214/ajr.18.20531] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Both 18F-FDG PET and perfusion MRI are commonly used techniques for posttreatment glioma surveillance. Using integrated PET-MRI, we assessed the rate of discordance between simultaneously acquired FDG PET images and dynamic contrast-enhanced (DCE) perfusion MR images and determined whether tumor genetics predicts discordance. MATERIALS AND METHODS Forty-one consecutive patients with high-grade gliomas (20 with grade IV gliomas and 21 with grade III gliomas) underwent a standardized tumor protocol performed using an integrated 3-T PET-MRI scanner. Quantitative measures of standardized uptake value, plasma volume, and permeability were obtained from segmented whole-tumor volumes of interest and targeted ROIs. ROC curve analysis and the Youden index were used to identify optimal cutoffs for FDG PET and DCE-MRI. Two-by-two contingency tables and percent agreement were used to assess accuracy and concordance. Twenty-six patients (63%) from the cohort underwent next-generation sequencing for tumor genetics. RESULTS The best-performing FDG PET and DCE-MRI cutoffs achieved sensitivities of 94% and 91%, respectively; specificities of 56% and 89%, respectively; and accuracies of 80% and 83%, respectively. FDG PET and DCE-MRI findings were discordant for 11 patients (27%), with DCE-MRI findings correct for six of these patients (55%). Tumor grade, tumor volume, bevacizumab exposure, and time since radiation predicted discordance between FDG PET and DCE-MRI findings, with an ROC AUC value of 0.78. Isocitrate dehydrogenase gene and receptor tyrosine kinase gene pathway mutations increased the ROC AUC value to 0.83. CONCLUSION FDG PET and DCE-MRI show comparable accuracy and sensitivity in identifying tumor progression. These modalities were shown to have discordant findings for more than a quarter of the patients assessed. Tumor genetics may contribute to perfusion-metabolism discordance, warranting further investigation.
Collapse
|
30
|
Sawlani V, Davies N, Patel M, Flintham R, Fong C, Heyes G, Cruickshank G, Steven N, Peet A, Hartley A, Benghiat H, Meade S, Sanghera P. Evaluation of Response to Stereotactic Radiosurgery in Brain Metastases Using Multiparametric Magnetic Resonance Imaging and a Review of the Literature. Clin Oncol (R Coll Radiol) 2018; 31:41-49. [PMID: 30274767 DOI: 10.1016/j.clon.2018.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/03/2018] [Accepted: 08/16/2018] [Indexed: 01/01/2023]
Abstract
AIMS Following stereotactic radiosurgery (SRS), brain metastases initially increase in size in up to a third of cases, suggesting treatment failure. Current imaging using structural magnetic resonance imaging (MRI) cannot differentiate between tumour recurrence and SRS-induced changes, creating difficulties with patient management. Combining multiparametric MRI techniques, which assess tissue physiological and metabolic information, has shown promise in answering this clinical question. MATERIALS AND METHODS Multiparametric MRI techniques, including spectroscopy, diffusion and perfusion imaging, were used for the differentiation of radiation-related changes and tumour recurrence after SRS for intracranial metastases in six cases. All patients presented with enlargement of the treated lesion, an increase in perilesional brain oedema and aggravation or appearance of neurological signs and symptoms from 7 to 29 weeks after primary treatment. RESULTS Multiparametric imaging helped to differentiate features of tumour progression (n = 4) from radiation-related changes (n = 2). A low apparent diffusion coefficient (ADC) <1000 × 10-6 mm2/s, high relative cerebral blood volume (rCBV) ratio > 2.1, high choline:creatine (Cho:Cr) ratio > 1.8 suggested tumour recurrence. A high ADC > 1000 × 10-6 mm2/s, low rCBV ratio < 2.1, Cho:Cr ratio < 1.8 suggested SRS-induced radiation changes. Multiparametric MRI diagnosis was confirmed by histology or radiological and clinical follow-up. CONCLUSION Multiparametric MRI was helpful in the early identification of radiation-related changes and tumour recurrence and may be useful for monitoring treatment changes in intracranial neoplasms after SRS treatment.
Collapse
Affiliation(s)
- V Sawlani
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| | - N Davies
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - M Patel
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - R Flintham
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - C Fong
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - G Heyes
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - G Cruickshank
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - N Steven
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - A Peet
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; Birmingham Children's Hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - A Hartley
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - H Benghiat
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - S Meade
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - P Sanghera
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
31
|
Vander Jagt TA, Davis LE, Thakur MD, Franz C, Pollock JM. Pseudoprogression of CNS metastatic disease of alveolar soft part sarcoma during anti-PDL1 treatment. Radiol Case Rep 2018; 13:882-885. [PMID: 29991973 PMCID: PMC6037874 DOI: 10.1016/j.radcr.2018.05.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/17/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors are increasingly used in treatment of metastatic renal cell carcinoma, melanoma, and nonsmall cell lung cancer, as well as in clinical trials for novel targets. We present a pediatric patient with metastatic alveolar soft part sarcoma who was treated with MPDL3280 (Atezolizumab), a monoclonal anti-programmed death ligand-1 antibody. Imaging results for the patient suggested disease progression of multiple brain metastases with stable systemic disease. The patient met response evaluation criteria in solid tumors (RECIST) criteria of progression of disease and was removed from treatment with MPDL3280. Subsequent surgical resection of the brain lesions revealed nonviable tumor with extensive lymphocytic infiltrates consistent with pseudoprogression. This case report adds to a growing number of reports that question reliance on RECIST criteria and suggest need for further refinement of RECIST or irRECIST during immune checkpoint inhibitor treatment for central nervous system metastatic lesions.
Collapse
Affiliation(s)
| | - Lara E Davis
- Department of Pediatric Hematology and Oncology; Knight Cancer Institute Division of Hematology and Medical Oncology, Oregon Health Science University, Portland, OR, USA.,Division of Oncology Biomarker Development, Genetech, South San Francisco, CA, USA
| | - Meghna Das Thakur
- Division of Oncology Biomarker Development, Genetech, South San Francisco, CA, USA
| | - Carl Franz
- Division of Chemistry and Biology, Lake Tahoe Community College, South Lake Tahoe, CA, USA
| | - Jeffery M Pollock
- Department of Radiology, Oregon Health Science University, Portland, OR, USA
| |
Collapse
|
32
|
Machireddy A, Thibault G, Huang W, Song X. Analysis of DCE-MRI for Early Prediction of Breast Cancer Therapy Response. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2018; 2018:682-685. [PMID: 30440488 DOI: 10.1109/embc.2018.8512301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Positive response to neoadjuvant chemotherapy (NACT) has been correlated to better long-term outcomes in breast cancer treatment. Early prediction of response to NACT can help modify the regimen for non-responding patients, sparing them of potential toxicities of ineffective therapies. It has been observed that tumor functions such as vascularization and vascular permeability change even before noticeable changes occur in the tumor size in response to the treatment. Therefore, it is essential to have reliable imaging based features to measure these changes. Texture analysis on parametric maps from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) has shown to be a good predictor of breast cancer response to NACT at an early stage. But hand crafted texture features might not be able to capture the rich spatio-temporal information in the parametric maps. In this work, we studied the ability of convolutional neural networks in predicting the response to NACT at an early stage.
Collapse
|
33
|
Bao S, Watanabe Y, Takahashi H, Tanaka H, Arisawa A, Matsuo C, Wu R, Fujimoto Y, Tomiyama N. Differentiating between Glioblastoma and Primary CNS Lymphoma Using Combined Whole-tumor Histogram Analysis of the Normalized Cerebral Blood Volume and the Apparent Diffusion Coefficient. Magn Reson Med Sci 2018; 18:53-61. [PMID: 29848919 PMCID: PMC6326759 DOI: 10.2463/mrms.mp.2017-0135] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose: This study aimed to determine whether whole-tumor histogram analysis of normalized cerebral blood volume (nCBV) and apparent diffusion coefficient (ADC) for contrast-enhancing lesions can be used to differentiate between glioblastoma (GBM) and primary central nervous system lymphoma (PCNSL). Methods: From 20 patients, 9 with PCNSL and 11 with GBM without any hemorrhagic lesions, underwent MRI, including diffusion-weighted imaging and dynamic susceptibility contrast perfusion-weighted imaging before surgery. Histogram analysis of nCBV and ADC from whole-tumor voxels in contrast-enhancing lesions was performed. An unpaired t-test was used to compare the mean values for each type of tumor. A multivariate logistic regression model (LRM) was performed to classify GBM and PCNSL using the best parameters of ADC and nCBV. Results: All nCBV histogram parameters of GBMs were larger than those of PCNSLs, but only average nCBV was statistically significant after Bonferroni correction. Meanwhile, ADC histogram parameters were also larger in GBM compared to those in PCNSL, but these differences were not statistically significant. According to receiver operating characteristic curve analysis, the nCBV average and ADC 25th percentile demonstrated the largest area under the curve with values of 0.869 and 0.838, respectively. The LRM combining these two parameters differentiated between GBM and PCNSL with a higher area under the curve value (Logit (P) = −21.12 + 10.00 × ADC 25th percentile (10−3 mm2/s) + 5.420 × nCBV mean, P < 0.001). Conclusion: Our results suggest that whole-tumor histogram analysis of nCBV and ADC combined can be a valuable objective diagnostic method for differentiating between GBM and PCNSL.
Collapse
Affiliation(s)
- Shixing Bao
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine
| | - Yoshiyuki Watanabe
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine
| | - Hiroto Takahashi
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine
| | - Hisashi Tanaka
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine
| | - Atsuko Arisawa
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine
| | - Chisato Matsuo
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine
| | - Rongli Wu
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine
| | - Yasunori Fujimoto
- Department of Neurosurgery, Osaka University Graduate School of Medicine
| | - Noriyuki Tomiyama
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine
| |
Collapse
|
34
|
Thust SC, van den Bent MJ, Smits M. Pseudoprogression of brain tumors. J Magn Reson Imaging 2018; 48:571-589. [PMID: 29734497 PMCID: PMC6175399 DOI: 10.1002/jmri.26171] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 04/07/2018] [Indexed: 12/11/2022] Open
Abstract
This review describes the definition, incidence, clinical implications, and magnetic resonance imaging (MRI) findings of pseudoprogression of brain tumors, in particular, but not limited to, high-grade glioma. Pseudoprogression is an important clinical problem after brain tumor treatment, interfering not only with day-to-day patient care but also the execution and interpretation of clinical trials. Radiologically, pseudoprogression is defined as a new or enlarging area(s) of contrast agent enhancement, in the absence of true tumor growth, which subsides or stabilizes without a change in therapy. The clinical definitions of pseudoprogression have been quite variable, which may explain some of the differences in reported incidences, which range from 9-30%. Conventional structural MRI is insufficient for distinguishing pseudoprogression from true progressive disease, and advanced imaging is needed to obtain higher levels of diagnostic certainty. Perfusion MRI is the most widely used imaging technique to diagnose pseudoprogression and has high reported diagnostic accuracy. Diagnostic performance of MR spectroscopy (MRS) appears to be somewhat higher, but MRS is less suitable for the routine and universal application in brain tumor follow-up. The combination of MRS and diffusion-weighted imaging and/or perfusion MRI seems to be particularly powerful, with diagnostic accuracy reaching up to or even greater than 90%. While diagnostic performance can be high with appropriate implementation and interpretation, even a combination of techniques, however, does not provide 100% accuracy. It should also be noted that most studies to date are small, heterogeneous, and retrospective in nature. Future improvements in diagnostic accuracy can be expected with harmonization of acquisition and postprocessing, quantitative MRI and computer-aided diagnostic technology, and meticulous evaluation with clinical and pathological data. LEVEL OF EVIDENCE 3 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2018.
Collapse
Affiliation(s)
- Stefanie C. Thust
- Lysholm Neuroradiology DepartmentNational Hospital for Neurology and NeurosurgeryLondonUK
- Department of Brain Rehabilitation and RepairUCL Institute of NeurologyLondonUK
- Imaging DepartmentUniversity College London HospitalLondonUK
| | - Martin J. van den Bent
- Department of NeurologyThe Brain Tumor Centre at Erasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MCUniversity Medical Centre RotterdamRotterdamThe Netherlands
| |
Collapse
|
35
|
Horvath-Rizea D, Surov A, Hoffmann KT, Garnov N, Vörkel C, Kohlhof-Meinecke P, Ganslandt O, Bäzner H, Gihr GA, Kalman M, Henkes E, Henkes H, Schob S. The value of whole lesion ADC histogram profiling to differentiate between morphologically indistinguishable ring enhancing lesions-comparison of glioblastomas and brain abscesses. Oncotarget 2018; 9:18148-18159. [PMID: 29719596 PMCID: PMC5915063 DOI: 10.18632/oncotarget.24454] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 01/30/2018] [Indexed: 12/17/2022] Open
Abstract
Background Morphologically similar appearing ring enhancing lesions in the brain parenchyma can be caused by a number of distinct pathologies, however, they consistently represent life-threatening conditions. The two most frequently encountered diseases manifesting as such are glioblastoma multiforme (GBM) and brain abscess (BA), each requiring disparate therapeutical approaches. As a result of their morphological resemblance, essential treatment might be significantly delayed or even ommited, in case results of conventional imaging remain inconclusive. Therefore, our study aimed to investigate, whether ADC histogram profiling reliably can distinguish between both entities, thus enhancing the differential diagnostic process and preventing treatment failure in this highly critical context. Methods 103 patients (51 BA, 52 GBM) with histopathologically confirmed diagnosis were enrolled. Pretreatment diffusion weighted imaging (DWI) was obtained in a 1.5T system using b values of 0, 500, and 1000 s/mm2. Whole lesion ADC volumes were analyzed using a histogram-based approach. Statistical analysis was performed using SPSS version 23. Results All investigated parameters were statistically different in comparison of both groups. Most importantly, ADCp10 was able to differentiate reliably between BA and GBM with excellent accuracy (0.948) using a cutpoint value of 70 × 10−5 mm2 × s−1. Conclusions ADC whole lesion histogram profiling provides a valuable tool to differentiate between morphologically indistinguishable mass lesions. Among the investigated parameters, the 10th percentile of the ADC volume distinguished best between GBM and BA.
Collapse
Affiliation(s)
| | - Alexey Surov
- Clinic for Diagnostic and Interventional Radiology, University Hospital Leipzig, Leipzig, Germany
| | - Karl-Titus Hoffmann
- Department for Neuroradiology, University Hospital Leipzig, Leipzig, Germany
| | | | - Cathrin Vörkel
- Clinic for Diagnostic and Interventional Radiology, University Hospital Leipzig, Leipzig, Germany
| | | | - Oliver Ganslandt
- Clinic for Neurosurgery, Katharinenhospital Stuttgart, Stuttgart, Germany
| | - Hansjörg Bäzner
- Clinic for Neurology, Katherinenhospital Stuttgart, Stuttgart, Germany
| | | | - Marcell Kalman
- Clinic for Neuroradiology, Katharinenhospital Stuttgart, Stuttgart, Germany
| | - Elina Henkes
- Clinic for Neuroradiology, Katharinenhospital Stuttgart, Stuttgart, Germany
| | - Hans Henkes
- Clinic for Neuroradiology, Katharinenhospital Stuttgart, Stuttgart, Germany
| | - Stefan Schob
- Department for Neuroradiology, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
36
|
Salama GR, Heier LA, Patel P, Ramakrishna R, Magge R, Tsiouris AJ. Diffusion Weighted/Tensor Imaging, Functional MRI and Perfusion Weighted Imaging in Glioblastoma-Foundations and Future. Front Neurol 2018; 8:660. [PMID: 29403420 PMCID: PMC5786563 DOI: 10.3389/fneur.2017.00660] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/22/2017] [Indexed: 01/20/2023] Open
Abstract
In this article, we review the basics of diffusion tensor imaging and functional MRI, their current utility in preoperative neurosurgical mapping, and their limitations. We also discuss potential future applications, including implementation of resting state functional MRI. We then discuss perfusion and diffusion-weighted imaging and their application in advanced neuro-oncologic practice. We explain how these modalities can be helpful in guiding surgical biopsies and differentiating recurrent tumor from treatment related changes.
Collapse
Affiliation(s)
- Gayle R Salama
- Department of Neuroradiology, Weill Cornell Medical College, New York, NY, United States
| | - Linda A Heier
- Department of Neuroradiology, Weill Cornell Medical College, New York, NY, United States
| | - Praneil Patel
- Department of Neuroradiology, Weill Cornell Medical College, New York, NY, United States
| | - Rohan Ramakrishna
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY, United States
| | - Rajiv Magge
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | | |
Collapse
|
37
|
Dongas J, Asahina AT, Bacchi S, Patel S. Magnetic Resonance Perfusion Imaging in the Diagnosis of High-Grade Glioma Progression and Treatment-Related Changes: A Systematic Review. ACTA ACUST UNITED AC 2018. [DOI: 10.4236/ojmn.2018.83024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
38
|
Treatment-related changes in glioblastoma: a review on the controversies in response assessment criteria and the concepts of true progression, pseudoprogression, pseudoresponse and radionecrosis. Clin Transl Oncol 2017; 20:939-953. [DOI: 10.1007/s12094-017-1816-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 11/27/2017] [Indexed: 10/18/2022]
|
39
|
Zakhari N, Taccone MS, Torres C, Chakraborty S, Sinclair J, Woulfe J, Jansen GH, Nguyen TB. Diagnostic Accuracy of Centrally Restricted Diffusion in the Differentiation of Treatment-Related Necrosis from Tumor Recurrence in High-Grade Gliomas. AJNR Am J Neuroradiol 2017; 39:260-264. [PMID: 29217742 DOI: 10.3174/ajnr.a5485] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/17/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND PURPOSE Centrally restricted diffusion has been demonstrated in recurrent high-grade gliomas treated with bevacizumab. Our purpose was to assess the accuracy of centrally restricted diffusion in the diagnosis of radiation necrosis in high-grade gliomas not treated with bevacizumab. MATERIALS AND METHODS In this prospective study, we enrolled patients with high-grade gliomas who developed a new ring-enhancing necrotic lesion and who underwent re-resection. The presence of a centrally restricted diffusion within the ring-enhancing lesion was assessed visually on diffusion trace images and by ADC measurements on 3T preoperative diffusion tensor examination. The percentage of tumor recurrence and radiation necrosis in each surgical specimen was defined histopathologically. The association between centrally restricted diffusion and radiation necrosis was assessed using the Fisher exact test. Differences in ADC and the ADC ratio between the groups were assessed via the Mann-Whitney U test, and receiver operating characteristic curve analysis was performed. RESULTS Seventeen patients had re-resected ring-enhancing lesions: 8 cases of radiation necrosis and 9 cases of tumor recurrence. There was significant association between centrally restricted diffusion by visual assessment and radiation necrosis (P = .015) with a sensitivity of 75% and a specificity of 88.9%, a positive predictive value 85.7%, and a negative predictive value of 80% for the diagnosis of radiation necrosis. There was a statistically significant difference in the ADC and ADC ratio between radiation necrosis and tumor recurrence (P = .027). CONCLUSIONS The presence of centrally restricted diffusion in a new ring-enhancing lesion might indicate radiation necrosis rather than tumor recurrence in high-grade gliomas previously treated with standard chemoradiation without bevacizumab.
Collapse
Affiliation(s)
- N Zakhari
- From the Department of Radiology (N.Z., C.T., S.C., T.B.N.), Division of Neuroradiology, University of Ottawa, The Ottawa Hospital Civic and General Campus, Ottawa, Ontario, Canada
| | | | - C Torres
- From the Department of Radiology (N.Z., C.T., S.C., T.B.N.), Division of Neuroradiology, University of Ottawa, The Ottawa Hospital Civic and General Campus, Ottawa, Ontario, Canada
| | - S Chakraborty
- From the Department of Radiology (N.Z., C.T., S.C., T.B.N.), Division of Neuroradiology, University of Ottawa, The Ottawa Hospital Civic and General Campus, Ottawa, Ontario, Canada
| | | | - J Woulfe
- Department of Pathology and Laboratory Medicine (J.W., G.H.J.), University of Ottawa, The Ottawa Hospital Civic Campus, Ottawa, Ontario, Canada
| | - G H Jansen
- Department of Pathology and Laboratory Medicine (J.W., G.H.J.), University of Ottawa, The Ottawa Hospital Civic Campus, Ottawa, Ontario, Canada
| | - T B Nguyen
- From the Department of Radiology (N.Z., C.T., S.C., T.B.N.), Division of Neuroradiology, University of Ottawa, The Ottawa Hospital Civic and General Campus, Ottawa, Ontario, Canada
| |
Collapse
|
40
|
Nam JG, Kang KM, Choi SH, Lim WH, Yoo RE, Kim JH, Yun TJ, Sohn CH. Comparison between the Prebolus T1 Measurement and the Fixed T1 Value in Dynamic Contrast-Enhanced MR Imaging for the Differentiation of True Progression from Pseudoprogression in Glioblastoma Treated with Concurrent Radiation Therapy and Temozolomide Chemotherapy. AJNR Am J Neuroradiol 2017; 38:2243-2250. [PMID: 29074633 DOI: 10.3174/ajnr.a5417] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE Glioblastoma is the most common primary brain malignancy and differentiation of true progression from pseudoprogression is clinically important. Our purpose was to compare the diagnostic performance of dynamic contrast-enhanced pharmacokinetic parameters using the fixed T1 and measured T1 on differentiating true from pseudoprogression of glioblastoma after chemoradiation with temozolomide. MATERIALS AND METHODS This retrospective study included 37 patients with histopathologically confirmed glioblastoma with new enhancing lesions after temozolomide chemoradiation defined as true progression (n = 15) or pseudoprogression (n = 22). Dynamic contrast-enhanced pharmacokinetic parameters, including the volume transfer constant, the rate transfer constant, the blood plasma volume per unit volume, and the extravascular extracellular space per unit volume, were calculated by using both the fixed T1 of 1000 ms and measured T1 by using the multiple flip-angle method. Intra- and interobserver reproducibility was assessed by using the intraclass correlation coefficient. Dynamic contrast-enhanced pharmacokinetic parameters were compared between the 2 groups by using univariate and multivariate analysis. The diagnostic performance was evaluated by receiver operating characteristic analysis and leave-one-out cross validation. RESULTS The intraclass correlation coefficients of all the parameters from both T1 values were fair to excellent (0.689-0.999). The volume transfer constant and rate transfer constant from the fixed T1 were significantly higher in patients with true progression (P = .048 and .010, respectively). Multivariate analysis revealed that the rate transfer constant from the fixed T1 was the only independent variable (OR, 1.77 × 105) and showed substantial diagnostic power on receiver operating characteristic analysis (area under the curve, 0.752; P = .002). The sensitivity and specificity on leave-one-out cross validation were 73.3% (11/15) and 59.1% (13/20), respectively. CONCLUSIONS The dynamic contrast-enhanced parameter of rate transfer constant from the fixed T1 acted as a preferable marker to differentiate true progression from pseudoprogression.
Collapse
Affiliation(s)
- J G Nam
- From the Department of Radiology (J.G.N., K.M.K., S.H.C., W.H.L., R.-E.Y., J.-H.K., T.J.Y., C.-H.S.), Seoul National University Hospital, Seoul, Korea
| | - K M Kang
- From the Department of Radiology (J.G.N., K.M.K., S.H.C., W.H.L., R.-E.Y., J.-H.K., T.J.Y., C.-H.S.), Seoul National University Hospital, Seoul, Korea
| | - S H Choi
- From the Department of Radiology (J.G.N., K.M.K., S.H.C., W.H.L., R.-E.Y., J.-H.K., T.J.Y., C.-H.S.), Seoul National University Hospital, Seoul, Korea
- Center for Nanoparticle Research, Institute for Basic Science (S.H.C., C.-H.S.)
- School of Chemical and Biological Engineering (S.H.C.), Seoul National University, Seoul, Korea
| | - W H Lim
- From the Department of Radiology (J.G.N., K.M.K., S.H.C., W.H.L., R.-E.Y., J.-H.K., T.J.Y., C.-H.S.), Seoul National University Hospital, Seoul, Korea
| | - R-E Yoo
- From the Department of Radiology (J.G.N., K.M.K., S.H.C., W.H.L., R.-E.Y., J.-H.K., T.J.Y., C.-H.S.), Seoul National University Hospital, Seoul, Korea
| | - J-H Kim
- From the Department of Radiology (J.G.N., K.M.K., S.H.C., W.H.L., R.-E.Y., J.-H.K., T.J.Y., C.-H.S.), Seoul National University Hospital, Seoul, Korea
| | - T J Yun
- From the Department of Radiology (J.G.N., K.M.K., S.H.C., W.H.L., R.-E.Y., J.-H.K., T.J.Y., C.-H.S.), Seoul National University Hospital, Seoul, Korea
| | - C-H Sohn
- From the Department of Radiology (J.G.N., K.M.K., S.H.C., W.H.L., R.-E.Y., J.-H.K., T.J.Y., C.-H.S.), Seoul National University Hospital, Seoul, Korea
- Center for Nanoparticle Research, Institute for Basic Science (S.H.C., C.-H.S.)
| |
Collapse
|
41
|
Meeus EM, Zarinabad N, Manias KA, Novak J, Rose HEL, Dehghani H, Foster K, Morland B, Peet AC. Diffusion-weighted MRI and intravoxel incoherent motion model for diagnosis of pediatric solid abdominal tumors. J Magn Reson Imaging 2017; 47:1475-1486. [PMID: 29159937 PMCID: PMC6001424 DOI: 10.1002/jmri.25901] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/06/2017] [Indexed: 12/24/2022] Open
Abstract
Background Pediatric retroperitoneal tumors in the renal bed are often large and heterogeneous, and their diagnosis based on conventional imaging alone is not possible. More advanced imaging methods, such as diffusion‐weighted (DW) MRI and the use of intravoxel incoherent motion (IVIM), have the potential to provide additional biomarkers that could facilitate their noninvasive diagnosis. Purpose To assess the use of an IVIM model for diagnosis of childhood malignant abdominal tumors and discrimination of benign from malignant lesions. Study Type Retrospective. Population Forty‐two pediatric patients with abdominal lesions (n = 32 malignant, n = 10 benign), verified by histopathology. Field Strength/Sequence 1.5T MRI system and a DW‐MRI sequence with six b‐values (0, 50, 100, 150, 600, 1000 s/mm2). Assessment Parameter maps of apparent diffusion coefficient (ADC), and IVIM maps of slow diffusion coefficient (D), fast diffusion coefficient (D*), and perfusion fraction (f) were computed using a segmented fitting model. Histograms were constructed for whole‐tumor regions of each parameter. Statistical Tests Comparison of histogram parameters of and their diagnostic performance was determined using Kruskal–Wallis, Mann–Whitney U, and receiver‐operating characteristic (ROC) analysis. Results IVIM parameters D* and f were significantly higher in neuroblastoma compared to Wilms' tumors (P < 0.05). The ROC analysis showed that the best diagnostic performance was achieved with D* 90th percentile (area under the curve [AUC] = 0.935; P = 0.002; cutoff value = 32,376 × 10−6 mm2/s) and f mean values (AUC = 1.00; P < 0.001; cutoff value = 14.7) in discriminating between neuroblastoma (n = 11) and Wilms' tumors (n = 8). Discrimination between tumor types was not possible with IVIM D or ADC parameters. Malignant tumors revealed significantly lower ADC, D, and higher D* values than in benign lesions (all P < 0.05). Data Conclusion IVIM perfusion parameters could distinguish between malignant childhood tumor types, providing potential imaging biomarkers for their diagnosis. Level of Evidence: 4 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2018;47:1475–1486.
Collapse
Affiliation(s)
- Emma M Meeus
- Physical Sciences of Imaging in Biomedical Sciences (PSIBS) Doctoral Training Centre, University of Birmingham, UK.,Institute of Cancer and Genomic Sciences, University of Birmingham, UK.,Department of Oncology, Birmingham Children's Hospital, Birmingham, UK
| | - Niloufar Zarinabad
- Institute of Cancer and Genomic Sciences, University of Birmingham, UK.,Department of Oncology, Birmingham Children's Hospital, Birmingham, UK
| | - Karen A Manias
- Institute of Cancer and Genomic Sciences, University of Birmingham, UK.,Department of Oncology, Birmingham Children's Hospital, Birmingham, UK
| | - Jan Novak
- Institute of Cancer and Genomic Sciences, University of Birmingham, UK.,Department of Oncology, Birmingham Children's Hospital, Birmingham, UK
| | - Heather E L Rose
- Institute of Cancer and Genomic Sciences, University of Birmingham, UK.,Department of Oncology, Birmingham Children's Hospital, Birmingham, UK
| | - Hamid Dehghani
- Physical Sciences of Imaging in Biomedical Sciences (PSIBS) Doctoral Training Centre, University of Birmingham, UK.,School of Computer Science, University of Birmingham, UK
| | - Katharine Foster
- Department of Radiology, Birmingham Children's Hospital, Birmingham, UK
| | - Bruce Morland
- Department of Oncology, Birmingham Children's Hospital, Birmingham, UK
| | - Andrew C Peet
- Institute of Cancer and Genomic Sciences, University of Birmingham, UK.,Department of Oncology, Birmingham Children's Hospital, Birmingham, UK
| |
Collapse
|
42
|
Meng J, Zhu L, Zhu L, Xie L, Wang H, Liu S, Yan J, Liu B, Guan Y, He J, Ge Y, Zhou Z, Yang X. Whole-lesion ADC histogram and texture analysis in predicting recurrence of cervical cancer treated with CCRT. Oncotarget 2017; 8:92442-92453. [PMID: 29190929 PMCID: PMC5696195 DOI: 10.18632/oncotarget.21374] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/28/2017] [Indexed: 01/30/2023] Open
Abstract
PURPOSE To explore the value of whole-lesion apparent diffusion coefficient (ADC) histogram and texture analysis in predicting tumor recurrence of advanced cervical cancer treated with concurrent chemo-radiotherapy (CCRT). METHODS 36 women with pathologically confirmed advanced cervical squamous carcinomas were enrolled in this prospective study. 3.0 T pelvic MR examinations including diffusion weighted imaging (b = 0, 800 s/mm2) were performed before CCRT (pre-CCRT) and at the end of 2nd week of CCRT (mid-CCRT). ADC histogram and texture features were derived from the whole volume of cervical cancers. RESULTS With a mean follow-up of 25 months (range, 11 ∼ 43), 10/36 (27.8%) patients ended with recurrence. Pre-CCRT 75th, 90th, correlation, autocorrelation and mid-CCRT ADCmean, 10th, 25th, 50th, 75th, 90th, autocorrelation can effectively differentiate the recurrence from nonrecurrence group with area under the curve ranging from 0.742 to 0.850 (P values range, 0.001 ∼ 0.038). CONCLUSIONS Pre- and mid-treatment whole-lesion ADC histogram and texture analysis hold great potential in predicting tumor recurrence of advanced cervical cancer treated with CCRT.
Collapse
Affiliation(s)
- Jie Meng
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Lijing Zhu
- The Comprehensive Cancer Centre of Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Li Zhu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Li Xie
- The Comprehensive Cancer Centre of Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Huanhuan Wang
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Song Liu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jing Yan
- The Comprehensive Cancer Centre of Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yue Guan
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - Jian He
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yun Ge
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - Zhengyang Zhou
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xiaofeng Yang
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, USA
| |
Collapse
|
43
|
Park JE, Han K, Sung YS, Chung MS, Koo HJ, Yoon HM, Choi YJ, Lee SS, Kim KW, Shin Y, An S, Cho HM, Park SH. Selection and Reporting of Statistical Methods to Assess Reliability of a Diagnostic Test: Conformity to Recommended Methods in a Peer-Reviewed Journal. Korean J Radiol 2017; 18:888-897. [PMID: 29089821 PMCID: PMC5639154 DOI: 10.3348/kjr.2017.18.6.888] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022] Open
Abstract
Objective To evaluate the frequency and adequacy of statistical analyses in a general radiology journal when reporting a reliability analysis for a diagnostic test. Materials and Methods Sixty-three studies of diagnostic test accuracy (DTA) and 36 studies reporting reliability analyses published in the Korean Journal of Radiology between 2012 and 2016 were analyzed. Studies were judged using the methodological guidelines of the Radiological Society of North America-Quantitative Imaging Biomarkers Alliance (RSNA-QIBA), and COnsensus-based Standards for the selection of health Measurement INstruments (COSMIN) initiative. DTA studies were evaluated by nine editorial board members of the journal. Reliability studies were evaluated by study reviewers experienced with reliability analysis. Results Thirty-one (49.2%) of the 63 DTA studies did not include a reliability analysis when deemed necessary. Among the 36 reliability studies, proper statistical methods were used in all (5/5) studies dealing with dichotomous/nominal data, 46.7% (7/15) of studies dealing with ordinal data, and 95.2% (20/21) of studies dealing with continuous data. Statistical methods were described in sufficient detail regarding weighted kappa in 28.6% (2/7) of studies and regarding the model and assumptions of intraclass correlation coefficient in 35.3% (6/17) and 29.4% (5/17) of studies, respectively. Reliability parameters were used as if they were agreement parameters in 23.1% (3/13) of studies. Reproducibility and repeatability were used incorrectly in 20% (3/15) of studies. Conclusion Greater attention to the importance of reporting reliability, thorough description of the related statistical methods, efforts not to neglect agreement parameters, and better use of relevant terminology is necessary.
Collapse
Affiliation(s)
- Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Kyunghwa Han
- Department of Radiology, Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Yu Sub Sung
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Mi Sun Chung
- Department of Radiology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 06973, Korea
| | - Hyun Jung Koo
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Hee Mang Yoon
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Young Jun Choi
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Seung Soo Lee
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Kyung Won Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Youngbin Shin
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Suah An
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Hyo-Min Cho
- Korea Research Institute of Standards and Science, Daejeon 34113, Korea
| | - Seong Ho Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
44
|
Villanueva-Meyer JE, Mabray MC, Cha S. Current Clinical Brain Tumor Imaging. Neurosurgery 2017; 81:397-415. [PMID: 28486641 PMCID: PMC5581219 DOI: 10.1093/neuros/nyx103] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 02/23/2017] [Indexed: 01/12/2023] Open
Abstract
Neuroimaging plays an ever evolving role in the diagnosis, treatment planning, and post-therapy assessment of brain tumors. This review provides an overview of current magnetic resonance imaging (MRI) methods routinely employed in the care of the brain tumor patient. Specifically, we focus on advanced techniques including diffusion, perfusion, spectroscopy, tractography, and functional MRI as they pertain to noninvasive characterization of brain tumors and pretreatment evaluation. The utility of both structural and physiological MRI in the post-therapeutic brain evaluation is also reviewed with special attention to the challenges presented by pseudoprogression and pseudoresponse.
Collapse
Affiliation(s)
- Javier E. Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, Neuroradiology Section, University of California San Francisco, San Francisco, California
| | - Marc C. Mabray
- Department of Radiology and Biomedical Imaging, Neuroradiology Section, University of California San Francisco, San Francisco, California
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, Neuroradiology Section, University of California San Francisco, San Francisco, California
| |
Collapse
|
45
|
Incidence of Tumour Progression and Pseudoprogression in High-Grade Gliomas: a Systematic Review and Meta-Analysis. Clin Neuroradiol 2017; 28:401-411. [PMID: 28466127 PMCID: PMC6105173 DOI: 10.1007/s00062-017-0584-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 04/04/2017] [Indexed: 12/29/2022]
Abstract
Background High-grade gliomas are the most common primary brain tumours. Pseudoprogression describes the false appearance of radiation-induced progression on MRI. A distinction should be made from true tumour progression to correctly plan treatment. However, there is wide variation of reported pseudoprogression. We thus aimed to establish the incidence of pseudoprogression and tumour progression in high-grade glioma patients with a systematic review and meta-analysis. Methods We searched PubMed, Embase and Web of Science on the incidence of pseudoprogression and tumour progression in adult high-grade glioma patients from 2005, the latest on 8 October 2014. Histology or imaging follow-up was used as reference standard. Extracted data included number of patients with worsening of imaging findings on T1 postcontrast or T2/FLAIR, pseudoprogression and tumour progression. Study quality was assessed. Heterogeneity was tested with I2. Pooling of the results was done with random models using Metaprop in STATA (StataCorp. Stata Statistical Software. College Station, TX: StataCorp LP). Results We identified 73 studies. MRI progression occurred in 2603 patients. Of these, 36% (95% confidence interval [CI] 33–40%) demonstrated pseudoprogression, 60% (95%CI 56–64%) tumour progression and unknown outcome was present in the remaining 4% of the patients (range 1–37%). Conclusion This meta-analysis demonstrated for the first time a notably high pooled incidence of pseudoprogression in patients with a form of progression across the available literature. This highlighted the full extent of the problem of the currently conventional MRI-based Response Assessment in Neuro-Oncology (RANO) criteria for treatment evaluation in high-grade gliomas. This underscores the need for more accurate treatment evaluation using advanced imaging to improve diagnostic accuracy and therapeutic approach. Electronic supplementary material The online version of this article (doi: 10.1007/s00062-017-0584-x) contains supplementary material, which is available to authorized users. It contains the characteristics of the included studies (supplementary table 1) and a full search strategy (see supplementary search strategy).
Collapse
|
46
|
MRI in Glioma Immunotherapy: Evidence, Pitfalls, and Perspectives. J Immunol Res 2017; 2017:5813951. [PMID: 28512646 PMCID: PMC5415864 DOI: 10.1155/2017/5813951] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/06/2017] [Accepted: 03/02/2017] [Indexed: 01/14/2023] Open
Abstract
Pseudophenomena, that is, imaging alterations due to therapy rather than tumor evolution, have an important impact on the management of glioma patients and the results of clinical trials. RANO (response assessment in neurooncology) criteria, including conventional MRI (cMRI), addressed the issues of pseudoprogression after radiotherapy and concomitant chemotherapy and pseudoresponse during antiangiogenic therapy of glioblastomas (GBM) and other gliomas. The development of cancer immunotherapy forced the identification of further relevant response criteria, summarized by the iRANO working group in 2015. In spite of this, the unequivocal definition of glioma progression by cMRI remains difficult particularly in the setting of immunotherapy approaches provided by checkpoint inhibitors and dendritic cells. Advanced MRI (aMRI) may in principle address this unmet clinical need. Here, we discuss the potential contribution of different aMRI techniques and their indications and pitfalls in relation to biological and imaging features of glioma and immune system interactions.
Collapse
|
47
|
Abstract
OPINION STATEMENT With advances in treatments and survival of patients with glioblastoma (GBM), it has become apparent that conventional imaging sequences have significant limitations both in terms of assessing response to treatment and monitoring disease progression. Both 'pseudoprogression' after chemoradiation for newly diagnosed GBM and 'pseudoresponse' after anti-angiogenesis treatment for relapsed GBM are well-recognised radiological entities. This in turn has led to revision of response criteria away from the standard MacDonald criteria, which depend on the two-dimensional measurement of contrast-enhancing tumour, and which have been the primary measure of radiological response for over three decades. A working party of experts published RANO (Response Assessment in Neuro-oncology Working Group) criteria in 2010 which take into account signal change on T2/FLAIR sequences as well as the contrast-enhancing component of the tumour. These have recently been modified for immune therapies, which are associated with specific issues related to the timing of radiological response. There has been increasing interest in quantification and validation of physiological and metabolic parameters in GBM over the last 10 years utilising the wide range of advanced imaging techniques available on standard MRI platforms. Previously, MRI would provide structural information only on the anatomical location of the tumour and the presence or absence of a disrupted blood-brain barrier. Advanced MRI sequences include proton magnetic resonance spectroscopy (MRS), vascular imaging (perfusion/permeability) and diffusion imaging (diffusion weighted imaging/diffusion tensor imaging) and are now routinely available. They provide biologically relevant functional, haemodynamic, cellular, metabolic and cytoarchitectural information and are being evaluated in clinical trials to determine whether they offer superior biomarkers of early treatment response than conventional imaging, when correlated with hard survival endpoints. Multiparametric imaging, incorporating different combinations of these modalities, improves accuracy over single imaging modalities but has not been widely adopted due to the amount of post-processing analysis required, lack of clinical trial data, lack of radiology training and wide variations in threshold values. New techniques including diffusion kurtosis and radiomics will offer a higher level of quantification but will require validation in clinical trial settings. Given all these considerations, it is clear that there is an urgent need to incorporate advanced techniques into clinical trial design to avoid the problems of under or over assessment of treatment response.
Collapse
|
48
|
van Dijken BRJ, van Laar PJ, Holtman GA, van der Hoorn A. Diagnostic accuracy of magnetic resonance imaging techniques for treatment response evaluation in patients with high-grade glioma, a systematic review and meta-analysis. Eur Radiol 2017; 27:4129-4144. [PMID: 28332014 PMCID: PMC5579204 DOI: 10.1007/s00330-017-4789-9] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/01/2017] [Accepted: 02/23/2017] [Indexed: 01/04/2023]
Abstract
Objective Treatment response assessment in high-grade gliomas uses contrast enhanced T1-weighted MRI, but is unreliable. Novel advanced MRI techniques have been studied, but the accuracy is not well known. Therefore, we performed a systematic meta-analysis to assess the diagnostic accuracy of anatomical and advanced MRI for treatment response in high-grade gliomas. Methods Databases were searched systematically. Study selection and data extraction were done by two authors independently. Meta-analysis was performed using a bivariate random effects model when ≥5 studies were included. Results Anatomical MRI (five studies, 166 patients) showed a pooled sensitivity and specificity of 68% (95%CI 51–81) and 77% (45–93), respectively. Pooled apparent diffusion coefficients (seven studies, 204 patients) demonstrated a sensitivity of 71% (60–80) and specificity of 87% (77–93). DSC-perfusion (18 studies, 708 patients) sensitivity was 87% (82–91) with a specificity of 86% (77–91). DCE-perfusion (five studies, 207 patients) sensitivity was 92% (73–98) and specificity was 85% (76–92). The sensitivity of spectroscopy (nine studies, 203 patients) was 91% (79–97) and specificity was 95% (65–99). Conclusion Advanced techniques showed higher diagnostic accuracy than anatomical MRI, the highest for spectroscopy, supporting the use in treatment response assessment in high-grade gliomas. Key points • Treatment response assessment in high-grade gliomas with anatomical MRI is unreliable • Novel advanced MRI techniques have been studied, but diagnostic accuracy is unknown • Meta-analysis demonstrates that advanced MRI showed higher diagnostic accuracy than anatomical MRI • Highest diagnostic accuracy for spectroscopy and perfusion MRI • Supports the incorporation of advanced MRI in high-grade glioma treatment response assessment Electronic supplementary material The online version of this article (doi:10.1007/s00330-017-4789-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bart R J van Dijken
- University Medical Center Groningen Department of Radiology, University of Groningen, Hanzeplein 1, P. O. Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Peter Jan van Laar
- University Medical Center Groningen Department of Radiology, University of Groningen, Hanzeplein 1, P. O. Box 30.001, 9700 RB, Groningen, The Netherlands
- University Medical Center Groningen, Center for Medical Imaging-North East Netherlands, University of Groningen, Groningen, The Netherlands
| | - Gea A Holtman
- University Medical Center Groningen, Department of General Practice, University of Groningen, Groningen, The Netherlands
| | - Anouk van der Hoorn
- University Medical Center Groningen Department of Radiology, University of Groningen, Hanzeplein 1, P. O. Box 30.001, 9700 RB, Groningen, The Netherlands.
- University Medical Center Groningen, Center for Medical Imaging-North East Netherlands, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
49
|
Wagner F, Hakami YA, Warnock G, Fischer G, Huellner MW, Veit-Haibach P. Comparison of Contrast-Enhanced CT and [18F]FDG PET/CT Analysis Using Kurtosis and Skewness in Patients with Primary Colorectal Cancer. Mol Imaging Biol 2017; 19:795-803. [DOI: 10.1007/s11307-017-1066-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
50
|
Carceller F, Jerome NP, Miyazaki K, Collins DJ, Orton MR, d'Arcy JA, Wallace T, Moreno L, Pearson ADJ, Zacharoulis S, Leach MO, Marshall LV, Koh DM. Feasibility and applicability of diffusion-weighted and dynamic contrast-enhanced magnetic resonance imaging in routine assessments of children with high-grade gliomas. Pediatr Blood Cancer 2017; 64:279-283. [PMID: 27615273 DOI: 10.1002/pbc.26216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/26/2016] [Accepted: 07/26/2016] [Indexed: 12/28/2022]
Abstract
Diffusion-weighted (DW) and dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) have been used as imaging biomarkers in adults with high-grade gliomas (HGGs). We incorporated free-breathing DW-MRI and DCE-MRI, at a single time point, in the routine follow-up of five children (median age 9 years, range 8-15) with histologically confirmed HGG within a prospective imaging study. It was feasible to incorporate DW-MRI and DCE-MRI in routine assessments of children with HGG. DW and DCE parameters were repeatable in paediatric HGG. Higher median ADC100-1000 significantly correlated with longer survival in our sample.
Collapse
Affiliation(s)
- Fernando Carceller
- Paediatric & Adolescent Drug Development Team, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Neil P Jerome
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - Keiko Miyazaki
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - David J Collins
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - Matthew R Orton
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - James A d'Arcy
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - Toni Wallace
- Radiology Department, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Lucas Moreno
- Paediatric & Adolescent Drug Development Team, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
- Clinical Trials Unit, Paediatric Oncology Department, Hospital Niño Jesús, Madrid, Spain
| | - Andrew D J Pearson
- Paediatric & Adolescent Drug Development Team, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Stergios Zacharoulis
- Paediatric & Adolescent Drug Development Team, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Martin O Leach
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - Lynley V Marshall
- Paediatric & Adolescent Drug Development Team, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Dow-Mu Koh
- Radiology Department, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|