1
|
Li Y, Li C, Wei Y, Price S, Schönlieb CB, Chen X. Multi-objective Bayesian optimization with enhanced features for adaptively improved glioblastoma partitioning and survival prediction. Comput Med Imaging Graph 2024; 116:102420. [PMID: 39079409 DOI: 10.1016/j.compmedimag.2024.102420] [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: 12/01/2023] [Revised: 03/30/2024] [Accepted: 07/17/2024] [Indexed: 09/02/2024]
Abstract
Glioblastoma, an aggressive brain tumor prevalent in adults, exhibits heterogeneity in its microstructures and vascular patterns. The delineation of its subregions could facilitate the development of region-targeted therapies. However, current unsupervised learning techniques for this task face challenges in reliability due to fluctuations of clustering algorithms, particularly when processing data from diverse patient cohorts. Furthermore, stable clustering results do not guarantee clinical meaningfulness. To establish the clinical relevance of these subregions, we will perform survival predictions using radiomic features extracted from them. Following this, achieving a balance between outcome stability and clinical relevance presents a significant challenge, further exacerbated by the extensive time required for hyper-parameter tuning. In this study, we introduce a multi-objective Bayesian optimization (MOBO) framework, which leverages a Feature-enhanced Auto-Encoder (FAE) and customized losses to assess both the reproducibility of clustering algorithms and the clinical relevance of their outcomes. Specifically, we embed the entirety of these processes within the MOBO framework, modeling both using distinct Gaussian Processes (GPs). The proposed MOBO framework can automatically balance the trade-off between the two criteria by employing bespoke stability and clinical significance losses. Our approach efficiently optimizes all hyper-parameters, including the FAE architecture and clustering parameters, within a few steps. This not only accelerates the process but also consistently yields robust MRI subregion delineations and provides survival predictions with strong statistical validation.
Collapse
Affiliation(s)
- Yifan Li
- Department of Computer Science, University of Bath, Bath, UK.
| | - Chao Li
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| | - Yiran Wei
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| | - Stephen Price
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| | - Carola-Bibiane Schönlieb
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, UK.
| | - Xi Chen
- Department of Computer Science, University of Bath, Bath, UK.
| |
Collapse
|
2
|
Aznarez-Sanado M, Romero-Garcia R, Li C, Morris RC, Price SJ, Manly T, Santarius T, Erez Y, Hart MG, Suckling J. Brain tumour microstructure is associated with post-surgical cognition. Sci Rep 2024; 14:5646. [PMID: 38454017 PMCID: PMC10920778 DOI: 10.1038/s41598-024-55130-5] [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: 06/14/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Abstract
Brain tumour microstructure is potentially predictive of changes following treatment to cognitive functions subserved by the functional networks in which they are embedded. To test this hypothesis, intra-tumoural microstructure was quantified from diffusion-weighted MRI to identify which tumour subregions (if any) had a greater impact on participants' cognitive recovery after surgical resection. Additionally, we studied the role of tumour microstructure in the functional interaction between the tumour and the rest of the brain. Sixteen patients (22-56 years, 7 females) with brain tumours located in or near speech-eloquent areas of the brain were included in the analyses. Two different approaches were adopted for tumour segmentation from a multishell diffusion MRI acquisition: the first used a two-dimensional four group partition of feature space, whilst the second used data-driven clustering with Gaussian mixture modelling. For each approach, we assessed the capability of tumour microstructure to predict participants' cognitive outcomes after surgery and the strength of association between the BOLD signal of individual tumour subregions and the global BOLD signal. With both methodologies, the volumes of partially overlapped subregions within the tumour significantly predicted cognitive decline in verbal skills after surgery. We also found that these particular subregions were among those that showed greater functional interaction with the unaffected cortex. Our results indicate that tumour microstructure measured by MRI multishell diffusion is associated with cognitive recovery after surgery.
Collapse
Affiliation(s)
- Maite Aznarez-Sanado
- School of Education and Psychology, University of Navarra, 31009, Pamplona, Spain
| | - Rafael Romero-Garcia
- Department of Medical Physiology and Biophysics, Instituto de Biomedicina de Sevilla (IBiS), HUVR/CSIC/Universidad de Sevilla/CIBERSAM, ISCIII, 41013, Sevilla, Spain.
- Department of Psychiatry, University of Cambridge, Herchel Smith Bldg, Robinson Way, Cambridge, CB2 0SZ, UK.
| | - Chao Li
- Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
- Department of Applied Mathematics and Theoretical Physics, The Centre for Mathematical Imaging in Healthcare, Cambridge, CB3 0WA, UK
- School of Medicine & School of Science and Engineering, University of Dundee, Dundee, DD1 4HN, UK
| | - Rob C Morris
- Academic Neurosurgery Division, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Stephen J Price
- Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Thomas Manly
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, CB2 7EF, UK
| | - Thomas Santarius
- Academic Neurosurgery Division, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Yaara Erez
- Faculty of Engineering, Bar-Ilan University, 5290002, Ramat Gan, Israel
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Michael G Hart
- St George's, University of London and St George's University Hospitals NHS Foundation Trust, Institute of Molecular and Clinical Sciences, Neurosciences Research Centre, Cranmer Terrace, London, SW17 0RE, UK
| | - John Suckling
- Department of Psychiatry, University of Cambridge, Herchel Smith Bldg, Robinson Way, Cambridge, CB2 0SZ, UK
- Cambridge and Peterborough NHS Foundation Trust, Cambridge, CB21 5EF, UK
| |
Collapse
|
3
|
Wang Z, Guan F, Duan W, Guo Y, Pei D, Qiu Y, Wang M, Xing A, Liu Z, Yu B, Zheng H, Liu X, Yan D, Ji Y, Cheng J, Yan J, Zhang Z. Diffusion tensor imaging-based machine learning for IDH wild-type glioblastoma stratification to reveal the biological underpinning of radiomic features. CNS Neurosci Ther 2023; 29:3339-3350. [PMID: 37222229 PMCID: PMC10580329 DOI: 10.1111/cns.14263] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/09/2023] [Accepted: 05/03/2023] [Indexed: 05/25/2023] Open
Abstract
INTRODUCTION This study addresses the lack of systematic investigation into the prognostic value of hand-crafted radiomic features derived from diffusion tensor imaging (DTI) in isocitrate dehydrogenase (IDH) wild-type glioblastoma (GBM), as well as the limited understanding of the biological interpretation of individual DTI radiomic features and metrics. AIMS To develop and validate a DTI-based radiomic model for predicting prognosis in patients with IDH wild-type GBM and reveal the biological underpinning of individual DTI radiomic features and metrics. RESULTS The DTI-based radiomic signature was an independent prognostic factor (p < 0.001). Incorporating the radiomic signature into a clinical model resulted in a radiomic-clinical nomogram that predicted survival better than either the radiomic model or clinical model alone, with a better calibration and classification accuracy. Four categories of pathways (synapse, proliferation, DNA damage response, and complex cellular functions) were significantly correlated with the DTI-based radiomic features and DTI metrics. CONCLUSION The prognostic radiomic features derived from DTI are driven by distinct pathways involved in synapse, proliferation, DNA damage response, and complex cellular functions of GBM.
Collapse
Affiliation(s)
- Zilong Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Fangzhan Guan
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Wenchao Duan
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yu Guo
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Dongling Pei
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuning Qiu
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Minkai Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Aoqi Xing
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhongyi Liu
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Bin Yu
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Hongwei Zheng
- Department of MRIThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xianzhi Liu
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Dongming Yan
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuchen Ji
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jingliang Cheng
- Department of MRIThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jing Yan
- Department of MRIThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhenyu Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
4
|
Wei Y, Chen X, Zhu L, Zhang L, Schonlieb CB, Price S, Li C. Multi-Modal Learning for Predicting the Genotype of Glioma. IEEE TRANSACTIONS ON MEDICAL IMAGING 2023; 42:3167-3178. [PMID: 37022918 DOI: 10.1109/tmi.2023.3244038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The isocitrate dehydrogenase (IDH) gene mutation is an essential biomarker for the diagnosis and prognosis of glioma. It is promising to better predict glioma genotype by integrating focal tumor image and geometric features with brain network features derived from MRI. Convolutional neural networks show reasonable performance in predicting IDH mutation, which, however, cannot learn from non-Euclidean data, e.g., geometric and network data. In this study, we propose a multi-modal learning framework using three separate encoders to extract features of focal tumor image, tumor geometrics and global brain networks. To mitigate the limited availability of diffusion MRI, we develop a self-supervised approach to generate brain networks from anatomical multi-sequence MRI. Moreover, to extract tumor-related features from the brain network, we design a hierarchical attention module for the brain network encoder. Further, we design a bi-level multi-modal contrastive loss to align the multi-modal features and tackle the domain gap at the focal tumor and global brain. Finally, we propose a weighted population graph to integrate the multi-modal features for genotype prediction. Experimental results on the testing set show that the proposed model outperforms the baseline deep learning models. The ablation experiments validate the performance of different components of the framework. The visualized interpretation corresponds to clinical knowledge with further validation. In conclusion, the proposed learning framework provides a novel approach for predicting the genotype of glioma.
Collapse
|
5
|
Salvalaggio A, Pini L, Gaiola M, Velco A, Sansone G, Anglani M, Fekonja L, Chioffi F, Picht T, Thiebaut de Schotten M, Zagonel V, Lombardi G, D’Avella D, Corbetta M. White Matter Tract Density Index Prediction Model of Overall Survival in Glioblastoma. JAMA Neurol 2023; 80:1222-1231. [PMID: 37747720 PMCID: PMC10520843 DOI: 10.1001/jamaneurol.2023.3284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/07/2023] [Indexed: 09/26/2023]
Abstract
Importance The prognosis of overall survival (OS) in patients with glioblastoma (GBM) may depend on the underlying structural connectivity of the brain. Objective To examine the association between white matter tracts affected by GBM and patients' OS by means of a new tract density index (TDI). Design, Setting, and Participants This prognostic study in patients with a histopathologic diagnosis of GBM examined a discovery cohort of 112 patients who underwent surgery between February 1, 2015, and November 30, 2020 (follow-up to May 31, 2023), in Italy and 70 patients in a replicative cohort (n = 70) who underwent surgery between September 1, 2012, and November 30, 2015 (follow-up to May 31, 2023), in Germany. Statistical analyses were performed from June 1, 2021, to May 31, 2023. Thirteen and 12 patients were excluded from the discovery and the replicative sets, respectively, because of magnetic resonance imaging artifacts. Exposure The density of white matter tracts encompassing GBM. Main Outcomes and Measures Correlation, linear regression, Cox proportional hazards regression, Kaplan-Meier, and prediction analysis were used to assess the association between the TDI and OS. Results were compared with common prognostic factors of GBM, including age, performance status, O6-methylguanine-DNA methyltransferase methylation, and extent of surgery. Results In the discovery cohort (n = 99; mean [SD] age, 62.2 [11.5] years; 29 female [29.3%]; 70 male [70.7%]), the TDI was significantly correlated with OS (r = -0.34; P < .001). This association was more stable compared with other prognostic factors. The TDI showed a significant regression pattern (Cox: hazard ratio, 0.28 [95% CI, 0.02-0.55; P = .04]; linear: t = -2.366; P = .02). and a significant Kaplan-Meier stratification of patients as having lower or higher OS based on the TDI (log-rank test = 4.52; P = .03). Results were confirmed in the replicative cohort (n = 58; mean [SD] age, 58.5 [11.1] years, 14 female [24.1%]; 44 male [75.9%]). High (24-month cutoff) and low (18-month cutoff) OS was predicted based on the TDI computed in the discovery cohort (accuracy = 87%). Conclusions and Relevance In this study, GBMs encompassing regions with low white matter tract density were associated with longer OS. These findings indicate that the TDI is a reliable presurgical outcome predictor that may be considered in clinical trials and clinical practice. These findings support a framework in which the outcome of GBM depends on the patient's brain organization.
Collapse
Affiliation(s)
- Alessandro Salvalaggio
- Clinica Neurologica, Department of Neuroscience, University of Padova, Padova, Italy
- Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Lorenzo Pini
- Clinica Neurologica, Department of Neuroscience, University of Padova, Padova, Italy
- Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Matteo Gaiola
- Clinica Neurologica, Department of Neuroscience, University of Padova, Padova, Italy
| | - Aron Velco
- Clinica Neurologica, Department of Neuroscience, University of Padova, Padova, Italy
| | - Giulio Sansone
- Clinica Neurologica, Department of Neuroscience, University of Padova, Padova, Italy
| | | | - Lucius Fekonja
- Department of Neurosurgery, Charité Universitätsmedizin Berlin, Berlin, Germany
- Cluster of Excellence “Matters of Activity. Image Space Material,” Humboldt University, Berlin, Germany
| | - Franco Chioffi
- Division of Neurosurgery, Azienda Ospedaliera Università di Padova, Padova, Italy
| | - Thomas Picht
- Department of Neurosurgery, Charité Universitätsmedizin Berlin, Berlin, Germany
- Cluster of Excellence “Matters of Activity. Image Space Material,” Humboldt University, Berlin, Germany
| | - Michel Thiebaut de Schotten
- Brain Connectivity and Behaviour Laboratory, Sorbonne Universities, Paris, France
- Groupe d’Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives-UMR 5293, CNRS, CEA University of Bordeaux, Bordeaux, France
| | - Vittorina Zagonel
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Domenico D’Avella
- Academic Neurosurgery, Department of Neurosciences, University of Padova, Padova, Italy
| | - Maurizio Corbetta
- Clinica Neurologica, Department of Neuroscience, University of Padova, Padova, Italy
- Padova Neuroscience Center, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Fondazione Biomedica, Padova, Italy
| |
Collapse
|
6
|
Wang K, Li Z, Wang H, Liu S, Pan M, Wang M, Wang S, Song Z. Improving brain tumor segmentation with anatomical prior-informed pre-training. Front Med (Lausanne) 2023; 10:1211800. [PMID: 37771979 PMCID: PMC10525322 DOI: 10.3389/fmed.2023.1211800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/21/2023] [Indexed: 09/30/2023] Open
Abstract
Introduction Precise delineation of glioblastoma in multi-parameter magnetic resonance images is pivotal for neurosurgery and subsequent treatment monitoring. Transformer models have shown promise in brain tumor segmentation, but their efficacy heavily depends on a substantial amount of annotated data. To address the scarcity of annotated data and improve model robustness, self-supervised learning methods using masked autoencoders have been devised. Nevertheless, these methods have not incorporated the anatomical priors of brain structures. Methods This study proposed an anatomical prior-informed masking strategy to enhance the pre-training of masked autoencoders, which combines data-driven reconstruction with anatomical knowledge. We investigate the likelihood of tumor presence in various brain structures, and this information is then utilized to guide the masking procedure. Results Compared with random masking, our method enables the pre-training to concentrate on regions that are more pertinent to downstream segmentation. Experiments conducted on the BraTS21 dataset demonstrate that our proposed method surpasses the performance of state-of-the-art self-supervised learning techniques. It enhances brain tumor segmentation in terms of both accuracy and data efficiency. Discussion Tailored mechanisms designed to extract valuable information from extensive data could enhance computational efficiency and performance, resulting in increased precision. It's still promising to integrate anatomical priors and vision approaches.
Collapse
Affiliation(s)
- Kang Wang
- Digital Medical Research Center, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Key Lab of Medical Image Computing and Computer Assisted Intervention, Fudan University, Shanghai, China
| | - Zeyang Li
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haoran Wang
- Digital Medical Research Center, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Key Lab of Medical Image Computing and Computer Assisted Intervention, Fudan University, Shanghai, China
| | - Siyu Liu
- Digital Medical Research Center, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Key Lab of Medical Image Computing and Computer Assisted Intervention, Fudan University, Shanghai, China
| | - Mingyuan Pan
- Radiation Oncology Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Manning Wang
- Digital Medical Research Center, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Key Lab of Medical Image Computing and Computer Assisted Intervention, Fudan University, Shanghai, China
| | - Shuo Wang
- Digital Medical Research Center, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Key Lab of Medical Image Computing and Computer Assisted Intervention, Fudan University, Shanghai, China
| | - Zhijian Song
- Digital Medical Research Center, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Key Lab of Medical Image Computing and Computer Assisted Intervention, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Collet S, Guillamo JS, Berro DH, Chakhoyan A, Constans JM, Lechapt-Zalcman E, Derlon JM, Hatt M, Visvikis D, Guillouet S, Perrio C, Bernaudin M, Valable S. Simultaneous Mapping of Vasculature, Hypoxia, and Proliferation Using Dynamic Susceptibility Contrast MRI, 18F-FMISO PET, and 18F-FLT PET in Relation to Contrast Enhancement in Newly Diagnosed Glioblastoma. J Nucl Med 2021; 62:1349-1356. [PMID: 34016725 PMCID: PMC8724903 DOI: 10.2967/jnumed.120.249524] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 01/14/2021] [Indexed: 11/16/2022] Open
Abstract
Conventional MRI plays a key role in the management of patients with high-grade glioma, but multiparametric MRI and PET tracers could provide further information to better characterize tumor metabolism and heterogeneity by identifying regions having a high risk of recurrence. In this study, we focused on proliferation, hypervascularization, and hypoxia, all factors considered indicative of poor prognosis. They were assessed by measuring uptake of 18F-3'-deoxy-3'-18F-fluorothymidine (18F-FLT), relative cerebral blood volume (rCBV) maps, and uptake of 18F-fluoromisonidazole (18F-FMISO), respectively. For each modality, the volumes and high-uptake subvolumes (hot spots) were semiautomatically segmented and compared with the contrast enhancement (CE) volume on T1-weighted gadolinium-enhanced (T1w-Gd) images, commonly used in the management of patients with glioblastoma. Methods: Dynamic susceptibility contrast-enhanced MRI (31 patients), 18F-FLT PET (20 patients), or 18F-FMISO PET (20 patients), for a total of 31 patients, was performed on preoperative glioblastoma patients. Volumes and hot spots were segmented on SUV maps for 18F-FLT PET (using the fuzzy locally adaptive bayesian algorithm) and 18F-FMISO PET (using a mean contralateral image + 3.3 SDs) and on rCBV maps (using a mean contralateral image + 1.96 SDs) for dynamic susceptibility contrast-enhanced MRI and overlaid on T1w-Gd images. For each modality, the percentages of the peripheral volumes and the peripheral hot spots outside the CE volume were calculated. Results: All tumors showed highly proliferated, hypervascularized, and hypoxic regions. The images also showed pronounced heterogeneity of both tracers regarding their uptake and rCBV maps, within each individual patient. Overlaid volumes on T1w-Gd images showed that some proliferative, hypervascularized, and hypoxic regions extended beyond the CE volume but with marked differences between patients. The ranges of peripheral volume outside the CE volume were 1.6%-155.5%, 1.5%-89.5%, and 3.1%-78.0% for 18F-FLT, rCBV, and 18F-FMISO, respectively. All patients had hyperproliferative hot spots outside the CE volume, whereas hypervascularized and hypoxic hot spots were detected mainly within the enhancing region. Conclusion: Spatial analysis of multiparametric maps with segmented volumes and hot spots provides valuable information to optimize the management and treatment of patients with glioblastoma.
Collapse
Affiliation(s)
- Solène Collet
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
- Radiophysics Department, Centre François Baclesse, Caen, France
| | - Jean-Sébastien Guillamo
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
- Department of Neurology, CHU de Caen, Caen, France
- Department of Neurology, CHU de Nimes, Nimes, France
| | - David Hassanein Berro
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
- Department of Neurosurgery, CHU de Caen, Caen, France
| | - Ararat Chakhoyan
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| | - Jean-Marc Constans
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
- Department of Neuroradiology, CHU de Caen, Caen, France
| | - Emmanuèle Lechapt-Zalcman
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
- Department of Pathology, CHU de Caen, Caen, France
- Department of Neuropathology, GHU Paris Psychiatry and Neuroscience, Paris, France
| | - Jean-Michel Derlon
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, University of Brest, Brest, France; and
| | | | - Stéphane Guillouet
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, GIP Cyceron, Caen, France
| | - Cécile Perrio
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, GIP Cyceron, Caen, France
| | - Myriam Bernaudin
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| | - Samuel Valable
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France;
| |
Collapse
|
8
|
Yan J, Zhao Y, Chen Y, Wang W, Duan W, Wang L, Zhang S, Ding T, Liu L, Sun Q, Pei D, Zhan Y, Zhao H, Sun T, Sun C, Wang W, Liu Z, Hong X, Wang X, Guo Y, Li W, Cheng J, Liu X, Lv X, Li ZC, Zhang Z. Deep learning features from diffusion tensor imaging improve glioma stratification and identify risk groups with distinct molecular pathway activities. EBioMedicine 2021; 72:103583. [PMID: 34563923 PMCID: PMC8479635 DOI: 10.1016/j.ebiom.2021.103583] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND To develop and validate a deep learning signature (DLS) from diffusion tensor imaging (DTI) for predicting overall survival in patients with infiltrative gliomas, and to investigate the biological pathways underlying the developed DLS. METHODS The DLS was developed based on a deep learning cohort (n = 688). The key pathways underlying the DLS were identified on a radiogenomics cohort with paired DTI and RNA-seq data (n=78), where the prognostic value of the pathway genes was validated in public databases (TCGA, n = 663; CGGA, n = 657). FINDINGS The DLS was associated with survival (log-rank P < 0.001) and was an independent predictor (P < 0.001). Incorporating the DLS into existing risk system resulted in a deep learning nomogram predicting survival better than either the DLS or the clinicomolecular nomogram alone, with a better calibration and classification accuracy (net reclassification improvement 0.646, P < 0.001). Five kinds of pathways (synaptic transmission, calcium signaling, glutamate secretion, axon guidance, and glioma pathways) were significantly correlated with the DLS. Average expression value of pathway genes showed prognostic significance in our radiogenomics cohort and TCGA/CGGA cohorts (log-rank P < 0.05). INTERPRETATION DTI-derived DLS can improve glioma stratification by identifying risk groups with dysregulated biological pathways that contributed to survival outcomes. Therapies inhibiting neuron-to-brain tumor synaptic communication may be more effective in high-risk glioma defined by DTI-derived DLS. FUNDING A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Glioma Multidisciplinary Research Group, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanshen Zhao
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yinsheng Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Weiwei Wang
- Glioma Multidisciplinary Research Group, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenchao Duan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shenghai Zhang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Tianqing Ding
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lei Liu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qiuchang Sun
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dongling Pei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yunbo Zhan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haibiao Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tao Sun
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chen Sun
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenqing Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuanke Hong
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiangxiang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaofei Lv
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Zhi-Cheng Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; National Innovation Center for Advanced Medical Devices, Shenzhen, China.
| | - Zhenyu Zhang
- Glioma Multidisciplinary Research Group, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
9
|
Hill L, Bruns J, Zustiak SP. Hydrogel matrix presence and composition influence drug responses of encapsulated glioblastoma spheroids. Acta Biomater 2021; 132:437-447. [PMID: 34010694 DOI: 10.1016/j.actbio.2021.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 12/26/2022]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive brain tumor with median patient survival of 12-15 months. To facilitate treatment development, bioengineered GBM models that adequately recapitulate the in vivo tumor microenvironment are needed. Matrix-encapsulated multicellular spheroids represent such model because they recapitulate solid tumor characteristics, such as dimensionality, cell-cell, and cell-matrix interactions. Yet, there is no consensus as to which matrix properties are key to improving the predictive capacity of spheroid-based drug screening platforms. We used a hydrogel-encapsulated GBM spheroid model, where matrix properties were independently altered to investigate their effect on GBM spheroid characteristics and drug responsiveness. We focused on hydrogel degradability, tuned via enzymatically degradable crosslinkers, and hydrogel adhesiveness, tuned via integrin ligands. We observed increased cellular infiltration of GBM spheroids and increased resistance to temozolomide in degradable, adhesive hydrogels compared to spheroids in non-degradable, non-adhesive hydrogels or to free-floating spheroids. Further, a higher infiltration index was noted for spheroids in adhesive compared to non-adhesive degradable hydrogels. For spheroids in degradable hydrogels, we determined that infiltrating cells were more susceptible to temozolomide compared to cells in the spheroid core. The temozolomide susceptibility of the infiltrating cells was independent of integrin adhesion. We could not attribute differential drug responses to differential cellular proliferation or to limited drug penetration into the hydrogel matrix. Our results suggest that cell-matrix interactions guide GBM spheroid drug responsiveness and that further elucidation of these interactions could enable the engineering of more predictive drug screening platforms. STATEMENT OF SIGNIFICANCE: Glioblastoma multiforme (GBM) multicellular spheroids hold promise for drug screening and development as they better mimic in vivo cellular responses to therapeutics compared to monolayer cultures. Traditional spheroid models lack an external extracellular matrix (ECM) and fail to mimic the mechanical, physical, and biochemical cues seen in the GBM microenvironment. While embedding spheroids in hydrogel matrices has been shown to better recapitulate the tumor microenvironment, there is still limited understanding as to the key matrix properties that govern spheroid responsiveness to drugs. Here we decoupled and independently altered matrix properties such as degradability, via an enzymatically degradable peptide crosslinker, and cell adhesion, via an adhesive ligand, giving further insight into what matrix properties contribute to GBM chemoresistance.
Collapse
|
10
|
Lin M, Wynne JF, Zhou B, Wang T, Lei Y, Curran WJ, Liu T, Yang X. Artificial intelligence in tumor subregion analysis based on medical imaging: A review. J Appl Clin Med Phys 2021; 22:10-26. [PMID: 34164913 PMCID: PMC8292694 DOI: 10.1002/acm2.13321] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/17/2021] [Accepted: 05/22/2021] [Indexed: 12/20/2022] Open
Abstract
Medical imaging is widely used in the diagnosis and treatment of cancer, and artificial intelligence (AI) has achieved tremendous success in medical image analysis. This paper reviews AI-based tumor subregion analysis in medical imaging. We summarize the latest AI-based methods for tumor subregion analysis and their applications. Specifically, we categorize the AI-based methods by training strategy: supervised and unsupervised. A detailed review of each category is presented, highlighting important contributions and achievements. Specific challenges and potential applications of AI in tumor subregion analysis are discussed.
Collapse
Affiliation(s)
- Mingquan Lin
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Jacob F. Wynne
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Boran Zhou
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Tonghe Wang
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Yang Lei
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Walter J. Curran
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Tian Liu
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Xiaofeng Yang
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
11
|
Radiology in the lead: towards radiological profiling for precision medicine in glioblastoma patients? Editorial comment on Glioblastoma patients with a moderate vascular profile benefit the most from MGMT methylation. Eur Radiol 2021; 31:1736-1737. [PMID: 33427947 DOI: 10.1007/s00330-020-07588-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/02/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022]
|
12
|
Zhang I, Lépine P, Han C, Lacalle-Aurioles M, Chen CXQ, Haag R, Durcan TM, Maysinger D. Nanotherapeutic Modulation of Human Neural Cells and Glioblastoma in Organoids and Monocultures. Cells 2020; 9:cells9112434. [PMID: 33171886 PMCID: PMC7695149 DOI: 10.3390/cells9112434] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
Inflammatory processes in the brain are orchestrated by microglia and astrocytes in response to activators such as pathogen-associated molecular patterns, danger-associated molecular patterns and some nanostructures. Microglia are the primary immune responders in the brain and initiate responses amplified by astrocytes through intercellular signaling. Intercellular communication between neural cells can be studied in cerebral organoids, co-cultures or in vivo. We used human cerebral organoids and glioblastoma co-cultures to study glia modulation by dendritic polyglycerol sulfate (dPGS). dPGS is an extensively studied nanostructure with inherent anti-inflammatory properties. Under inflammatory conditions, lipocalin-2 levels in astrocytes are markedly increased and indirectly enhanced by soluble factors released from hyperactive microglia. dPGS is an effective anti-inflammatory modulator of these markers. Our results show that dPGS can enter neural cells in cerebral organoids and glial cells in monocultures in a time-dependent manner. dPGS markedly reduces lipocalin-2 abundance in the neural cells. Glioblastoma tumoroids of astrocytic origin respond to activated microglia with enhanced invasiveness, whereas conditioned media from dPGS-treated microglia reduce tumoroid invasiveness. Considering that many nanostructures have only been tested in cancer cells and rodent models, experiments in human 3D cerebral organoids and co-cultures are complementary in vitro models to evaluate nanotherapeutics in the pre-clinical setting. Thoroughly characterized organoids and standardized procedures for their preparation are prerequisites to gain information of translational value in nanomedicine. This study provides data for a well-characterized dendrimer (dPGS) that modulates the activation state of human microglia implicated in brain tumor invasiveness.
Collapse
Affiliation(s)
- Issan Zhang
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada;
| | - Paula Lépine
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Chanshuai Han
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - María Lacalle-Aurioles
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Carol X.-Q. Chen
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany;
| | - Thomas M. Durcan
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada; (P.L.); (C.H.); (M.L.-A.); (C.X.-Q.C.); (T.M.D.)
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada;
- Correspondence: ; Tel.: +1-514-398-1264
| |
Collapse
|
13
|
Ammer LM, Vollmann-Zwerenz A, Ruf V, Wetzel CH, Riemenschneider MJ, Albert NL, Beckhove P, Hau P. The Role of Translocator Protein TSPO in Hallmarks of Glioblastoma. Cancers (Basel) 2020; 12:cancers12102973. [PMID: 33066460 PMCID: PMC7602186 DOI: 10.3390/cancers12102973] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The translocator protein (TSPO) has been under extensive investigation as a specific marker in positron emission tomography (PET) to visualize brain lesions following injury or disease. In recent years, TSPO is increasingly appreciated as a potential novel therapeutic target in cancer. In Glioblastoma (GBM), the most malignant primary brain tumor, TSPO expression levels are strongly elevated and scientific evidence accumulates, hinting at a pivotal role of TSPO in tumorigenesis and glioma progression. The aim of this review is to summarize the current literature on TSPO with respect to its role both in diagnostics and especially with regard to the critical hallmarks of cancer postulated by Hanahan and Weinberg. Overall, our review contributes to a better understanding of the functional significance of TSPO in Glioblastoma and draws attention to TSPO as a potential modulator of treatment response and thus an important factor that may influence the clinical outcome of GBM. Abstract Glioblastoma (GBM) is the most fatal primary brain cancer in adults. Despite extensive treatment, tumors inevitably recur, leading to an average survival time shorter than 1.5 years. The 18 kDa translocator protein (TSPO) is abundantly expressed throughout the body including the central nervous system. The expression of TSPO increases in states of inflammation and brain injury due to microglia activation. Not least due to its location in the outer mitochondrial membrane, TSPO has been implicated with a broad spectrum of functions. These include the regulation of proliferation, apoptosis, migration, as well as mitochondrial functions such as mitochondrial respiration and oxidative stress regulation. TSPO is frequently overexpressed in GBM. Its expression level has been positively correlated to WHO grade, glioma cell proliferation, and poor prognosis of patients. Several lines of evidence indicate that TSPO plays a functional part in glioma hallmark features such as resistance to apoptosis, invasiveness, and proliferation. This review provides a critical overview of how TSPO could regulate several aspects of tumorigenesis in GBM, particularly in the context of the hallmarks of cancer proposed by Hanahan and Weinberg in 2011.
Collapse
Affiliation(s)
- Laura-Marie Ammer
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany; (L.-M.A.); (A.V.-Z.)
| | - Arabel Vollmann-Zwerenz
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany; (L.-M.A.); (A.V.-Z.)
| | - Viktoria Ruf
- Center for Neuropathology and Prion Research, Ludwig Maximilians University of Munich, 81377 Munich, Germany;
| | - Christian H. Wetzel
- Molecular Neurosciences, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany;
| | | | - Nathalie L. Albert
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, 81377 Munich, Germany;
| | - Philipp Beckhove
- Regensburg Center for Interventional Immunology (RCI) and Department Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Peter Hau
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany; (L.-M.A.); (A.V.-Z.)
- Correspondence:
| |
Collapse
|
14
|
Sravya P, Nimbalkar VP, Kanuri NN, Sugur H, Verma BK, Kundu P, Rao S, Uday Krishna AS, Somanna S, Kondaiah P, Arivazhagan A, Santosh V. Low mitochondrial DNA copy number is associated with poor prognosis and treatment resistance in glioblastoma. Mitochondrion 2020; 55:154-163. [PMID: 33045388 DOI: 10.1016/j.mito.2020.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/27/2020] [Accepted: 10/05/2020] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Mitochondrial DNA (mtDNA) content in several solid tumors was found to be lower than in their normal counterparts. However, there is paucity of literature on the clinical significance of mtDNA content in glioblastoma and its effect on treatment response. Hence, we studied the prognostic significance of mtDNA content in glioblastoma tumor tissue and the effect of mtDNA depletion in glioblastoma cells on response to treatment. MATERIALS AND METHODS 130 newly diagnosed glioblastomas, 32 paired newly diagnosed and recurrent glioblastomas and 35 non-neoplastic brain tissues were utilized for the study. mtDNA content in the patient tumor tissue was assessed and compared with known biomarkers and patient survival. mtDNA was chemically depleted in malignant glioma cell lines, U87, LN229. The biology and treatment response of parent and depleted cells were compared. RESULTS Lower range of mtDNA copy number in glioblastoma was associated with poor overall survival (p = 0.01), progression free survival (p = 0.04) and also with wild type IDH (p = 0.02). In recurrent glioblastoma, mtDNA copy number was higher than newly diagnosed glioblastoma in the patients who received RT (p = 0.01). mtDNA depleted U87 and LN229 cells showed higher survival fraction post radiation exposure when compared to parent lines. The IC50 of TMZ was also higher for mtDNA depleted U87 and LN229 cells. The depleted cells formed more neurospheres than their parent counterparts, thus showing increased stemness of mtDNA depleted cells. CONCLUSION Low mtDNA copy number in glioblastoma is associated with poor patient survival and treatment resistance in cell lines possibly by impacting stemness of the glioblastoma cells.
Collapse
Affiliation(s)
- Palavalasa Sravya
- Department of Clinical Neurosciences, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Vidya Prasad Nimbalkar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Nandaki Nag Kanuri
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Harsha Sugur
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Brijesh Kumar Verma
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Paramita Kundu
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Shilpa Rao
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - A S Uday Krishna
- Department of Radiation Oncology, KIDWAI Memorial Institute of Oncology, Bengaluru, India
| | - Sampath Somanna
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Paturu Kondaiah
- Department of Molecular Reproduction Development and Genetics, Indian Institute of Science, Bengaluru, India
| | - Arimappamagan Arivazhagan
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bengaluru, India.
| | - Vani Santosh
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, India.
| |
Collapse
|
15
|
Predicting Survival in Glioblastoma Patients Using Diffusion MR Imaging Metrics-A Systematic Review. Cancers (Basel) 2020; 12:cancers12102858. [PMID: 33020420 PMCID: PMC7600641 DOI: 10.3390/cancers12102858] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/20/2022] Open
Abstract
Simple Summary An accurate survival analysis is crucial for disease management in glioblastoma (GBM) patients. Due to the ability of the diffusion MRI techniques of providing a quantitative assessment of GBM tumours, an ever-growing number of studies aimed at investigating the role of diffusion MRI metrics in survival prediction of GBM patients. Since the role of diffusion MRI in prediction and evaluation of survival outcomes has not been fully addressed and results are often controversial or unsatisfactory, we performed this systematic review in order to collect, summarize and evaluate all studies evaluating the role of diffusion MRI metrics in predicting survival in GBM patients. We found that quantitative diffusion MRI metrics provide useful information for predicting survival outcomes in GBM patients, mainly in combination with other clinical and multimodality imaging parameters. Abstract Despite advances in surgical and medical treatment of glioblastoma (GBM), the medium survival is about 15 months and varies significantly, with occasional longer survivors and individuals whose tumours show a significant response to therapy with respect to others. Diffusion MRI can provide a quantitative assessment of the intratumoral heterogeneity of GBM infiltration, which is of clinical significance for targeted surgery and therapy, and aimed at improving GBM patient survival. So, the aim of this systematic review is to assess the role of diffusion MRI metrics in predicting survival of patients with GBM. According to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement, a systematic literature search was performed to identify original articles since 2010 that evaluated the association of diffusion MRI metrics with overall survival (OS) and progression-free survival (PFS). The quality of the included studies was evaluated using the QUIPS tool. A total of 52 articles were selected. The most examined metrics were associated with the standard Diffusion Weighted Imaging (DWI) (34 studies) and Diffusion Tensor Imaging (DTI) models (17 studies). Our findings showed that quantitative diffusion MRI metrics provide useful information for predicting survival outcomes in GBM patients, mainly in combination with other clinical and multimodality imaging parameters.
Collapse
|
16
|
Abstract
Magnetic resonance imaging (MRI) has been the cornerstone of imaging of brain tumors in the past 4 decades. Conventional MRI remains the workhorse for neuro-oncologic imaging, not only for basic information such as location, extent, and navigation but also able to provide information regarding proliferation and infiltration, angiogenesis, hemorrhage, and more. More sophisticated MRI sequences have extended the ability to assess and quantify these features; for example, permeability and perfusion acquisitions can assess blood-brain barrier disruption and angiogenesis, diffusion techniques can assess cellularity and infiltration, and spectroscopy can address metabolism. Techniques such as fMRI and diffusion fiber tracking can be helpful in diagnostic planning for resection and radiation therapy, and more sophisticated iterations of these techniques can extend our understanding of neurocognitive effects of these tumors and associated treatment responses and effects. More recently, MRI has been used to go beyond such morphological, physiological, and functional characteristics to assess the tumor microenvironment. The current review highlights multiple recent and emerging approaches in MRI to characterize the tumor microenvironment.
Collapse
|
17
|
Rahmat R, Brochu F, Li C, Sinha R, Price SJ, Jena R. Semi-automated construction of patient individualised clinical target volumes for radiotherapy treatment of glioblastoma utilising diffusion tensor decomposition maps. Br J Radiol 2020; 93:20190441. [PMID: 31944147 PMCID: PMC7362908 DOI: 10.1259/bjr.20190441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 12/09/2019] [Accepted: 01/09/2020] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVES Glioblastoma multiforme (GBM) is a highly infiltrative primary brain tumour with an aggressive clinical course. Diffusion tensor imaging (DT-MRI or DTI) is a recently developed technique capable of visualising subclinical tumour spread into adjacent brain tissue. Tensor decomposition through p and q maps can be used for planning of treatment. Our objective was to develop a tool to automate the segmentation of DTI decomposed p and q maps in GBM patients in order to inform construction of radiotherapy target volumes. METHODS Chan-Vese level set model is applied to segment the p map using the q map as its initial starting point. The reason of choosing this model is because of the robustness of this model on either conventional MRI or only DTI. The method was applied on a data set consisting of 50 patients having their gross tumour volume delineated on their q map and Chan-Vese level set model uses these superimposed masks to incorporate the infiltrative edges. RESULTS The expansion of tumour boundary from q map to p map is clearly visible in all cases and the Dice coefficient (DC) showed a mean similarity of 74% across all 50 patients between the manually segmented ground truth p map and the level set automatic segmentation. CONCLUSION Automated segmentation of the tumour infiltration boundary using DTI and tensor decomposition is possible using Chan-Vese level set methods to expand q map to p map. We have provided initial validation of this technique against manual contours performed by experienced clinicians. ADVANCES IN KNOWLEDGE This novel automated technique to generate p maps has the potential to individualise radiation treatment volumes and act as a decision support tool for the treating oncologist.
Collapse
Affiliation(s)
- Roushanak Rahmat
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | | | - Chao Li
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Rohitashwa Sinha
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Stephen John Price
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Raj Jena
- Oncology Centre, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
18
|
Muñoz-Hidalgo L, San-Miguel T, Megías J, Monleón D, Navarro L, Roldán P, Cerdá-Nicolás M, López-Ginés C. Somatic copy number alterations are associated with EGFR amplification and shortened survival in patients with primary glioblastoma. Neoplasia 2019; 22:10-21. [PMID: 31751860 PMCID: PMC6864306 DOI: 10.1016/j.neo.2019.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/16/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary tumor of the central nervous system. With no effective therapy, the prognosis for patients is terrible poor. It is highly heterogeneous and EGFR amplification is its most frequent molecular alteration. In this light, we aimed to examine the genetic heterogeneity of GBM and to correlate it with the clinical characteristics of the patients. For that purpose, we analyzed the status of EGFR and the somatic copy number alterations (CNAs) of a set of tumor suppressor genes and oncogenes. Thus, we found GBMs with high level of EGFR amplification, low level and with no EGFR amplification. Highly amplified tumors showed histological features of aggressiveness. Interestingly, accumulation of CNAs, as a measure of tumor mutational burden, was frequent and significantly associated to shortened survival. EGFR-amplified GBMs displayed both a higher number of concrete CNAs and a higher global tumor mutational burden than their no EGFR-amplified counterparts. In addition to genetic changes previously described in GBM, we found PARK2 and LARGE1 CNAs associated to EGFR amplification. The set of genes analyzed allowed us to explore relevant signaling pathways on GBM. Both PARK2 and LARGE1 are related to receptor tyrosine kinase/PI3K/PTEN/AKT/mTOR-signaling pathway. Finally, we found an association between the molecular pathways altered, EGFR amplification and a poor outcome. Our results underline the potential interest of categorizing GBM according to their EGFR amplification level and the usefulness of assessing the tumor mutational burden. These approaches would open new knowledge possibilities related to GBM biology and therapy.
Collapse
Affiliation(s)
| | - Teresa San-Miguel
- INCLIVA Research Institute, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain; Department of Pathology, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain.
| | - Javier Megías
- INCLIVA Research Institute, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain; Department of Pathology, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Daniel Monleón
- INCLIVA Research Institute, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain; Department of Pathology, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Lara Navarro
- Consortium Hospital General Universitario de Valencia, Av. Tres cruces, 2, 46014 Valencia, Spain
| | - Pedro Roldán
- Department of Neurosurgery, Hospital Clínico Universitario de Valencia, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain
| | - Miguel Cerdá-Nicolás
- INCLIVA Research Institute, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain; Department of Pathology, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Concha López-Ginés
- INCLIVA Research Institute, Av. Blasco Ibáñez, 17, 46010 Valencia, Spain; Department of Pathology, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| |
Collapse
|
19
|
Li C, Wang S, Serra A, Torheim T, Yan JL, Boonzaier NR, Huang Y, Matys T, McLean MA, Markowetz F, Price SJ. Multi-parametric and multi-regional histogram analysis of MRI: modality integration reveals imaging phenotypes of glioblastoma. Eur Radiol 2019; 29:4718-4729. [PMID: 30707277 PMCID: PMC6682853 DOI: 10.1007/s00330-018-5984-z] [Citation(s) in RCA: 5] [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: 11/02/2018] [Accepted: 12/18/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Integrating multiple imaging modalities is crucial for MRI data interpretation. The purpose of this study is to determine whether a previously proposed multi-view approach can effectively integrate the histogram features from multi-parametric MRI and whether the selected features can offer incremental prognostic values over clinical variables. METHODS Eighty newly-diagnosed glioblastoma patients underwent surgery and chemoradiotherapy. Histogram features of diffusion and perfusion imaging were extracted from contrast-enhancing (CE) and non-enhancing (NE) regions independently. An unsupervised patient clustering was performed by the multi-view approach. Kaplan-Meier and Cox proportional hazards regression analyses were performed to evaluate the relevance of patient clustering to survival. The metabolic signatures of patient clusters were compared using multi-voxel spectroscopy analysis. The prognostic values of histogram features were evaluated by survival and ROC curve analyses. RESULTS Two patient clusters were generated, consisting of 53 and 27 patients respectively. Cluster 2 demonstrated better overall survival (OS) (p = 0.007) and progression-free survival (PFS) (p < 0.001) than Cluster 1. Cluster 2 displayed lower N-acetylaspartate/creatine ratio in NE region (p = 0.040). A higher mean value of anisotropic diffusion in NE region was associated with worse OS (hazard ratio [HR] = 1.40, p = 0.020) and PFS (HR = 1.36, p = 0.031). The seven features selected by this approach showed significantly incremental value in predicting 12-month OS (p = 0.020) and PFS (p = 0.022). CONCLUSIONS The multi-view clustering method can provide an effective integration of multi-parametric MRI. The histogram features selected may be used as potential prognostic markers. KEY POINTS • Multi-parametric magnetic resonance imaging captures multi-faceted tumor physiology. • Contrast-enhancing and non-enhancing tumor regions represent different tumor components with distinct clinical relevance. • Multi-view data analysis offers a method which can effectively select and integrate multi-parametric and multi-regional imaging features.
Collapse
Affiliation(s)
- Chao Li
- Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Box 167 Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.
- Department of Neurosurgery, Shanghai General Hospital (originally named "Shanghai First People's Hospital"), Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- The Centre for Mathematical Imaging in Healthcare, Department of Pure Mathematics and Mathematical Statistics, University of Cambridge, Cambridge, UK.
| | - Shuo Wang
- The Centre for Mathematical Imaging in Healthcare, Department of Pure Mathematics and Mathematical Statistics, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Institute of Biosciences and Medical Technologies (BioMediTech), Tampere, Finland
- NeuRoNe Lab, DISA-MIS, University of Salerno, Fisciano, SA, Italy
| | - Turid Torheim
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge, UK
| | - Jiun-Lin Yan
- Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Box 167 Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- Department of Neurosurgery, Chang Gung Memorial Hospital, Keelung, Taiwan
- Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Natalie R Boonzaier
- Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Box 167 Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- Developmental Imaging and Biophysics Section, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Yuan Huang
- The Centre for Mathematical Imaging in Healthcare, Department of Pure Mathematics and Mathematical Statistics, University of Cambridge, Cambridge, UK
| | - Tomasz Matys
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Mary A McLean
- Department of Radiology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Florian Markowetz
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge, UK
| | - Stephen J Price
- Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Box 167 Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|