1
|
Zhou L, Udayakumar D, Wang Y, Pinho MC, Wagner BC, Youssef M, Maldjian JA, Madhuranthakam AJ. Repeatability and Reproducibility of Pseudocontinuous Arterial Spin-Labeling-Measured Brain Perfusion in Healthy Volunteers and Patients with Glioblastoma. AJNR Am J Neuroradiol 2025; 46:973-982. [PMID: 39443151 DOI: 10.3174/ajnr.a8551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND PURPOSE Arterial spin-labeling (ASL) MRI has gained recognition as a quantitative perfusion imaging method for managing patients with brain tumors. Limited studies have so far investigated the reproducibility of ASL-derived perfusion in these patients. This study aimed to evaluate intrasession repeatability and intersession reproducibility of perfusion measurements using 3D pseudocontinuous ASL (pCASL) with TSE Cartesian acquisition with spiral profile reordering (TSE-CASPR) in healthy volunteers (HV) and patients with glioblastoma (GBM) at 3T and to compare them against 3D pCASL with gradient and spin echo (GRASE). MATERIALS AND METHODS This prospective study (NCT03922984) was approved by the institutional review board, and written informed consent was obtained from all subjects. HV underwent repeat pCASL evaluations 2-4 weeks apart between November 2021 and October 2022. Patients with GBM were recruited for longitudinal MRI from September 2019 to February 2023. Intrasession repeatability (HV and GBM) and intersession reproducibility (HV only) of pCASL were assessed using linear regression, Bland-Altman analyses, the intraclass correlation coefficient (ICC) with 95% CI, and within-subject coefficients of variation (wsCV). RESULTS Twenty HV (9 men; mean age, 25.1 [SD, 1.7] years; range, 23-30 years) and 21 patients with GBM (15 men; mean age, 59.8 [SD, 14.3] years; range, 28-81 years) were enrolled. In imaging sessions, 3D pCASL-measured perfusion with TSE-CASPR and GRASE, respectively, achieved high R 2 values (0.88-0.95; 0.93-0.96), minimal biases (-0.46-0.81; -0.08-0.35 mL/100 g/min), high ICCs [95% CI], 0.96-0.98 [0.94-0.98]; 0.96-0.98 [0.92-0.99]), and low wsCV (6.64%-9.07%; 5.20%-8.16%) in HV (n = 20) and patients with GBM (n = 21). Across imaging sessions, 3D pCASL in HV (n = 20) achieved high R 2 values (0.71; 0.82), minimal biases (-1.2; -0.90 mL/100 g/min), high ICC [95% CI] values (0.85 [0.81-0.89]; 0.90 [0.87-0.93]), and low wsCV values (13.82%; 9.98%). CONCLUSIONS Our study demonstrated excellent intrasession repeatability of 3D pCASL-measured cerebral perfusion in HV and patients with GBM and good-to-excellent intersession reproducibility in HV. 3D pCASL with GRASE performed slightly better than 3D pCASL with TSE-CASPR in HV; however, in patients with GBM, 3D pCASL with TSE-CASPR showed better performance in tumor regions with a nearly 2-fold higher SNR. ASL-measured perfusion could serve as a noncontrast quantitative imaging biomarker to facilitate the management of patients with GBM.
Collapse
Affiliation(s)
- Limin Zhou
- From the Department of Radiology (L.Z., D.U., YW., M.C.P., B.C.W. J.A.M. A.J.M.), Utah Southwestern Medical Center, Dallas, Texas
| | - Durga Udayakumar
- From the Department of Radiology (L.Z., D.U., YW., M.C.P., B.C.W. J.A.M. A.J.M.), Utah Southwestern Medical Center, Dallas, Texas
- Advanced Imaging Research Center (D.U., M.C.P., J.A.M., A.J.M.), Utah Southwestern Medical Center, Dallas, Texas
| | - Yiming Wang
- From the Department of Radiology (L.Z., D.U., YW., M.C.P., B.C.W. J.A.M. A.J.M.), Utah Southwestern Medical Center, Dallas, Texas
| | - Marco C Pinho
- From the Department of Radiology (L.Z., D.U., YW., M.C.P., B.C.W. J.A.M. A.J.M.), Utah Southwestern Medical Center, Dallas, Texas
- Advanced Imaging Research Center (D.U., M.C.P., J.A.M., A.J.M.), Utah Southwestern Medical Center, Dallas, Texas
| | - Benjamin C Wagner
- From the Department of Radiology (L.Z., D.U., YW., M.C.P., B.C.W. J.A.M. A.J.M.), Utah Southwestern Medical Center, Dallas, Texas
| | - Michael Youssef
- Departments of Neurology (M.Y.), UT Southwestern Medical Center, Dallas, Texas
- Department of Hematology and Oncology (M.Y.), Utah Southwestern Medical Center, Dallas, Texas
| | - Joseph A Maldjian
- From the Department of Radiology (L.Z., D.U., YW., M.C.P., B.C.W. J.A.M. A.J.M.), Utah Southwestern Medical Center, Dallas, Texas
- Advanced Imaging Research Center (D.U., M.C.P., J.A.M., A.J.M.), Utah Southwestern Medical Center, Dallas, Texas
| | - Ananth J Madhuranthakam
- From the Department of Radiology (L.Z., D.U., YW., M.C.P., B.C.W. J.A.M. A.J.M.), Utah Southwestern Medical Center, Dallas, Texas
- Advanced Imaging Research Center (D.U., M.C.P., J.A.M., A.J.M.), Utah Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
2
|
Familiar AM, Khalili N, Khalili N, Schuman C, Grove E, Viswanathan K, Seidlitz J, Alexander-Bloch A, Zapaishchykova A, Kann BH, Vossough A, Storm PB, Resnick AC, Kazerooni AF, Nabavizadeh A. Empowering Data Sharing in Neuroscience: A Deep Learning Deidentification Method for Pediatric Brain MRIs. AJNR Am J Neuroradiol 2025; 46:964-972. [PMID: 39532533 DOI: 10.3174/ajnr.a8581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND PURPOSE Privacy concerns, such as identifiable facial features within brain scans, have hindered the availability of pediatric neuroimaging data sets for research. Consequently, pediatric neuroscience research lags adult counterparts, particularly in rare disease and under-represented populations. The removal of face regions (image defacing) can mitigate this; however, existing defacing tools often fail with pediatric cases and diverse image types, leaving a critical gap in data accessibility. Given recent National Institutes of Health data sharing mandates, novel solutions are a critical need. MATERIALS AND METHODS To develop an artificial intelligence (AI)-powered tool for automatic defacing of pediatric brain MRIs, deep learning methodologies (nnU-Net) were used by using a large, diverse multi-institutional data set of clinical radiology images. This included multiparametric MRIs (T1-weighted [T1W], T1W-contrast-enhanced, T2-weighted [T2W], T2W-FLAIR) with 976 total images from 208 patients with brain tumor (Children's Brain Tumor Network, CBTN) and 36 clinical control patients (Scans with Limited Imaging Pathology, SLIP) ranging in age from 7 days to 21 years old. RESULTS Face and ear removal accuracy for withheld testing data were the primary measure of model performance. Potential influences of defacing on downstream research usage were evaluated with standard image processing and AI-based pipelines. Group-level statistical trends were compared between original (nondefaced) and defaced images. Across image types, the model had high accuracy for removing face regions (mean accuracy, 98%; n=98 subjects/392 images), with lower performance for removal of ears (73%). Analysis of global and regional brain measures (SLIP cohort) showed minimal differences between original and defaced outputs (mean r S = 0.93, all P < .0001). AI-generated whole brain and tumor volumes (CBTN cohort) and temporalis muscle metrics (volume, cross-sectional area, centile scores; SLIP cohort) were not significantly affected by image defacing (all r S > 0.9, P < .0001). CONCLUSIONS The defacing model demonstrates efficacy in removing facial regions across multiple MRI types and exhibits minimal impact on downstream research usage. A software package with the trained model is freely provided for wider use and further development (pediatric-auto-defacer; https://github.com/d3b-center/pediatric-auto-defacer-public). By offering a solution tailored to pediatric cases and multiple MRI sequences, this defacing tool will expedite research efforts and promote broader adoption of data sharing practices within the neuroscience community.
Collapse
Affiliation(s)
- Ariana M Familiar
- From the Center for Data-Driven Discovery in Biomedicine (Db) (A.M.F., Neda K., Nastaran K., K.V., A.V., P.B.S., A.C.R., A.F.K., A.N.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Neurosurgery (A.M.F., Neda K., Nastaran K., K.V., P.B.S., A.C.R., A.F.K), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Neda Khalili
- From the Center for Data-Driven Discovery in Biomedicine (Db) (A.M.F., Neda K., Nastaran K., K.V., A.V., P.B.S., A.C.R., A.F.K., A.N.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Neurosurgery (A.M.F., Neda K., Nastaran K., K.V., P.B.S., A.C.R., A.F.K), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Nastaran Khalili
- From the Center for Data-Driven Discovery in Biomedicine (Db) (A.M.F., Neda K., Nastaran K., K.V., A.V., P.B.S., A.C.R., A.F.K., A.N.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Neurosurgery (A.M.F., Neda K., Nastaran K., K.V., P.B.S., A.C.R., A.F.K), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Cassidy Schuman
- School of Engineering and Applied Science (C.S., E.G.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Evan Grove
- School of Engineering and Applied Science (C.S., E.G.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Karthik Viswanathan
- From the Center for Data-Driven Discovery in Biomedicine (Db) (A.M.F., Neda K., Nastaran K., K.V., A.V., P.B.S., A.C.R., A.F.K., A.N.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Neurosurgery (A.M.F., Neda K., Nastaran K., K.V., P.B.S., A.C.R., A.F.K), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jakob Seidlitz
- Department of Child and Adolescent Psychiatry and Behavioral Science (J.S., A.A.-B.), The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Psychiatry (J.S., A.A.-B.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Aaron Alexander-Bloch
- Department of Child and Adolescent Psychiatry and Behavioral Science (J.S., A.A.-B.), The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Psychiatry (J.S., A.A.-B.), University of Pennsylvania, Philadelphia, Pennsylvania
- Lifespan Brain Institute at the Children's Hospital of Philadelphia and University of Pennsylvania (A.A.-B.), Philadelphia, Pennsylvania
| | - Anna Zapaishchykova
- Artificial Intelligence in Medicine (AIM) Program (A.Z., B.H.K.), Mass General Brigham, Harvard Medical School, Boston, Massachusetts
- Department of Radiation Oncology (A.Z., B.H.K.), Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Benjamin H Kann
- Artificial Intelligence in Medicine (AIM) Program (A.Z., B.H.K.), Mass General Brigham, Harvard Medical School, Boston, Massachusetts
- Department of Radiation Oncology (A.Z., B.H.K.), Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Arastoo Vossough
- From the Center for Data-Driven Discovery in Biomedicine (Db) (A.M.F., Neda K., Nastaran K., K.V., A.V., P.B.S., A.C.R., A.F.K., A.N.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Radiology (A.V.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Radiology, Perelman School of Medicine (A.V., A.N.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Phillip B Storm
- From the Center for Data-Driven Discovery in Biomedicine (Db) (A.M.F., Neda K., Nastaran K., K.V., A.V., P.B.S., A.C.R., A.F.K., A.N.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Neurosurgery (A.M.F., Neda K., Nastaran K., K.V., P.B.S., A.C.R., A.F.K), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Adam C Resnick
- From the Center for Data-Driven Discovery in Biomedicine (Db) (A.M.F., Neda K., Nastaran K., K.V., A.V., P.B.S., A.C.R., A.F.K., A.N.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Neurosurgery (A.M.F., Neda K., Nastaran K., K.V., P.B.S., A.C.R., A.F.K), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Anahita Fathi Kazerooni
- From the Center for Data-Driven Discovery in Biomedicine (Db) (A.M.F., Neda K., Nastaran K., K.V., A.V., P.B.S., A.C.R., A.F.K., A.N.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Neurosurgery (A.M.F., Neda K., Nastaran K., K.V., P.B.S., A.C.R., A.F.K), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- AI2D Center for AI and Data Science for Integrated Diagnostics (A.F.K.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ali Nabavizadeh
- From the Center for Data-Driven Discovery in Biomedicine (Db) (A.M.F., Neda K., Nastaran K., K.V., A.V., P.B.S., A.C.R., A.F.K., A.N.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Radiology, Perelman School of Medicine (A.V., A.N.), University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
3
|
Namdar K, Wagner MW, Kudus K, Hawkins C, Tabori U, Ertl-Wagner BB, Khalvati F. Improving Deep Learning Models for Pediatric Low-Grade Glioma Tumours Molecular Subtype Identification Using MRI-based 3D Probability Distributions of Tumour Location. Can Assoc Radiol J 2025; 76:313-323. [PMID: 39544176 DOI: 10.1177/08465371241296834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024] Open
Abstract
Purpose: Pediatric low-grade gliomas (pLGG) are the most common brain tumour in children, and the molecular diagnosis of pLGG enables targeted treatment. We use MRI-based Convolutional Neural Networks (CNNs) for molecular subtype identification of pLGG and augment the models using tumour location probability maps. Materials and Methods: MRI FLAIR sequences of 214 patients (110 male, mean age of 8.54 years, 143 BRAF fused and 71 BRAF V600E mutated pLGG tumours) from January 2000 to December 2018 were included in this retrospective REB-approved study. Tumour segmentations (volumes of interest-VOIs) were provided by a pediatric neuroradiology fellow and verified by a pediatric neuroradiologist. Patients were randomly split into development and test sets with an 80/20 ratio. The 3D binary VOI masks for each class in the development set were combined to derive the probability density functions of tumour location. Three pipelines for molecular diagnosis of pLGG were developed: location-based, CNN-based, and hybrid. The experiment was repeated 100 times each with different model initializations and data splits, and the Areas Under the Receiver Operating Characteristic Curve (AUROC) was calculated, and Student's t-test was conducted. Results: The location-based classifier achieved an AUROC of 77.9, 95% confidence interval (CI) (76.8, 79.0). CNN-based classifiers achieved an AUROC of 86.1, 95% CI (85.0, 87.3), while the tumour-location-guided CNNs outperformed the other classifiers with an average AUROC of 88.64, 95% CI (87.6, 89.7), which was statistically significant (P-value .0018). Conclusion: Incorporating tumour location probability maps into CNN models led to significant improvements for molecular subtype identification of pLGG.
Collapse
Affiliation(s)
- Khashayar Namdar
- Division of Neuroradiology, Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children (SickKids), Toronto, ON, Canada
- Neurosciences & Mental Health Research Program, SickKids Research Institute, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
| | - Matthias W Wagner
- Division of Neuroradiology, Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children (SickKids), Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Augsburg, Augsburg, Germany
| | - Kareem Kudus
- Division of Neuroradiology, Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children (SickKids), Toronto, ON, Canada
- Neurosciences & Mental Health Research Program, SickKids Research Institute, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Cynthia Hawkins
- Department of Paediatric Laboratory Medicine, Division of Pathology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Uri Tabori
- Department of Neurooncology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Birgit B Ertl-Wagner
- Division of Neuroradiology, Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children (SickKids), Toronto, ON, Canada
- Neurosciences & Mental Health Research Program, SickKids Research Institute, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Farzad Khalvati
- Division of Neuroradiology, Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children (SickKids), Toronto, ON, Canada
- Neurosciences & Mental Health Research Program, SickKids Research Institute, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Yue L, Pan Y, Li W, Mao J, Hong B, Gu Z, Liu M, Shen D, Xiao S. Predicting cognitive decline: Deep-learning reveals subtle brain changes in pre-MCI stage. J Prev Alzheimers Dis 2025; 12:100079. [PMID: 39920001 DOI: 10.1016/j.tjpad.2025.100079] [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: 10/31/2024] [Revised: 12/22/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND Mild cognitive impairment (MCI) and preclinical MCI (e.g., subjective cognitive decline, SCD) are considered risk states of dementia, such as Alzheimer's Disease (AD). However, it is challenging to accurately predict conversion from normal cognition (NC) to MCI, which is important for early detection and intervention. Since neuropathological changes may have occurred in the brain many years before clinical AD, we sought to detect the subtle brain changes in the pre-MCI stage using a deep-learning method based on structural Magnetic Resonance Imaging (MRI). OBJECTIVES To discover early structural neuroimaging changes that differentiate between stable and progressive cognitive status, and to establish a predictive model for MCI conversion. DESIGN, SETTING AND PARTICIPANTS We first created a unique deep-learning framework for pre-AD conversion prediction through the Alzheimer's Disease Neuroimaging Initiative-1 (ADNI-1) database (n = 845). Then, we tested the model on ADNI-2 (n = 321, followed 3 years) and our private study (n = 109), the China Longitudinal Aging Study (CLAS), to validate the rationality for pre-MCI conversion prediction. The CLAS is a 7-year community-based cohort study in Shanghai. Our framework consisted of two steps: 1) a single-ROI-based network (SRNet) for identifying informative regions in the brain, and 2) a multi-ROI-based network (MRNet) for pre-AD conversion prediction. We then utilized these "ROI-based deep learning" neural networks to create a composite score using advanced algorithm-building. We coined this score as the Progressive Index (PI), which serves as a metric for assessing the propensity of AD conversion. Ultimately, we employed the PI to gauge its predictive capability for MCI conversion in both ADNI-2 and CLAS datasets. MEASUREMENTS We primarily utilized baseline T1-weighted MRI scans to identify the most discriminative brain regions and subsequently developed the PI in both training and validation datasets. We compared the PI across different cognitive groups and conducted logistic regression models along with their AUCs, adjusting for education level, gender, neuropsychological test scores, and the presence of comorbid conditions. RESULTS We trained the SRNet and MRNet using 845 subjects from ADNI-1 with baseline MRI data, in which AD and progressive MCI (converting to AD within 3 years) patients were considered as positive samples, while NC and stable MCI (remaining stable for 3 years) subjects were considered as negative samples. The convolutional neural networks identified the top 10 regions of interest (ROIs) for distinguishing progressive from stable cases. These key brain regions included the hippocampus, amygdala, temporal lobe, insula, and anterior cerebellum. A total of 321 subjects from ADNI-2, including 209 NC (18 progressive NC (pNC), 113 stable NC (sNC), and 78 remaining NC (rNC)) and 112 SCD (11 pSCD, 5 sSCD, and 96 rSCD), as well as 109 subjects from CLAS, including 17 sNC, 16 pNC, 52 sSCD and 24 pSCD participated in the test set, separately. We found that the PI score effectively sorted all subjects by their stages (stable vs progressive). Furthermore, the PI score demonstrated excellent discrimination between the two outcomes in the CLAS data(p<0.001), even after controlling for age, gender, education level, depression symptoms, anxiety symptoms, somatic diseases, and baseline MoCA score. Better performance for prediction progression to MCI in CLAS was obtained when the PI score was combined with clinical measures (AUC=0.812; 95 %CI: 0.725-0.900). CONCLUSIONS This study effectively predicted the progression to MCI among order individuals at normal cognition state by deep learning algorithm with MRI scans. Exploring the key brain alterations during the very early stages, specifically the transition from NC to MCI, based on deep learning methods holds significant potential for further research and contributes to a deeper understanding of disease mechanisms.
Collapse
Affiliation(s)
- Ling Yue
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China
| | - Yongsheng Pan
- School of Computer Science and Engineering, Northwestern Polytechnical University, 127 West Youyi Road, 710072, Xi'an, China
| | - Wei Li
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China
| | - Junyan Mao
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China
| | - Bo Hong
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China
| | - Zhen Gu
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China
| | - Mingxia Liu
- Department of Radiology and BRIC, University of North Carolina at Chapel Hill, 130 Mason Farm Road, Chapel Hill, NC 27599, USA.
| | - Dinggang Shen
- School of Biomedical Engineering & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai United Imaging Intelligence Co., Ltd., Shanghai 200232, China; Shanghai Clinical Research and Trial Center, Shanghai, 201210, China.
| | - Shifu Xiao
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, 200032, Shanghai, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, 600 South Wanping Road, 200032, Shanghai, China.
| |
Collapse
|
5
|
Zhu Q, Liu B, Cui ZX, Cao C, Yan X, Liu Y, Cheng J, Zhou Y, Zhu Y, Wang H, Zeng H, Liang D. PEARL: Cascaded Self-Supervised Cross-Fusion Learning for Parallel MRI Acceleration. IEEE J Biomed Health Inform 2025; 29:3086-3097. [PMID: 38147421 DOI: 10.1109/jbhi.2023.3347355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Supervised deep learning (SDL) methodology holds promise for accelerated magnetic resonance imaging (AMRI) but is hampered by the reliance on extensive training data. Some self-supervised frameworks, such as deep image prior (DIP), have emerged, eliminating the explicit training procedure but often struggling to remove noise and artifacts under significant degradation. This work introduces a novel self-supervised accelerated parallel MRI approach called PEARL, leveraging a multiple-stream joint deep decoder with two cross-fusion schemes to accurately reconstruct one or more target images from compressively sampled k-space. Each stream comprises cascaded cross-fusion sub-block networks (SBNs) that sequentially perform combined upsampling, 2D convolution, joint attention, ReLU activation and batch normalization (BN). Among them, combined upsampling and joint attention facilitate mutual learning between multiple-stream networks by integrating multi-parameter priors in both additive and multiplicative manners. Long-range unified skip connections within SBNs ensure effective information propagation between distant cross-fusion layers. Additionally, incorporating dual-normalized edge-orientation similarity regularization into the training loss enhances detail reconstruction and prevents overfitting. Experimental results consistently demonstrate that PEARL outperforms the existing state-of-the-art (SOTA) self-supervised AMRI technologies in various MRI cases. Notably, 5-fold$\sim$6-fold accelerated acquisition yields a 1$\%$ $\sim$ 2$\%$ improvement in SSIM$_{\mathsf{ROI}}$ and a 3$\%$ $\sim$ 6$\%$ improvement in PSNR$_{\mathsf{ROI}}$, along with a significant 15$\%$ $\sim$ 20$\%$ reduction in RLNE$_{\mathsf{ROI}}$.
Collapse
|
6
|
Hoffmann G, Preibisch C, Günther M, Mahroo A, van Osch MJP, Václavů L, Metz M, Jung K, Zimmer C, Wiestler B, Kaczmarz S. Noninvasive blood-brain barrier integrity mapping in patients with high-grade glioma and metastasis by multi-echo time-encoded arterial spin labeling. Magn Reson Med 2025; 93:2086-2098. [PMID: 39777739 PMCID: PMC11893035 DOI: 10.1002/mrm.30415] [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: 08/01/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE In brain tumors, disruption of the blood-brain barrier (BBB) indicates malignancy. Clinical assessment is qualitative; quantitative evaluation is feasible using the K2 leakage parameter from dynamic susceptibility contrast MRI. However, contrast agent-based techniques are limited in patients with renal dysfunction and insensitive to subtle impairments. Assessing water transport times across the BBB (Tex) by multi-echo arterial spin labeling promises to detect BBB impairments noninvasively and potentially more sensitively. We hypothesized that reduced Tex indicates impaired BBB. Furthermore, we assumed higher sensitivity for Tex than dynamic susceptibility contrast-based K2, because arterial spin labeling uses water as a freely diffusible tracer. METHODS We acquired 3T MRI data from 28 patients with intraparenchymal brain tumors (World Health Organization Grade 3 & 4 gliomas [n = 17] or metastases [n = 11]) and 17 age-matched healthy controls. The protocol included multi-echo and single-echo Hadamard-encoded arterial spin labeling, dynamic susceptibility contrast, and conventional clinical imaging. Tex was calculated using a T2-dependent multi-compartment model. Areas of contrast-enhancing tissue, edema, and normal-appearing tissue were automatically segmented, and parameter values were compared across volumes of interest and between patients and healthy controls. RESULTS Tex was significantly reduced (-20.3%) in contrast-enhancing tissue compared with normal-appearing gray matter and correlated well with |K2| (r = -0.347). Compared with healthy controls, Tex was significantly lower in tumor patients' normal-appearing gray matter (Tex,tumor = 0.141 ± 0.032 s vs. Tex,HC = 0.172 ± 0.036 s) and normal-appearing white matter (Tex,tumor = 0.116 ± 0.015 vs. Tex,HC = 0.127 ± 0.017 s), whereas |K2| did not differ significantly. Receiver operating characteristic analysis showed a larger area under the curve for Tex (0.784) than K2 (0.604). CONCLUSION Tex is sensitive to pathophysiologically impaired BBB. It agrees with contrast agent-based K2 in contrast-enhancing tissue and indicates sensitivity to subtle leakage.
Collapse
Affiliation(s)
- Gabriel Hoffmann
- School of Medicine and Health, Institute for Diagnostic and Interventional NeuroradiologyTechnical University of MunichMunichGermany
- School of Medicine and Health, TUM‐Neuroimaging CenterTechnical University of MunichMunichGermany
| | - Christine Preibisch
- School of Medicine and Health, Institute for Diagnostic and Interventional NeuroradiologyTechnical University of MunichMunichGermany
- School of Medicine and Health, TUM‐Neuroimaging CenterTechnical University of MunichMunichGermany
- School of Medicine and Health, Clinic of NeurologyTechnical University of MunichMunichGermany
| | - Matthias Günther
- Fraunhofer Institute for Digital Medicine MEVIS, MR PhysicsBremenGermany
- MR‐Imaging and SpectroscopyUniversity of BremenBremenGermany
- MediriHeidelbergGermany
| | - Amnah Mahroo
- Fraunhofer Institute for Digital Medicine MEVIS, MR PhysicsBremenGermany
| | - Matthias J. P. van Osch
- Department of Radiology, C.J. Gorter MRI CenterLeiden University Medical CenterLeidenThe Netherlands
- Leiden Institute of Brain and CognitionLeiden UniversityLeidenThe Netherlands
| | - Lena Václavů
- Department of Radiology, C.J. Gorter MRI CenterLeiden University Medical CenterLeidenThe Netherlands
| | - Marie‐Christin Metz
- School of Medicine and Health, Institute for Diagnostic and Interventional NeuroradiologyTechnical University of MunichMunichGermany
| | - Kirsten Jung
- School of Medicine and Health, Institute for Diagnostic and Interventional NeuroradiologyTechnical University of MunichMunichGermany
| | - Claus Zimmer
- School of Medicine and Health, Institute for Diagnostic and Interventional NeuroradiologyTechnical University of MunichMunichGermany
- School of Medicine and Health, TUM‐Neuroimaging CenterTechnical University of MunichMunichGermany
| | - Benedikt Wiestler
- School of Medicine and Health, Institute for Diagnostic and Interventional NeuroradiologyTechnical University of MunichMunichGermany
- TranslaTUMTechnical University of MunichMunichGermany
| | - Stephan Kaczmarz
- School of Medicine and Health, Institute for Diagnostic and Interventional NeuroradiologyTechnical University of MunichMunichGermany
- School of Medicine and Health, TUM‐Neuroimaging CenterTechnical University of MunichMunichGermany
- Philips GmbH Market DACHHamburgGermany
| |
Collapse
|
7
|
Ramos-Cabrer P, Cabrera-Zubizarreta A, Padro D, Matute-González M, Rodríguez-Antigüedad A, Matute C. Reversible reduction in brain myelin content upon marathon running. Nat Metab 2025; 7:697-703. [PMID: 40128612 PMCID: PMC12021653 DOI: 10.1038/s42255-025-01244-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 02/12/2025] [Indexed: 03/26/2025]
Abstract
Here we use magnetic resonance imaging to study the impact of marathon running on brain structure in humans. We show that the signal for myelin water fraction-a surrogate of myelin content-is substantially reduced upon marathon running in specific brain regions involved in motor coordination and sensory and emotional integration, but recovers within two months. These findings suggest that brain myelin content is temporarily and reversibly diminished by severe exercise, a finding consistent with recent evidence from rodent studies that suggest that myelin lipids may act as glial energy reserves in extreme metabolic conditions.
Collapse
Affiliation(s)
- Pedro Ramos-Cabrer
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Alberto Cabrera-Zubizarreta
- Neuroradiology Department, MRI Unit, HT Médica, Jaén, Spain
- Osatek Magnetic Resonance Imaging Unit, Galdakao Hospital, Galdakao, Spain
| | - Daniel Padro
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain
| | | | - Alfredo Rodríguez-Antigüedad
- Department of Neurology, Cruces University Hospital, University of the Basque Country (UPV/EHU), Barakaldo, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU) and CIBERNED-Instituto Carlos III, Leioa, Spain
- Biobizkaia Health Research Institute, Barkaldo, Spain
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country (UPV/EHU) and CIBERNED-Instituto Carlos III, Leioa, Spain.
- Biobizkaia Health Research Institute, Barkaldo, Spain.
| |
Collapse
|
8
|
Häger W, Toma-Dașu I, Astaraki M, Lazzeroni M. Role of modeled high-grade glioma cell invasion and survival on the prediction of tumor progression after radiotherapy. Phys Med Biol 2025; 70:065017. [PMID: 40043359 DOI: 10.1088/1361-6560/adbcf4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Objective.Glioblastoma (GBM) prognosis remains poor despite progress in radiotherapy and imaging techniques. Tumor recurrence has been attributed to the widespread tumor invasion of normal tissue. Since the complete extension of invasion is undetectable on imaging, it is not deliberately treated. To improve the treatment outcome, models have been developed to predict tumor invasion based standard imaging data. This study aimed to investigate whether a tumor invasion model, together with the predicted number of surviving cells after radiotherapy, could predict tumor progression post-treatment.Approach.A tumor invasion model was applied to 56 cases of GBMs treated with radiotherapy. The invasion was quantified as the volume encompassed by the 100 cells mm-3isocontour (V100). A new metric, cell-volume-product, was defined as the product of the volume with cell density greater than a threshold value (in cells mm-3), and the number of surviving cells within that volume, post-treatment. Tumor progression was assessed at 20 ± 10 d and 90 ± 20 d after treatment. Correlations between the disease progression and the gross tumor volume (GTV),V100, and cell-volume-product, were determined using receiver operating characteristic curves.Main results.For the early follow-up time, the correlation between GTV and tumor progression was not statistically significant (p= 0.684). However, statistically significant correlations with progression were found betweenV100and cell-volume-product with a cell threshold of 10-6cells mm-3with areas-under-the-curve of 0.69 (p= 0.023) and 0.66 (p= 0.045), respectively. No significant correlations were found for the late follow-up time.Significance.Modeling tumor spread otherwise undetectable on conventional imaging, as well as radiobiological model predictions of cell survival after treatment, may provide useful information regarding the likelihood of tumor progression at an early follow-up time point, which could potentially lead to improved treatment decisions for patients with GBMs.
Collapse
Affiliation(s)
- Wille Häger
- Department of Physics, Stockholm University, Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Iuliana Toma-Dașu
- Department of Physics, Stockholm University, Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Mehdi Astaraki
- Department of Biomedical Engineering and Health Systems, Royal Institute of Technology, Huddinge, Sweden
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Marta Lazzeroni
- Department of Physics, Stockholm University, Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
9
|
Ohara EY, Vigneshwaran V, Souza R, Vamosi FG, Wilms M, Forkert ND. Dimensionality reduction in 3D causal deep learning for neuroimage generation: an evaluation study. J Med Imaging (Bellingham) 2025; 12:024506. [PMID: 40276097 PMCID: PMC12014944 DOI: 10.1117/1.jmi.12.2.024506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 03/06/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Purpose Causal deep learning (DL) using normalizing flows allows the generation of true counterfactual images, which is relevant for many medical applications such as explainability of decisions, image harmonization, and in-silico studies. However, such models are computationally expensive when applied directly to high-resolution 3D images and, therefore, require image dimensionality reduction (DR) to efficiently process the data. The goal of this work was to compare how different DR methods affect counterfactual neuroimage generation. Approach Five DR techniques [2D principal component analysis (PCA), 2.5D PCA, 3D PCA, autoencoder, and Vector Quantised-Variational AutoEncoder] were applied to 23,692 3D brain images to create low-dimensional representations for causal DL model training. Convolutional neural networks were used to quantitatively evaluate age and sex changes on the counterfactual neuroimages. Age alterations were measured using the mean absolute error (MAE), whereas sex changes were assessed via classification accuracy. Results The 2.5D PCA technique achieved the lowest MAE of 4.16 when changing the age variable of an original image. When sex was changed, the autoencoder embedding led to the highest classification accuracy of 97.84% while also significantly impacting the age variable predictions, increasing the MAE to 5.24 years. Overall, 3D PCA provided the best balance, with an age prediction MAE of 4.57 years while maintaining 94.01% sex classification accuracy when altering the age variable and 94.73% sex classification accuracy and the lowest age prediction MAE (3.84 years) when altering the sex variable. Conclusions 3D PCA appears to be the best-suited DR method for causal neuroimage analysis.
Collapse
Affiliation(s)
- Erik Y. Ohara
- University of Calgary, Biomedical Engineering Graduate Program, Calgary, Alberta, Canada
- University of Calgary, Department of Radiology, Calgary, Alberta, Canada
| | | | - Raissa Souza
- University of Calgary, Biomedical Engineering Graduate Program, Calgary, Alberta, Canada
- University of Calgary, Department of Radiology, Calgary, Alberta, Canada
- University of Calgary, Hotchkiss Brain Institute, Calgary, Alberta, Canada
- University of Calgary, Alberta Children’s Hospital Research Institute, Calgary, Alberta, Canada
| | - Finn G. Vamosi
- University of Calgary, Department of Computer Science, Calgary, Alberta, Canada
| | - Matthias Wilms
- University of Calgary, Department of Radiology, Calgary, Alberta, Canada
- University of Calgary, Hotchkiss Brain Institute, Calgary, Alberta, Canada
- University of Calgary, Alberta Children’s Hospital Research Institute, Calgary, Alberta, Canada
- University of Calgary, Departments of Pediatrics and Community Health Sciences, Calgary, Alberta, Canada
| | - Nils D. Forkert
- University of Calgary, Department of Radiology, Calgary, Alberta, Canada
- University of Calgary, Hotchkiss Brain Institute, Calgary, Alberta, Canada
- University of Calgary, Alberta Children’s Hospital Research Institute, Calgary, Alberta, Canada
| |
Collapse
|
10
|
Fathi Kazerooni A, Akbari H, Hu X, Bommineni V, Grigoriadis D, Toorens E, Sako C, Mamourian E, Ballinger D, Sussman R, Singh A, Verginadis II, Dahmane N, Koumenis C, Binder ZA, Bagley SJ, Mohan S, Hatzigeorgiou A, O'Rourke DM, Ganguly T, De S, Bakas S, Nasrallah MP, Davatzikos C. The radiogenomic and spatiogenomic landscapes of glioblastoma and their relationship to oncogenic drivers. COMMUNICATIONS MEDICINE 2025; 5:55. [PMID: 40025245 PMCID: PMC11873127 DOI: 10.1038/s43856-025-00767-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/12/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Glioblastoma is a highly heterogeneous brain tumor, posing challenges for precision therapies and patient stratification in clinical trials. Understanding how genetic mutations influence tumor imaging may improve patient management and treatment outcomes. This study investigates the relationship between imaging features, spatial patterns of tumor location, and genetic alterations in IDH-wildtype glioblastoma, as well as the likely sequence of mutational events. METHODS We conducted a retrospective analysis of 357 IDH-wildtype glioblastomas with pre-operative multiparametric MRI and targeted genetic sequencing data. Radiogenomic signatures and spatial distribution maps were generated for key mutations in genes such as EGFR, PTEN, TP53, and NF1 and their corresponding pathways. Machine and deep learning models were used to identify imaging biomarkers and stratify tumors based on their genetic profiles and molecular heterogeneity. RESULTS Here, we show that glioblastoma mutations produce distinctive imaging signatures, which are more pronounced in tumors with less molecular heterogeneity. These signatures provide insights into how mutations affect tumor characteristics such as neovascularization, cell density, invasion, and vascular leakage. We also found that tumor location and spatial distribution correlate with genetic profiles, revealing associations between tumor regions and specific oncogenic drivers. Additionally, imaging features reflect the cross-sectionally inferred evolutionary trajectories of glioblastomas. CONCLUSIONS This study establishes clinically accessible imaging biomarkers that capture the molecular composition and oncogenic drivers of glioblastoma. These findings have potential implications for noninvasive tumor profiling, personalized therapies, and improved patient stratification in clinical trials.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Center for Data-Driven Discovery in Biomedicine (D3b), Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamed Akbari
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, USA
| | - Xiaoju Hu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ, USA
| | - Vikas Bommineni
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
| | - Dimitris Grigoriadis
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Erik Toorens
- Penn Genomic Analysis Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chiharu Sako
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dominique Ballinger
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robyn Sussman
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashish Singh
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nadia Dahmane
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Bagley
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Artemis Hatzigeorgiou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- Hellenic Pasteur Institute, Athens, Greece
| | - Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Tapan Ganguly
- Penn Genomic Analysis Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Subhajyoti De
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ, USA
| | - Spyridon Bakas
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - MacLean P Nasrallah
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Fiszer J, Ciupek D, Malawski M, Pieciak T. Validation of ten federated learning strategies for multi-contrast image-to-image MRI data synthesis from heterogeneous sources. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.09.637305. [PMID: 39990397 PMCID: PMC11844418 DOI: 10.1101/2025.02.09.637305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Deep learning (DL)-based image synthesis has recently gained enormous interest in medical imaging, allowing for generating multi-contrast data and therefore, the recovery of missing samples from interrupted or artefact-distorted acquisitions. However, the accuracy of DL models heavily relies on the representativeness of the training datasets naturally characterized by their distributions, experimental setups or preprocessing schemes. These complicate generalizing DL models across multi-site heterogeneous data sets while maintaining the confidentiality of the data. One of the possible solutions is to employ federated learning (FL), which enables the collaborative training of a DL model in a decentralized manner, demanding the involved sites to share only the characteristics of the models without transferring their sensitive medical data. The paper presents a DL-based magnetic resonance (MR) data translation in a FL way. We introduce a new aggregation strategy called FedBAdam that couples two state-of-the-art methods with complementary strengths by incorporating momentum in the aggregation scheme and skipping the batch normalization layers. The work comprehensively validates 10 FL-based strategies for an image-to-image multi-contrast MR translation, considering healthy and tumorous brain scans from five different institutions. Our study has revealed that the FedBAdam shows superior results in terms of mean squared error and structural similarity index over personalized methods, like the FedMRI, and standard FL-based aggregation techniques, such as the FedAvg or FedProx, considering multi-site multi-vendor heterogeneous environment. The FedBAdam has prevented the overfitting of the model and gradually reached the optimal model parameters, exhibiting no oscillations.
Collapse
Affiliation(s)
- Jan Fiszer
- Sano Centre for Computational Medicine, Kraków, Poland
- AGH University of Science and Technology, Kraków, Poland
| | | | - Maciej Malawski
- Sano Centre for Computational Medicine, Kraków, Poland
- AGH University of Science and Technology, Kraków, Poland
| | - Tomasz Pieciak
- Laboratorio de Procesado de Imagen (LPI), ETSI Telecomunicación, Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
12
|
Safari M, Eidex Z, Chang CW, Qiu RL, Yang X. Advancing MRI Reconstruction: A Systematic Review of Deep Learning and Compressed Sensing Integration. ARXIV 2025:arXiv:2501.14158v2. [PMID: 39975448 PMCID: PMC11838702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Magnetic resonance imaging (MRI) is a non-invasive imaging modality and provides comprehensive anatomical and functional insights into the human body. However, its long acquisition times can lead to patient discomfort, motion artifacts, and limiting real-time applications. To address these challenges, strategies such as parallel imaging have been applied, which utilize multiple receiver coils to speed up the data acquisition process. Additionally, compressed sensing (CS) is a method that facilitates image reconstruction from sparse data, significantly reducing image acquisition time by minimizing the amount of data collection needed. Recently, deep learning (DL) has emerged as a powerful tool for improving MRI reconstruction. It has been integrated with parallel imaging and CS principles to achieve faster and more accurate MRI reconstructions. This review comprehensively examines DL-based techniques for MRI reconstruction. We categorize and discuss various DL-based methods, including end-to-end approaches, unrolled optimization, and federated learning, highlighting their potential benefits. Our systematic review highlights significant contributions and underscores the potential of DL in MRI reconstruction. Additionally, we summarize key results and trends in DL-based MRI reconstruction, including quantitative metrics, the dataset, acceleration factors, and the progress of and research interest in DL techniques over time. Finally, we discuss potential future directions and the importance of DL-based MRI reconstruction in advancing medical imaging. To facilitate further research in this area, we provide a GitHub repository that includes up-to-date DL-based MRI reconstruction publications and public datasets-https://github.com/mosaf/Awesome-DL-based-CS-MRI.
Collapse
Affiliation(s)
- Mojtaba Safari
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States of America
| | - Zach Eidex
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States of America
| | - Chih-Wei Chang
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States of America
| | - Richard L.J. Qiu
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States of America
| | - Xiaofeng Yang
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States of America
| |
Collapse
|
13
|
Solak N, Ferreira A, Luijten G, Puladi B, Alves V, Egger J. GBM-Reservoir: Brain tumor (Glioblastoma Multiforme) MRI dataset collection with ground truth segmentation masks. Data Brief 2025; 58:111287. [PMID: 39911270 PMCID: PMC11795548 DOI: 10.1016/j.dib.2025.111287] [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: 12/18/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
In this article, we present a brain tumor database collection comprising 23,049 samples, with each sample including four different types of MRI brain scans: FLAIR, T1, T1ce, and T2. Additionally, one or two segmentation masks (ground truth) are provided for each sample. The first mask is the raw output from the registration process and is provided for all samples, while the second mask, provided particularly for synthetic samples, is a post-processed version of the first, designed to simplify interpretation and optimize it for network training. These samples have been acquired via registration process of 438 samples available at the moment of registration from the original dataset provided by the BraTS 2022 Challenge. Registering each pair of existing brain scans results in two additional scans that retain a similar brain shape while featuring varying tumor locations. Consequently, by registering all possible pairs, a dataset originally consisting of n samples can be expanded to n2 samples. The original dataset was collected from different institutions under standard clinical conditions, but with different equipment and imaging protocols. As a result, the image quality is heterogeneous, reflecting the diversity of clinical practices across institutions. This dataset can be utilized for various tasks, such as developing fully automated segmentation algorithms for new, unseen brain tumor cases, particularly through deep learning-based approaches, since ground truth is provided for each sample.
Collapse
Affiliation(s)
- Naida Solak
- Institute for AI in Medicine (IKIM), University Hospital Essen (UKE), Ruhrgebiet, Essen, Germany
- Graz University of Technology (TU Graz), Graz, Styria, Austria
- Computer Algorithms for Medicine Laboratory (Café Lab), Graz, Styria, Austria
| | - André Ferreira
- Institute for AI in Medicine (IKIM), University Hospital Essen (UKE), Ruhrgebiet, Essen, Germany
- Center Algoritmi / LASI, University of Minho, Braga, Portugal
- Institute of Medical Informatics, University Hospital RWTH Aachen, Aachen, Germany
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Gijs Luijten
- Institute for AI in Medicine (IKIM), University Hospital Essen (UKE), Ruhrgebiet, Essen, Germany
- Graz University of Technology (TU Graz), Graz, Styria, Austria
- Computer Algorithms for Medicine Laboratory (Café Lab), Graz, Styria, Austria
- Center for Virtual and Extended Reality in Medicine, University Medicine Essen, Essen, Germany
| | - Behrus Puladi
- Institute of Medical Informatics, University Hospital RWTH Aachen, Aachen, Germany
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Victor Alves
- Center Algoritmi / LASI, University of Minho, Braga, Portugal
| | - Jan Egger
- Institute for AI in Medicine (IKIM), University Hospital Essen (UKE), Ruhrgebiet, Essen, Germany
- Graz University of Technology (TU Graz), Graz, Styria, Austria
- Computer Algorithms for Medicine Laboratory (Café Lab), Graz, Styria, Austria
- Center for Virtual and Extended Reality in Medicine, University Medicine Essen, Essen, Germany
| |
Collapse
|
14
|
Woodley MMO, Zhao Q, Goldston DB, Michael AM, Clark DB, Brown SA, Nooner KB. Adverse childhood experiences and post-traumatic stress impacts on brain connectivity and alcohol use in adolescence. Child Neuropsychol 2025:1-21. [PMID: 39819312 DOI: 10.1080/09297049.2025.2451799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
The current study investigated the relationship between adverse childhood experiences (ACEs), post-traumatic stress disorder (PTSD) symptoms, within-network resting-state functional connectivity (rs-FC), and alcohol use during adolescence using functional magnetic resonance imaging (fMRI) data from the National Consortium on Alcohol and Neurodevelopment in Adolescence study (NCANDA; N = 687). Significant rs-FC differences emerged that linked participant ACEs, PTSD symptoms, and alcohol use problems. Participants with ACEs compared to those without had diminished rs-FC within the default mode, salience, and medial frontoparietal networks (p ≤ 0.005). Further reduction in rs-FC within the default mode and medial frontoparietal networks (p ≤ 0.005) was found when PTSD symptoms were present in addition to ACEs. Findings suggest that PTSD symptoms are associated with lower within network rs-FC beyond exposure to ACEs, and some of these rs-FC changes were associated with worsened alcohol use problems (i.e. withdrawal symptoms). These findings highlight the importance of addressing PTSD symptoms in adolescents with a history of ACEs as it may mitigate problematic changes in brain connectivity and reduce the risk of developing alcohol use problems.
Collapse
Affiliation(s)
- Mary Milo O Woodley
- Department of Psychology, University of North Carolina Wilmington, Wilmington, NC, USA
| | - Qingyu Zhao
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - David B Goldston
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NY, USA
| | - Andrew M Michael
- Duke Institute for Brain Sciences, Duke University, Durham, NY, USA
| | - Duncan B Clark
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sandra A Brown
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Department of Psychology, University of California San Diego, La Jolla, CA, USA
| | - Kate B Nooner
- Department of Psychology, University of North Carolina Wilmington, Wilmington, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NY, USA
| |
Collapse
|
15
|
Knittel JJ, Hoskin JL, Hoyt DJ, Abdo JA, Foldes EL, McElvogue MM, Oliver CM, Keesler DA, Fife TD, Barranco FD, Smith KA, McComb JG, Borzage MT, King KS. Automated Detection of Normal Pressure Hydrocephalus Using CT Imaging for Calculating the Ventricle-to-Subarachnoid Volume Ratio. AJNR Am J Neuroradiol 2025; 46:141-146. [PMID: 39746816 PMCID: PMC11735426 DOI: 10.3174/ajnr.a8451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/15/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND AND PURPOSE Normal pressure hydrocephalus (NPH) is a diagnostic challenge because its clinical symptoms and imaging appearance resemble normal aging and other forms of dementia. Identifying NPH is essential so that patients can receive timely treatment to improve gait distortion and quality of life. An automated marker of NPH was developed and evaluated on clinical CT images, and its utility was assessed in a large patient cohort. MATERIALS AND METHODS A retrospective review was conducted of CT images from 306 tap test-responsive patients with NPH between January 2015 and January 2022. Control CT images were obtained from patients in the emergency department who were evaluated for headache and had unremarkable CT findings between June 2021 and August 2022. The ventricle-to-subarachnoid volume ratio (VSR) was automatically calculated by the imaging software and used as a predictor of NPH in linear regression modeling with adjustment for age and sex. The correlations of VSR with age, sex, and the receiver operating characteristic were computed. RESULTS VSR was significantly greater in patients with NPH than controls (P < .001). Importantly, VSR was not significantly correlated with age (P = .56, R2 = 0.001). VSR identifies NPH with a sensitivity and specificity of 94.1% and 92.5%, respectively, with an area under the receiver operating characteristic curve of 0.99 (95% CI 0.975-0.995). CONCLUSIONS Automated assessment of the VSR on head CT images identified probable NPH with 93% accuracy. The assessment of a large cohort of patients with NPH supports the generalizability of clinical screening of CT images. Moreover, the results support the utility of ventricle-to-sulcal concordance often used by radiologists but not currently a part of the accepted guidelines for imaging markers of NPH.
Collapse
Affiliation(s)
- Jacob J Knittel
- From the Creighton University School of Medicine (J.J.K., J.A.A., C.M.O.), Phoenix, Arizona
| | - Justin L Hoskin
- Department of Neurology (J.L.H., T.D.F.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Dylan J Hoyt
- Department of Radiology (D.J.H., D.A.K.), St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Jonathan A Abdo
- From the Creighton University School of Medicine (J.J.K., J.A.A., C.M.O.), Phoenix, Arizona
| | - Emily L Foldes
- Department of Neuroradiology (E.L.F., M.M.M., K.S.K.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Molly M McElvogue
- Department of Neuroradiology (E.L.F., M.M.M., K.S.K.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Clay M Oliver
- From the Creighton University School of Medicine (J.J.K., J.A.A., C.M.O.), Phoenix, Arizona
| | - Daniel A Keesler
- Department of Neurology (J.L.H., T.D.F.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Terry D Fife
- Department of Neurology (J.L.H., T.D.F.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - F David Barranco
- Department of Neurosurgery (F.D.B., K.A.S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Kris A Smith
- Department of Neurosurgery (F.D.B., K.A.S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - J Gordon McComb
- Department of Neurosurgery (J.G.M.), Children's Hospital of Los Angeles, Los Angeles, California
| | - Matthew T Borzage
- Division of Neonatology (M.T.B.), Children's Hospital of Los Angeles, Los Angeles, California
| | - Kevin S King
- Department of Neuroradiology (E.L.F., M.M.M., K.S.K.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| |
Collapse
|
16
|
Wang L, Sun Y, Seidlitz J, Bethlehem RAI, Alexander-Bloch A, Dorfschmidt L, Li G, Elison JT, Lin W, Wang L. A lifespan-generalizable skull-stripping model for magnetic resonance images that leverages prior knowledge from brain atlases. Nat Biomed Eng 2025:10.1038/s41551-024-01337-w. [PMID: 39779813 DOI: 10.1038/s41551-024-01337-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
In magnetic resonance imaging of the brain, an imaging-preprocessing step removes the skull and other non-brain tissue from the images. But methods for such a skull-stripping process often struggle with large data heterogeneity across medical sites and with dynamic changes in tissue contrast across lifespans. Here we report a skull-stripping model for magnetic resonance images that generalizes across lifespans by leveraging personalized priors from brain atlases. The model consists of a brain extraction module that provides an initial estimation of the brain tissue on an image, and a registration module that derives a personalized prior from an age-specific atlas. The model is substantially more accurate than state-of-the-art skull-stripping methods, as we show with a large and diverse dataset of 21,334 lifespans acquired from 18 sites with various imaging protocols and scanners, and it generates naturally consistent and seamless lifespan changes in brain volume, faithfully charting the underlying biological processes of brain development and ageing.
Collapse
Affiliation(s)
- Limei Wang
- Developing Brain Computing Lab, Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| | - Yue Sun
- Developing Brain Computing Lab, Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| | - Jakob Seidlitz
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| | | | - Aaron Alexander-Bloch
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| | - Lena Dorfschmidt
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| | - Gang Li
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jed T Elison
- Institute of Child Development, University of Minnesota, Minneapolis, MN, USA
| | - Weili Lin
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Li Wang
- Developing Brain Computing Lab, Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
17
|
Griessmair M, Schramm S, Ziegenfeuter J, Canisius J, Jung K, Delbridge C, Schmidt-Graf F, Mitsdoerffer M, Zimmer C, Meyer B, Metz MC, Wiestler B. Advanced imaging reveals enhanced malignancy in glioblastomas involving the subventricular zone: evidence of increased infiltrative growth and perfusion. J Neurooncol 2025; 171:343-350. [PMID: 39387957 PMCID: PMC11695386 DOI: 10.1007/s11060-024-04849-2] [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: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Glioblastoma's infiltrative growth and heterogeneity are influenced by neural, molecular, genetic, and immunological factors, with the precise origin of these tumors remaining elusive. Neurogenic zones might serve as the tumor stem cells' nest, with tumors in contact with these zones exhibiting worse outcomes and more aggressive growth patterns. This study aimed to determine if these characteristics are reflected in advanced imaging, specifically diffusion and perfusion data. METHODS In this monocentric retrospective study, 137 glioblastoma therapy-naive patients (IDH-wildtype, grade 4) with advanced preoperative MRI, including perfusion and diffusion imaging, were analyzed. Tumors and neurogenic zones were automatically segmented. Advanced imaging metrics, including cerebral blood volume (CBV) from perfusion imaging, tissue volume mask (TVM), and free water corrected fractional anisotropy (FA-FWE) from diffusion imaging, were extracted. RESULTS SVZ infiltration positively correlated with CBV, indicating higher perfusion in tumors. Significant CBV differences were noted between high and low SVZ infiltration cases at specific percentiles. Negative correlation was observed with TVM and positive correlation with FA-FWE, suggesting more infiltrative tumor growth. Significant differences in TVM and FA-FWE values were found between high and low SVZ infiltration cases. DISCUSSION Glioblastomas with SVZ infiltration exhibit distinct imaging characteristics, including higher perfusion and lower cell density per voxel, indicating a more infiltrative growth and higher vascularization. Stem cell-like characteristics in SVZ-infiltrating cells could explain the increased infiltration and aggressive behavior. Understanding these imaging and biological correlations could enhance the understanding of glioblastoma evolution.
Collapse
Affiliation(s)
- Michael Griessmair
- Dept. of Neuroradiology, Klinikum Rechts der Isar, TU Munich, Ismaningerstr. 22, 81675, Munich, Germany.
| | - Severin Schramm
- Dept. of Neuroradiology, Klinikum Rechts der Isar, TU Munich, Ismaningerstr. 22, 81675, Munich, Germany
| | - Julian Ziegenfeuter
- Dept. of Neuroradiology, Klinikum Rechts der Isar, TU Munich, Ismaningerstr. 22, 81675, Munich, Germany
| | - Julian Canisius
- Dept. of Neuroradiology, Klinikum Rechts der Isar, TU Munich, Ismaningerstr. 22, 81675, Munich, Germany
| | - Kirsten Jung
- Dept. of Neuroradiology, Klinikum Rechts der Isar, TU Munich, Ismaningerstr. 22, 81675, Munich, Germany
| | | | | | - Meike Mitsdoerffer
- Dept. of Neurology, Klinikum Rechts der Isar, TU Munich, 81675, Munich, Germany
| | - Claus Zimmer
- Dept. of Neuroradiology, Klinikum Rechts der Isar, TU Munich, Ismaningerstr. 22, 81675, Munich, Germany
| | - Bernhard Meyer
- Dept. of Neurosurgery, Klinikum Rechts der Isar, TU Munich, 81675, Munich, Germany
| | - Marie-Christin Metz
- Dept. of Neuroradiology, Klinikum Rechts der Isar, TU Munich, Ismaningerstr. 22, 81675, Munich, Germany
| | - Benedikt Wiestler
- Dept. of Neuroradiology, Klinikum Rechts der Isar, TU Munich, Ismaningerstr. 22, 81675, Munich, Germany
- TranslaTUM, TU Munich, 81675, Munich, Germany
| |
Collapse
|
18
|
Moawad AW, Janas A, Baid U, Ramakrishnan D, Saluja R, Ashraf N, Maleki N, Jekel L, Yordanov N, Fehringer P, Gkampenis A, Amiruddin R, Manteghinejad A, Adewole M, Albrecht J, Anazodo U, Aneja S, Anwar SM, Bergquist T, Chiang V, Chung V, Conte GM, Dako F, Eddy J, Ezhov I, Khalili N, Farahani K, Iglesias JE, Jiang Z, Johanson E, Kazerooni AF, Kofler F, Krantchev K, LaBella D, Van Leemput K, Li HB, Linguraru MG, Liu X, Meier Z, Menze BH, Moy H, Osenberg K, Piraud M, Reitman Z, Shinohara RT, Wang C, Wiestler B, Wiggins W, Shafique U, Willms K, Avesta A, Bousabarah K, Chakrabarty S, Gennaro N, Holler W, Kaur M, LaMontagne P, Lin M, Lost J, Marcus DS, Maresca R, Merkaj S, Cassinelli Pedersen G, von Reppert M, Sotiras A, Teytelboym O, Tillmans N, Westerhoff M, Youssef A, Godfrey D, Floyd S, Rauschecker A, Villanueva-Meyer J, Pflüger I, Cho J, Bendszus M, Brugnara G, Cramer J, Perez-Carillo GJG, Johnson DR, Kam A, Kwan BYM, Lai L, Lall NU, Memon F, Krycia M, Patro SN, Petrovic B, So TY, Thompson G, Wu L, Schrickel EB, Bansal A, Barkhof F, Besada C, Chu S, Druzgal J, Dusoi A, Farage L, Feltrin F, et alMoawad AW, Janas A, Baid U, Ramakrishnan D, Saluja R, Ashraf N, Maleki N, Jekel L, Yordanov N, Fehringer P, Gkampenis A, Amiruddin R, Manteghinejad A, Adewole M, Albrecht J, Anazodo U, Aneja S, Anwar SM, Bergquist T, Chiang V, Chung V, Conte GM, Dako F, Eddy J, Ezhov I, Khalili N, Farahani K, Iglesias JE, Jiang Z, Johanson E, Kazerooni AF, Kofler F, Krantchev K, LaBella D, Van Leemput K, Li HB, Linguraru MG, Liu X, Meier Z, Menze BH, Moy H, Osenberg K, Piraud M, Reitman Z, Shinohara RT, Wang C, Wiestler B, Wiggins W, Shafique U, Willms K, Avesta A, Bousabarah K, Chakrabarty S, Gennaro N, Holler W, Kaur M, LaMontagne P, Lin M, Lost J, Marcus DS, Maresca R, Merkaj S, Cassinelli Pedersen G, von Reppert M, Sotiras A, Teytelboym O, Tillmans N, Westerhoff M, Youssef A, Godfrey D, Floyd S, Rauschecker A, Villanueva-Meyer J, Pflüger I, Cho J, Bendszus M, Brugnara G, Cramer J, Perez-Carillo GJG, Johnson DR, Kam A, Kwan BYM, Lai L, Lall NU, Memon F, Krycia M, Patro SN, Petrovic B, So TY, Thompson G, Wu L, Schrickel EB, Bansal A, Barkhof F, Besada C, Chu S, Druzgal J, Dusoi A, Farage L, Feltrin F, Fong A, Fung SH, Gray RI, Ikuta I, Iv M, Postma AA, Mahajan A, Joyner D, Krumpelman C, Letourneau-Guillon L, Lincoln CM, Maros ME, Miller E, Morón FEA, Nimchinsky EA, Ozsarlak O, Patel U, Rohatgi S, Saha A, Sayah A, Schwartz ED, Shih R, Shiroishi MS, Small JE, Tanwar M, Valerie J, Weinberg BD, White ML, Young R, Zohrabian VM, Azizova A, Brüßeler MMT, Ghonim M, Ghonim M, Okar A, Pasquini L, Sharifi Y, Singh G, Sollmann N, Soumala T, Taherzadeh M, Vollmuth P, Foltyn-Dumitru M, Malhotra A, Abayazeed AH, Dellepiane F, Lohmann P, Pérez-García VM, Elhalawani H, de Verdier MC, Al-Rubaiey S, Armindo RD, Ashraf K, Asla MM, Badawy M, Bisschop J, Lomer NB, Bukatz J, Chen J, Cimflova P, Corr F, Crawley A, Deptula L, Elakhdar T, Shawali IH, Faghani S, Frick A, Gulati V, Haider MA, Hierro F, Dahl RH, Jacobs SM, Hsieh KCJ, Kandemirli SG, Kersting K, Kida L, Kollia S, Koukoulithras I, Li X, Abouelatta A, Mansour A, Maria-Zamfirescu RC, Marsiglia M, Mateo-Camacho YS, McArthur M, McDonnell O, McHugh M, Moassefi M, Morsi SM, Munteanu A, Nandolia KK, Naqvi SR, Nikanpour Y, Alnoury M, Nouh AMA, Pappafava F, Patel MD, Petrucci S, Rawie E, Raymond S, Roohani B, Sabouhi S, Sanchez-Garcia LM, Shaked Z, Suthar PP, Altes T, Isufi E, Dhemesh Y, Gass J, Thacker J, Tarabishy AR, Turner B, Vacca S, Vilanilam GK, Warren D, Weiss D, Worede F, Yousry S, Lerebo W, Aristizabal A, Karargyris A, Kassem H, Pati S, Sheller M, Link KEE, Calabrese E, Tahon NH, Nada A, Velichko YS, Bakas S, Rudie JD, Aboian M. The Brain Tumor Segmentation - Metastases (BraTS-METS) Challenge 2023: Brain Metastasis Segmentation on Pre-treatment MRI. ARXIV 2024:arXiv:2306.00838v3. [PMID: 37396600 PMCID: PMC10312806] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The translation of AI-generated brain metastases (BM) segmentation into clinical practice relies heavily on diverse, high-quality annotated medical imaging datasets. The BraTS-METS 2023 challenge has gained momentum for testing and benchmarking algorithms using rigorously annotated internationally compiled real-world datasets. This study presents the results of the segmentation challenge and characterizes the challenging cases that impacted the performance of the winning algorithms. Untreated brain metastases on standard anatomic MRI sequences (T1, T2, FLAIR, T1PG) from eight contributed international datasets were annotated in stepwise method: published UNET algorithms, student, neuroradiologist, final approver neuroradiologist. Segmentations were ranked based on lesion-wise Dice and Hausdorff distance (HD95) scores. False positives (FP) and false negatives (FN) were rigorously penalized, receiving a score of 0 for Dice and a fixed penalty of 374 for HD95. The mean scores for the teams were calculated. Eight datasets comprising 1303 studies were annotated, with 402 studies (3076 lesions) released on Synapse as publicly available datasets to challenge competitors. Additionally, 31 studies (139 lesions) were held out for validation, and 59 studies (218 lesions) were used for testing. Segmentation accuracy was measured as rank across subjects, with the winning team achieving a LesionWise mean score of 7.9. The Dice score for the winning team was 0.65 ± 0.25. Common errors among the leading teams included false negatives for small lesions and misregistration of masks in space. The Dice scores and lesion detection rates of all algorithms diminished with decreasing tumor size, particularly for tumors smaller than 100 mm3. In conclusion, algorithms for BM segmentation require further refinement to balance high sensitivity in lesion detection with the minimization of false positives and negatives. The BraTS-METS 2023 challenge successfully curated well-annotated, diverse datasets and identified common errors, facilitating the translation of BM segmentation across varied clinical environments and providing personalized volumetric reports to patients undergoing BM treatment.
Collapse
Affiliation(s)
| | - Anastasia Janas
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Ujjwal Baid
- Division of Computational Pathology, Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Divya Ramakrishnan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Rachit Saluja
- Department of Electrical and Computer Engineering, Cornell University and Cornell Tech, New York, NY, USA
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Nader Ashraf
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Nazanin Maleki
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Leon Jekel
- DKFZ Division of Translational Neurooncology at the WTZ, German Cancer Consortium, DKTK Partner Site, University Hospital Essen, Essen, Germany
| | - Nikolay Yordanov
- Faculty of Medicine, Medical University - Sofia, Sofia, Bulgaria
| | - Pascal Fehringer
- Faculty of Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | | | - Raisa Amiruddin
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Maruf Adewole
- Medical Artificial Intelligence Lab, Crestview Radiology, Lagos, Nigeria
| | | | - Udunna Anazodo
- Medical Artificial Intelligence Lab, Crestview Radiology, Lagos, Nigeria
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Sanjay Aneja
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Syed Muhammad Anwar
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, D.C., USA
| | | | - Veronica Chiang
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | | | | | - Farouk Dako
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | | | - Ivan Ezhov
- Department of Informatics, Technical University Munich, Germany
| | - Nastaran Khalili
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Keyvan Farahani
- Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Juan Eugenio Iglesias
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Zhifan Jiang
- Children’s National Hospital, Washington, D.C., USA
| | - Elaine Johanson
- PrecisionFDA, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Florian Kofler
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
| | - Kiril Krantchev
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Dominic LaBella
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Koen Van Leemput
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Denmark
| | - Hongwei Bran Li
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Marius George Linguraru
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, D.C., USA
- Departments of Radiology and Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, D.C., USA
| | - Xinyang Liu
- Children’s National Hospital, Washington, D.C., USA
| | | | - Bjoern H Menze
- Biomedical Image Analysis & Machine Learning, Department of Quantitative Biomedicine, University of Zurich, Switzerland
| | - Harrison Moy
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Klara Osenberg
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | | | - Zachary Reitman
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Russell Takeshi Shinohara
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | - Chunhao Wang
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Benedikt Wiestler
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
| | | | - Umber Shafique
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA
| | - Klara Willms
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Arman Avesta
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Radiology, Neuroradiology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Satrajit Chakrabarty
- Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO, USA
- GE HealthCare, San Ramon, CA, USA
| | - Nicolo Gennaro
- Department of Radiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | | | | | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Jan Lost
- Department of Neurosurgery, Heinrich-Heine University, Moorenstrasse 5, Dusseldorf, Germany
| | - Daniel S. Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ryan Maresca
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | | | | | | | - Aristeidis Sotiras
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Niklas Tillmans
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Dusseldorf, Germany
| | | | | | - Devon Godfrey
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Scott Floyd
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Andreas Rauschecker
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA, USA
| | - Javier Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA, USA
| | - Irada Pflüger
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jaeyoung Cho
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Gianluca Brugnara
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Justin Cramer
- Department of Radiology, Mayo Clinic, Phoenix, AZ, USA
| | | | | | - Anthony Kam
- Loyola University Medical Center, Hines, IL, USA
| | | | - Lillian Lai
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | | | - Fatima Memon
- Carolina Radiology Associates, Myrtle Beach, SC, USA
- McLeod Regional Medical Center, Florence, SC, USA
- Medical University of South Carolina, Charleston, SC, USA
| | - Mark Krycia
- Carolina Radiology Associates, Myrtle Beach, SC, USA
| | | | | | - Tiffany Y. So
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Gerard Thompson
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Department of Clinical Neurosciences, NHS Lothian, Edinburgh, United Kingdom
| | - Lei Wu
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - E. Brooke Schrickel
- Department of Radiology, Ohio State University College of Medicine, Columbus, OH, USA
| | - Anu Bansal
- Albert Einstein Medical Center, Hartford, CT, USA
| | - Frederik Barkhof
- Amsterdam UMC, location Vrije Universiteir, Netherlands
- University College London, United Kingdom
| | | | - Sammy Chu
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Jason Druzgal
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA
| | | | - Luciano Farage
- Centro Universitario Euro-Americana (UNIEURO), Brasília, DF, Brazil
| | - Fabricio Feltrin
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amy Fong
- Southern District Health Board, Dunedin, New Zealand
| | - Steve H. Fung
- Department of Radiology, Houston Methodist, Houston, TX, USA
| | - R. Ian Gray
- University of Tennessee Medical Center, Knoxville, TN, USA
| | - Ichiro Ikuta
- Department of Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Michael Iv
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Alida A. Postma
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands
| | - Amit Mahajan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - David Joyner
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA
| | - Chase Krumpelman
- Department of Radiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | | | - Christie M Lincoln
- Department of Neuroradiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Mate E. Maros
- Departments of Neuroradiology & Biomedical Informatics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Elka Miller
- Department of Diagnostic and Interventional Radiology, SickKids Hospital, University of Toronto, Canada
| | | | | | - Ozkan Ozsarlak
- Department of Radiology, AZ Monica, Antwerp Area, Belgium
| | - Uresh Patel
- Medicolegal Imaging Experts LLC, Mercer Island, WA, USA
| | - Saurabh Rohatgi
- Department of Radiology, Neuroradiology, Massachusetts General Hospital, Boston, MA, USA
| | - Atin Saha
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Anousheh Sayah
- MedStar Georgetown University Hospital, Washington, D.C., USA
| | - Eric D. Schwartz
- Department of Radiology, St.Elizabeth’s Medical Center, Boston, MA, USA
- Department of Radiology, Tufts University School of Medicine, Boston, MA, USA
| | - Robert Shih
- Walter Reed National Military Medical Center, Bethesda, MD, USA
| | | | - Juan E. Small
- Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Manoj Tanwar
- Department of Radiology, University of Alabama, Birmingham, AL, USA
| | - Jewels Valerie
- Department of Radiology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Brent D. Weinberg
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | | | - Robert Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vahe M. Zohrabian
- Northwell Health, Zucker Hofstra School of Medicine at Northwell, North Shore University Hospital, Hempstead, New York, NY, USA
| | - Aynur Azizova
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | | | - Mohanad Ghonim
- Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Mohamed Ghonim
- Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Abdullah Okar
- University of Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Luca Pasquini
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yasaman Sharifi
- Department of Radiology, Iran University of Medical Sciences, Tehran, Iran
| | - Gagandeep Singh
- Columbia University Irving Medical Center, New York, NY, USA
| | - Nico Sollmann
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | | | - Philipp Vollmuth
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
- Department of Medical Image Computing, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Ajay Malhotra
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | | | - Francesco Dellepiane
- Functional and Interventional Neuroradiology Unit, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-4), Research Center Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Víctor M. Pérez-García
- Mathematical Oncology Laboratory & Department of Mathematics, University of Castilla-La Mancha, Spain
| | - Hesham Elhalawani
- Department of Radiation Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Correia de Verdier
- Department of Surgical Sciences, Section of Neuroradiology, Uppsala University, Sweden
- Department of Radiology, University of California San Diego, CA, USA
| | - Sanaria Al-Rubaiey
- Charité-Universitätsmedizin Berlin (Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Berlin, Germany
| | - Rui Duarte Armindo
- Department of Neuroradiology, Western Lisbon Hospital Centre (CHLO), Portugal
| | | | | | - Mohamed Badawy
- Diagnostic Radiology Department, Wayne State University, Detroit, MI
| | - Jeroen Bisschop
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Jan Bukatz
- Charité-Universitätsmedizin Berlin (Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Berlin, Germany
| | - Jim Chen
- Department of Radiology/Division of Neuroradiology, San Diego Veterans Administration Medical Center/UC San Diego Health System, San Diego, CA, USA
| | - Petra Cimflova
- Department of Radiology, University of Calgary, Calgary, Canada
| | - Felix Corr
- EDU Institute of Higher Education, Villa Bighi, Chaplain’s House, Kalkara, Malta
| | | | - Lisa Deptula
- Ross University School of Medicine, Bridgetown, Barbados
| | | | | | | | - Alexandra Frick
- Department of Neurosurgery, Vivantes Klinikum Neukölln, Berlin, Germany
| | | | | | - Fátima Hierro
- Neuroradiology Department, Pedro Hispano Hospital, Matosinhos, Portugal
| | - Rasmus Holmboe Dahl
- Department of Radiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | | | | | - Sedat G. Kandemirli
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Katharina Kersting
- Charité-Universitätsmedizin Berlin (Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Berlin, Germany
| | - Laura Kida
- Charité-Universitätsmedizin Berlin (Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Berlin, Germany
| | - Sofia Kollia
- National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | | | - Xiao Li
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | | | | | - Ruxandra-Catrinel Maria-Zamfirescu
- Charité-Universitätsmedizin Berlin (Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Berlin, Germany
| | - Marcela Marsiglia
- Department of Radiology, Brigham and Women’s Hospital, Massachusetts General Hospital, Boston, MA, USA
| | | | - Mark McArthur
- Department of Radiological Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Maire McHugh
- Department of Radiology Manchester NHS Foundation Trust, North West School of Radiology, Manchester, United Kingdom
| | - Mana Moassefi
- Artificial Intelligence Lab, Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Khanak K. Nandolia
- Department of Radiodiagnosis, All India Institute of Medical Sciences Rishikesh, India
| | - Syed Raza Naqvi
- Windsor Regional Hospital, Western University, Ontario, Canada
| | - Yalda Nikanpour
- Artificial Intelligence & Informatics, Mayo Clinic, Rochester, MN, USA
| | - Mostafa Alnoury
- Department of Radiology, University of Pennsylvania, PA, USA
| | | | - Francesca Pappafava
- Department of Medicine and Surgery, Università degli Studi di Perugia, Italy
| | - Markand D. Patel
- Department of Neuroradiology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Samantha Petrucci
- Department of Radiology and Biomedical Imaging, University of California San Francisco, CA, USA
| | - Eric Rawie
- Department of Radiology, Michigan Medicine, Ann Arbor, MI, USA
| | - Scott Raymond
- Department of Radiology, University of Vermont Medical Center, Burlington, VT, USA
| | - Borna Roohani
- University of Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sadeq Sabouhi
- Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Zoe Shaked
- Charité-Universitätsmedizin Berlin (Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Berlin, Germany
| | | | - Talissa Altes
- Radiology Department, University of Missouri, Columbia, MO, USA
| | - Edvin Isufi
- Radiology Department, University of Missouri, Columbia, MO, USA
| | - Yaseen Dhemesh
- Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jaime Gass
- Radiology Department, University of Missouri, Columbia, MO, USA
| | | | - Abdul Rahman Tarabishy
- Department of NeuroRadiology, Rockefeller Neuroscience Institute, West Virginia University. Morgantown, WV, USA
| | | | - Sebastiano Vacca
- University of Cagliari, School of Medicine and Surgery, Cagliari, Italy
| | | | - Daniel Warren
- Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - David Weiss
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Fikadu Worede
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Wondwossen Lerebo
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | - Sarthak Pati
- Division of Computational Pathology, Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN, USA
- MLCommons, San Francisco, CA, USA
- Center For Federated Learning in Medicine, Indiana University, Indianapolis, IN, USA
| | - Micah Sheller
- MLCommons, San Francisco, CA, USA
- Intel Corporation, Hillsboro, OR, USA
| | | | - Evan Calabrese
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | | | - Ayman Nada
- Radiology Department, University of Missouri, Columbia, MO, USA
| | - Yuri S. Velichko
- Department of Radiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Spyridon Bakas
- Division of Computational Pathology, Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA
- Department of Neurological Surgery, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Jeffrey D. Rudie
- Department of Radiology, University of California San Diego, CA, USA
- Department of Radiology, Scripps Clinic Medical Group, CA, USA
| | - Mariam Aboian
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
19
|
Morales AM, Jones SA, Carlson B, Kliamovich D, Dehoney J, Simpson BL, Dominguez-Savage KA, Hernandez KO, Lopez DA, Baker FC, Clark DB, Goldston DB, Luna B, Nooner KB, Muller-Oehring EM, Tapert SF, Thompson WK, Nagel BJ. Associations between mesolimbic connectivity, and alcohol use from adolescence to adulthood. Dev Cogn Neurosci 2024; 70:101478. [PMID: 39577156 PMCID: PMC11617707 DOI: 10.1016/j.dcn.2024.101478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/18/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024] Open
Abstract
Dopaminergic projections from the ventral tegmental area (VTA) to limbic regions play a key role in the initiation and maintenance of substance use; however, the relationship between mesolimbic resting-state functional connectivity (RSFC) and alcohol use during development remains unclear. We examined the associations between alcohol use and VTA RSFC to subcortical structures in 796 participants (12-21 years old at baseline, 51 % female) across 9 waves of longitudinal data from the National Consortium on Alcohol and Neurodevelopment in Adolescence. Linear mixed effects models included interactions between age, sex, and alcohol use, and best fitting models were selected using log-likelihood ratio tests. Results demonstrated a positive association between alcohol use and VTA RSFC to the nucleus accumbens. Age was associated with VTA RSFC to the amygdala and hippocampus, and an age-by-alcohol use interaction on VTA-globus pallidus connectivity was driven by a positive association between alcohol and VTA-globus pallidus RSFC in adolescence, but not adulthood. On average, male participants exhibited greater VTA RSFC to the amygdala, nucleus accumbens, caudate, hippocampus, globus pallidus, and thalamus. Differences in VTA RSFC related to age, sex, and alcohol, may inform our understanding of neurobiological risk and resilience for alcohol use and other psychiatric disorders.
Collapse
Affiliation(s)
- Angelica M Morales
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States.
| | - Scott A Jones
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States
| | - Birgitta Carlson
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States
| | - Dakota Kliamovich
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States
| | - Joseph Dehoney
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Brooke L Simpson
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States
| | | | - Kristina O Hernandez
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States
| | - Daniel A Lopez
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States
| | - Fiona C Baker
- Center for Health Sciences, SRI International, Menlo Park, CA, United States
| | - Duncan B Clark
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - David B Goldston
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, United States
| | - Beatriz Luna
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kate B Nooner
- Department of Psychology, University of North Carolina Wilmington, Wilmington, NC, United States
| | - Eva M Muller-Oehring
- Center for Health Sciences, SRI International, Menlo Park, CA, United States; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Susan F Tapert
- Department of Psychiatry, University of California, San Diego, CA, United States
| | | | - Bonnie J Nagel
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
20
|
Li HB, Conte GM, Hu Q, Anwar SM, Kofler F, Ezhov I, van Leemput K, Piraud M, Diaz M, Cole B, Calabrese E, Rudie J, Meissen F, Adewole M, Janas A, Kazerooni AF, LaBella D, Moawad AW, Farahani K, Eddy J, Bergquist T, Chung V, Shinohara RT, Dako F, Wiggins W, Reitman Z, Wang C, Liu X, Jiang Z, Familiar A, Johanson E, Meier Z, Davatzikos C, Freymann J, Kirby J, Bilello M, Fathallah-Shaykh HM, Wiest R, Kirschke J, Colen RR, Kotrotsou A, Lamontagne P, Marcus D, Milchenko M, Nazeri A, Weber MA, Mahajan A, Mohan S, Mongan J, Hess C, Cha S, Villanueva-Meyer J, Colak E, Crivellaro P, Jakab A, Albrecht J, Anazodo U, Aboian M, Yu T, Chung V, Bergquist T, Eddy J, Albrecht J, Baid U, Bakas S, Linguraru MG, Menze B, Iglesias JE, Wiestler B. The Brain Tumor Segmentation (BraTS) Challenge 2023: Brain MR Image Synthesis for Tumor Segmentation (BraSyn). ARXIV 2024:arXiv:2305.09011v6. [PMID: 37608932 PMCID: PMC10441440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Automated brain tumor segmentation methods have become well-established and reached performance levels offering clear clinical utility. These methods typically rely on four input magnetic resonance imaging (MRI) modalities: T1-weighted images with and without contrast enhancement, T2-weighted images, and FLAIR images. However, some sequences are often missing in clinical practice due to time constraints or image artifacts, such as patient motion. Consequently, the ability to substitute missing modalities and gain segmentation performance is highly desirable and necessary for the broader adoption of these algorithms in the clinical routine. In this work, we present the establishment of the Brain MR Image Synthesis Benchmark (BraSyn) in conjunction with the Medical Image Computing and Computer-Assisted Intervention (MICCAI) 2023. The primary objective of this challenge is to evaluate image synthesis methods that can realistically generate missing MRI modalities when multiple available images are provided. The ultimate aim is to facilitate automated brain tumor segmentation pipelines. The image dataset used in the benchmark is diverse and multi-modal, created through collaboration with various hospitals and research institutions.
Collapse
Affiliation(s)
- Hongwei Bran Li
- University of Zurich, Switzerland
- Department of Informatics, Technical University Munich, Germany
- Klinikum rechts der Isar, Technical University of Munich, Germany
| | | | - Qingqiao Hu
- University of Zurich, Switzerland
- Department of Informatics, Technical University Munich, Germany
- Klinikum rechts der Isar, Technical University of Munich, Germany
- Mayo Clinic, Rochester, USA
- Helmholtz AI, Helmholtz Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Germany
- Children's National Hospital, Washington DC, USA and George Washington University, USA
- Finnish Center for Artificial Intelligence, Finland
- Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, USA
- Medical Artificial Intelligence (MAI) Lab, Crestview Radiology, Lagos, Nigeria
- Children's National Hospital, Washington DC, USA
- George Washington University, Washington DC, USA
- Center for AI and Data Science for Integrated Diagnostics (AI2D) I& Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Yale University, New Haven, CT, USA
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
- Duke University Medical Center, Department of Radiation Oncology, USA
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
- Mercy Catholic Medical Center, Darby, PA, USA
- Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
- Sage Bionetworks, USA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, USA
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Duke University Medical Center, Department of Radiology, USA
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Denmark
- Montreal Neurological Institute (MNI), McGill University, Montreal, QC, Canada
- PrecisionFDA, U.S. Food and Drug Administration, Silver Spring, MD, USA
- Booz Allen Hamilton, McLean, VA, USA
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
- Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, USA
- Institute for Surgical Technology and Biomechanics, University of Bern, Bern, Switzerland
- Support Centre for Advanced Neuroimaging Inselspital, Institute for Diagnostic and Interventional Neuroradiology, Bern University Hospital, Bern, Switzerland
- University of Pittsburgh Medical Center, PA, USA
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Psychology, Washington University, St. Louis, MO, USA
- Neuroimaging Informatics and Analysis Center, Washington University, St. Louis, MO, USA
- Department of Radiology, Washington University, St. Louis, MO, USA
- Institute of Diagnostic and Interventional Radiology, Pediatric Radiology and Neuroradiology, University Medical Center Rostock, Ernst-Heydemann-Str. 6, 18057 Rostock, Germany
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
- University of California San Francisco, CA, USA
- University of Toronto, Toronto, ON, Canada
- University of Debrecen, Hungary Stony Brook University, NY, USA
| | - Syed Muhammad Anwar
- Children's National Hospital, Washington DC, USA
- George Washington University, Washington DC, USA
| | - Florian Kofler
- Helmholtz AI, Helmholtz Munich, Germany
- Department of Informatics, Technical University Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Ivan Ezhov
- Department of Informatics, Technical University Munich, Germany
| | | | | | | | | | - Evan Calabrese
- Duke University Medical Center, Department of Radiology, USA
- University of California San Francisco, CA, USA
| | - Jeff Rudie
- University of California San Francisco, CA, USA
| | - Felix Meissen
- Department of Informatics, Technical University Munich, Germany
| | - Maruf Adewole
- Medical Artificial Intelligence (MAI) Lab, Crestview Radiology, Lagos, Nigeria
| | | | - Anahita Fathi Kazerooni
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
- Center for AI and Data Science for Integrated Diagnostics (AI2D) I& Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Dominic LaBella
- Duke University Medical Center, Department of Radiation Oncology, USA
| | | | - Keyvan Farahani
- Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | | | | | | | - Russell Takeshi Shinohara
- Center for AI and Data Science for Integrated Diagnostics (AI2D) I& Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, USA
| | - Farouk Dako
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Walter Wiggins
- Duke University Medical Center, Department of Radiology, USA
| | - Zachary Reitman
- Duke University Medical Center, Department of Radiation Oncology, USA
| | - Chunhao Wang
- Duke University Medical Center, Department of Radiation Oncology, USA
| | - Xinyang Liu
- Children's National Hospital, Washington DC, USA
- George Washington University, Washington DC, USA
| | - Zhifan Jiang
- Children's National Hospital, Washington DC, USA
- George Washington University, Washington DC, USA
| | - Ariana Familiar
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Elaine Johanson
- PrecisionFDA, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | | | - Christos Davatzikos
- Center for AI and Data Science for Integrated Diagnostics (AI2D) I& Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Freymann
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
- Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Justin Kirby
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
- Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Michel Bilello
- Center for AI and Data Science for Integrated Diagnostics (AI2D) I& Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Roland Wiest
- Institute for Surgical Technology and Biomechanics, University of Bern, Bern, Switzerland
- Support Centre for Advanced Neuroimaging Inselspital, Institute for Diagnostic and Interventional Neuroradiology, Bern University Hospital, Bern, Switzerland
| | - Jan Kirschke
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
| | - Rivka R Colen
- University of Pittsburgh Medical Center, PA, USA
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aikaterini Kotrotsou
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Daniel Marcus
- Neuroimaging Informatics and Analysis Center, Washington University, St. Louis, MO, USA
- Department of Radiology, Washington University, St. Louis, MO, USA
| | - Mikhail Milchenko
- Neuroimaging Informatics and Analysis Center, Washington University, St. Louis, MO, USA
- Department of Radiology, Washington University, St. Louis, MO, USA
| | - Arash Nazeri
- Department of Radiology, Washington University, St. Louis, MO, USA
| | - Marc-André Weber
- Institute of Diagnostic and Interventional Radiology, Pediatric Radiology and Neuroradiology, University Medical Center Rostock, Ernst-Heydemann-Str. 6, 18057 Rostock, Germany
| | - Abhishek Mahajan
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Suyash Mohan
- Center for AI and Data Science for Integrated Diagnostics (AI2D) I& Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Mongan
- University of California San Francisco, CA, USA
| | | | - Soonmee Cha
- University of California San Francisco, CA, USA
| | | | | | | | - Andras Jakab
- University of Debrecen, Hungary Stony Brook University, NY, USA
| | | | - Udunna Anazodo
- Montreal Neurological Institute (MNI), McGill University, Montreal, QC, Canada
| | | | - Thomas Yu
- Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, USA
| | | | | | | | | | - Ujjwal Baid
- Center for AI and Data Science for Integrated Diagnostics (AI2D) I& Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Spyridon Bakas
- Center for AI and Data Science for Integrated Diagnostics (AI2D) I& Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
21
|
Hussain MA, LaMay D, Grant E, Ou Y. Deep learning of structural MRI predicts fluid, crystallized, and general intelligence. Sci Rep 2024; 14:27935. [PMID: 39537706 PMCID: PMC11561325 DOI: 10.1038/s41598-024-78157-0] [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: 03/10/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Can brain structure predict human intelligence? T1-weighted structural brain magnetic resonance images (sMRI) have been correlated with intelligence. However, the population-level association does not fully account for individual variability in intelligence. To address this, studies have emerged recently to predict individual subject's intelligence or neurocognitive scores. However, they are mostly on predicting fluid intelligence (the ability to solve new problems). Studies are lacking to predict crystallized intelligence (the ability to accumulate knowledge) or general intelligence (fluid and crystallized intelligence combined). This study tests whether deep learning of sMRI can predict an individual subject's verbal, comprehensive, and full-scale intelligence quotients (VIQ, PIQ, and FSIQ), which reflect fluid and crystallized intelligence. We performed a comprehensive set of 432 experiments, using different input image channels, six deep learning models, and two outcome settings, in 850 healthy and autistic subjects 6-64 years of age. Our findings indicate a statistically significant potential of T1-weighted sMRI in predicting intelligence, with a Pearson correlation exceeding 0.21 (p < 0.001). Interestingly, we observed that an increase in the complexity of deep learning models does not necessarily translate to higher accuracy in intelligence prediction. The interpretations of our 2D and 3D CNNs, based on GradCAM, align well with the Parieto-Frontal Integration Theory (P-FIT), reinforcing the theory's suggestion that human intelligence is a result of interactions among various brain regions, including the occipital, temporal, parietal, and frontal lobes. These promising results invite further studies and open new questions in the field.
Collapse
Affiliation(s)
- Mohammad Arafat Hussain
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, 401 Park Drive, Boston, MA, 02115, USA
| | - Danielle LaMay
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, 401 Park Drive, Boston, MA, 02115, USA
- Khoury College of Computer and Information Science, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Ellen Grant
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, 401 Park Drive, Boston, MA, 02115, USA
- Department of Radiology, Harvard Medical School, 401 Park Drive, Boston, MA, 02115, USA
| | - Yangming Ou
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, 401 Park Drive, Boston, MA, 02115, USA.
- Department of Radiology, Harvard Medical School, 401 Park Drive, Boston, MA, 02115, USA.
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, 401 Park Drive, Boston, MA, 02115, USA.
| |
Collapse
|
22
|
Sun X, Li S, Ma C, Fang W, Jing X, Yang C, Li H, Zhang X, Ge C, Liu B, Li Z. Glioma subtype prediction based on radiomics of tumor and peritumoral edema under automatic segmentation. Sci Rep 2024; 14:27471. [PMID: 39523433 PMCID: PMC11551193 DOI: 10.1038/s41598-024-79344-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
Comprehensive and non-invasive preoperative molecular diagnosis is important for prognostic and therapy decision-making in adult-type diffuse gliomas. We employed a deep learning method for automatic segmentation of brain gliomas directly from conventional magnetic resonance imaging (MRI) scans of the tumor core and peritumoral edema regions based on available glioma MRI data provided in the BraTS2021. Three-dimensional volumes of interest were segmented from 424 cases of glioma imaging data retrospectively obtained from two medical centers using the segmentation method and radiomic features were extracted. We developed a subtype prediction model based on extracted radiomic features and analyzed significance and correlations between glioma morphological characteristics and pathological features using data from patients with adult-type diffuse glioma. The automated segmentation achieved mean Dice scores of 0.884 and 0.889 for the tumor core and whole tumor, respectively. The area under the receiver operating characteristic curve for the prediction of adult-type diffuse gliomas subtypes was 0.945. "Glioblastoma, IDH-wildtype", "Astrocytoma, IDH-mutant", and "Oligodendroglioma, IDH-mutant, 1p/19q-coded" showed AUCs of 0.96, 0.914, and 0.961, respectively, for subtype prediction. Glioma morphological characteristics, molecular and pathological levels, and clinical data showed significant differences and correlations. An automatic segmentation model for gliomas based on 3D U-Nets was developed, and the prediction model for gliomas built using the parameters obtained from the automatic segmentation model showed high overall performance.
Collapse
Affiliation(s)
- Xiangyu Sun
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China
| | - Sirui Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan, China
| | - Chao Ma
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China
| | - Wei Fang
- Wuhan Zhongke Industrial Research Institute of Medical Science Co., Ltd., Wuhan, China
| | - Xin Jing
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Chao Yang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China
| | - Huan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuchang District, Wuhan, China
| | - Xu Zhang
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Chuanbin Ge
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Bo Liu
- Wuhan United Imaging Healthcare Surgical Technology Co., Ltd., Wuhan, China
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.125 Donghu Road, WuChang, Wuhan, 430062, China.
| |
Collapse
|
23
|
Stanley EAM, Souza R, Winder AJ, Gulve V, Amador K, Wilms M, Forkert ND. Towards objective and systematic evaluation of bias in artificial intelligence for medical imaging. J Am Med Inform Assoc 2024; 31:2613-2621. [PMID: 38942737 PMCID: PMC11491635 DOI: 10.1093/jamia/ocae165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024] Open
Abstract
OBJECTIVE Artificial intelligence (AI) models trained using medical images for clinical tasks often exhibit bias in the form of subgroup performance disparities. However, since not all sources of bias in real-world medical imaging data are easily identifiable, it is challenging to comprehensively assess their impacts. In this article, we introduce an analysis framework for systematically and objectively investigating the impact of biases in medical images on AI models. MATERIALS AND METHODS Our framework utilizes synthetic neuroimages with known disease effects and sources of bias. We evaluated the impact of bias effects and the efficacy of 3 bias mitigation strategies in counterfactual data scenarios on a convolutional neural network (CNN) classifier. RESULTS The analysis revealed that training a CNN model on the datasets containing bias effects resulted in expected subgroup performance disparities. Moreover, reweighing was the most successful bias mitigation strategy for this setup. Finally, we demonstrated that explainable AI methods can aid in investigating the manifestation of bias in the model using this framework. DISCUSSION The value of this framework is showcased in our findings on the impact of bias scenarios and efficacy of bias mitigation in a deep learning model pipeline. This systematic analysis can be easily expanded to conduct further controlled in silico trials in other investigations of bias in medical imaging AI. CONCLUSION Our novel methodology for objectively studying bias in medical imaging AI can help support the development of clinical decision-support tools that are robust and responsible.
Collapse
Affiliation(s)
- Emma A M Stanley
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
- Department of Radiology, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Raissa Souza
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
- Department of Radiology, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Anthony J Winder
- Department of Radiology, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Vedant Gulve
- Department of Radiology, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Kimberly Amador
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
- Department of Radiology, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Matthias Wilms
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Department of Pediatrics, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Nils D Forkert
- Department of Radiology, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
- Department of Electrical and Software Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
| |
Collapse
|
24
|
Gao M, Cheng J, Qiu A, Zhao D, Wang J, Liu J. Magnetic resonance imaging (MRI)-based intratumoral and peritumoral radiomics for prognosis prediction in glioma patients. Clin Radiol 2024; 79:e1383-e1393. [PMID: 39218720 DOI: 10.1016/j.crad.2024.08.005] [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: 02/20/2024] [Revised: 06/30/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
AIM The purpose of this study was to identify robust radiological features from intratumoral and peritumoral regions, evaluate MRI protocols, and machine learning methods for overall survival stratification of glioma patients, and explore the relationship between radiological features and the tumour microenvironment. MATERIAL AND METHODS A retrospective analysis was conducted on 163 glioma patients, divided into a training set (n=113) and a testing set (n=50). For each patient, 2135 features were extracted from clinical MRI. Feature selection was performed using the Minimum Redundancy Maximum Relevance method and the Random Forest (RF) algorithm. Prognostic factors were assessed using the Cox proportional hazards model. Four machine learning models (RF, Logistic Regression, Support Vector Machine, and XGBoost) were trained on clinical and radiological features from tumour and peritumoral regions. Model evaluations on the testing set used receiver operating characteristic curves. RESULTS Among the 163 patients, 96 had an overall survival (OS) of less than three years postsurgery, while 67 had an OS of more than three years. Univariate Cox regression in the validation set indicated that age (p=0.003) and tumour grade (p<0.001) were positively associated with the risk of death within three years postsurgery. The final predictive model incorporated 13 radiological and 7 clinical features. The RF model, combining intratumor and peritumor radiomics, achieved the best predictive performance (AUC = 0.91; ACC = 0.86), outperforming single-region models. CONCLUSION Combined intratumoral and peritumoral radiomics can improve survival prediction and have potential as a practical imaging biomarker to guide clinical decision-making.
Collapse
Affiliation(s)
- M Gao
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - J Cheng
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China; Institute of Guizhou Aerospace Measuring and Testing Technology, Guiyang, China
| | - A Qiu
- Department of Biomedical Engineering, The Johns Hopkins University, MD, USA; Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - D Zhao
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.
| | - J Wang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China.
| | - J Liu
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, China; Department of Radiology Quality Control Center, Changsha, China.
| |
Collapse
|
25
|
Paschali M, Zhao Q, Sassoon SA, Pfefferbaum A, Sullivan EV, Pohl KM. Multi-domain predictors of grip strength differentiate individuals with and without alcohol use disorder. Addict Biol 2024; 29:e70007. [PMID: 39532141 PMCID: PMC11556900 DOI: 10.1111/adb.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/11/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Grip strength is considered one of the simplest and reliable indices of general health. Although motor ability and strength are commonly affected in people with alcohol use disorder (AUD), factors predictive of grip strength decline in AUD have not been investigated. Here, we employed a data-driven analysis predicting grip strength from measurements in 53 controls and 110 AUD participants, 53 of whom were comorbid with HIV infection. Controls and AUD were matched on sex, age, and body mass index. Measurements included commonly available metrics of brain structure, neuropsychological functioning, behavioural status, haematological and health status, and demographics. Based on 5-fold stratified cross-validation, a machine learning approach predicted grip strength separately for each cohort. The strongest (top 10%) predictors of grip were then tested against grip strength with correlational analysis. Leading grip strength predictors for both cohorts were cerebellar volume and mean corpuscular haemoglobin concentration. Predictors specific to controls were Backwards Digit Span, precentral gyrus volume, diastolic blood pressure, and mean platelet volume, which together significantly predicted grip strength (R2 = 0.255, p = 0.001). Unique predictors for AUD were comorbidity for HIV infection, social functioning, insular volume, and platelet count, which together significantly predicted grip strength (R2 = 0.162, p = 0.002). These cohort-specific predictors were doubly dissociated. Salient predictors of grip strength differed by diagnosis with only modest overlap. The constellation of cohort-specific predictive measurements of compromised grip strength provides insight into brain, behavioural, and physiological factors that may signal subtly affected yet treatable processes of physical decline and frailty.
Collapse
Affiliation(s)
- Magdalini Paschali
- Department of Psychiatry & Behavioral SciencesStanford UniversityStanfordCAUSA
- Department of RadiologyStanford UniversityStanfordCAUSA
| | - Qingyu Zhao
- Department of RadiologyWeill Cornell MedicineNew YorkNYUSA
| | - Stephanie A. Sassoon
- Department of Psychiatry & Behavioral SciencesStanford UniversityStanfordCAUSA
- Centefr for Health SciencesSRI InternationalMenlo ParkCAUSA
| | - Adolf Pfefferbaum
- Department of Psychiatry & Behavioral SciencesStanford UniversityStanfordCAUSA
- Centefr for Health SciencesSRI InternationalMenlo ParkCAUSA
| | - Edith V. Sullivan
- Department of Psychiatry & Behavioral SciencesStanford UniversityStanfordCAUSA
| | - Kilian M. Pohl
- Department of Psychiatry & Behavioral SciencesStanford UniversityStanfordCAUSA
- Department of Electrical EngineeringStanford UniversityStanfordCAUSA
| |
Collapse
|
26
|
Bakas S, Vollmuth P, Galldiks N, Booth TC, Aerts HJWL, Bi WL, Wiestler B, Tiwari P, Pati S, Baid U, Calabrese E, Lohmann P, Nowosielski M, Jain R, Colen R, Ismail M, Rasool G, Lupo JM, Akbari H, Tonn JC, Macdonald D, Vogelbaum M, Chang SM, Davatzikos C, Villanueva-Meyer JE, Huang RY. Artificial Intelligence for Response Assessment in Neuro Oncology (AI-RANO), part 2: recommendations for standardisation, validation, and good clinical practice. Lancet Oncol 2024; 25:e589-e601. [PMID: 39481415 PMCID: PMC12007431 DOI: 10.1016/s1470-2045(24)00315-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 11/02/2024]
Abstract
Technological advancements have enabled the extended investigation, development, and application of computational approaches in various domains, including health care. A burgeoning number of diagnostic, predictive, prognostic, and monitoring biomarkers are continuously being explored to improve clinical decision making in neuro-oncology. These advancements describe the increasing incorporation of artificial intelligence (AI) algorithms, including the use of radiomics. However, the broad applicability and clinical translation of AI are restricted by concerns about generalisability, reproducibility, scalability, and validation. This Policy Review intends to serve as the leading resource of recommendations for the standardisation and good clinical practice of AI approaches in health care, particularly in neuro-oncology. To this end, we investigate the repeatability, reproducibility, and stability of AI in response assessment in neuro-oncology in studies on factors affecting such computational approaches, and in publicly available open-source data and computational software tools facilitating these goals. The pathway for standardisation and validation of these approaches is discussed with the view of trustworthy AI enabling the next generation of clinical trials. We conclude with an outlook on the future of AI-enabled neuro-oncology.
Collapse
Affiliation(s)
- Spyridon Bakas
- Department of Pathology & Laboratory Medicine, Division of Computational Pathology, Indiana University, Indianopolis, IN, USA; Department of Radiology & Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN, USA; Department of Neurological Surgery, School of Medicine, Indiana University, Indianapolis, IN, USA; Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianopolis, IN, USA; Department of Computer Science, Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, IN, USA.
| | - Philipp Vollmuth
- Division for Computational Radiology and Clinical AI, Clinic for Neuroradiology, University Hospital Bonn, Bonn, Germany; Faculty of Medicine, University of Bonn, Bonn, Germany; Division for Medical Image Computing, German Cancer Research Center, Heidelberg, Germany
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
| | - Thomas C Booth
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK; Department of Neuroradiology, King's College Hospital NHS Foundation Trust, London, UK
| | - Hugo J W L Aerts
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Artificial Intelligence in Medicine Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA; Radiology and Nuclear Medicine, Maastricht University, Maastricht, Netherlands
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benedikt Wiestler
- Department of Neuroradiology, University Hospital, Technical University of Munich, Munich, Germany
| | - Pallavi Tiwari
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Sarthak Pati
- Department of Pathology & Laboratory Medicine, Division of Computational Pathology, Indiana University, Indianopolis, IN, USA
| | - Ujjwal Baid
- Department of Pathology & Laboratory Medicine, Division of Computational Pathology, Indiana University, Indianopolis, IN, USA; Department of Radiology & Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianopolis, IN, USA
| | - Evan Calabrese
- Department of Radiology, School of Medicine, Duke University, Durham, NC, USA
| | - Philipp Lohmann
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Martha Nowosielski
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Rajan Jain
- Department of Radiology and Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Rivka Colen
- Department of Radiology, Neuroradiology Division, Center for Artificial Intelligence Innovation in Medical Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marwa Ismail
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Ghulam Rasool
- Department of Machine Learning, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Hamed Akbari
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, USA
| | - Joerg C Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany; German Cancer Consortium, Partner Site Munich, Munich, Germany
| | | | - Michael Vogelbaum
- Department of Neuro-Oncology, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA; Department of Neurosurgery, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA; H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Susan M Chang
- Department of Neurological Surgery, Division of Neuro-Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Christos Davatzikos
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Artificial Intelligence for Integrated Diagnostics and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Pfefferbaum A, Zahr NM, Sassoon SA, Fama R, Saranathan M, Pohl KM, Sullivan EV. Aging, HIV infection, and alcohol exert synergist effects on regional thalamic volumes resulting in functional impairment. Neuroimage Clin 2024; 44:103684. [PMID: 39423567 PMCID: PMC11513528 DOI: 10.1016/j.nicl.2024.103684] [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: 06/29/2024] [Revised: 09/23/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVE Pharmacologically-treated people living with HIV infection have near-normal life spans with more than 50 % living into at-risk age for dementia and a disproportionate number relative to uninfected people engaging in unhealthy drinking. Accelerated aging in HIV occurs in some brain structures including the multinucleated thalamus. Unknown is whether aging with HIV affects thalamic nuclei and associated functions differentially and whether the common comorbidity of alcohol use disorder (AUD) + HIV accelerates aging. METHODS This mixed cross-sectional/longitudinal design examined 216 control, 69 HIV, and 74 HIV + AUD participants, age 25-75 years old at initial visit, examined 1-8 times. MRI thalamic volumetry, parcellated using THalamus Optimized Multi-Atlas Segmentation (THOMAS), identified 10 nuclei grouped into 4 functional regions for correlation with age and measures of neuropsychological, clinical, and hematological status. RESULTS Aging in the control group was best modeled with quadratic functions in the Anterior and Ventral regions and with linear functions in the Medial and Posterior regions. Relative to controls, age-related decline was even steeper in the Anterior and Ventral regions of the HIV group and in the Anterior region of the comorbid group. Anterior volumes of each HIV group declined significantly faster after age 50 (HIV = -2.4 %/year; HIV + AUD = -2.8 %/year) than that of controls (-1.8 %/year). Anterior and Ventral volumes were significantly smaller in the HIV + AUD than HIV-only group when controlling for infection factors. Although compared with controls HIV + AUD declined faster than HIV alone, the two HIV groups did not differ significantly from each other in aging rates. Declining Attention/Working Memory and Motor Skills performance correlated with Anterior and Posterior volume declines in the HIV + AUD group. CONCLUSIONS Regional thalamic volumetry detected normal aging declines, differential and accelerated volume losses in HIV, relations between age-related nuclear and performance declines, and exacerbation of volume declines in comorbid AUD contributing to functional deficits.
Collapse
Affiliation(s)
- Adolf Pfefferbaum
- Center for Health Sciences, SRI International, Menlo Park, CA, United States; Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Natalie M Zahr
- Center for Health Sciences, SRI International, Menlo Park, CA, United States; Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Stephanie A Sassoon
- Center for Health Sciences, SRI International, Menlo Park, CA, United States; Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Rosemary Fama
- Center for Health Sciences, SRI International, Menlo Park, CA, United States; Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Manojkumar Saranathan
- Department of Radiology, University of Massachusetts Chan School of Medicine, Worcester, MA, United States
| | - Kilian M Pohl
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Edith V Sullivan
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
28
|
Romme CJA, Stanley EAM, Mouches P, Wilms M, Pike GB, Metz LM, Forkert ND. Analysis and visualization of the effect of multiple sclerosis on biological brain age. Front Neurol 2024; 15:1423485. [PMID: 39450049 PMCID: PMC11499186 DOI: 10.3389/fneur.2024.1423485] [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: 04/25/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction The rate of neurodegeneration in multiple sclerosis (MS) is an important biomarker for disease progression but can be challenging to quantify. The brain age gap, which quantifies the difference between a patient's chronological and their estimated biological brain age, might be a valuable biomarker of neurodegeneration in patients with MS. Thus, the aim of this study was to investigate the value of an image-based prediction of the brain age gap using a deep learning model and compare brain age gap values between healthy individuals and patients with MS. Methods A multi-center dataset consisting of 5,294 T1-weighted magnetic resonance images of the brain from healthy individuals aged between 19 and 89 years was used to train a convolutional neural network (CNN) for biological brain age prediction. The trained model was then used to calculate the brain age gap in 195 patients with relapsing remitting MS (20-60 years). Additionally, saliency maps were generated for healthy subjects and patients with MS to identify brain regions that were deemed important for the brain age prediction task by the CNN. Results Overall, the application of the CNN revealed accelerated brain aging with a larger brain age gap for patients with MS with a mean of 6.98 ± 7.18 years in comparison to healthy test set subjects (0.23 ± 4.64 years). The brain age gap for MS patients was weakly to moderately correlated with age at disease onset (ρ = -0.299, p < 0.0001), EDSS score (ρ = 0.206, p = 0.004), disease duration (ρ = 0.162, p = 0.024), lesion volume (ρ = 0.630, p < 0.0001), and brain parenchymal fraction (ρ = -0.718, p < 0.0001). The saliency maps indicated significant differences in the lateral ventricle (p < 0.0001), insula (p < 0.0001), third ventricle (p < 0.0001), and fourth ventricle (p = 0.0001) in the right hemisphere. In the left hemisphere, the inferior lateral ventricle (p < 0.0001) and the third ventricle (p < 0.0001) showed significant differences. Furthermore, the Dice similarity coefficient showed the highest overlap of salient regions between the MS patients and the oldest healthy subjects, indicating that neurodegeneration is accelerated in this patient cohort. Discussion In conclusion, the results of this study show that the brain age gap is a valuable surrogate biomarker to measure disease progression in patients with multiple sclerosis.
Collapse
Affiliation(s)
- Catharina J. A. Romme
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Emma A. M. Stanley
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Pauline Mouches
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Matthias Wilms
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - G. Bruce Pike
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Luanne M. Metz
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nils D. Forkert
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
29
|
Kamel P, Khalid M, Steger R, Kanhere A, Kulkarni P, Parekh V, Yi PH, Gandhi D, Bodanapally U. Dual Energy CT for Deep Learning-Based Segmentation and Volumetric Estimation of Early Ischemic Infarcts. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024:10.1007/s10278-024-01294-5. [PMID: 39384719 DOI: 10.1007/s10278-024-01294-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/13/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
Ischemic changes are not visible on non-contrast head CT until several hours after infarction, though deep convolutional neural networks have shown promise in the detection of subtle imaging findings. This study aims to assess if dual-energy CT (DECT) acquisition can improve early infarct visibility for machine learning. The retrospective dataset consisted of 330 DECTs acquired up to 48 h prior to confirmation of a DWI positive infarct on MRI between 2016 and 2022. Infarct segmentation maps were generated from the MRI and co-registered to the CT to serve as ground truth for segmentation. A self-configuring 3D nnU-Net was trained for segmentation on (1) standard 120 kV mixed-images (2) 190 keV virtual monochromatic images and (3) 120 kV + 190 keV images as dual channel inputs. Algorithm performance was assessed with Dice scores with paired t-tests on a test set. Global aggregate Dice scores were 0.616, 0.645, and 0.665 for standard 120 kV images, 190 keV, and combined channel inputs respectively. Differences in overall Dice scores were statistically significant with highest performance for combined channel inputs (p < 0.01). Small but statistically significant differences were observed for infarcts between 6 and 12 h from last-known-well with higher performance for larger infarcts. Volumetric accuracy trended higher with combined inputs but differences were not statistically significant (p = 0.07). Supplementation of standard head CT images with dual-energy data provides earlier and more accurate segmentation of infarcts for machine learning particularly between 6 and 12 h after last-known-well.
Collapse
Affiliation(s)
- Peter Kamel
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA.
- University of Maryland Medical Intelligent Imaging (UM2ii) Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA.
| | - Mazhar Khalid
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rachel Steger
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Adway Kanhere
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
- University of Maryland Medical Intelligent Imaging (UM2ii) Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
| | - Pranav Kulkarni
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
- University of Maryland Medical Intelligent Imaging (UM2ii) Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
| | - Vishwa Parekh
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
- University of Maryland Medical Intelligent Imaging (UM2ii) Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
| | - Paul H Yi
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
- University of Maryland Medical Intelligent Imaging (UM2ii) Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
| | - Dheeraj Gandhi
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
| | - Uttam Bodanapally
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD, 21201, USA
| |
Collapse
|
30
|
Ziegenfeuter J, Delbridge C, Bernhardt D, Gempt J, Schmidt-Graf F, Hedderich D, Griessmair M, Thomas M, Meyer HS, Zimmer C, Meyer B, Combs SE, Yakushev I, Metz MC, Wiestler B. Resolving spatial response heterogeneity in glioblastoma. Eur J Nucl Med Mol Imaging 2024; 51:3685-3695. [PMID: 38837060 PMCID: PMC11445274 DOI: 10.1007/s00259-024-06782-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024]
Abstract
PURPOSE Spatial intratumoral heterogeneity poses a significant challenge for accurate response assessment in glioblastoma. Multimodal imaging coupled with advanced image analysis has the potential to unravel this response heterogeneity. METHODS Based on automated tumor segmentation and longitudinal registration with follow-up imaging, we categorized contrast-enhancing voxels of 61 patients with suspected recurrence of glioblastoma into either true tumor progression (TP) or pseudoprogression (PsP). To allow the unbiased analysis of semantically related image regions, adjacent voxels with similar values of cerebral blood volume (CBV), FET-PET, and contrast-enhanced T1w were automatically grouped into supervoxels. We then extracted first-order statistics as well as texture features from each supervoxel. With these features, a Random Forest classifier was trained and validated employing a 10-fold cross-validation scheme. For model evaluation, the area under the receiver operating curve, as well as classification performance metrics were calculated. RESULTS Our image analysis pipeline enabled reliable spatial assessment of tumor response. The predictive model reached an accuracy of 80.0% and a macro-weighted AUC of 0.875, which takes class imbalance into account, in the hold-out samples from cross-validation on supervoxel level. Analysis of feature importances confirmed the significant role of FET-PET-derived features. Accordingly, TP- and PsP-labeled supervoxels differed significantly in their 10th and 90th percentile, as well as the median of tumor-to-background normalized FET-PET. However, CBV- and T1c-related features also relevantly contributed to the model's performance. CONCLUSION Disentangling the intratumoral heterogeneity in glioblastoma holds immense promise for advancing precise local response evaluation and thereby also informing more personalized and localized treatment strategies in the future.
Collapse
Affiliation(s)
- Julian Ziegenfeuter
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675, München, Germany.
| | - Claire Delbridge
- Department of Pathology, Technical University of Munich, 81675, München, Germany
| | - Denise Bernhardt
- Department of Radiation Oncology, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
| | - Jens Gempt
- Department of Neurosurgery, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Friederike Schmidt-Graf
- Department of Neurology, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
| | - Dennis Hedderich
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
| | - Michael Griessmair
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
| | - Marie Thomas
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
| | - Hanno S Meyer
- Department of Neurosurgery, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Claus Zimmer
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
| | - Igor Yakushev
- Department of Nuclear Medicine, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
| | - Marie-Christin Metz
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
| | - Benedikt Wiestler
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675, München, Germany
- TranslaTUM, Technical University of Munich, 81675, München, Germany
| |
Collapse
|
31
|
Lorkiewicz SA, Müller-Oehring EM, Baker FC, Elkins BV, Schulte T. A longitudinal study of the relationship between alcohol-related blackouts and attenuated structural brain development. Dev Cogn Neurosci 2024; 69:101448. [PMID: 39307082 PMCID: PMC11440320 DOI: 10.1016/j.dcn.2024.101448] [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: 06/07/2024] [Revised: 08/21/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
PURPOSE Alcohol-related blackouts (ARBs) are common in adolescents and emerging adults. ARBs may also be indicative of persistent, alcohol-related neurocognitive changes. This study explored ARBs as a predictor of altered structural brain development and associated cognitive correlates. METHODS Longitudinal growth curve modeling estimated trajectories of brain volume across 6 years in participants from the National Consortium on Alcohol and Neurodevelopment in Adolescence (NCANDA) study (n = 800, 213 with lifetime ARB history). While controlling for demographics and overall alcohol use, ARB history was analyzed as a predictor of brain volume growth in regions associated with alcohol-related cognitive change. Post hoc analyses examined whether ARBs moderated relationships between brain morphology and cognition. RESULTS ARBs significantly predicted attenuated development of fusiform gyrus and hippocampal volume at unique timepoints compared to overall alcohol use. Alcohol use without ARBs significantly predicted attenuated fusiform and hippocampal growth at earlier and later timepoints, respectively. Despite altered development in regions associated with memory, ARBs did not significantly moderate relationships between brain volume and cognitive performance. CONCLUSION ARBs and overall alcohol use predicted altered brain development in the fusiform gyrus and hippocampus at different timepoints, suggesting ARBs represent a unique marker of neurocognitive risk in younger drinkers.
Collapse
Affiliation(s)
- Sara A Lorkiewicz
- Palo Alto University, Clinical Psychology, Palo Alto, CA, USA; Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Eva M Müller-Oehring
- SRI International, Neuroscience Program, Menlo Park, CA, USA; Stanford University School of Medicine, Psychiatry and Behavioral Sciences, Stanford, CA, USA; Stanford University School of Medicine, Department of Neurology and Neurological Sciences, Stanford, CA, USA
| | - Fiona C Baker
- SRI International, Neuroscience Program, Menlo Park, CA, USA; Brain Function Research Group, School of Physiology, University of Witwatersrand, Johannesburg, South Africa
| | - Brionne V Elkins
- University of Texas Medical Branch, Department of Neurology, Galveston, TX, USA
| | - Tilman Schulte
- Palo Alto University, Clinical Psychology, Palo Alto, CA, USA; SRI International, Neuroscience Program, Menlo Park, CA, USA.
| |
Collapse
|
32
|
Sullivan EV, Zahr NM, Zhao Q, Pohl KM, Sassoon SA, Pfefferbaum A. Contributions of Cerebral White Matter Hyperintensities to Postural Instability in Aging With and Without Alcohol Use Disorder. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:998-1009. [PMID: 38569932 PMCID: PMC11442683 DOI: 10.1016/j.bpsc.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/29/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Both postural instability and brain white matter hyperintensities (WMHs) are noted markers of normal aging and alcohol use disorder (AUD). Here, we questioned what variables contribute to the sway path-WMH relationship in individuals with AUD and healthy control participants. METHODS The data comprised 404 balance platform sessions, yielding sway path length and magnetic resonance imaging data acquired cross-sectionally or longitudinally in 102 control participants and 158 participants with AUD ages 25 to 80 years. Balance sessions were typically conducted on the same day as magnetic resonance imaging fluid-attenuated inversion recovery acquisitions, permitting WMH volume quantification. Factors considered in multiple regression analyses as potential contributors to the relationship between WMH volumes and postural instability were age, sex, socioeconomic status, education, pedal 2-point discrimination, systolic and diastolic blood pressure, body mass index, depressive symptoms, total alcohol consumed in the past year, and race. RESULTS Initial analysis identified diagnosis, age, sex, and race as significant contributors to observed sway path-WMH relationships. Inclusion of these factors as predictors in multiple regression analyses substantially attenuated the sway path-WMH relationships in both AUD and healthy control groups. Women, irrespective of diagnosis or race, had shorter sway paths than men. Black participants, irrespective of diagnosis or sex, had shorter sway paths than non-Black participants despite having modestly larger WMH volumes than non-Black participants, which is possibly a reflection of the younger age of the Black sample. CONCLUSIONS Longer sway paths were related to larger WMH volumes in healthy men and women with and without AUD. Critically, however, age almost fully accounted for these associations.
Collapse
Affiliation(s)
- Edith V Sullivan
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, California.
| | - Natalie M Zahr
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, California; Center for Health Sciences, SRI International, Menlo Park, California
| | - Qingyu Zhao
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Kilian M Pohl
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, California
| | - Stephanie A Sassoon
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, California; Center for Health Sciences, SRI International, Menlo Park, California
| | - Adolf Pfefferbaum
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, California; Center for Health Sciences, SRI International, Menlo Park, California
| |
Collapse
|
33
|
Duman A, Sun X, Thomas S, Powell JR, Spezi E. Reproducible and Interpretable Machine Learning-Based Radiomic Analysis for Overall Survival Prediction in Glioblastoma Multiforme. Cancers (Basel) 2024; 16:3351. [PMID: 39409970 PMCID: PMC11476262 DOI: 10.3390/cancers16193351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/20/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
PURPOSE To develop and validate an MRI-based radiomic model for predicting overall survival (OS) in patients diagnosed with glioblastoma multiforme (GBM), utilizing a retrospective dataset from multiple institutions. MATERIALS AND METHODS Pre-treatment MRI images of 289 GBM patients were collected. From each patient's tumor volume, 660 radiomic features (RFs) were extracted and subjected to robustness analysis. The initial prognostic model with minimum RFs was subsequently enhanced by including clinical variables. The final clinical-radiomic model was derived through repeated three-fold cross-validation on the training dataset. Performance evaluation included assessment of concordance index (C-Index), integrated area under curve (iAUC) alongside patient stratification into low and high-risk groups for overall survival (OS). RESULTS The final prognostic model, which has the highest level of interpretability, utilized primary gross tumor volume (GTV) and one MRI modality (T2-FLAIR) as a predictor and integrated the age variable with two independent, robust RFs, achieving moderately good discriminatory performance (C-Index [95% confidence interval]: 0.69 [0.62-0.75]) with significant patient stratification (p = 7 × 10-5) on the validation cohort. Furthermore, the trained model exhibited the highest iAUC at 11 months (0.81) in the literature. CONCLUSION We identified and validated a clinical-radiomic model for stratification of patients into low and high-risk groups based on OS in patients with GBM using a multicenter retrospective dataset. Future work will focus on the use of deep learning-based features, with recently standardized convolutional filters on OS tasks.
Collapse
Affiliation(s)
- Abdulkerim Duman
- School of Engineering, Cardiff University, Cardiff CF24 3AA, UK;
| | - Xianfang Sun
- School of Computer Science and Informatics, Cardiff University, Cardiff CF24 4AG, UK;
| | - Solly Thomas
- Maidstone and Tunbridge Wells NHS Trust, Kent ME16 9QQ, UK;
| | - James R. Powell
- Department of Oncology, Velindre University NHS Trust, Cardiff CF14 2TL, UK;
| | - Emiliano Spezi
- School of Engineering, Cardiff University, Cardiff CF24 3AA, UK;
| |
Collapse
|
34
|
Buchner JA, Kofler F, Mayinger M, Christ SM, Brunner TB, Wittig A, Menze B, Zimmer C, Meyer B, Guckenberger M, Andratschke N, El Shafie RA, Debus J, Rogers S, Riesterer O, Schulze K, Feldmann HJ, Blanck O, Zamboglou C, Ferentinos K, Bilger-Zähringer A, Grosu AL, Wolff R, Piraud M, Eitz KA, Combs SE, Bernhardt D, Rueckert D, Wiestler B, Peeken JC. Radiomics-based prediction of local control in patients with brain metastases following postoperative stereotactic radiotherapy. Neuro Oncol 2024; 26:1638-1650. [PMID: 38813990 PMCID: PMC11376458 DOI: 10.1093/neuonc/noae098] [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: 12/23/2023] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Surgical resection is the standard of care for patients with large or symptomatic brain metastases (BMs). Despite improved local control after adjuvant stereotactic radiotherapy, the risk of local failure (LF) persists. Therefore, we aimed to develop and externally validate a pre-therapeutic radiomics-based prediction tool to identify patients at high LF risk. METHODS Data were collected from A Multicenter Analysis of Stereotactic Radiotherapy to the Resection Cavity of BMs (AURORA) retrospective study (training cohort: 253 patients from 2 centers; external test cohort: 99 patients from 5 centers). Radiomic features were extracted from the contrast-enhancing BM (T1-CE MRI sequence) and the surrounding edema (T2-FLAIR sequence). Different combinations of radiomic and clinical features were compared. The final models were trained on the entire training cohort with the best parameter set previously determined by internal 5-fold cross-validation and tested on the external test set. RESULTS The best performance in the external test was achieved by an elastic net regression model trained with a combination of radiomic and clinical features with a concordance index (CI) of 0.77, outperforming any clinical model (best CI: 0.70). The model effectively stratified patients by LF risk in a Kaplan-Meier analysis (P < .001) and demonstrated an incremental net clinical benefit. At 24 months, we found LF in 9% and 74% of the low and high-risk groups, respectively. CONCLUSIONS A combination of clinical and radiomic features predicted freedom from LF better than any clinical feature set alone. Patients at high risk for LF may benefit from stricter follow-up routines or intensified therapy.
Collapse
Affiliation(s)
- Josef A Buchner
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Florian Kofler
- Department of Informatics, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Helmholtz AI, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Michael Mayinger
- Department of Radiation Oncology, University of Zurich, Zurich, Switzerland
| | - Sebastian M Christ
- Department of Radiation Oncology, University of Zurich, Zurich, Switzerland
| | - Thomas B Brunner
- Department of Radiation Oncology, University Hospital Magdeburg, Magdeburg, Germany
| | - Andrea Wittig
- Department of Radiotherapy and Radiation Oncology, University Hospital Jena, Friedrich-Schiller University, Jena, Germany
| | - Bjoern Menze
- Department of Informatics, Technical University of Munich, Munich, Germany
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Claus Zimmer
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | | | - Rami A El Shafie
- Department of Radiation Oncology, University Medical Center Göttingen, Göttingen, Germany
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jürgen Debus
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Susanne Rogers
- Radiation Oncology Center KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Oliver Riesterer
- Radiation Oncology Center KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Katrin Schulze
- Department of Radiation Oncology, General Hospital Fulda, Fulda, Germany
| | - Horst J Feldmann
- Department of Radiation Oncology, General Hospital Fulda, Fulda, Germany
| | - Oliver Blanck
- Saphir Radiosurgery Center Frankfurt and Northern Germany, Kiel, Germany
- Department of Radiation Oncology, University Medical Center Schleswig Holstein, Kiel, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, German Oncology Center, European University of Cyprus, Limassol, Cyprus
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- Department of Radiation Oncology, University of Freiburg - Medical Center, Freiburg, Germany
| | - Konstantinos Ferentinos
- Department of Radiation Oncology, German Oncology Center, European University of Cyprus, Limassol, Cyprus
| | - Angelika Bilger-Zähringer
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- Department of Radiation Oncology, University of Freiburg - Medical Center, Freiburg, Germany
| | - Anca L Grosu
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- Department of Radiation Oncology, University of Freiburg - Medical Center, Freiburg, Germany
| | - Robert Wolff
- Saphir Radiosurgery Center Frankfurt and Northern Germany, Kiel, Germany
- Department of Neurosurgery, University Hospital Frankfurt, Frankfurt, Germany
| | - Marie Piraud
- Helmholtz AI, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Kerstin A Eitz
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Center Munich, Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Center Munich, Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany
| | - Denise Bernhardt
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany
| | - Daniel Rueckert
- Institute for Artificial Intelligence and Informatics in Medicine, Technical University of Munich, Munich, Germany
| | - Benedikt Wiestler
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jan C Peeken
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Center Munich, Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
35
|
Schilling L, Singleton SP, Tozlu C, Hédo M, Zhao Q, Pohl KM, Jamison K, Kuceyeski A. Sex-specific differences in brain activity dynamics of youth with a family history of substance use disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.610959. [PMID: 39282344 PMCID: PMC11398379 DOI: 10.1101/2024.09.03.610959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
An individual's risk of substance use disorder (SUD) is shaped by a complex interplay of potent biosocial factors. Current neurodevelopmental models posit vulnerability to SUD in youth is due to an overreactive reward system and reduced inhibitory control. Having a family history of SUD is a particularly strong risk factor, yet few studies have explored its impact on brain function and structure prior to substance exposure. Herein, we utilized a network control theory approach to quantify sex-specific differences in brain activity dynamics in youth with and without a family history of SUD, drawn from a large cohort of substance-naïve youth from the Adolescent Brain Cognitive Development Study. We summarize brain dynamics by calculating transition energy, which probes the ease with which a whole brain, region or network drives the brain towards a specific spatial pattern of activation (i.e., brain state). Our findings reveal that a family history of SUD is associated with alterations in the brain's dynamics wherein: i) independent of sex, certain regions' transition energies are higher in those with a family history of SUD and ii) there exist sex-specific differences in SUD family history groups at multiple levels of transition energy (global, network, and regional). Family history-by-sex effects reveal that energetic demand is increased in females with a family history of SUD and decreased in males with a family history of SUD, compared to their same-sex counterparts with no SUD family history. Specifically, we localize these effects to higher energetic demands of the default mode network in females with a family history of SUD and lower energetic demands of attention networks in males with a family history of SUD. These results suggest a family history of SUD may increase reward saliency in males and decrease efficiency of top-down inhibitory control in females. This work could be used to inform personalized intervention strategies that may target differing cognitive mechanisms that predispose individuals to the development of SUD.
Collapse
Affiliation(s)
- Louisa Schilling
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | | | - Ceren Tozlu
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Marie Hédo
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Qingyu Zhao
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Kilian M Pohl
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Keith Jamison
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Amy Kuceyeski
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
36
|
Zhuang Z, Lin J, Wan Z, Weng J, Yuan Z, Xie Y, Liu Z, Xie P, Mao S, Wang Z, Wang X, Huang M, Luo Y, Yu H. Radiogenomic profiling of global DNA methylation associated with molecular phenotypes and immune features in glioma. BMC Med 2024; 22:352. [PMID: 39218882 PMCID: PMC11367996 DOI: 10.1186/s12916-024-03573-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The radiogenomic analysis has provided valuable imaging biomarkers with biological insights for gliomas. The radiogenomic markers for molecular profile such as DNA methylation remain to be uncovered to assist the molecular diagnosis and tumor treatment. METHODS We apply the machine learning approaches to identify the magnetic resonance imaging (MRI) features that are associated with molecular profiles in 146 patients with gliomas, and the fitting models for each molecular feature (MoRad) are developed and validated. To provide radiological annotations for the molecular profiles, we devise two novel approaches called radiomic oncology (RO) and radiomic set enrichment analysis (RSEA). RESULTS The generated MoRad models perform well for profiling each molecular feature with radiomic features, including mutational, methylation, transcriptional, and protein profiles. Among them, the MoRad models have a remarkable performance in quantitatively mapping global DNA methylation. With RO and RSEA approaches, we find that global DNA methylation could be reflected by the heterogeneity in volumetric and textural features of enhanced regions in T2-weighted MRI. Finally, we demonstrate the associations of global DNA methylation with clinicopathological, molecular, and immunological features, including histological grade, mutations of IDH and ATRX, MGMT methylation, multiple methylation-high subtypes, tumor-infiltrating lymphocytes, and long-term survival outcomes. CONCLUSIONS Global DNA methylation is highly associated with radiological profiles in glioma. Radiogenomic global methylation is an imaging-based quantitative molecular biomarker that is associated with specific consensus molecular subtypes and immune features.
Collapse
Affiliation(s)
- Zhuokai Zhuang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Jinxin Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Zixiao Wan
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Jingrong Weng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Ze Yuan
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Yumo Xie
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Zongchao Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cancer Epidemiology, Peking University Cancer Institute, Beijing, 100142, China
| | - Peiyi Xie
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Siyue Mao
- Image and Minimally Invasive Intervention Center, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zongming Wang
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaolin Wang
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Meijin Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Yanxin Luo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China
| | - Huichuan Yu
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China.
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Guangzhou, Guangdong, China.
| |
Collapse
|
37
|
Kwak S, Akbari H, Garcia JA, Mohan S, Dicker Y, Sako C, Matsumoto Y, Nasrallah MP, Shalaby M, O’Rourke DM, Shinohara RT, Liu F, Badve C, Barnholtz-Sloan JS, Sloan AE, Lee M, Jain R, Cepeda S, Chakravarti A, Palmer JD, Dicker AP, Shukla G, Flanders AE, Shi W, Woodworth GF, Davatzikos C. Predicting peritumoral glioblastoma infiltration and subsequent recurrence using deep-learning-based analysis of multi-parametric magnetic resonance imaging. J Med Imaging (Bellingham) 2024; 11:054001. [PMID: 39220048 PMCID: PMC11363410 DOI: 10.1117/1.jmi.11.5.054001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/16/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose Glioblastoma (GBM) is the most common and aggressive primary adult brain tumor. The standard treatment approach is surgical resection to target the enhancing tumor mass, followed by adjuvant chemoradiotherapy. However, malignant cells often extend beyond the enhancing tumor boundaries and infiltrate the peritumoral edema. Traditional supervised machine learning techniques hold potential in predicting tumor infiltration extent but are hindered by the extensive resources needed to generate expertly delineated regions of interest (ROIs) for training models on tissue most and least likely to be infiltrated. Approach We developed a method combining expert knowledge and training-based data augmentation to automatically generate numerous training examples, enhancing the accuracy of our model for predicting tumor infiltration through predictive maps. Such maps can be used for targeted supra-total surgical resection and other therapies that might benefit from intensive yet well-targeted treatment of infiltrated tissue. We apply our method to preoperative multi-parametric magnetic resonance imaging (mpMRI) scans from a subset of 229 patients of a multi-institutional consortium (Radiomics Signatures for Precision Diagnostics) and test the model on subsequent scans with pathology-proven recurrence. Results Leave-one-site-out cross-validation was used to train and evaluate the tumor infiltration prediction model using initial pre-surgical scans, comparing the generated prediction maps with follow-up mpMRI scans confirming recurrence through post-resection tissue analysis. Performance was measured by voxel-wised odds ratios (ORs) across six institutions: University of Pennsylvania (OR: 9.97), Ohio State University (OR: 14.03), Case Western Reserve University (OR: 8.13), New York University (OR: 16.43), Thomas Jefferson University (OR: 8.22), and Rio Hortega (OR: 19.48). Conclusions The proposed model demonstrates that mpMRI analysis using deep learning can predict infiltration in the peri-tumoral brain region for GBM patients without needing to train a model using expert ROI drawings. Results for each institution demonstrate the model's generalizability and reproducibility.
Collapse
Affiliation(s)
- Sunwoo Kwak
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Philadelphia, Pennsylvania, United States
| | - Hamed Akbari
- Santa Clara University, School of Engineering, Department of Bioengineering, Santa Clara, California, United States
| | - Jose A. Garcia
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Philadelphia, Pennsylvania, United States
| | - Suyash Mohan
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Philadelphia, Pennsylvania, United States
| | - Yehuda Dicker
- Columbia University, School of Engineering, Department of Computer Science, New York, United States
| | - Chiharu Sako
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Philadelphia, Pennsylvania, United States
| | - Yuji Matsumoto
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology, Philadelphia, Pennsylvania, United States
| | - MacLean P. Nasrallah
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, Perelman School of Medicine, Department of Pathology and Laboratory Medicine, Philadelphia, Pennsylvania, United States
| | - Mahmoud Shalaby
- Mercy Catholic Medical Center, Department of Radiology, Philadelphia, Pennsylvania, United States
| | - Donald M. O’Rourke
- University of Pennsylvania, Perelman School of Medicine, Department of Neurosurgery, Philadelphia, Pennsylvania, United States
| | - Russel T. Shinohara
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, Perelman School of Medicine, Department of Biostatistics and Epidemiology, Philadelphia, Pennsylvania, United States
| | - Fang Liu
- University of Pennsylvania, Perelman School of Medicine, Department of Biostatistics and Epidemiology, Philadelphia, Pennsylvania, United States
| | - Chaitra Badve
- Case Western Reserve University, University Hospitals Cleveland Medical Center, Department of Radiology, Cleveland, Ohio, United States
| | - Jill S. Barnholtz-Sloan
- National Cancer Institute, Center for Biomedical Informatics and Information Technology, Division of Cancer Epidemiology and Genetics, Bethesda, Maryland, United States
| | - Andrew E. Sloan
- Piedmont Healthcare, Division of Neuroscience, Atlanta, Georgia, United States
| | - Matthew Lee
- NYU Grossman School of Medicine, Department of Radiology, New York, United States
| | - Rajan Jain
- NYU Grossman School of Medicine, Department of Radiology, New York, United States
- NYU Grossman School of Medicine, Department of Neurosurgery, New York, United States
| | | | - Arnab Chakravarti
- Ohio State University Wexner Medical Center, Department of Radiation Oncology, Columbus, Ohio, United States
| | - Joshua D. Palmer
- Ohio State University Wexner Medical Center, Department of Radiation Oncology, Columbus, Ohio, United States
| | - Adam P. Dicker
- Thomas Jefferson University, Sidney Kimmel Cancer Center, Department of Radiation Oncology, Philadelphia, Pennsylvania, United States
| | - Gaurav Shukla
- Thomas Jefferson University, Sidney Kimmel Cancer Center, Department of Radiation Oncology, Philadelphia, Pennsylvania, United States
| | - Adam E. Flanders
- Thomas Jefferson University, Sidney Kimmel Cancer Center, Department of Radiation Oncology, Philadelphia, Pennsylvania, United States
| | - Wenyin Shi
- Thomas Jefferson University, Sidney Kimmel Cancer Center, Department of Radiation Oncology, Philadelphia, Pennsylvania, United States
| | - Graeme F. Woodworth
- University of Maryland, School of Medicine, Department of Neurosurgery, Baltimore, Maryland, United States
| | - Christos Davatzikos
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, Perelman School of Medicine, Center for Biomedical Image Computing and Analytics, Philadelphia, Pennsylvania, United States
| |
Collapse
|
38
|
Kudus K, Wagner MW, Namdar K, Bennett J, Nobre L, Tabori U, Hawkins C, Ertl-Wagner BB, Khalvati F. Beyond hand-crafted features for pretherapeutic molecular status identification of pediatric low-grade gliomas. Sci Rep 2024; 14:19102. [PMID: 39154039 PMCID: PMC11330469 DOI: 10.1038/s41598-024-69870-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024] Open
Abstract
The use of targeted agents in the treatment of pediatric low-grade gliomas (pLGGs) relies on the determination of molecular status. It has been shown that genetic alterations in pLGG can be identified non-invasively using MRI-based radiomic features or convolutional neural networks (CNNs). We aimed to build and assess a combined radiomics and CNN non-invasive pLGG molecular status identification model. This retrospective study used the tumor regions, manually segmented from T2-FLAIR MR images, of 336 patients treated for pLGG between 1999 and 2018. We designed a CNN and Random Forest radiomics model, along with a model relying on a combination of CNN and radiomic features, to predict the genetic status of pLGG. Additionally, we investigated whether CNNs could predict radiomic feature values from MR images. The combined model (mean AUC: 0.824) outperformed the radiomics model (0.802) and CNN (0.764). The differences in model performance were statistically significant (p-values < 0.05). The CNN was able to learn predictive radiomic features such as surface-to-volume ratio (average correlation: 0.864), and difference matrix dependence non-uniformity normalized (0.924) well but was unable to learn others such as run-length matrix variance (- 0.017) and non-uniformity normalized (- 0.042). Our results show that a model relying on both CNN and radiomic-based features performs better than either approach separately in differentiating the genetic status of pLGGs, and that CNNs are unable to express all handcrafted features.
Collapse
Affiliation(s)
- Kareem Kudus
- Neurosciences & Mental Health Research Program, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Matthias W Wagner
- Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children, Toronto, Canada
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Augsburg, Augsburg, Germany
| | - Khashayar Namdar
- Neurosciences & Mental Health Research Program, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Julie Bennett
- Division of Hematology and Oncology, The Hospital for Sick Children, Toronto, Canada
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
- Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Liana Nobre
- Department of Paediatrics, University of Alberta, Edmonton, Canada
- Division of Immunology, Hematology/Oncology and Palliative Care, Stollery Children's Hospital, Edmonton, Canada
| | - Uri Tabori
- Division of Hematology and Oncology, The Hospital for Sick Children, Toronto, Canada
| | - Cynthia Hawkins
- Paediatric Laboratory Medicine, Division of Pathology, The Hospital for Sick Children, Toronto, Canada
| | - Birgit Betina Ertl-Wagner
- Neurosciences & Mental Health Research Program, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children, Toronto, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Canada
| | - Farzad Khalvati
- Neurosciences & Mental Health Research Program, The Hospital for Sick Children, Toronto, Canada.
- Institute of Medical Science, University of Toronto, Toronto, Canada.
- Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children, Toronto, Canada.
- Department of Medical Imaging, University of Toronto, Toronto, Canada.
- Department of Computer Science, University of Toronto, Toronto, Canada.
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada.
| |
Collapse
|
39
|
Jimenez-Garcia A, Garcia HF, Cardenas-Pena DA, Cardenas-Bedoya W, Porras-Hurtado GL, Orozco-Gutierrez AA. Unsupervised Anomaly Detection by Learning Elastic Transformations Within an Autoencoder Approach. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40039758 DOI: 10.1109/embc53108.2024.10781622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Machine learning has approached magnetic resonance image (MRI) analysis using multiple techniques, including deep supervised learning methodologies, where lesions such as tumors or features associated with defined pathologies have been identified satisfactorily. However, many of these models require a certain amount of labeled data which access is usually restricted, due to the protection of health information. Furthermore, those methodologies are focused only on the pathologies considered in the training process, causing the model to ignore other kinds of brain lesions. In this paper, an unsupervised anomaly detection methodology is developed to model healthy structures and detect anomaly regions in MRI. Random elastic transform patches are applied over the whole volume to quantify and optimize the reconstruction performance during training, leading to effective anomaly region detection. The experimental results show competitive results with common state-of-the-art unsupervised approaches.
Collapse
|
40
|
Zeng Q, Tian Z, Dong F, Shi F, Xu P, Zhang J, Ling C, Guo Z. Multi-parameter MRI radiomic features may contribute to predict progression-free survival in patients with WHO grade II meningiomas. Front Oncol 2024; 14:1246730. [PMID: 39007097 PMCID: PMC11239420 DOI: 10.3389/fonc.2024.1246730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Aim This study aims to investigate the potential value of radiomic features from multi-parameter MRI in predicting progression-free survival (PFS) of patients with WHO grade II meningiomas. Methods Kaplan-Meier survival curves were used for survival analysis of clinical features. A total of 851 radiomic features were extracted based on tumor region segmentation from each sequence, and Max-Relevance and Min-Redundancy (mRMR) algorithm was applied to filter and select radiomic features. Bagged AdaBoost, Stochastic Gradient Boosting, Random Forest, and Neural Network models were built based on selected features. Discriminative abilities of models were evaluated using receiver operating characteristics (ROC) and area under the curve (AUC). Results Our study enrolled 164 patients with WHO grade II meningiomas. Female gender (p=0.023), gross total resection (GTR) (p<0.001), age <68 years old (p=0.023), and edema index <2.3 (p=0.006) are protective factors for PFS in these patients. Both the Bagged AdaBoost model and the Neural Network model achieved the best performance on test set with an AUC of 0.927 (95% CI, Bagged AdaBoost: 0.834-1.000; Neural Network: 0.836-1.000). Conclusion The Bagged AdaBoost model and the Neural Network model based on radiomic features demonstrated decent predictive ability for PFS in patients with WHO grade II meningiomas who underwent operation using preoperative multi-parameter MR images, thus bringing benefit for patient prognosis prediction in clinical practice. Our study emphasizes the importance of utilizing advanced imaging techniques such as radiomics to improve personalized treatment strategies for meningiomas by providing more accurate prognostic information that can guide clinicians toward better decision-making processes when treating their patients' conditions effectively while minimizing risks associated with unnecessary interventions or treatments that may not be beneficial.
Collapse
Affiliation(s)
- Qiang Zeng
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Zhongyu Tian
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Fei Dong
- Department of Radiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Feina Shi
- Department of Neurology, Sir Runrun Shaw Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Penglei Xu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Chenhan Ling
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Zhige Guo
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
- Department of Neurosurgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
41
|
Appiah R, Pulletikurthi V, Esquivel-Puentes HA, Cabrera C, Hasan NI, Dharmarathne S, Gomez LJ, Castillo L. Brain tumor detection using proper orthogonal decomposition integrated with deep learning networks. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 250:108167. [PMID: 38669717 DOI: 10.1016/j.cmpb.2024.108167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/11/2024] [Accepted: 04/06/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND AND OBJECTIVE The central organ of the human nervous system is the brain, which receives and sends stimuli to the various parts of the body to engage in daily activities. Uncontrolled growth of brain cells can result in tumors which affect the normal functions of healthy brain cells. An automatic reliable technique for detecting tumors is imperative to assist medical practitioners in the timely diagnosis of patients. Although machine learning models are being used, with minimal data availability to train, development of low-order based models integrated with machine learning are a tool for reliable detection. METHODS In this study, we focus on comparing a low-order model such as proper orthogonal decomposition (POD) coupled with convolutional neural network (CNN) on 2D images from magnetic resonance imaging (MRI) scans to effectively identify brain tumors. The explainability of the coupled POD-CNN prediction output as well as the state-of-the-art pre-trained transfer learning models such as MobileNetV2, Inception-v3, ResNet101, and VGG-19 were explored. RESULTS The results showed that CNN predicted tumors with an accuracy of 99.21% whereas POD-CNN performed better with about 1/3rd of computational time at an accuracy of 95.88%. Explainable AI with SHAP showed MobileNetV2 has better prediction in identifying the tumor boundaries. CONCLUSIONS Integration of POD with CNN is carried for the first time to detect brain tumor detection with minimal MRI scan data. This study facilitates low-model approaches in machine learning to improve the accuracy and performance of tumor detection.
Collapse
Affiliation(s)
- Rita Appiah
- School of Nuclear Engineering, Purdue University, West Lafayette, IN 47906, USA.
| | | | | | - Cristiano Cabrera
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, USA
| | - Nahian I Hasan
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN 47906, USA
| | - Suranga Dharmarathne
- R.B. Annis School of Engineering, University of Indianapolis, Indianapolis, IN 46227, USA
| | - Luis J Gomez
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN 47906, USA
| | - Luciano Castillo
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47906, USA
| |
Collapse
|
42
|
Kazerooni AF, Khalili N, Liu X, Haldar D, Jiang Z, Anwar SM, Albrecht J, Adewole M, Anazodo U, Anderson H, Bagheri S, Baid U, Bergquist T, Borja AJ, Calabrese E, Chung V, Conte GM, Dako F, Eddy J, Ezhov I, Familiar A, Farahani K, Haldar S, Iglesias JE, Janas A, Johansen E, Jones BV, Kofler F, LaBella D, Lai HA, Van Leemput K, Li HB, Maleki N, McAllister AS, Meier Z, Menze B, Moawad AW, Nandolia KK, Pavaine J, Piraud M, Poussaint T, Prabhu SP, Reitman Z, Rodriguez A, Rudie JD, Shaikh IS, Shah LM, Sheth N, Shinohara RT, Tu W, Viswanathan K, Wang C, Ware JB, Wiestler B, Wiggins W, Zapaishchykova A, Aboian M, Bornhorst M, de Blank P, Deutsch M, Fouladi M, Hoffman L, Kann B, Lazow M, Mikael L, Nabavizadeh A, Packer R, Resnick A, Rood B, Vossough A, Bakas S, Linguraru MG. The Brain Tumor Segmentation (BraTS) Challenge 2023: Focus on Pediatrics (CBTN-CONNECT-DIPGR-ASNR-MICCAI BraTS-PEDs). ARXIV 2024:arXiv:2305.17033v7. [PMID: 37292481 PMCID: PMC10246083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Pediatric tumors of the central nervous system are the most common cause of cancer-related death in children. The five-year survival rate for high-grade gliomas in children is less than 20%. Due to their rarity, the diagnosis of these entities is often delayed, their treatment is mainly based on historic treatment concepts, and clinical trials require multi-institutional collaborations. The MICCAI Brain Tumor Segmentation (BraTS) Challenge is a landmark community benchmark event with a successful history of 12 years of resource creation for the segmentation and analysis of adult glioma. Here we present the CBTN-CONNECT-DIPGR-ASNR-MICCAI BraTS-PEDs 2023 challenge, which represents the first BraTS challenge focused on pediatric brain tumors with data acquired across multiple international consortia dedicated to pediatric neuro-oncology and clinical trials. The BraTS-PEDs 2023 challenge focuses on benchmarking the development of volumentric segmentation algorithms for pediatric brain glioma through standardized quantitative performance evaluation metrics utilized across the BraTS 2023 cluster of challenges. Models gaining knowledge from the BraTS-PEDs multi-parametric structural MRI (mpMRI) training data will be evaluated on separate validation and unseen test mpMRI dataof high-grade pediatric glioma. The CBTN-CONNECT-DIPGR-ASNR-MICCAI BraTS-PEDs 2023 challenge brings together clinicians and AI/imaging scientists to lead to faster development of automated segmentation techniques that could benefit clinical trials, and ultimately the care of children with brain tumors.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
- Center for AI & Data Science for Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Nastaran Khalili
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xinyang Liu
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington DC, USA
| | - Debanjan Haldar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Thomas Jefferson University Hospital, PA, USA
| | - Zhifan Jiang
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington DC, USA
| | - Syed Muhammed Anwar
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington DC, USA
| | | | - Maruf Adewole
- Medical Artificial Intelligence (MAI) Lab, Crestview Radiology, Lagos, Nigeria
| | - Udunna Anazodo
- Montreal Neurological Institute (MNI), McGill University, Montreal, QC, Canada
| | - Hannah Anderson
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sina Bagheri
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ujjwal Baid
- Center for AI & Data Science for Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Austin J. Borja
- Department of Neurosurgery at the University of Southern California, CA, USA
| | - Evan Calabrese
- Department of Radiology, Duke University Medical Center, USA
| | | | | | - Farouk Dako
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ivan Ezhov
- Department of Informatics, Technical University Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Germany
| | - Ariana Familiar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Keyvan Farahani
- Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shuvanjan Haldar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Biomedical Engineering Rutgers University, New Brunswick, NJ, USA
| | - Juan Eugenio Iglesias
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | | | - Elaine Johansen
- PrecisionFDA, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | | | | | - Dominic LaBella
- Department of Radiation Oncology, Duke University Medical Center, USA
| | - Hollie Anne Lai
- Department of Radiology, Children’s Health Orange County, CA, USA
| | - Koen Van Leemput
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Denmark
| | - Hongwei Bran Li
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | - Bjoern Menze
- Biomedical Image Analysis & Machine Learning, Department of Quantitative Biomedicine, University of Zurich, Switzerland
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
| | | | - Khanak K Nandolia
- Department of Diagnostic and Interventional Radiology, All India Institute of Medical Sciences, Rishikesh, India
| | - Julija Pavaine
- Department of Paediatric Radiology, Royal Manchester Children’s Hospital, Manchester University Hospitals NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | | | - Tina Poussaint
- Dana-Farber Brigham Cancer Center & Boston Children’s Hospital, Boston, MA, USA
| | - Sanjay P Prabhu
- Division of Neuroradiology, Department of Radiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zachary Reitman
- Department of Radiation Oncology, Duke University Medical Center, USA
| | - Andres Rodriguez
- Department of Diagnostic & Interventional Imaging, Neuroradiology section. The University of Texas Health Sciences Center at Houston
| | | | | | | | - Nakul Sheth
- Department of Radiology, The Children’s Hospital of Philadelphia, PA, USA
| | - Russel Taki Shinohara
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, USA
| | - Wenxin Tu
- College of Arts and Sciences, University of Pennsylvania, PA, USA
| | - Karthik Viswanathan
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Chunhao Wang
- Department of Radiation Oncology, Duke University Medical Center, USA
| | - Jeffrey B Ware
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benedikt Wiestler
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
| | - Walter Wiggins
- Department of Radiology, Duke University Medical Center, USA
| | - Anna Zapaishchykova
- Dana-Farber Brigham Cancer Center & Boston Children’s Hospital, Boston, MA, USA
| | | | - Miriam Bornhorst
- Brain Tumor Institute, Children’s National Hospital, Washington, DC, USA
| | - Peter de Blank
- Brain Tumor Center, Cincinnati Children’s Hospital, Cincinnati, OH, USA
| | - Michelle Deutsch
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, OH, US
| | - Maryam Fouladi
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, OH, US
| | - Lindsey Hoffman
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, AZ, US
| | - Benjamin Kann
- Dana-Farber Brigham Cancer Center & Boston Children’s Hospital, Boston, MA, USA
| | - Margot Lazow
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, OH, US
| | - Leonie Mikael
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, OH, US
| | - Ali Nabavizadeh
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roger Packer
- Brain Tumor Institute, Children’s National Hospital, Washington, DC, USA
| | - Adam Resnick
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian Rood
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC, USA
| | - Arastoo Vossough
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, The Children’s Hospital of Philadelphia, PA, USA
| | - Spyridon Bakas
- Center for AI & Data Science for Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marius George Linguraru
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington DC, USA
- Departments of Radiology and Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
43
|
LaBella D, Khanna O, McBurney-Lin S, Mclean R, Nedelec P, Rashid AS, Tahon NH, Altes T, Baid U, Bhalerao R, Dhemesh Y, Floyd S, Godfrey D, Hilal F, Janas A, Kazerooni A, Kent C, Kirkpatrick J, Kofler F, Leu K, Maleki N, Menze B, Pajot M, Reitman ZJ, Rudie JD, Saluja R, Velichko Y, Wang C, Warman PI, Sollmann N, Diffley D, Nandolia KK, Warren DI, Hussain A, Fehringer JP, Bronstein Y, Deptula L, Stein EG, Taherzadeh M, Portela de Oliveira E, Haughey A, Kontzialis M, Saba L, Turner B, Brüßeler MMT, Ansari S, Gkampenis A, Weiss DM, Mansour A, Shawali IH, Yordanov N, Stein JM, Hourani R, Moshebah MY, Abouelatta AM, Rizvi T, Willms K, Martin DC, Okar A, D'Anna G, Taha A, Sharifi Y, Faghani S, Kite D, Pinho M, Haider MA, Alonso-Basanta M, Villanueva-Meyer J, Rauschecker AM, Nada A, Aboian M, Flanders A, Bakas S, Calabrese E. A multi-institutional meningioma MRI dataset for automated multi-sequence image segmentation. Sci Data 2024; 11:496. [PMID: 38750041 PMCID: PMC11096318 DOI: 10.1038/s41597-024-03350-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024] Open
Abstract
Meningiomas are the most common primary intracranial tumors and can be associated with significant morbidity and mortality. Radiologists, neurosurgeons, neuro-oncologists, and radiation oncologists rely on brain MRI for diagnosis, treatment planning, and longitudinal treatment monitoring. However, automated, objective, and quantitative tools for non-invasive assessment of meningiomas on multi-sequence MR images are not available. Here we present the BraTS Pre-operative Meningioma Dataset, as the largest multi-institutional expert annotated multilabel meningioma multi-sequence MR image dataset to date. This dataset includes 1,141 multi-sequence MR images from six sites, each with four structural MRI sequences (T2-, T2/FLAIR-, pre-contrast T1-, and post-contrast T1-weighted) accompanied by expert manually refined segmentations of three distinct meningioma sub-compartments: enhancing tumor, non-enhancing tumor, and surrounding non-enhancing T2/FLAIR hyperintensity. Basic demographic data are provided including age at time of initial imaging, sex, and CNS WHO grade. The goal of releasing this dataset is to facilitate the development of automated computational methods for meningioma segmentation and expedite their incorporation into clinical practice, ultimately targeting improvement in the care of meningioma patients.
Collapse
Affiliation(s)
- Dominic LaBella
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Omaditya Khanna
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Shan McBurney-Lin
- Center for Intelligent Imaging (ci2), Department of Radiology & Biomedical Imaging, University of California San Francisco (UCSF), San Francisco, CA, USA
| | | | - Pierre Nedelec
- Center for Intelligent Imaging (ci2), Department of Radiology & Biomedical Imaging, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Arif S Rashid
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Ujjwal Baid
- Division of Computational Pathology, Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Radhika Bhalerao
- Center for Intelligent Imaging (ci2), Department of Radiology & Biomedical Imaging, University of California San Francisco (UCSF), San Francisco, CA, USA
| | | | - Scott Floyd
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Devon Godfrey
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | | | | | - Anahita Kazerooni
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Collin Kent
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - John Kirkpatrick
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Florian Kofler
- Helmholtz AI, Helmholtz Munich, Neuherberg, Germany
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Tech nical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kevin Leu
- Center for Intelligent Imaging (ci2), Department of Radiology & Biomedical Imaging, University of California San Francisco (UCSF), San Francisco, CA, USA
| | | | | | - Maxence Pajot
- Center for Intelligent Imaging (ci2), Department of Radiology & Biomedical Imaging, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Zachary J Reitman
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey D Rudie
- Department of Radiology, University of California San Diego, San Diego, CA, USA
| | - Rachit Saluja
- Department of Radiology, Cornell University, Ithaca, NY, USA
| | - Yury Velichko
- Department of Radiology, Northwestern University, Evanston, IL, USA
| | - Chunhao Wang
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Pranav I Warman
- Duke University Medical Center, School of Medicine, Durham, NC, USA
| | - Nico Sollmann
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | - Khanak K Nandolia
- Department of Diagnostic and Interventional Radiology, All India Institute of Medical Sciences, Rishikesh, India
| | - Daniel I Warren
- Department of Neuroradiology, Washington University, St. Louis, MO, USA
| | - Ali Hussain
- University of Rochester Medical Center, Rochester, NY, USA
| | - John Pascal Fehringer
- Faculty of Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | | | - Lisa Deptula
- Ross University School of Medicine, Bridgetown, Barbados
| | - Evan G Stein
- Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
| | | | | | - Aoife Haughey
- Department of Neuroradiology, JDMI, University of Toronto, Toronto, TO, Canada
| | | | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria of Cagliari-Polo di Monserrato, Cagliari, Italy
| | | | | | | | | | - David Maximilian Weiss
- Department of Neuroradiology, University Hospital Essen, Essen, North Rhine-Westphalia, Germany
| | | | - Islam H Shawali
- Department of Radiology, Kasr Alainy, Cairo University, Cairo, Egypt
| | - Nikolay Yordanov
- Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Joel M Stein
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roula Hourani
- Department of Radiology, American University of Beirut Medical center, Beirut, Lebanon
| | | | | | - Tanvir Rizvi
- Department of Radiology and Medical Imaging, University of Virginia Health, Charlottesville, VA, USA
| | | | - Dann C Martin
- Department of Radiology and Radiologic Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Abdullah Okar
- Faculty of Medicine, Hamburg University, Hamburg, Germany
| | - Gennaro D'Anna
- Neuroimaging Unit, ASST Ovest Milanese, Legnano, Milan, Italy
| | - Ahmed Taha
- University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yasaman Sharifi
- Department of Radiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shahriar Faghani
- Radiology Informatics Lab, Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Dominic Kite
- Department of Radiology, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Marco Pinho
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Michelle Alonso-Basanta
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Javier Villanueva-Meyer
- Center for Intelligent Imaging (ci2), Department of Radiology & Biomedical Imaging, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Andreas M Rauschecker
- Center for Intelligent Imaging (ci2), Department of Radiology & Biomedical Imaging, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Ayman Nada
- University of Missouri, Columbia, MO, USA
| | - Mariam Aboian
- Department of Radiology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Adam Flanders
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Spyridon Bakas
- Division of Computational Pathology, Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN, USA
- Department of Neurological Surgery, School of Medicine, Indiana University, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Evan Calabrese
- Department of Radiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
44
|
Koçak B, Yüzkan S, Mutlu S, Karagülle M, Kala A, Kadıoğlu M, Solak S, Sunman Ş, Temiz ZH, Ganiyusufoğlu AK. Influence of image preprocessing on the segmentation-based reproducibility of radiomic features: in vivo experiments on discretization and resampling parameters. Diagn Interv Radiol 2024; 30:152-162. [PMID: 38073244 PMCID: PMC11095065 DOI: 10.4274/dir.2023.232543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/14/2023] [Indexed: 05/15/2024]
Abstract
PURPOSE To systematically investigate the impact of image preprocessing parameters on the segmentation-based reproducibility of magnetic resonance imaging (MRI) radiomic features. METHODS The MRI scans of 50 patients were included from the multi-institutional Brain Tumor Segmentation 2021 public glioma dataset. Whole tumor volumes were manually segmented by two independent readers, with the participation of eight readers. Radiomic features were extracted from two sequences: T2-weighted (T2) and contrast-enhanced T1-weighted (T1ce). Two methods were considered for discretization: bin count (i.e., relative discretization) and bin width (i.e., absolute discretization). Ten discretization (five for each method) and five resampling parameters were varied while other parameters were fixed. The intraclass correlation coefficient (ICC) was used for reliability analysis based on two commonly used cut-off values (0.75 and 0.90). RESULTS Image preprocessing parameters had a significant impact on the segmentation-based reproducibility of radiomic features. The bin width method yielded more reproducible features than the bin count method. In discretization experiments using the bin width on both sequences, according to the ICC cut-off values of 0.75 and 0.90, the rate of reproducible features ranged from 70% to 84% and from 35% to 57%, respectively, with an increasing percentage trend as parameter values decreased (from 84 to 5 for T2; 100 to 6 for T1ce). In the resampling experiments, these ranged from 53% to 74% and from 10% to 20%, respectively, with an increasing percentage trend from lower to higher parameter values (physical voxel size; from 1 x 1 x 1 to 2 x 2 x 2 mm3). CONCLUSION The segmentation-based reproducibility of radiomic features appears to be substantially influenced by discretization and resampling parameters. Our findings indicate that the bin width method should be used for discretization and lower bin width and higher resampling values should be used to allow more reproducible features.
Collapse
Affiliation(s)
- Burak Koçak
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Sabahattin Yüzkan
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Samet Mutlu
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Mehmet Karagülle
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Ahmet Kala
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Mehmet Kadıoğlu
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Sıla Solak
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Şeyma Sunman
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Zişan Hayriye Temiz
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Ali Kürşad Ganiyusufoğlu
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| |
Collapse
|
45
|
Chirokoff V, Pohl KM, Berthoz S, Fatseas M, Misdrahi D, Serre F, Auriacombe M, Pfefferbaum A, Sullivan EV, Chanraud S. Multi-level prediction of substance use: Interaction of white matter integrity, resting-state connectivity and inhibitory control measured repeatedly in every-day life. Addict Biol 2024; 29:e13400. [PMID: 38706091 PMCID: PMC11070496 DOI: 10.1111/adb.13400] [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: 10/03/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024]
Abstract
Substance use disorders are characterized by inhibition deficits related to disrupted connectivity in white matter pathways, leading via interaction to difficulties in resisting substance use. By combining neuroimaging with smartphone-based ecological momentary assessment (EMA), we questioned how biomarkers moderate inhibition deficits to predict use. Thus, we aimed to assess white matter integrity interaction with everyday inhibition deficits and related resting-state network connectivity to identify multi-dimensional predictors of substance use. Thirty-eight patients treated for alcohol, cannabis or tobacco use disorder completed 1 week of EMA to report substance use five times and complete Stroop inhibition testing twice daily. Before EMA tracking, participants underwent resting state functional MRI and diffusion tensor imaging (DTI) scanning. Regression analyses were conducted between mean Stroop performances and whole-brain fractional anisotropy (FA) in white matter. Moderation testing was conducted between mean FA within significant clusters as moderator and the link between momentary Stroop performance and use as outcome. Predictions between FA and resting-state connectivity strength in known inhibition-related networks were assessed using mixed modelling. Higher FA values in the anterior corpus callosum and bilateral anterior corona radiata predicted higher mean Stroop performance during the EMA week and stronger functional connectivity in occipital-frontal-cerebellar regions. Integrity in these regions moderated the link between inhibitory control and substance use, whereby stronger inhibition was predictive of the lowest probability of use for the highest FA values. In conclusion, compromised white matter structural integrity in anterior brain systems appears to underlie impairment in inhibitory control functional networks and compromised ability to refrain from substance use.
Collapse
Affiliation(s)
- Valentine Chirokoff
- Univ. Bordeaux, INCIA CNRS‐UMR 5287BordeauxFrance
- EPHEPSL Research UniversityParisFrance
| | - Kilian M. Pohl
- Department of Psychiatry & Behavioral SciencesStanford University School of MedicineStanfordCaliforniaUSA
| | - Sylvie Berthoz
- Univ. Bordeaux, INCIA CNRS‐UMR 5287BordeauxFrance
- Department of Psychiatry for Adolescents and Young AdultsInstitut Mutualiste MontsourisParisFrance
| | - Melina Fatseas
- Univ. Bordeaux, INCIA CNRS‐UMR 5287BordeauxFrance
- CH Charles PerrensBordeauxFrance
- CHU BordeauxBordeauxFrance
| | - David Misdrahi
- Univ. Bordeaux, INCIA CNRS‐UMR 5287BordeauxFrance
- CH Charles PerrensBordeauxFrance
| | - Fuschia Serre
- CNRS UMR 6033 – Sleep, Addiction and Neuropsychiatry (SANPSY)University of BordeauxBordeauxFrance
| | - Marc Auriacombe
- CH Charles PerrensBordeauxFrance
- CNRS UMR 6033 – Sleep, Addiction and Neuropsychiatry (SANPSY)University of BordeauxBordeauxFrance
| | - Adolf Pfefferbaum
- Department of Psychiatry & Behavioral SciencesStanford University School of MedicineStanfordCaliforniaUSA
- Center for Health SciencesSRI InternationalMenlo ParkCaliforniaUSA
| | - Edith V. Sullivan
- Department of Psychiatry & Behavioral SciencesStanford University School of MedicineStanfordCaliforniaUSA
| | - Sandra Chanraud
- Univ. Bordeaux, INCIA CNRS‐UMR 5287BordeauxFrance
- EPHEPSL Research UniversityParisFrance
| |
Collapse
|
46
|
Bathla G, Dhruba DD, Liu Y, Le NH, Soni N, Zhang H, Mohan S, Roberts-Wolfe D, Rathore S, Sonka M, Priya S, Agarwal A. Differentiation Between Glioblastoma and Metastatic Disease on Conventional MRI Imaging Using 3D-Convolutional Neural Networks: Model Development and Validation. Acad Radiol 2024; 31:2041-2049. [PMID: 37977889 DOI: 10.1016/j.acra.2023.10.044] [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: 08/13/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023]
Abstract
RATIONALE AND OBJECTIVES Imaging-based differentiation between glioblastoma (GB) and brain metastases (BM) remains challenging. Our aim was to evaluate the performance of 3D-convolutional neural networks (CNN) to address this binary classification problem. MATERIALS AND METHODS T1-CE, T2WI, and FLAIR 3D-segmented masks of 307 patients (157 GB and 150 BM) were generated post resampling, co-registration normalization and semi-automated 3D-segmentation and used for internal model development. Subsequent external validation was performed on 59 cases (27 GB and 32 BM) from another institution. Four different mask-sequence combinations were evaluated using area under the curve (AUC), precision, recall and F1-scores. Diagnostic performance of a neuroradiologist and a general radiologist, both without and with the model output available, was also assessed. RESULTS 3D-model using the T1-CE tumor mask (TM) showed the highest performance [AUC 0.93 (95% CI 0.858-0.995)] on the external test set, followed closely by the model using T1-CE TM and FLAIR mask of peri-tumoral region (PTR) [AUC of 0.91 (95% CI 0.834-0.986)]. Models using T2WI masks showed robust performance on the internal dataset but lower performance on the external set. Both neuroradiologist and general radiologist showed improved performance with model output provided [AUC increased from 0.89 to 0.968 (p = 0.06) and from 0.78 to 0.965 (p = 0.007) respectively], the latter being statistically significant. CONCLUSION 3D-CNNs showed robust performance for differentiating GB from BMs, with T1-CE TM, either alone or combined with FLAIR-PTR masks. Availability of model output significantly improved the accuracy of the general radiologist.
Collapse
Affiliation(s)
- Girish Bathla
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA (G.B., N.S., S.P.); Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA (G.B.)
| | - Durjoy Deb Dhruba
- Electrical and Computer Engineering, University of Iowa, Iowa City, Iowa, USA (D.D.D.).
| | - Yanan Liu
- College of Engineering, University of Iowa, Iowa City, Iowa, USA (Y.L., N.H.L., H.Z., M.S.)
| | - Nam H Le
- College of Engineering, University of Iowa, Iowa City, Iowa, USA (Y.L., N.H.L., H.Z., M.S.)
| | - Neetu Soni
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA (G.B., N.S., S.P.); Department of Radiology, Mayo Clinic, Jacksonville, Florida, USA (N.S., A.A.)
| | - Honghai Zhang
- College of Engineering, University of Iowa, Iowa City, Iowa, USA (Y.L., N.H.L., H.Z., M.S.)
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA (S.M., D.R.W.)
| | - Douglas Roberts-Wolfe
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA (S.M., D.R.W.)
| | - Saima Rathore
- Senior research scientist, Avid Radiopharmaceuticals, Philadelphia, Pennsylvania, USA (S.R.)
| | - Milan Sonka
- College of Engineering, University of Iowa, Iowa City, Iowa, USA (Y.L., N.H.L., H.Z., M.S.)
| | - Sarv Priya
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA (G.B., N.S., S.P.)
| | - Amit Agarwal
- Department of Radiology, Mayo Clinic, Jacksonville, Florida, USA (N.S., A.A.)
| |
Collapse
|
47
|
Safari M, Eidex Z, Chang CW, Qiu RL, Yang X. Fast MRI Reconstruction Using Deep Learning-based Compressed Sensing: A Systematic Review. ARXIV 2024:arXiv:2405.00241v1. [PMID: 38745700 PMCID: PMC11092677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Magnetic resonance imaging (MRI) has revolutionized medical imaging, providing a non-invasive and highly detailed look into the human body. However, the long acquisition times of MRI present challenges, causing patient discomfort, motion artifacts, and limiting real-time applications. To address these challenges, researchers are exploring various techniques to reduce acquisition time and improve the overall efficiency of MRI. One such technique is compressed sensing (CS), which reduces data acquisition by leveraging image sparsity in transformed spaces. In recent years, deep learning (DL) has been integrated with CS-MRI, leading to a new framework that has seen remarkable growth. DL-based CS-MRI approaches are proving to be highly effective in accelerating MR imaging without compromising image quality. This review comprehensively examines DL-based CS-MRI techniques, focusing on their role in increasing MR imaging speed. We provide a detailed analysis of each category of DL-based CS-MRI including end-to-end, unroll optimization, self-supervised, and federated learning. Our systematic review highlights significant contributions and underscores the exciting potential of DL in CS-MRI. Additionally, our systematic review efficiently summarizes key results and trends in DL-based CS-MRI including quantitative metrics, the dataset used, acceleration factors, and the progress of and research interest in DL techniques over time. Finally, we discuss potential future directions and the importance of DL-based CS-MRI in the advancement of medical imaging. To facilitate further research in this area, we provide a GitHub repository that includes up-to-date DL-based CS-MRI publications and publicly available datasets - https://github.com/mosaf/Awesome-DL-based-CS-MRI.
Collapse
Affiliation(s)
- Mojtaba Safari
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States of America
| | - Zach Eidex
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States of America
| | - Chih-Wei Chang
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States of America
| | - Richard L.J. Qiu
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States of America
| | - Xiaofeng Yang
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States of America
| |
Collapse
|
48
|
Jung K, Kempter J, Prokop G, Herrmann T, Griessmair M, Kim SH, Delbridge C, Meyer B, Bernhardt D, Combs SE, Zimmer C, Wiestler B, Schmidt-Graf F, Metz MC. Quantitative Assessment of Tumor Contact with Neurogenic Zones and Its Effects on Survival: Insights beyond Traditional Predictors. Cancers (Basel) 2024; 16:1743. [PMID: 38730694 PMCID: PMC11083354 DOI: 10.3390/cancers16091743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
So far, the cellular origin of glioblastoma (GBM) needs to be determined, with prevalent theories suggesting emergence from transformed endogenous stem cells. Adult neurogenesis primarily occurs in two brain regions: the subventricular zone (SVZ) and the subgranular zone (SGZ) of the hippocampal dentate gyrus. Whether the proximity of GBM to these neurogenic niches affects patient outcome remains uncertain. Previous studies often rely on subjective assessments, limiting the reliability of those results. In this study, we assessed the impact of GBM's relationship with the cortex, SVZ and SGZ on clinical variables using fully automated segmentation methods. In 177 glioblastoma patients, we calculated optimal cutpoints of minimal distances to the SVZ and SGZ to distinguish poor from favorable survival. The impact of tumor contact with neurogenic zones on clinical parameters, such as overall survival, multifocality, MGMT promotor methylation, Ki-67 and KPS score was also examined by multivariable regression analysis, chi-square test and Mann-Whitney-U. The analysis confirmed shorter survival in tumors contacting the SVZ with an optimal cutpoint of 14 mm distance to the SVZ, separating poor from more favorable survival. In contrast, tumor contact with the SGZ did not negatively affect survival. We did not find significant correlations with multifocality or MGMT promotor methylation in tumors contacting the SVZ, as previous studies discussed. These findings suggest that the spatial relationship between GBM and neurogenic niches needs to be assessed differently. Objective measurements disprove prior assumptions, warranting further research on this topic.
Collapse
Affiliation(s)
- Kirsten Jung
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (T.H.); (M.G.); (S.-H.K.); (C.Z.); (B.W.); (M.-C.M.)
| | - Johanna Kempter
- Department of Neurology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (J.K.); (G.P.); (F.S.-G.)
| | - Georg Prokop
- Department of Neurology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (J.K.); (G.P.); (F.S.-G.)
| | - Tim Herrmann
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (T.H.); (M.G.); (S.-H.K.); (C.Z.); (B.W.); (M.-C.M.)
| | - Michael Griessmair
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (T.H.); (M.G.); (S.-H.K.); (C.Z.); (B.W.); (M.-C.M.)
| | - Su-Hwan Kim
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (T.H.); (M.G.); (S.-H.K.); (C.Z.); (B.W.); (M.-C.M.)
| | - Claire Delbridge
- Department of Pathology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany;
| | - Bernhard Meyer
- Department of Neurosurgery, School of Medicine and Health, Technical University of Munich, 81675 München, Germany
| | - Denise Bernhardt
- Department of Radiation Oncology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (D.B.); (S.E.C.)
| | - Stephanie E. Combs
- Department of Radiation Oncology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (D.B.); (S.E.C.)
| | - Claus Zimmer
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (T.H.); (M.G.); (S.-H.K.); (C.Z.); (B.W.); (M.-C.M.)
| | - Benedikt Wiestler
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (T.H.); (M.G.); (S.-H.K.); (C.Z.); (B.W.); (M.-C.M.)
- TranslaTUM, Technical University of Munich, 81675 München, Germany
| | - Friederike Schmidt-Graf
- Department of Neurology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (J.K.); (G.P.); (F.S.-G.)
| | - Marie-Christin Metz
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (T.H.); (M.G.); (S.-H.K.); (C.Z.); (B.W.); (M.-C.M.)
| |
Collapse
|
49
|
Kobets AJ, Alavi SAN, Ahmad SJ, Castillo A, Young D, Minuti A, Altschul DJ, Zhu M, Abbott R. Volumetric segmentation in the context of posterior fossa-related pathologies: a systematic review. Neurosurg Rev 2024; 47:170. [PMID: 38637466 PMCID: PMC11026186 DOI: 10.1007/s10143-024-02366-4] [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: 01/28/2024] [Revised: 03/04/2024] [Accepted: 03/16/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Segmentation tools continue to advance, evolving from manual contouring to deep learning. Researchers have utilized segmentation to study a myriad of posterior fossa-related conditions, such as Chiari malformation, trigeminal neuralgia, post-operative pediatric cerebellar mutism syndrome, and Crouzon syndrome. Herein, we present a summary of the current literature on segmentation of the posterior fossa. The review highlights the various segmentation techniques, and their respective strengths and weaknesses, employed along with objectives and outcomes of the various studies reported in the literature. METHODS A literature search was conducted in PubMed, Embase, Cochrane, and Web of Science up to November 2023 for articles on segmentation techniques of posterior fossa. The two senior authors searched through databases based on the keywords of the article separately and then enrolled joint articles that met the inclusion and exclusion criteria. RESULTS The initial search identified 2205 articles. After applying inclusion and exclusion criteria, 77 articles were selected for full-text review after screening of titles/abstracts. 52 articles were ultimately included in the review. Segmentation techniques included manual, semi-automated, and fully automated (atlas-based, convolutional neural networks). The most common pathology investigated was Chiari malformation. CONCLUSIONS Various forms of segmentation techniques have been used to assess posterior fossa volumes/pathologies and each has its advantages and disadvantages. We discuss these nuances and summarize the current state of literature in the context of posterior fossa-associated pathologies.
Collapse
Affiliation(s)
- Andrew J Kobets
- Department of Neurological Surgery, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, NY, 10467, USA
| | - Seyed Ahmad Naseri Alavi
- Department of Neurological Surgery, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, NY, 10467, USA.
| | | | | | | | | | - David J Altschul
- Department of Neurological Surgery, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, NY, 10467, USA
| | - Michael Zhu
- Albert Einstein College of Medicine, New York City, USA
| | - Rick Abbott
- Department of Neurological Surgery, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, NY, 10467, USA
| |
Collapse
|
50
|
Wiestler B, Bison B, Behrens L, Tüchert S, Metz M, Griessmair M, Jakob M, Schlegel PG, Binder V, von Luettichau I, Metzler M, Johann P, Hau P, Frühwald M. Human-Level Differentiation of Medulloblastoma from Pilocytic Astrocytoma: A Real-World Multicenter Pilot Study. Cancers (Basel) 2024; 16:1474. [PMID: 38672556 PMCID: PMC11048511 DOI: 10.3390/cancers16081474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Medulloblastoma and pilocytic astrocytoma are the two most common pediatric brain tumors with overlapping imaging features. In this proof-of-concept study, we investigated using a deep learning classifier trained on a multicenter data set to differentiate these tumor types. We developed a patch-based 3D-DenseNet classifier, utilizing automated tumor segmentation. Given the heterogeneity of imaging data (and available sequences), we used all individually available preoperative imaging sequences to make the model robust to varying input. We compared the classifier to diagnostic assessments by five readers with varying experience in pediatric brain tumors. Overall, we included 195 preoperative MRIs from children with medulloblastoma (n = 69) or pilocytic astrocytoma (n = 126) across six university hospitals. In the 64-patient test set, the DenseNet classifier achieved a high AUC of 0.986, correctly predicting 62/64 (97%) diagnoses. It misclassified one case of each tumor type. Human reader accuracy ranged from 100% (expert neuroradiologist) to 80% (resident). The classifier performed significantly better than relatively inexperienced readers (p < 0.05) and was on par with pediatric neuro-oncology experts. Our proof-of-concept study demonstrates a deep learning model based on automated tumor segmentation that can reliably preoperatively differentiate between medulloblastoma and pilocytic astrocytoma, even in heterogeneous data.
Collapse
Affiliation(s)
- Benedikt Wiestler
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675 Munich, Germany (M.G.)
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, 81675 Munich, Germany
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
| | - Brigitte Bison
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
- KIONET, Kinderonkologisches Netzwerk Bayern
- Diagnostic and Interventional Neuroradiology, Faculty of Medicine, University Hospital Augsburg, 86156 Augsburg, Germany; (B.B.); (L.B.)
- Neuroradiological Reference Center for the Pediatric Brain Tumor (HIT) Studies of the German Society of Pediatric Oncology and Hematology, Faculty of Medicine, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Lars Behrens
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
- KIONET, Kinderonkologisches Netzwerk Bayern
- Diagnostic and Interventional Neuroradiology, Faculty of Medicine, University Hospital Augsburg, 86156 Augsburg, Germany; (B.B.); (L.B.)
- Neuroradiological Reference Center for the Pediatric Brain Tumor (HIT) Studies of the German Society of Pediatric Oncology and Hematology, Faculty of Medicine, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Stefanie Tüchert
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Marie Metz
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675 Munich, Germany (M.G.)
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
| | - Michael Griessmair
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, 81675 Munich, Germany (M.G.)
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
| | - Marcus Jakob
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
- KIONET, Kinderonkologisches Netzwerk Bayern
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, 93053 Regensburg, Germany;
| | - Paul-Gerhardt Schlegel
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
- KIONET, Kinderonkologisches Netzwerk Bayern
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital Würzburg, 97080 Würzburg, Germany;
| | - Vera Binder
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
- KIONET, Kinderonkologisches Netzwerk Bayern
- Department of Pediatrics, Dr. Von Hauner Children’s Hospital, University Hospital, LMU Munich, 80539 Munich, Germany;
| | - Irene von Luettichau
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
- KIONET, Kinderonkologisches Netzwerk Bayern
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Kinderklinik München Schwabing, Children’s Cancer Research Center, TUM School of Medicine and Health, Technical University of Munich, 80333 Munich, Germany;
| | - Markus Metzler
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
- KIONET, Kinderonkologisches Netzwerk Bayern
- Pediatric Oncology and Hematology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany;
| | - Pascal Johann
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
- KIONET, Kinderonkologisches Netzwerk Bayern
- Swabian Children’s Cancer Center, Pediatrics and Adolescent Medicine, University Hospital Augsburg, 86156 Augsburg, Germany; (P.J.); (M.F.)
| | - Peter Hau
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Michael Frühwald
- Study Groups on CNS Tumors Within the Bavarian Cancer Research Center (BZKF)
- KIONET, Kinderonkologisches Netzwerk Bayern
- Swabian Children’s Cancer Center, Pediatrics and Adolescent Medicine, University Hospital Augsburg, 86156 Augsburg, Germany; (P.J.); (M.F.)
| |
Collapse
|