1
|
Ghosh A, Li H, Towbin A, Turpin B, Trout A. T2-weighted MRI radiomics for the prediction of pediatric and young adult rhabdomyosarcoma alveolar subtype and distant metastasis: a pilot study. Pediatr Radiol 2025:10.1007/s00247-025-06205-6. [PMID: 40100409 DOI: 10.1007/s00247-025-06205-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025]
Abstract
INTRODUCTION Rhabdomyosarcomas are the most common soft tissue sarcoma in children. While treatment outcomes have improved, risk-based therapy classification relies on staging and tumor subtypes for therapeutic planning. OBJECTIVE This study investigated the utility of T2-weighted MR radiomics features and machine learning models in identifying the presence of distant metastasis and alveolar histological subtypes at baseline imaging in children diagnosed with rhabdomyosarcoma. MATERIALS AND METHODS This retrospective cross-sectional study utilized MRIs from 86 patients, 49 (median age (IQR) 59 months (37-161), alveolar subtype=15, distant metastasis=9) of whom had been imaged at outside imaging centers (training set); and 37 (median age 52 months (24-164), alveolar subtype=14, distant metastasis=8) of whom were imaged at our institution (holdout validation set). Radiomic features were extracted from T2-weighted images. We selected features that demonstrated intra-scan repeatability and used maximum relevance and minimum redundancy supervised feature selection to identify the 50 most important features. Lasso logistic regression and support vector machine (SVM) classifiers were trained to predict binary outcomes. The median of all predictions for a given patient was used as patient-level predictions. DeLong's test compared the area under the receiver operating characteristic curves (AUC). Cut-offs obtained by maximizing the Youden index were evaluated on an external validation set, and accuracy metrics were reported. RESULTS On the validation set, the Lasso and SVM classifiers obtained patient level AUCs of 0.76 (95% CI 0.59-0.94) and 0.73 (0.54-0.92), respectively, in predicting alveolar subtype, with the Lasso regressor obtaining 71.4% (41.9-91.6) sensitivity and 60.9% (38.5-80.3) specificity. When predicting the presence of distant metastasis, the Lasso and SVM classifier had AUCs of 0.81 (0.67-0.95) and 0.77 (0.58-0.97), respectively. There were no differences between model performance (P>0.05). A total of 12 and 18 features had nonzero coefficients in the Lasso regressors for predicting alveolar subtype and tumor metastasis, respectively. CONCLUSION MRI radiomics from baseline T2-weighted MRI demonstrated potential in predicting alveolar subtype and distant metastatic disease at presentation. Larger studies are needed to explore multinomial multiclass models for better prognostication of pediatric rhabdomyosarcomas.
Collapse
Affiliation(s)
- Adarsh Ghosh
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA.
- Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA.
| | - Hailong Li
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | | | - Brian Turpin
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Andrew Trout
- Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| |
Collapse
|
2
|
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
|
3
|
Pogosbekyan E, Zakharova N, Batalov A, Shevchenko A, Fadeeva L, Bykanov A, Tyurina A, Chekhonin I, Galstyan S, Pitskhelauri D, Pronin I, Usachev D. Individual Brain Tumor Invasion Mapping Based on Diffusion Kurtosis Imaging. Sovrem Tekhnologii Med 2025; 17:81-90. [PMID: 40071079 PMCID: PMC11892574 DOI: 10.17691/stm2025.17.1.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Indexed: 03/14/2025] Open
Abstract
The aim of the investigation is to develop and implement an algorithm for image analysis in brain tumors (glioblastoma and metastasis) based on diffusion kurtosis MRI images (DKI) for the assessment of anisotropic changes in brain tissues in the directions from the tumor to the intact (as shown by the standard MRI data) white matter, which will enable generating individual tumor invasion maps. Materials and Methods A healthy volunteer and two patients (one with glioblastoma and the other with a single metastasis of small cell lung cancer) were examined by DKI obtaining 12 parametric kurtosis maps for each participant. Results During the investigation, we have developed an algorithm of DKI analysis and plotting the profile of tissue parameters in the direction from the tumor towards the unaffected white matter according to the data of standard MRI. Changes of the DKI indicators along the trajectories built using the proposed algorithm in the perifocal zone of glioblastoma and metastasis have been compared in this work. We obtained not only changes in the parameters (gradients in trajectory plots) but also a visual reflection (on color maps) of a known pathomorphology of the process - no significant gradients of DKI parameters were detected in the perifocal metastasis edema, since there was a pure vasogenic edema and no infiltrative component. In glioblastoma, gradients of DKI parameters were found not only in the zone of perifocal edema but beyond the zone of MR signal as well, which is believed to reflect diffusion disorders along the white matter fibers and different degrees of brain tissue infiltration by glioblastoma cells. Conclusion The developed algorithm of DKI analysis in brain tumors makes it possible to determine the degree of changes in the tissue microstructure in the perifocal zone of brain glioblastoma relative to the metastasis. The study aimed at obtaining individual maps of tumor invasion, which will be applied in planning neurosurgical and radiation treatment and for predicting directions of further growth of malignant gliomas.
Collapse
Affiliation(s)
- E.L. Pogosbekyan
- Medical Physicist, Department of Neuroradiology; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - N.E. Zakharova
- MD, DSc, Professor of the Russian Academy of Sciences, Chief Researcher, Department of Neuroradiology; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - A.I. Batalov
- MD, PhD, Researcher, Department of Neuroradiology; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - A.M. Shevchenko
- Radiologist, Department of Neuroradiology; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - L.M. Fadeeva
- Leading Engineer, Department of Neuroradiology; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - A.E. Bykanov
- MD, PhD, Researcher, Neurosurgery Department No.7; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - A.N. Tyurina
- MD, PhD, Researcher, Department of Neuroradiology; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - I.V. Chekhonin
- MD, PhD, Radiologist, Department of Neuroradiology; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - S.A. Galstyan
- Pathologist, Department of Pathology; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - D.I. Pitskhelauri
- MD, DSc, Professor, Head of Neurosurgery Department No.7; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - I.N. Pronin
- MD, DSc, Professor, Academician of the Russian Academy of Sciences, Head of the Department of Neuroradiology; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| | - D.Yu. Usachev
- MD, DSc, Professor, Academician of the Russian Academy of Sciences, Director; N.N. Burdenko National Medical Research Center of Neurosurgery of the Ministry of Health of the Russian Federation, 16, 4 Tverskaya-Yamskaya St., Moscow, 125047, Russia
| |
Collapse
|
4
|
Turrisi R, Pati S, Pioggia G, Tartarisco G. Adapting to evolving MRI data: A transfer learning approach for Alzheimer's disease prediction. Neuroimage 2025; 307:121016. [PMID: 39826774 DOI: 10.1016/j.neuroimage.2025.121016] [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: 07/25/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Integrating 3D magnetic resonance imaging (MRI) with machine learning has shown promising results in healthcare, especially in detecting Alzheimer's Disease (AD). However, changes in MRI technologies and acquisition protocols often yield limited data, leading to potential overfitting. This study explores Transfer Learning (TL) approaches to enhance AD diagnosis using a Baseline model consisting of a 3D-Convolutional Neural Network trained on 80 3T MRI scans. Two scenarios are explored: (A) utilizing historical data to address changes in MRI acquisitions (from 1.5T to 3T MRI), and (B) adapting 2D models pre-trained on ImageNet (ResNet18, ResNet50, ResNet101) for 3D image processing when historical data is unavailable. In both scenarios, two modeling approaches are tested. The General Approach involves distinct feature extraction and classification steps, using Radiomic features and TL-based features evaluated with six classifiers. The Deep Approach integrates these steps by fine-tuning the pre-trained models for AD diagnosis. In scenario (A), TL significantly boosts the Baseline's accuracy from 63% to 99%. In scenario (B), Radiomic features better represents 3D MRI than TL-features in the General Approach. Nonetheless, fine-tuning models pre-trained on natural images can increase the Baseline's accuracy by up to 12 percentage points, achieving an overall accuracy of 83%.
Collapse
Affiliation(s)
- Rosanna Turrisi
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), Messina, Italy; Machine Learning Genoa Center (MaLGa), University of Genoa, Genoa, Italy.
| | - Sarthak Pati
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis IN, USA; Medical Research Group, MLCommons, San Francisco CA, USA
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), Messina, Italy
| | - Gennaro Tartarisco
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), Messina, Italy
| |
Collapse
|
5
|
Boché A, Landras A, Morel M, Kellouche S, Carreiras F, Lambert A. Phenomics Demonstrates Cytokines Additive Induction of Epithelial to Mesenchymal Transition. J Cell Physiol 2025; 240:e31491. [PMID: 39565461 PMCID: PMC11747948 DOI: 10.1002/jcp.31491] [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: 04/30/2024] [Revised: 10/09/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024]
Abstract
Epithelial to mesenchymal transition (EMT) is highly plastic with a programme where cells lose adhesion and become more motile. EMT heterogeneity is one of the factors for disease progression and chemoresistance in cancer. Omics characterisations are costly and challenging to use. We developed single cell phenomics with easy to use wide-field fluorescence microscopy. We analyse over 70,000 cells and combined 53 features. Our simplistic pipeline allows efficient tracking of EMT plasticity, with a single statistical metric. We discriminate four high EMT plasticity cancer cell lines along the EMT spectrum. We test two cytokines, inducing EMT in all cell lines, alone or in combination. The single cell EMT metrics demonstrate the additive effect of cytokines combination on EMT independently of cell line EMT spectrum. The effects of cytokines are also observed at the front of migration during wound healing assay. Single cell phenomics is uniquely suited to characterise the cellular heterogeneity in response to complex microenvironment and show potential for drug testing assays.
Collapse
Affiliation(s)
- Alphonse Boché
- Equipe de Recherche sur les Relations Matrice Extracellulaire‐Cellules, ERRMECe, (EA1391), Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I‐MAT (FD4122)CY Cergy Paris UniversitéNeuville sur OiseVal d'OiseFrance
| | - Alexandra Landras
- Equipe de Recherche sur les Relations Matrice Extracellulaire‐Cellules, ERRMECe, (EA1391), Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I‐MAT (FD4122)CY Cergy Paris UniversitéNeuville sur OiseVal d'OiseFrance
| | - Mathieu Morel
- PASTEUR, Department of Chemistry, École Normale SupérieurePSL University, Sorbonne Université, CNRSParisFrance
| | - Sabrina Kellouche
- Equipe de Recherche sur les Relations Matrice Extracellulaire‐Cellules, ERRMECe, (EA1391), Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I‐MAT (FD4122)CY Cergy Paris UniversitéNeuville sur OiseVal d'OiseFrance
| | - Franck Carreiras
- Equipe de Recherche sur les Relations Matrice Extracellulaire‐Cellules, ERRMECe, (EA1391), Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I‐MAT (FD4122)CY Cergy Paris UniversitéNeuville sur OiseVal d'OiseFrance
| | - Ambroise Lambert
- Equipe de Recherche sur les Relations Matrice Extracellulaire‐Cellules, ERRMECe, (EA1391), Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I‐MAT (FD4122)CY Cergy Paris UniversitéNeuville sur OiseVal d'OiseFrance
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
Villanueva-Meyer JE, Bakas S, Tiwari P, Lupo JM, Calabrese E, Davatzikos C, Bi WL, Ismail M, Akbari H, Lohmann P, Booth TC, Wiestler B, Aerts HJWL, Rasool G, Tonn JC, Nowosielski M, Jain R, Colen RR, Pati S, Baid U, Vollmuth P, Macdonald D, Vogelbaum MA, Chang SM, Huang RY, Galldiks N. Artificial Intelligence for Response Assessment in Neuro Oncology (AI-RANO), part 1: review of current advancements. Lancet Oncol 2024; 25:e581-e588. [PMID: 39481414 DOI: 10.1016/s1470-2045(24)00316-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 11/02/2024]
Abstract
The development, application, and benchmarking of artificial intelligence (AI) tools to improve diagnosis, prognostication, and therapy in neuro-oncology are increasing at a rapid pace. This Policy Review provides an overview and critical assessment of the work to date in this field, focusing on diagnostic AI models of key genomic markers, predictive AI models of response before and after therapy, and differentiation of true disease progression from treatment-related changes, which is a considerable challenge based on current clinical care in neuro-oncology. Furthermore, promising future directions, including the use of AI for automated response assessment in neuro-oncology, are discussed.
Collapse
Affiliation(s)
- Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA; Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
| | - Spyridon Bakas
- Division of Computational Pathology, Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Biostatistics & Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA; Department of Computer Science, Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, IN, USA
| | - Pallavi Tiwari
- Department of Radiology and Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Evan Calabrese
- Duke University Center for Artificial Intelligence in Radiology, Department of Radiology, Duke University, Durham, NC, USA
| | - Christos Davatzikos
- Center for Artificial Intelligence and 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
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Marwa Ismail
- Department of Radiology and Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| | - Hamed Akbari
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-4), Research Center Juelich (FZJ), Juelich, Germany; Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Thomas C Booth
- Department of Neuroradiology, Ruskin Wing, King's College Hospital NHS Foundation Trust, London, UK; School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK; London Regional Cancer Program, London, UK
| | - Benedikt Wiestler
- Department of Neuroradiology, University Hospital, Technical University of Munich, Munich, Germany
| | - Hugo J W L Aerts
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA; Radiology and Nuclear Medicine, CARIM & GROW, Maastricht University, Maastricht, Netherlands
| | - Ghulam Rasool
- Department of Machine Learning, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Joerg C Tonn
- Department of Neurosurgery, Ludwig Maximilians University, Munich, Germany and German Cancer Consortium (DKTK), Partner Site Munich, Germany
| | - Martha Nowosielski
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Rajan Jain
- Department of Radiology and Department of Neurosurgery, New York University Langone Health, New York, NY, USA
| | - Rivka R Colen
- Radiology and Nuclear Medicine, CARIM & GROW, Maastricht University, Maastricht, Netherlands
| | - Sarthak Pati
- Division of Computational Pathology, Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ujjwal Baid
- Division of Computational Pathology, Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Philipp Vollmuth
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - David Macdonald
- Department of Neuro-Oncology, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Michael A Vogelbaum
- Department of Neurosurgery, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA; Department of Machine Learning, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Susan M Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-4), Research Center Juelich (FZJ), Juelich, Germany; Department of Neurology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| |
Collapse
|
10
|
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
|
11
|
Gongala S, Garcia JA, Korakavi N, Patil N, Akbari H, Sloan A, Barnholtz-Sloan JS, Sun J, Griffith B, Poisson LM, Booth TC, Jain R, Mohan S, Nasralla MP, Bakas S, Tippareddy C, Puig J, Palmer JD, Shi W, Colen RR, Sotiras A, Ahn SS, Park YW, Davatzikos C, Badve C. Sex-Specific Differences in Patients with IDH1-Wild-Type Grade 4 Glioma in the ReSPOND Consortium. AJNR Am J Neuroradiol 2024; 45:1299-1307. [PMID: 38684319 PMCID: PMC11392364 DOI: 10.3174/ajnr.a8319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/17/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND AND PURPOSE Understanding sex-based differences in patients with glioblastoma is necessary for accurate personalized treatment planning to improve patient outcomes. Our purpose was to investigate sex-specific differences in molecular, clinical, and radiologic tumor parameters, as well as survival outcomes in patients with glioblastoma, isocitrate dehydrogenase-1 wild-type (IDH1-WT), grade 4. MATERIALS AND METHODS Retrospective data of 1832 patients with glioblastoma, IDH1-WT with comprehensive information on tumor parameters was acquired from the Radiomics Signatures for Precision Oncology in Glioblastoma consortium. Data imputation was performed for missing values. Sex-based differences in tumor parameters, such as age, molecular parameters, preoperative Karnofsky performance score (KPS), tumor volumes, epicenter, and laterality were assessed through nonparametric tests. Spatial atlases were generated by using preoperative MRI maps to visualize tumor characteristics. Survival time analysis was performed through log-rank tests and Cox proportional hazard analyses. RESULTS Glioblastoma was diagnosed at a median age of 64 years in women compared with 61.9 years in men (false discovery rate [FDR] = 0.003). Men had a higher KPS (above 80) as compared with women (60.4% women versus 69.7% men, FDR = 0.044). Women had lower tumor volumes in enhancing (16.7 cm3 versus 20.6 cm3 in men, FDR = 0.001), necrotic core (6.18 cm3 versus 7.76 cm3 in men, FDR = 0.001), and edema regions (46.9 cm3 versus 59.2 cm3 in men, FDR = 0.0001). The right temporal region was the most common tumor epicenter in the overall population. Right as well as left temporal lobes were more frequently involved in men. There were no sex-specific differences in survival outcomes and mortality ratios. Higher age, unmethylated O6-methylguanine-DNA-methyltransferase promoter and undergoing subtotal resection increased the mortality risk in both men and women. CONCLUSIONS Our study demonstrates significant sex-based differences in clinical and radiologic tumor parameters of patients with glioblastoma. Sex is not an independent prognostic factor for survival outcomes and the tumor parameters influencing patient outcomes are identical for men and women.
Collapse
Affiliation(s)
- Sree Gongala
- From the Department of Radiology (S.G., N.K., J.S., C.T., C.B.), Case Western Reserve University School of Medicine, Cleveland, Ohio
- Department of Radiology (S.G., N.K., C.T., C.B.), University Hospitals of Cleveland, Cleveland, Ohio
| | - Jose A Garcia
- Center for Biomedical Image Computing and Analytics (CBICA) (J.A.G., C.D.), University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Radiology (J.A.G., S.M., C.D.), Division of Neuroradiology at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nisha Korakavi
- From the Department of Radiology (S.G., N.K., J.S., C.T., C.B.), Case Western Reserve University School of Medicine, Cleveland, Ohio
- Department of Radiology (S.G., N.K., C.T., C.B.), University Hospitals of Cleveland, Cleveland, Ohio
| | - Nirav Patil
- Department of Population and Quantitative Health Sciences (N.P.), Case Western Reserve University School of Medicine, Cleveland, Ohio
- University Hospitals Health System (N.P.), Research and Education Institute, Cleveland, Ohio
| | - Hamed Akbari
- Department of Bioengineering (H.A.), Santa Clara University, Santa Clara, California
| | - Andrew Sloan
- Neuroscience Service line (A.Sloan), Department of Neurosurgery, Piedmont Health, Atlanta, Georgia
- Department of Cancer Biology (A.Sloan), Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Jill S Barnholtz-Sloan
- Center for Biomedical Informatics and Information Technology (J.S.B.-S.), Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
- Trans-Divisional Research Program (J.S.B.-S.), Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Jessie Sun
- From the Department of Radiology (S.G., N.K., J.S., C.T., C.B.), Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Brent Griffith
- Department of Radiology (B.G.), Henry Ford Health, Detroit, Michigan
| | - Laila M Poisson
- Department of Radiology (L.M.P.), Wayne State University School of Medicine Henry Ford, Detroit, Michigan
| | - Thomas C Booth
- School of Biomedical Engineering & Imaging Sciences (L.M.P.), King's College London, London, United Kingdom
- Department of Neuroradiology (L.M.P.), King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Rajan Jain
- Departments of Radiology and Neurosurgery (R.J., M.P.N.), New York University Langone Health, New York, New York
| | - Suyash Mohan
- Department of Radiology (J.A.G., S.M., C.D.), Division of Neuroradiology at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - MacLean P Nasralla
- Center for AI and Data Science for Integrated Diagnostics (C.D), at the University of Pennsylvania, Philadelphia, Pennsylvania
- Departments of Radiology and Neurosurgery (R.J., M.P.N.), New York University Langone Health, New York, New York
- Department of Pathology and Laboratory Medicine (M.P.N.), at the University of Pennsylvania, Philadelphia, Pennsylvania
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center (M.P.N.), Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Spyridon Bakas
- Division of Computational Pathology (S.B.), Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, Indiana
- Department of Radiology and Imaging Sciences (S.B.), Indiana University, Indianapolis, Indiana
- Department of Neurological Surgery (S.B.), School of Medicine, Indiana University, Indianapolis, Indiana, Indiana University, Indianapolis, Indiana
| | - Charit Tippareddy
- From the Department of Radiology (S.G., N.K., J.S., C.T., C.B.), Case Western Reserve University School of Medicine, Cleveland, Ohio
- Department of Radiology (S.G., N.K., C.T., C.B.), University Hospitals of Cleveland, Cleveland, Ohio
| | - Josep Puig
- Radiology Department CDI (J.P.), Hospital Clinic of Barcelona, Barcelona, Spain
| | - Joshua D Palmer
- Department of Radiation Oncology and Neurosurgery (J.D.P.), The James Cancer Hospital at the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Wenyin Shi
- Department of Radiation Oncology (W.S.), Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Rivka R Colen
- Department of Radiology (R.R.C.), University of Pittsburgh, Pittsburgh, Pennsylvania
- Hillman Cancer Center (R.R.C.), University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Aristeidis Sotiras
- Department of Radiology (A.Sotiras), WA University School of Medicine, St. Louis, Missouri
- Institute for Informatics (A.Sotiras), Data Science & Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Sung Soo Ahn
- Department of Radiology (S.S.A., Y.W.P.), Section of Neuroradiology, Yonsei University Health System, Seoul, Republic of Korea
| | - Yae Won Park
- Department of Radiology (S.S.A., Y.W.P.), Section of Neuroradiology, Yonsei University Health System, Seoul, Republic of Korea
- Department of Radiology (J.A.G., S.M., C.D.), Division of Neuroradiology at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA) (J.A.G., C.D.), University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Radiology (J.A.G., S.M., C.D.), Division of Neuroradiology at the University of Pennsylvania, Philadelphia, Pennsylvania
- Center for AI and Data Science for Integrated Diagnostics (C.D), at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chaitra Badve
- From the Department of Radiology (S.G., N.K., J.S., C.T., C.B.), Case Western Reserve University School of Medicine, Cleveland, Ohio
- Department of Radiology (S.G., N.K., C.T., C.B.), University Hospitals of Cleveland, Cleveland, Ohio
| |
Collapse
|
12
|
Novak A, Hollowday M, Espinosa Morgado AT, Oke J, Shelmerdine S, Woznitza N, Metcalfe D, Costa ML, Wilson S, Kiam JS, Vaz J, Limphaibool N, Ventre J, Jones D, Greenhalgh L, Gleeson F, Welch N, Mistry A, Devic N, Teh J, Ather S. Evaluating the impact of artificial intelligence-assisted image analysis on the diagnostic accuracy of front-line clinicians in detecting fractures on plain X-rays (FRACT-AI): protocol for a prospective observational study. BMJ Open 2024; 14:e086061. [PMID: 39237277 PMCID: PMC11381697 DOI: 10.1136/bmjopen-2024-086061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/15/2024] [Indexed: 09/07/2024] Open
Abstract
INTRODUCTION Missed fractures are the most frequent diagnostic error attributed to clinicians in UK emergency departments and a significant cause of patient morbidity. Recently, advances in computer vision have led to artificial intelligence (AI)-enhanced model developments, which can support clinicians in the detection of fractures. Previous research has shown these models to have promising effects on diagnostic performance, but their impact on the diagnostic accuracy of clinicians in the National Health Service (NHS) setting has not yet been fully evaluated. METHODS AND ANALYSIS A dataset of 500 plain radiographs derived from Oxford University Hospitals (OUH) NHS Foundation Trust will be collated to include all bones except the skull, facial bones and cervical spine. The dataset will be split evenly between radiographs showing one or more fractures and those without. The reference ground truth for each image will be established through independent review by two senior musculoskeletal radiologists. A third senior radiologist will resolve disagreements between two primary radiologists. The dataset will be analysed by a commercially available AI tool, BoneView (Gleamer, Paris, France), and its accuracy for detecting fractures will be determined with reference to the ground truth diagnosis. We will undertake a multiple case multiple reader study in which clinicians interpret all images without AI support, then repeat the process with access to AI algorithm output following a 4-week washout. 18 clinicians will be recruited as readers from four hospitals in England, from six distinct clinical groups, each with three levels of seniority (early-stage, mid-stage and later-stage career). Changes in the accuracy, confidence and speed of reporting will be compared with and without AI support. Readers will use a secure web-based DICOM (Digital Imaging and Communications in Medicine) viewer (www.raiqc.com), allowing radiograph viewing and abnormality identification. Pooled analyses will be reported for overall reader performance as well as for subgroups including clinical role, level of seniority, pathological finding and difficulty of image. ETHICS AND DISSEMINATION The study has been approved by the UK Healthcare Research Authority (IRAS 310995, approved on 13 December 2022). The use of anonymised retrospective radiographs has been authorised by OUH NHS Foundation Trust. The results will be presented at relevant conferences and published in a peer-reviewed journal. TRIAL REGISTRATION NUMBERS This study is registered with ISRCTN (ISRCTN19562541) and ClinicalTrials.gov (NCT06130397). The paper reports the results of a substudy of STEDI2 (Simulation Training for Emergency Department Imaging Phase 2).
Collapse
Affiliation(s)
- Alex Novak
- Emergency Medicine Research Oxford, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Max Hollowday
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Jason Oke
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Susan Shelmerdine
- Clinical Radiology, Great Ormond Street Hospital for Children, London, UK
- Radiology, UCL GOSH ICH, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Nick Woznitza
- Radiology, University College London Hospitals NHS Foundation Trust, London, UK
- Canterbury Christ Church University, Canterbury Christ Church University, Canterbury, UK
| | - David Metcalfe
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Matthew L Costa
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), Oxford Trauma & Emergency Care (OxTEC), University of Oxford, Oxford, UK
| | - Sarah Wilson
- Frimley Health NHS Foundation Trust, Frimley, UK
| | - Jian Shen Kiam
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - James Vaz
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | | | | | | | - Fergus Gleeson
- Department of Oncology, University of Oxford, Oxford, UK
| | - Nick Welch
- Patient and Public Involvement Member, Oxford, UK
| | - Alpesh Mistry
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
- North West MSK Imaging, Liverpool, UK
| | - Natasa Devic
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - James Teh
- Nuffield Orthopaedic Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Sarim Ather
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
13
|
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
|
14
|
Sørensen PJ, Ladefoged CN, Larsen VA, Andersen FL, Nielsen MB, Poulsen HS, Carlsen JF, Hansen AE. Repurposing the Public BraTS Dataset for Postoperative Brain Tumour Treatment Response Monitoring. Tomography 2024; 10:1397-1410. [PMID: 39330751 PMCID: PMC11436089 DOI: 10.3390/tomography10090105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
The Brain Tumor Segmentation (BraTS) Challenge has been a main driver of the development of deep learning (DL) algorithms and provides by far the largest publicly available expert-annotated brain tumour dataset but contains solely preoperative examinations. The aim of our study was to facilitate the use of the BraTS dataset for training DL brain tumour segmentation algorithms for a postoperative setting. To this end, we introduced an automatic conversion of the three-label BraTS annotation protocol to a two-label annotation protocol suitable for postoperative brain tumour segmentation. To assess the viability of the label conversion, we trained a DL algorithm using both the three-label and the two-label annotation protocols. We assessed the models pre- and postoperatively and compared the performance with a state-of-the-art DL method. The DL algorithm trained using the BraTS three-label annotation misclassified parts of 10 out of 41 fluid-filled resection cavities in 72 postoperative glioblastoma MRIs, whereas the two-label model showed no such inaccuracies. The tumour segmentation performance of the two-label model both pre- and postoperatively was comparable to that of a state-of-the-art algorithm for tumour volumes larger than 1 cm3. Our study enables using the BraTS dataset as a basis for the training of DL algorithms for postoperative tumour segmentation.
Collapse
Affiliation(s)
- Peter Jagd Sørensen
- Department of Radiology, Centre of Diagnostic Investigation, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark; (V.A.L.); (M.B.N.); (J.F.C.); (A.E.H.)
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark;
- The DCCC Brain Tumor Center, 2100 Copenhagen, Denmark;
| | - Claes Nøhr Ladefoged
- Department of Clinical Physiology and Nuclear Medicine, Centre of Diagnostic Investigation, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, 2800 Lyngby, Denmark
| | - Vibeke Andrée Larsen
- Department of Radiology, Centre of Diagnostic Investigation, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark; (V.A.L.); (M.B.N.); (J.F.C.); (A.E.H.)
| | - Flemming Littrup Andersen
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark;
- Department of Clinical Physiology and Nuclear Medicine, Centre of Diagnostic Investigation, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Michael Bachmann Nielsen
- Department of Radiology, Centre of Diagnostic Investigation, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark; (V.A.L.); (M.B.N.); (J.F.C.); (A.E.H.)
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark;
| | | | - Jonathan Frederik Carlsen
- Department of Radiology, Centre of Diagnostic Investigation, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark; (V.A.L.); (M.B.N.); (J.F.C.); (A.E.H.)
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Adam Espe Hansen
- Department of Radiology, Centre of Diagnostic Investigation, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark; (V.A.L.); (M.B.N.); (J.F.C.); (A.E.H.)
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark;
- The DCCC Brain Tumor Center, 2100 Copenhagen, Denmark;
| |
Collapse
|
15
|
Wajid B, Jamil M, Awan FG, Anwar F, Anwar A. aXonica: A support package for MRI based Neuroimaging. BIOTECHNOLOGY NOTES (AMSTERDAM, NETHERLANDS) 2024; 5:120-136. [PMID: 39416698 PMCID: PMC11446389 DOI: 10.1016/j.biotno.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 10/19/2024]
Abstract
Magnetic Resonance Imaging (MRI) assists in studying the nervous system. MRI scans undergo significant processing before presenting the final images to medical practitioners. These processes are executed with ease due to excellent software pipelines. However, establishing software workstations is non-trivial and requires researchers in life sciences to be comfortable in downloading, installing, and scripting software that is non-user-friendly and may lack basic GUI. As researchers struggle with these skills, there is a dire need to develop software packages that can automatically install software pipelines speeding up building software workstations and laboratories. Previous solutions include NeuroDebian, BIDS Apps, Flywheel, QMENTA, Boutiques, Brainlife and Neurodesk. Overall, all these solutions complement each other. NeuroDebian covers neuroscience and has a wider scope, providing only 51 tools for MRI. Whereas, BIDS Apps is committed to the BIDS format, covering only 45 software related to MRI. Boutiques is more flexible, facilitating its pipelines to be easily installed as separate containers, validated, published, and executed. Whereas, both Flywheel and Qmenta are propriety, leaving four for users looking for 'free for use' tools, i.e., NeuroDebian, Brainlife, Neurodesk, and BIDS Apps. This paper presents an extensive survey of 317 tools published in MRI-based neuroimaging in the last ten years, along with 'aXonica,' an MRI-based neuroimaging support package that is unbiased towards any formatting standards and provides 130 applications, more than that of NeuroDebian (51), BIDS App (45), Flywheel (70), and Neurodesk (85). Using a technology stack that employs GUI as the front-end and shell scripted back-end, aXonica provides (i) 130 tools that span the entire MRI-based neuroimaging analysis, and allow the user to (ii) select the software of their choice, (iii) automatically resolve individual dependencies and (iv) installs them. Hence, aXonica can serve as an important resource for researchers and teachers working in the field of MRI-based Neuroimaging to (a) develop software workstations, and/or (b) install newer tools in their existing workstations.
Collapse
Affiliation(s)
- Bilal Wajid
- Dhanani School of Science and Engineering, Habib University, Karachi, Pakistan
- Muhammad Ibn Musa Al-Khwarizmi Research & Development Division, Sabz-Qalam, Lahore, Pakistan
| | - Momina Jamil
- Muhammad Ibn Musa Al-Khwarizmi Research & Development Division, Sabz-Qalam, Lahore, Pakistan
| | - Fahim Gohar Awan
- Department of Electrical Engineering, University of Engineering & Technology, Lahore, Pakistan
| | - Faria Anwar
- Out Patient Department, Mayo Hospital, Lahore, Pakistan
| | - Ali Anwar
- Department of Computer Science, University of Minnesota, Minneapolis, USA
| |
Collapse
|
16
|
Upadhyay VR, Ramesh V, Kumar H, Somagond YM, Priyadarsini S, Kuniyal A, Prakash V, Sahoo A. Phenomics in Livestock Research: Bottlenecks and Promises of Digital Phenotyping and Other Quantification Techniques on a Global Scale. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:380-393. [PMID: 39012961 DOI: 10.1089/omi.2024.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Bottlenecks in moving genomics to real-life applications also include phenomics. This is true not only for genomics medicine and public health genomics but also in ecology and livestock phenomics. This expert narrative review explores the intricate relationship between genetic makeup and observable phenotypic traits across various biological levels in the context of livestock research. We unpack and emphasize the significance of precise phenotypic data in selective breeding outcomes and examine the multifaceted applications of phenomics, ranging from improvement to assessing welfare, reproductive traits, and environmental adaptation in livestock. As phenotypic traits exhibit strong correlations, their measurement alongside specific biological outcomes provides insights into performance, overall health, and clinical endpoints like morbidity and disease. In addition, automated assessment of livestock holds potential for monitoring the dynamic phenotypic traits across various species, facilitating a deeper comprehension of how they adapt to their environment and attendant stressors. A key challenge in genetic improvement in livestock is predicting individuals with optimal fitness without direct measurement. Temporal predictions from unmanned aerial systems can surpass genomic predictions, offering in-depth data on livestock. In the near future, digital phenotyping and digital biomarkers may further unravel the genetic intricacies of stress tolerance, adaptation and welfare aspects of animals enabling the selection of climate-resilient and productive livestock. This expert review thus delves into challenges associated with phenotyping and discusses technological advancements shaping the future of biological research concerning livestock.
Collapse
Affiliation(s)
| | - Vikram Ramesh
- ICAR-National Research Centre on Mithun, Medziphema, Nagaland, India
| | - Harshit Kumar
- ICAR-National Research Centre on Mithun, Medziphema, Nagaland, India
| | - Y M Somagond
- ICAR-National Research Centre on Mithun, Medziphema, Nagaland, India
| | | | - Aruna Kuniyal
- ICAR-National Research Centre on Camel, Bikaner, Rajasthan, India
| | - Ved Prakash
- ICAR-National Research Centre on Camel, Bikaner, Rajasthan, India
| | - Artabandhu Sahoo
- ICAR-National Research Centre on Camel, Bikaner, Rajasthan, India
| |
Collapse
|
17
|
Kundal K, Rao KV, Majumdar A, Kumar N, Kumar R. Comprehensive benchmarking of CNN-based tumor segmentation methods using multimodal MRI data. Comput Biol Med 2024; 178:108799. [PMID: 38925087 DOI: 10.1016/j.compbiomed.2024.108799] [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: 02/07/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
Magnetic resonance imaging (MRI) has become an essential and a frontline technique in the detection of brain tumor. However, segmenting tumors manually from scans is laborious and time-consuming. This has led to an increasing trend towards fully automated methods for precise tumor segmentation in MRI scans. Accurate tumor segmentation is crucial for improved diagnosis, treatment, and prognosis. This study benchmarks and evaluates four widely used CNN-based methods for brain tumor segmentation CaPTk, 2DVNet, EnsembleUNets, and ResNet50. Using 1251 multimodal MRI scans from the BraTS2021 dataset, we compared the performance of these methods against a reference dataset of segmented images assisted by radiologists. This comparison was conducted using segmented images directly and further by radiomic features extracted from the segmented images using pyRadiomics. Performance was assessed using the Dice Similarity Coefficient (DSC) and Hausdorff Distance (HD). EnsembleUNets excelled, achieving a DSC of 0.93 and an HD of 18, outperforming the other methods. Further comparative analysis of radiomic features confirmed EnsembleUNets as the most precise segmentation method, surpassing other methods. EnsembleUNets recorded a Concordance Correlation Coefficient (CCC) of 0.79, a Total Deviation Index (TDI) of 1.14, and a Root Mean Square Error (RMSE) of 0.53, underscoring its superior performance. We also performed validation on an independent dataset of 611 samples (UPENN-GBM), which further supported the accuracy of EnsembleUNets, with a DSC of 0.85 and an HD of 17.5. These findings provide valuable insight into the efficacy of EnsembleUNets, supporting informed decisions for accurate brain tumor segmentation.
Collapse
Affiliation(s)
- Kavita Kundal
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, 502284, India
| | - K Venkateswara Rao
- Department of Neurosurgical Oncology, Basavatarakam Indo American Cancer Hospital & Research Institute, Hyderabad, Telangana, 500034, India
| | - Arunabha Majumdar
- Department of Mathematics, Indian Institute of Technology Hyderabad, Kandi, Telangana, 502284, India
| | - Neeraj Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, 502284, India; Department of Liberal Arts, Indian Institute of Technology Hyderabad, Kandi, Telangana, 502284, India
| | - Rahul Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, 502284, India.
| |
Collapse
|
18
|
Boelders SM, De Baene W, Postma E, Gehring K, Ong LL. Predicting Cognitive Functioning for Patients with a High-Grade Glioma: Evaluating Different Representations of Tumor Location in a Common Space. Neuroinformatics 2024; 22:329-352. [PMID: 38900230 PMCID: PMC11329426 DOI: 10.1007/s12021-024-09671-9] [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] [Accepted: 05/31/2024] [Indexed: 06/21/2024]
Abstract
Cognitive functioning is increasingly considered when making treatment decisions for patients with a brain tumor in view of a personalized onco-functional balance. Ideally, one can predict cognitive functioning of individual patients to make treatment decisions considering this balance. To make accurate predictions, an informative representation of tumor location is pivotal, yet comparisons of representations are lacking. Therefore, this study compares brain atlases and principal component analysis (PCA) to represent voxel-wise tumor location. Pre-operative cognitive functioning was predicted for 246 patients with a high-grade glioma across eight cognitive tests while using different representations of voxel-wise tumor location as predictors. Voxel-wise tumor location was represented using 13 different frequently-used population average atlases, 13 randomly generated atlases, and 13 representations based on PCA. ElasticNet predictions were compared between representations and against a model solely using tumor volume. Preoperative cognitive functioning could only partly be predicted from tumor location. Performances of different representations were largely similar. Population average atlases did not result in better predictions compared to random atlases. PCA-based representation did not clearly outperform other representations, although summary metrics indicated that PCA-based representations performed somewhat better in our sample. Representations with more regions or components resulted in less accurate predictions. Population average atlases possibly cannot distinguish between functionally distinct areas when applied to patients with a glioma. This stresses the need to develop and validate methods for individual parcellations in the presence of lesions. Future studies may test if the observed small advantage of PCA-based representations generalizes to other data.
Collapse
Affiliation(s)
- S M Boelders
- Department of Neurosurgery, Elisabeth-TweeSteden Hospital, Tilburg, The Netherlands
- Department of Cognitive Sciences and AI, Tilburg University, Tilburg, The Netherlands
| | - W De Baene
- Department of Cognitive Neuropsychology, Tilburg University Tilburg, Warandelaan 2, P. O. Box 90153, Tilburg, 5000 LE, The Netherlands
| | - E Postma
- Department of Cognitive Sciences and AI, Tilburg University, Tilburg, The Netherlands
| | - K Gehring
- Department of Neurosurgery, Elisabeth-TweeSteden Hospital, Tilburg, The Netherlands.
- Department of Cognitive Neuropsychology, Tilburg University Tilburg, Warandelaan 2, P. O. Box 90153, Tilburg, 5000 LE, The Netherlands.
| | - L L Ong
- Department of Cognitive Sciences and AI, Tilburg University, Tilburg, The Netherlands
| |
Collapse
|
19
|
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
|
20
|
Belue MJ, Harmon SA, Chappidi S, Zhuge Y, Tasci E, Jagasia S, Joyce T, Camphausen K, Turkbey B, Krauze AV. Diagnosing Progression in Glioblastoma-Tackling a Neuro-Oncology Problem Using Artificial-Intelligence-Derived Volumetric Change over Time on Magnetic Resonance Imaging to Examine Progression-Free Survival in Glioblastoma. Diagnostics (Basel) 2024; 14:1374. [PMID: 39001264 PMCID: PMC11241823 DOI: 10.3390/diagnostics14131374] [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: 05/06/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive and the most common primary brain tumor, defined by nearly uniform rapid progression despite the current standard of care involving maximal surgical resection followed by radiation therapy (RT) and temozolomide (TMZ) or concurrent chemoirradiation (CRT), with an overall survival (OS) of less than 30% at 2 years. The diagnosis of tumor progression in the clinic is based on clinical assessment and the interpretation of MRI of the brain using Response Assessment in Neuro-Oncology (RANO) criteria, which suffers from several limitations including a paucity of precise measures of progression. Given that imaging is the primary modality that generates the most quantitative data capable of capturing change over time in the standard of care for GBM, this renders it pivotal in optimizing and advancing response criteria, particularly given the lack of biomarkers in this space. In this study, we employed artificial intelligence (AI)-derived MRI volumetric parameters using the segmentation mask output of the nnU-Net to arrive at four classes (background, edema, non-contrast enhancing tumor (NET), and contrast-enhancing tumor (CET)) to determine if dynamic changes in AI volumes detected throughout therapy can be linked to PFS and clinical features. We identified associations between MR imaging AI-generated volumes and PFS independently of tumor location, MGMT methylation status, and the extent of resection while validating that CET and edema are the most linked to PFS with patient subpopulations separated by district rates of change throughout the disease. The current study provides valuable insights for risk stratification, future RT treatment planning, and treatment monitoring in neuro-oncology.
Collapse
Affiliation(s)
- Mason J. Belue
- Artificial Intelligence Resource, Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD 20892, USA; (M.J.B.); (S.A.H.); (B.T.)
| | - Stephanie A. Harmon
- Artificial Intelligence Resource, Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD 20892, USA; (M.J.B.); (S.A.H.); (B.T.)
| | - Shreya Chappidi
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD 20892, USA; (S.C.); (Y.Z.); (S.J.); (T.J.); (K.C.)
- Department of Computer Science and Technology, University of Cambridge, 15 JJ Thomson Ave., Cambridge CB3 0FD, UK
| | - Ying Zhuge
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD 20892, USA; (S.C.); (Y.Z.); (S.J.); (T.J.); (K.C.)
| | - Erdal Tasci
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD 20892, USA; (S.C.); (Y.Z.); (S.J.); (T.J.); (K.C.)
| | - Sarisha Jagasia
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD 20892, USA; (S.C.); (Y.Z.); (S.J.); (T.J.); (K.C.)
| | - Thomas Joyce
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD 20892, USA; (S.C.); (Y.Z.); (S.J.); (T.J.); (K.C.)
| | - Kevin Camphausen
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD 20892, USA; (S.C.); (Y.Z.); (S.J.); (T.J.); (K.C.)
| | - Baris Turkbey
- Artificial Intelligence Resource, Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD 20892, USA; (M.J.B.); (S.A.H.); (B.T.)
| | - Andra V. Krauze
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD 20892, USA; (S.C.); (Y.Z.); (S.J.); (T.J.); (K.C.)
| |
Collapse
|
21
|
Horng H, Scott C, Winham S, Jensen M, Pantalone L, Mankowski W, Kerlikowske K, Vachon CM, Kontos D, Shinohara RT. Multivariate testing and effect size measures for batch effect evaluation in radiomic features. Sci Rep 2024; 14:13923. [PMID: 38886407 PMCID: PMC11183083 DOI: 10.1038/s41598-024-64208-z] [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: 01/28/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
While precision medicine applications of radiomics analysis are promising, differences in image acquisition can cause "batch effects" that reduce reproducibility and affect downstream predictive analyses. Harmonization methods such as ComBat have been developed to correct these effects, but evaluation methods for quantifying batch effects are inconsistent. In this study, we propose the use of the multivariate statistical test PERMANOVA and the Robust Effect Size Index (RESI) to better quantify and characterize batch effects in radiomics data. We evaluate these methods in both simulated and real radiomics features extracted from full-field digital mammography (FFDM) data. PERMANOVA demonstrated higher power than standard univariate statistical testing, and RESI was able to interpretably quantify the effect size of site at extremely large sample sizes. These methods show promise as more powerful and interpretable methods for the detection and quantification of batch effects in radiomics studies.
Collapse
Affiliation(s)
- Hannah Horng
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Radiology, Center for Biomedical Image Computing and Analysis (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn Statistics in Imaging Endeavor (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | | | | | | | - Lauren Pantalone
- Department of Radiology, Center for Biomedical Image Computing and Analysis (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Walter Mankowski
- Department of Radiology, Center for Biomedical Image Computing and Analysis (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | | | - Despina Kontos
- Department of Radiology, Center for Biomedical Image Computing and Analysis (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Innovation in Imaging Biomarkers and Integrated Diagnostics (CIMBID), Columbia University, New York, NY, 10027, USA
| | - Russell T Shinohara
- Department of Radiology, Center for Biomedical Image Computing and Analysis (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Statistics in Imaging Endeavor (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
22
|
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
|
23
|
Morrison S, Gatsonis C, Eloyan A, Steingrimsson JA. Survival analysis using deep learning with medical imaging. Int J Biostat 2024; 20:1-12. [PMID: 37312249 PMCID: PMC11074924 DOI: 10.1515/ijb-2022-0113] [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: 09/14/2022] [Accepted: 02/24/2023] [Indexed: 06/15/2023]
Abstract
There is widespread interest in using deep learning to build prediction models for medical imaging data. These deep learning methods capture the local structure of the image and require no manual feature extraction. Despite the importance of modeling survival in the context of medical data analysis, research on deep learning methods for modeling the relationship of imaging and time-to-event data is still under-developed. We provide an overview of deep learning methods for time-to-event outcomes and compare several deep learning methods to Cox model based methods through the analysis of a histology dataset of gliomas.
Collapse
Affiliation(s)
- Samantha Morrison
- Department of Biostatistics, School of Public Health, Brown University, Providence, RI, USA
| | - Constantine Gatsonis
- Department of Biostatistics, School of Public Health, Brown University, Providence, RI, USA
| | - Ani Eloyan
- Department of Biostatistics, School of Public Health, Brown University, Providence, RI, USA
| | - Jon Arni Steingrimsson
- Department of Biostatistics, School of Public Health, Brown University, Providence, RI, USA
| |
Collapse
|
24
|
Fatania K, Frood R, Mistry H, Short SC, O’Connor J, Scarsbrook AF, Currie S. Tumour Size and Overall Survival in a Cohort of Patients with Unifocal Glioblastoma: A Uni- and Multivariable Prognostic Modelling and Resampling Study. Cancers (Basel) 2024; 16:1301. [PMID: 38610979 PMCID: PMC11011077 DOI: 10.3390/cancers16071301] [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: 02/15/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Published models inconsistently associate glioblastoma size with overall survival (OS). This study aimed to investigate the prognostic effect of tumour size in a large cohort of patients diagnosed with GBM and interrogate how sample size and non-linear transformations may impact on the likelihood of finding a prognostic effect. In total, 279 patients with a IDH-wildtype unifocal WHO grade 4 GBM between 2014 and 2020 from a retrospective cohort were included. Uni-/multivariable association between core volume, whole volume (CV and WV), and diameter with OS was assessed with (1) Cox proportional hazard models +/- log transformation and (2) resampling with 1,000,000 repetitions and varying sample size to identify the percentage of models, which showed a significant effect of tumour size. Models adjusted for operation type and a diameter model adjusted for all clinical variables remained significant (p = 0.03). Multivariable resampling increased the significant effects (p < 0.05) of all size variables as sample size increased. Log transformation also had a large effect on the chances of a prognostic effect of WV. For models adjusted for operation type, 19.5% of WV vs. 26.3% log-WV (n = 50) and 69.9% WV and 89.9% log-WV (n = 279) were significant. In this large well-curated cohort, multivariable modelling and resampling suggest tumour volume is prognostic at larger sample sizes and with log transformation for WV.
Collapse
Affiliation(s)
- Kavi Fatania
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds General Infirmary, Leeds LS1 3EX, UK (A.F.S.); (S.C.)
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9TJ, UK;
| | - Russell Frood
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds General Infirmary, Leeds LS1 3EX, UK (A.F.S.); (S.C.)
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9TJ, UK;
| | - Hitesh Mistry
- Division of Cancer Sciences, The University of Manchester, Manchester M13 9PL, UK; (H.M.)
| | - Susan C. Short
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9TJ, UK;
- Department of Oncology, Leeds Teaching Hospitals NHS Trust, St James’s University Hospital, Leeds LS9 7TF, UK
| | - James O’Connor
- Division of Cancer Sciences, The University of Manchester, Manchester M13 9PL, UK; (H.M.)
- Department of Radiology, The Christie Hospital, Manchester M20 4BX, UK
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London SM2 5NG, UK
| | - Andrew F. Scarsbrook
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds General Infirmary, Leeds LS1 3EX, UK (A.F.S.); (S.C.)
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9TJ, UK;
| | - Stuart Currie
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds General Infirmary, Leeds LS1 3EX, UK (A.F.S.); (S.C.)
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9TJ, UK;
| |
Collapse
|
25
|
Guo J, Fathi Kazerooni A, Toorens E, Akbari H, Yu F, Sako C, Mamourian E, Shinohara RT, Koumenis C, Bagley SJ, Morrissette JJD, Binder ZA, Brem S, Mohan S, Lustig RA, O'Rourke DM, Ganguly T, Bakas S, Nasrallah MP, Davatzikos C. Integrating imaging and genomic data for the discovery of distinct glioblastoma subtypes: a joint learning approach. Sci Rep 2024; 14:4922. [PMID: 38418494 PMCID: PMC10902376 DOI: 10.1038/s41598-024-55072-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: 04/26/2023] [Accepted: 02/19/2024] [Indexed: 03/01/2024] Open
Abstract
Glioblastoma is a highly heterogeneous disease, with variations observed at both phenotypical and molecular levels. Personalized therapies would be facilitated by non-invasive in vivo approaches for characterizing this heterogeneity. In this study, we developed unsupervised joint machine learning between radiomic and genomic data, thereby identifying distinct glioblastoma subtypes. A retrospective cohort of 571 IDH-wildtype glioblastoma patients were included in the study, and pre-operative multi-parametric MRI scans and targeted next-generation sequencing (NGS) data were collected. L21-norm minimization was used to select a subset of 12 radiomic features from the MRI scans, and 13 key driver genes from the five main signal pathways most affected in glioblastoma were selected from the genomic data. Subtypes were identified using a joint learning approach called Anchor-based Partial Multi-modal Clustering on both radiomic and genomic modalities. Kaplan-Meier analysis identified three distinct glioblastoma subtypes: high-risk, medium-risk, and low-risk, based on overall survival outcome (p < 0.05, log-rank test; Hazard Ratio = 1.64, 95% CI 1.17-2.31, Cox proportional hazard model on high-risk and low-risk subtypes). The three subtypes displayed different phenotypical and molecular characteristics in terms of imaging histogram, co-occurrence of genes, and correlation between the two modalities. Our findings demonstrate the synergistic value of integrated radiomic signatures and molecular characteristics for glioblastoma subtyping. Joint learning on both modalities can aid in better understanding the molecular basis of phenotypical signatures of glioblastoma, and provide insights into the biological underpinnings of tumor formation and progression.
Collapse
Affiliation(s)
- Jun Guo
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anahita Fathi Kazerooni
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, 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
| | - Erik Toorens
- Penn Genomic Analysis Core, 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
| | - Fanyang Yu
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, USA
| | - Chiharu Sako
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Russell T Shinohara
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Penn Statistics in Imaging and Visualization (PennSIVE) Center, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Bagley
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer J D Morrissette
- Department of Pathology and Laboratory Medicine, 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
| | - Steven Brem
- 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
| | - Suyash Mohan
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert A Lustig
- 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
| | - 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
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, 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
- Division of Computational Pathology, Department of Pathology & Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - MacLean P Nasrallah
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, 3700 Hamilton Walk, 7Th Floor, Philadelphia, PA, 19104, USA.
- Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Kumar D, Suthar N. Predictive analytics and early intervention in healthcare social work: a scoping review. SOCIAL WORK IN HEALTH CARE 2024; 63:208-229. [PMID: 38349783 DOI: 10.1080/00981389.2024.2316700] [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: 06/15/2023] [Revised: 12/13/2023] [Accepted: 01/05/2024] [Indexed: 02/15/2024]
Abstract
This scoping review investigates the untapped potential of predictive analytics in healthcare social work, specifically targeting early intervention frameworks. Despite the escalating attention predictive analytics has garnered across multiple disciplines, its tailored application in social work remains notably sparse. This study endeavors to fill this lacuna by meticulously reviewing the extant literature and delineating the prospective advantages and inherent constraints of integrating predictive analytics into healthcare social work. The outcomes of this inquiry enrich the prevailing dialogue on the utility of predictive analytics in healthcare, offering indispensable perspectives for practitioners and policymakers in the social work domain.
Collapse
Affiliation(s)
- Dinesh Kumar
- Faculty of Business and Applied Arts, Lovely Professional University, Mittal School of Business, Phagwara, India
| | - Nidhi Suthar
- Administration, Pomento IT Services, Hisar, India
| |
Collapse
|
27
|
Yu F, Wang R, Chaudhari P, Davatzikos C. Investigating Causal Genetic Effects on Overall Survival of Glioblastoma Patients using Normalizing Flow and Structural Causal Model. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2024; 12927:129271F. [PMID: 38650741 PMCID: PMC11034818 DOI: 10.1117/12.3005434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Glioblastoma (GBM) is the most common and aggressive brain tumor with short overall survival (OS) of about 15 months. Understanding the causal factors affecting the patient survival is crucial for disease prognosis and treatment planning. Although previous efforts on survival prediction using multi-omics data has yielded useful predictive models, the causation of the correlated genetic risk factors has not been addressed. Recent advances in causal deep learning models enable the study of causality from complex dataset. In this paper, we leverage the recently proposed structural causal model (SCM) with normalizing flows parameterized by deep networks to perform the counterfactual query to investigate the causal relationship between gene mutation and OS with the presence of other confounders including sex, age and radiomics features. The query amounts to the question that what the survival days will be if the gene mutation status has been changed, i.e., from mutant to non-mutant and vice versa. The trained causal model will infer the counterfactual outcome given the intervention on specific gene mutation. We apply multivariate Cox-PH model to find the genes associated with survival, and investigate the causal genetic effect by comparing the original and counterfactual survival days in a bi-directional fashion. Particularly, the following two scenarios are considered: (1) intervention on a specific gene with non-mutant status to generate the counterfactual survival days as if the gene is mutant, with which the original survival days of the subjects with that mutant gene will be compared; (2) intervention on the gene with mutant status and perform the comparison with survival days of subjects with that non-mutant gene. Our experimental results show that no causation of two correlated genes (NF1, RB1) was revealed in the cohort (n=181), while their genetic effects on OS in terms of prolonging or shortening are generally in accordance with clinical findings.
Collapse
Affiliation(s)
- Fanyang Yu
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rongguang Wang
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pratik Chaudhari
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Tan SH, Guan CA, Bujang MA, Lai WH, Voon PJ, Sim EUH. Identification of phenomic data in the pathogenesis of cancers of the gastrointestinal (GI) tract in the UK biobank. Sci Rep 2024; 14:1997. [PMID: 38263244 PMCID: PMC10805853 DOI: 10.1038/s41598-024-52421-9] [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: 09/13/2023] [Accepted: 01/18/2024] [Indexed: 01/25/2024] Open
Abstract
Gastrointestinal (GI) cancers account for a significant incidence and mortality rates of cancers globally. Utilization of a phenomic data approach allows researchers to reveal the mechanisms and molecular pathogenesis of these conditions. We aimed to investigate the association between the phenomic features and GI cancers in a large cohort study. We included 502,369 subjects aged 37-73 years in the UK Biobank recruited since 2006, followed until the date of the first cancer diagnosis, date of death, or the end of follow-up on December 31st, 2016, whichever occurred first. Socio-demographic factors, blood chemistry, anthropometric measurements and lifestyle factors of participants collected at baseline assessment were analysed. Unvariable and multivariable logistic regression were conducted to determine the significant risk factors for the outcomes of interest, based on the odds ratio (OR) and 95% confidence intervals (CI). The analysis included a total of 441,141 participants, of which 7952 (1.8%) were incident GI cancer cases and 433,189 were healthy controls. A marker, cystatin C was associated with total and each gastrointestinal cancer (adjusted OR 2.43; 95% CI 2.23-2.64). In this cohort, compared to Asians, the Whites appeared to have a higher risk of developing gastrointestinal cancers. Several other factors were associated with distinct GI cancers. Cystatin C and race appear to be important features in GI cancers, suggesting some overlap in the molecular pathogenesis of GI cancers. Given the small proportion of Asians within the UK Biobank, the association between race and GI cancers requires further confirmation.
Collapse
Affiliation(s)
- Shirin Hui Tan
- Clinical Research Centre, Sarawak General Hospital, Ministry of Health Malaysia, Jalan Hospital, 93586, Kuching, Sarawak, Malaysia.
- Faculty of Resource Science and Technology, Universiti Malaysia Sarawak, 94300, Kota Samarahan, Malaysia.
| | - Catherina Anak Guan
- Clinical Research Centre, Sarawak General Hospital, Ministry of Health Malaysia, Jalan Hospital, 93586, Kuching, Sarawak, Malaysia
| | - Mohamad Adam Bujang
- Clinical Research Centre, Sarawak General Hospital, Ministry of Health Malaysia, Jalan Hospital, 93586, Kuching, Sarawak, Malaysia
| | - Wei Hong Lai
- Clinical Research Centre, Sarawak General Hospital, Ministry of Health Malaysia, Jalan Hospital, 93586, Kuching, Sarawak, Malaysia
| | - Pei Jye Voon
- Department of Radiotherapy, Oncology and Palliative Care, Sarawak General Hospital, Ministry of Health Malaysia, Jalan Hospital, 93586, Kuching, Sarawak, Malaysia
| | - Edmund Ui Hang Sim
- Faculty of Resource Science and Technology, Universiti Malaysia Sarawak, 94300, Kota Samarahan, Malaysia
| |
Collapse
|
29
|
Arrington D, Motley R, Colbert ZM, Lehman M, Ramachandran P. PAHPhysRAD: A Digital Imaging and Communications in Medicine Research Tool for Segmentation and Radiomic Feature Extraction. J Med Phys 2024; 49:12-21. [PMID: 38828062 PMCID: PMC11141757 DOI: 10.4103/jmp.jmp_120_23] [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: 09/11/2023] [Revised: 11/27/2023] [Accepted: 01/07/2024] [Indexed: 06/05/2024] Open
Abstract
Introduction Segmentation and analysis of organs at risks (OARs) and tumor volumes are integral concepts in the development of radiotherapy treatment plans and prediction of patients' treatment outcomes. Aims To develop a research tool, PAHPhysRAD, that can be used to semi- and fully automate segmentation of OARs. In addition, the proposed software seeks to extract 3214 radiomic features from tumor volumes and user-specified dose-volume parameters. Materials and Methods Developed within MATLAB, PAHPhysRAD provides a comprehensive suite of segmentation tools, including manual, semi-automatic, and automatic options. For semi-autosegmentation, meta AI's Segment Anything Model was incorporated using the bounding box methods. Autosegmentation of OARs and tumor volume are implemented through a module that enables the addition of models in Open Neural Network Exchange format. To validate the radiomic feature extraction module in PAHPhysRAD, radiomic features extracted from gross tumor volume of 15 non-small cell lung carcinoma patients were compared against the features extracted from 3D Slicer™. The dose-volume parameters extraction module was validated using the dose volume data extracted from 28 tangential field-based breast treatment planning datasets. The volume receiving ≥20 Gy (V20) for ipsilateral lung and the mean doses received by the heart and ipsilateral lung, were compared against the parameters extracted from Eclipse. Results The Wilcoxon signed-rank test revealed no significant difference between the majority of the radiomic features derived from PAHPhysRAD and 3D Slicer. The average mean lung and heart doses calculated in Eclipse were 5.51 ± 2.28 Gy and 1.64 ± 1.98 Gy, respectively. Similarly, the average mean lung and heart doses calculated in PAHPhysRAD were 5.45 ± 2.89 Gy and 1.67 ± 2.08 Gy, respectively. Conclusion The MATLAB-based graphical user interface, PAHPhysRAD, offers a user-friendly platform for viewing and analyzing medical scans with options to extract radiomic features and dose-volume parameters. Its versatility, compatibility, and potential for further development make it an asset in medical image analysis.
Collapse
Affiliation(s)
- Daniel Arrington
- Department of Radiation Oncology, Cancer Services, Princess Alexandra Hospital, Woolloongabba, Australia
| | - Ryan Motley
- Department of Radiation Oncology, Cancer Services, Princess Alexandra Hospital, Woolloongabba, Australia
| | - Zachery Morton Colbert
- Department of Radiation Oncology, Cancer Services, Princess Alexandra Hospital, Woolloongabba, Australia
| | - Margot Lehman
- Department of Radiation Oncology, Cancer Services, Princess Alexandra Hospital, Woolloongabba, Australia
| | - Prabhakar Ramachandran
- Department of Radiation Oncology, Cancer Services, Princess Alexandra Hospital, Woolloongabba, Australia
| |
Collapse
|
30
|
Saeed N, Ridzuan M, Alasmawi H, Sobirov I, Yaqub M. MGMT promoter methylation status prediction using MRI scans? An extensive experimental evaluation of deep learning models. Med Image Anal 2023; 90:102989. [PMID: 37827111 DOI: 10.1016/j.media.2023.102989] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/24/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023]
Abstract
The number of studies on deep learning for medical diagnosis is expanding, and these systems are often claimed to outperform clinicians. However, only a few systems have shown medical efficacy. From this perspective, we examine a wide range of deep learning algorithms for the assessment of glioblastoma - a common brain tumor in older adults that is lethal. Surgery, chemotherapy, and radiation are the standard treatments for glioblastoma patients. The methylation status of the MGMT promoter, a specific genetic sequence found in the tumor, affects chemotherapy's effectiveness. MGMT promoter methylation improves chemotherapy response and survival in several cancers. MGMT promoter methylation is determined by a tumor tissue biopsy, which is then genetically tested. This lengthy and invasive procedure increases the risk of infection and other complications. Thus, researchers have used deep learning models to examine the tumor from brain MRI scans to determine the MGMT promoter's methylation state. We employ deep learning models and one of the largest public MRI datasets of 585 participants to predict the methylation status of the MGMT promoter in glioblastoma tumors using MRI scans. We test these models using Grad-CAM, occlusion sensitivity, feature visualizations, and training loss landscapes. Our results show no correlation between these two, indicating that external cohort data should be used to verify these models' performance to assure the accuracy and reliability of deep learning systems in cancer diagnosis.
Collapse
Affiliation(s)
- Numan Saeed
- Department of Machine Learning, Mohamed bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates.
| | - Muhammad Ridzuan
- Department of Machine Learning, Mohamed bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
| | - Hussain Alasmawi
- Department of Machine Learning, Mohamed bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
| | - Ikboljon Sobirov
- Department of Computer Vision, Mohamed bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
| | - Mohammad Yaqub
- Department of Computer Vision, Mohamed bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
| |
Collapse
|
31
|
Ruffle JK, Mohinta S, Pombo G, Gray R, Kopanitsa V, Lee F, Brandner S, Hyare H, Nachev P. Brain tumour genetic network signatures of survival. Brain 2023; 146:4736-4754. [PMID: 37665980 PMCID: PMC10629773 DOI: 10.1093/brain/awad199] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/12/2023] [Accepted: 05/30/2023] [Indexed: 09/06/2023] Open
Abstract
Tumour heterogeneity is increasingly recognized as a major obstacle to therapeutic success across neuro-oncology. Gliomas are characterized by distinct combinations of genetic and epigenetic alterations, resulting in complex interactions across multiple molecular pathways. Predicting disease evolution and prescribing individually optimal treatment requires statistical models complex enough to capture the intricate (epi)genetic structure underpinning oncogenesis. Here, we formalize this task as the inference of distinct patterns of connectivity within hierarchical latent representations of genetic networks. Evaluating multi-institutional clinical, genetic and outcome data from 4023 glioma patients over 14 years, across 12 countries, we employ Bayesian generative stochastic block modelling to reveal a hierarchical network structure of tumour genetics spanning molecularly confirmed glioblastoma, IDH-wildtype; oligodendroglioma, IDH-mutant and 1p/19q codeleted; and astrocytoma, IDH-mutant. Our findings illuminate the complex dependence between features across the genetic landscape of brain tumours and show that generative network models reveal distinct signatures of survival with better prognostic fidelity than current gold standard diagnostic categories.
Collapse
Affiliation(s)
- James K Ruffle
- Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Samia Mohinta
- Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Guilherme Pombo
- Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Robert Gray
- Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Valeriya Kopanitsa
- Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Faith Lee
- Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Sebastian Brandner
- Division of Neuropathology and Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Harpreet Hyare
- Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Parashkev Nachev
- Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| |
Collapse
|
32
|
Zhou Z, Qian X, Hu J, Chen G, Zhang C, Zhu J, Dai Y. An artificial intelligence-assisted diagnosis modeling software (AIMS) platform based on medical images and machine learning: a development and validation study. Quant Imaging Med Surg 2023; 13:7504-7522. [PMID: 37969634 PMCID: PMC10644131 DOI: 10.21037/qims-23-20] [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: 01/15/2023] [Accepted: 06/12/2023] [Indexed: 11/17/2023]
Abstract
Background Supervised machine learning methods [both radiomics and convolutional neural network (CNN)-based deep learning] are usually employed to develop artificial intelligence models with medical images for computer-assisted diagnosis and prognosis of diseases. A classical machine learning-based modeling workflow involves a series of interconnected components and various algorithms, but this makes it challenging, tedious, and labor intensive for radiologists and researchers to build customized models for specific clinical applications if they lack expertise in machine learning methods. Methods We developed a user-friendly artificial intelligence-assisted diagnosis modeling software (AIMS) platform, which supplies standardized machine learning-based modeling workflows for computer-assisted diagnosis and prognosis systems with medical images. In contrast to other existing software platforms, AIMS contains both radiomics and CNN-based deep learning workflows, making it an all-in-one software platform for machine learning-based medical image analysis. The modular design of AIMS allows users to build machine learning models easily, test models comprehensively, and fairly compare the performance of different models in a specific application. The graphical user interface (GUI) enables users to process large numbers of medical images without programming or script addition. Furthermore, AIMS also provides a flexible image processing toolkit (e.g., semiautomatic segmentation, registration, morphological operations) to rapidly create lesion labels for multiphase analysis, multiregion analysis of an individual tumor (e.g., tumor mass and peritumor), and multimodality analysis. Results The functionality and efficiency of AIMS were demonstrated in 3 independent experiments in radiation oncology, where multiphase, multiregion, and multimodality analyses were performed, respectively. For clear cell renal cell carcinoma (ccRCC) Fuhrman grading with multiphase analysis (sample size =187), the area under the curve (AUC) value of the AIMS was 0.776; for ccRCC Fuhrman grading with multiregion analysis (sample size =177), the AUC value of the AIMS was 0.848; for prostate cancer Gleason grading with multimodality analysis (sample size =206), the AUC value of the AIMS was 0.980. Conclusions AIMS provides a user-friendly infrastructure for radiologists and researchers, lowering the barrier to building customized machine learning-based computer-assisted diagnosis models for medical image analysis.
Collapse
Affiliation(s)
- Zhiyong Zhou
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Xusheng Qian
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jisu Hu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Guangqiang Chen
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Caiyuan Zhang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianbing Zhu
- Suzhou Science & Technology Town Hospital, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Yakang Dai
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
- Suzhou Guoke Kangcheng Medical Technology Co., Ltd., Suzhou, China
| |
Collapse
|
33
|
Bianconi A, Rossi LF, Bonada M, Zeppa P, Nico E, De Marco R, Lacroce P, Cofano F, Bruno F, Morana G, Melcarne A, Ruda R, Mainardi L, Fiaschi P, Garbossa D, Morra L. Deep learning-based algorithm for postoperative glioblastoma MRI segmentation: a promising new tool for tumor burden assessment. Brain Inform 2023; 10:26. [PMID: 37801128 PMCID: PMC10558414 DOI: 10.1186/s40708-023-00207-6] [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: 03/31/2023] [Accepted: 09/16/2023] [Indexed: 10/07/2023] Open
Abstract
OBJECTIVE Clinical and surgical decisions for glioblastoma patients depend on a tumor imaging-based evaluation. Artificial Intelligence (AI) can be applied to magnetic resonance imaging (MRI) assessment to support clinical practice, surgery planning and prognostic predictions. In a real-world context, the current obstacles for AI are low-quality imaging and postoperative reliability. The aim of this study is to train an automatic algorithm for glioblastoma segmentation on a clinical MRI dataset and to obtain reliable results both pre- and post-operatively. METHODS The dataset used for this study comprises 237 (71 preoperative and 166 postoperative) MRIs from 71 patients affected by a histologically confirmed Grade IV Glioma. The implemented U-Net architecture was trained by transfer learning to perform the segmentation task on postoperative MRIs. The training was carried out first on BraTS2021 dataset for preoperative segmentation. Performance is evaluated using DICE score (DS) and Hausdorff 95% (H95). RESULTS In preoperative scenario, overall DS is 91.09 (± 0.60) and H95 is 8.35 (± 1.12), considering tumor core, enhancing tumor and whole tumor (ET and edema). In postoperative context, overall DS is 72.31 (± 2.88) and H95 is 23.43 (± 7.24), considering resection cavity (RC), gross tumor volume (GTV) and whole tumor (WT). Remarkably, the RC segmentation obtained a mean DS of 63.52 (± 8.90) in postoperative MRIs. CONCLUSIONS The performances achieved by the algorithm are consistent with previous literature for both pre-operative and post-operative glioblastoma's MRI evaluation. Through the proposed algorithm, it is possible to reduce the impact of low-quality images and missing sequences.
Collapse
Affiliation(s)
- Andrea Bianconi
- Neurosurgery, Department of Neuroscience, University of Turin, via Cherasco 15, 10126, Turin, Italy.
| | | | - Marta Bonada
- Neurosurgery, Department of Neuroscience, University of Turin, via Cherasco 15, 10126, Turin, Italy
| | - Pietro Zeppa
- Neurosurgery, Department of Neuroscience, University of Turin, via Cherasco 15, 10126, Turin, Italy
| | - Elsa Nico
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Raffaele De Marco
- Neurosurgery, Department of Neuroscience, University of Turin, via Cherasco 15, 10126, Turin, Italy
| | | | - Fabio Cofano
- Neurosurgery, Department of Neuroscience, University of Turin, via Cherasco 15, 10126, Turin, Italy
| | - Francesco Bruno
- Neurooncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Giovanni Morana
- Neuroradiology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Antonio Melcarne
- Neurosurgery, Department of Neuroscience, University of Turin, via Cherasco 15, 10126, Turin, Italy
| | - Roberta Ruda
- Neurooncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Luca Mainardi
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milan, Italy
| | - Pietro Fiaschi
- IRCCS Ospedale Policlinico S. Martino, Genoa, Italy
- Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Univeristy of Genoa, Genoa, Italy
| | - Diego Garbossa
- Neurosurgery, Department of Neuroscience, University of Turin, via Cherasco 15, 10126, Turin, Italy
| | - Lia Morra
- Dipartimento di Automatica e Informatica, Politecnico di Torino, Turin, Italy
| |
Collapse
|
34
|
Familiar AM, Kazerooni AF, Anderson H, Lubneuski A, Viswanathan K, Breslow R, Khalili N, Bagheri S, Haldar D, Kim MC, Arif S, Madhogarhia R, Nguyen TQ, Frenkel EA, Helili Z, Harrison J, Farahani K, Linguraru MG, Bagci U, Velichko Y, Stevens J, Leary S, Lober RM, Campion S, Smith AA, Morinigo D, Rood B, Diamond K, Pollack IF, Williams M, Vossough A, Ware JB, Mueller S, Storm PB, Heath AP, Waanders AJ, Lilly J, Mason JL, Resnick AC, Nabavizadeh A. A multi-institutional pediatric dataset of clinical radiology MRIs by the Children's Brain Tumor Network. ARXIV 2023:arXiv:2310.01413v1. [PMID: 38106459 PMCID: PMC10723526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Pediatric brain and spinal cancers remain the leading cause of cancer-related death in children. Advancements in clinical decision-support in pediatric neuro-oncology utilizing the wealth of radiology imaging data collected through standard care, however, has significantly lagged other domains. Such data is ripe for use with predictive analytics such as artificial intelligence (AI) methods, which require large datasets. To address this unmet need, we provide a multi-institutional, large-scale pediatric dataset of 23,101 multi-parametric MRI exams acquired through routine care for 1,526 brain tumor patients, as part of the Children's Brain Tumor Network. This includes longitudinal MRIs across various cancer diagnoses, with associated patient-level clinical information, digital pathology slides, as well as tissue genotype and omics data. To facilitate downstream analysis, treatment-naïve images for 370 subjects were processed and released through the NCI Childhood Cancer Data Initiative via the Cancer Data Service. Through ongoing efforts to continuously build these imaging repositories, our aim is to accelerate discovery and translational AI models with real-world data, to ultimately empower precision medicine for children.
Collapse
Affiliation(s)
- Ariana M. Familiar
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hannah Anderson
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aliaksandr Lubneuski
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Karthik Viswanathan
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rocky Breslow
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nastaran Khalili
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sina Bagheri
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Debanjan Haldar
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Meen Chul Kim
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sherjeel Arif
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rachel Madhogarhia
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Thinh Q. Nguyen
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth A. Frenkel
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Zeinab Helili
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jessica Harrison
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, 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
| | - Ulas Bagci
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yury Velichko
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jeffrey Stevens
- Department of Hematology and Oncology, Seattle Children’s, Seattle, WA, USA
| | - Sarah Leary
- Department of Hematology and Oncology, Seattle Children’s, Seattle, WA, USA
| | - Robert M. Lober
- Division of Neurosurgery, Dayton Children’s Hospital, Dayton, OH, USA
| | - Stephani Campion
- Department of Pediatric Hematology & Oncology, Orlando Health Arnold Palmer Hospital for Children, Orlando, FL, USA
| | - Amy A. Smith
- Department of Pediatric Hematology & Oncology, Orlando Health Arnold Palmer Hospital for Children, Orlando, FL, USA
| | - Denise Morinigo
- Department of Hematology-Oncology, Children’s National Hospital, Washington, DC, USA
| | - Brian Rood
- Department of Hematology-Oncology, Children’s National Hospital, Washington, DC, USA
| | - Kimberly Diamond
- Department of Pediatric Neurosurgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Ian F. Pollack
- Department of Pediatric Neurosurgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Melissa Williams
- Division of Hematology, Oncology, NeuroOncology, and Transplant, Ann & Robert H Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Arastoo Vossough
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeffrey B. Ware
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sabine Mueller
- Department of Neurology, Division of Child Neurology, University of San Francisco, San Francisco, CA, USA
| | - Phillip B. Storm
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Allison P. Heath
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Angela J. Waanders
- Division of Hematology, Oncology, NeuroOncology, and Transplant, Ann & Robert H Lurie Children’s Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jena Lilly
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jennifer L. Mason
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adam C. Resnick
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ali Nabavizadeh
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
35
|
Ghosh A, Yekeler E, Teixeira SR, Dalal D, States L. Role of MRI radiomics for the prediction of MYCN amplification in neuroblastomas. Eur Radiol 2023; 33:6726-6735. [PMID: 37178203 DOI: 10.1007/s00330-023-09628-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 02/18/2023] [Accepted: 02/26/2023] [Indexed: 05/15/2023]
Abstract
OBJECTIVES We evaluate MR radiomics and develop machine learning-based classifiers to predict MYCN amplification in neuroblastomas. METHODS A total of 120 patients with neuroblastomas and baseline MR imaging examination available were identified of whom 74 (mean age ± standard deviation [SD] of 6 years and 2 months ± 4 years and 9 months; 43 females and 31 males, 14 MYCN amplified) underwent imaging at our institution. This was therefore used to develop radiomics models. The model was tested in a cohort of children with the same diagnosis but imaged elsewhere (n = 46, mean age ± SD: 5 years 11 months ± 3 years 9 months, 26 females and 14 MYCN amplified). Whole tumour volumes of interest were adopted to extract first-order histogram and second-order radiomics features. Interclass correlation coefficient and maximum relevance and minimum redundancy algorithm were applied for feature selection. Logistic regression, support vector machine, and random forest were employed as the classifiers. Receiver operating characteristic (ROC) analysis was performed to evaluate the diagnostic accuracy of the classifiers on the external test set. RESULTS The logistic regression model and the random forest both showed an AUC of 0.75. The support vector machine classifier obtained an AUC of 0.78 on the test set with a sensitivity of 64% and a specificity of 72%. CONCLUSION The study provides preliminary retrospective evidence demonstrating the feasibility of MRI radiomics in predicting MYCN amplification in neuroblastomas. Future studies are needed to explore the correlation between other imaging features and genetic markers and to develop multiclass predictive models. KEY POINTS • MYCN amplification in neuroblastomas is an important determinant of disease prognosis. • Radiomics analysis of pre-treatment MR examinations can be used to predict MYCN amplification in neuroblastomas. • Radiomics machine learning models showed good generalisability to external test set, demonstrating reproducibility of the computational models.
Collapse
Affiliation(s)
- Adarsh Ghosh
- Department of Radiology, Cincinnati Children's Hospital and Medical Centre, Cincinnati, OH, USA.
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Ensar Yekeler
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sara Reis Teixeira
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Deepa Dalal
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lisa States
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
36
|
Cepeda S, García-García S, Arrese I, Herrero F, Escudero T, Zamora T, Sarabia R. The Río Hortega University Hospital Glioblastoma dataset: A comprehensive collection of preoperative, early postoperative and recurrence MRI scans (RHUH-GBM). Data Brief 2023; 50:109617. [PMID: 37808543 PMCID: PMC10551826 DOI: 10.1016/j.dib.2023.109617] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/10/2023] Open
Abstract
Glioblastoma, a highly aggressive primary brain tumor, is associated with poor patient outcomes. Although magnetic resonance imaging (MRI) plays a critical role in diagnosing, characterizing, and forecasting glioblastoma progression, public MRI repositories present significant drawbacks, including insufficient postoperative and follow-up studies as well as expert tumor segmentations. To address these issues, we present the "Río Hortega University Hospital Glioblastoma Dataset (RHUH-GBM)," a collection of multiparametric MRI images, volumetric assessments, molecular data, and survival details for glioblastoma patients who underwent total or near-total enhancing tumor resection. The dataset features expert-corrected segmentations of tumor subregions, offering valuable ground truth data for developing algorithms for postoperative and follow-up MRI scans.
Collapse
Affiliation(s)
- Santiago Cepeda
- Department of Neurosurgery, Río Hortega University Hospital, Dulzaina 2, 47012 Valladolid, Spain
| | - Sergio García-García
- Department of Neurosurgery, Río Hortega University Hospital, Dulzaina 2, 47012 Valladolid, Spain
| | - Ignacio Arrese
- Department of Neurosurgery, Río Hortega University Hospital, Dulzaina 2, 47012 Valladolid, Spain
| | - Francisco Herrero
- Department of Radiology, Río Hortega University Hospital, Dulzaina 2, 47012 Valladolid, Spain
| | - Trinidad Escudero
- Department of Radiology, Río Hortega University Hospital, Dulzaina 2, 47012 Valladolid, Spain
| | - Tomás Zamora
- Department of Pathology, Río Hortega University Hospital, Dulzaina 2, 47012 Valladolid, Spain
| | - Rosario Sarabia
- Department of Neurosurgery, Río Hortega University Hospital, Dulzaina 2, 47012 Valladolid, Spain
| |
Collapse
|
37
|
Perillo T, de Giorgi M, Papace UM, Serino A, Cuocolo R, Manto A. Current role of machine learning and radiogenomics in precision neuro-oncology. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:545-555. [PMID: 37720347 PMCID: PMC10501892 DOI: 10.37349/etat.2023.00151] [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: 12/20/2022] [Accepted: 04/20/2023] [Indexed: 09/19/2023] Open
Abstract
In the past few years, artificial intelligence (AI) has been increasingly used to create tools that can enhance workflow in medicine. In particular, neuro-oncology has benefited from the use of AI and especially machine learning (ML) and radiogenomics, which are subfields of AI. ML can be used to develop algorithms that dynamically learn from available medical data in order to automatically do specific tasks. On the other hand, radiogenomics can identify relationships between tumor genetics and imaging features, thus possibly giving new insights into the pathophysiology of tumors. Therefore, ML and radiogenomics could help treatment tailoring, which is crucial in personalized neuro-oncology. The aim of this review is to illustrate current and possible future applications of ML and radiomics in neuro-oncology.
Collapse
Affiliation(s)
- Teresa Perillo
- Department of Neuroradiology, “Umberto I” Hospital, 84014 Norcera Inferiore, Italy
| | - Marco de Giorgi
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80138 Naples, Italy
| | - Umberto Maria Papace
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80138 Naples, Italy
| | - Antonietta Serino
- Department of Neuroradiology, “Umberto I” Hospital, 84014 Norcera Inferiore, Italy
| | - Renato Cuocolo
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84084 Fisciano, Italy
| | - Andrea Manto
- Department of Neuroradiology, “Umberto I” Hospital, 84014 Norcera Inferiore, Italy
| |
Collapse
|
38
|
Rezaeijo SM, Chegeni N, Baghaei Naeini F, Makris D, Bakas S. Within-Modality Synthesis and Novel Radiomic Evaluation of Brain MRI Scans. Cancers (Basel) 2023; 15:3565. [PMID: 37509228 PMCID: PMC10377568 DOI: 10.3390/cancers15143565] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/27/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
One of the most common challenges in brain MRI scans is to perform different MRI sequences depending on the type and properties of tissues. In this paper, we propose a generative method to translate T2-Weighted (T2W) Magnetic Resonance Imaging (MRI) volume from T2-weight-Fluid-attenuated-Inversion-Recovery (FLAIR) and vice versa using Generative Adversarial Networks (GAN). To evaluate the proposed method, we propose a novel evaluation schema for generative and synthetic approaches based on radiomic features. For the evaluation purpose, we consider 510 pair-slices from 102 patients to train two different GAN-based architectures Cycle GAN and Dual Cycle-Consistent Adversarial network (DC2Anet). The results indicate that generative methods can produce similar results to the original sequence without significant change in the radiometric feature. Therefore, such a method can assist clinics to make decisions based on the generated image when different sequences are not available or there is not enough time to re-perform the MRI scans.
Collapse
Affiliation(s)
- Seyed Masoud Rezaeijo
- Department of Medical Physics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; (S.M.R.)
| | - Nahid Chegeni
- Department of Medical Physics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; (S.M.R.)
| | - Fariborz Baghaei Naeini
- Faculty of Engineering, Computing and the Environment, Kingston University, Penrhyn Road Campus, Kingston upon Thames, London KT1 2EE, UK; (F.B.N.); (D.M.)
| | - Dimitrios Makris
- Faculty of Engineering, Computing and the Environment, Kingston University, Penrhyn Road Campus, Kingston upon Thames, London KT1 2EE, UK; (F.B.N.); (D.M.)
| | - Spyridon Bakas
- Faculty of Engineering, Computing and the Environment, Kingston University, Penrhyn Road Campus, Kingston upon Thames, London KT1 2EE, UK; (F.B.N.); (D.M.)
- Richards Medical Research Laboratories, Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Floor 7, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| |
Collapse
|
39
|
Adewole M, Rudie JD, Gbdamosi A, Toyobo O, Raymond C, Zhang D, Omidiji O, Akinola R, Suwaid MA, Emegoakor A, Ojo N, Aguh K, Kalaiwo C, Babatunde G, Ogunleye A, Gbadamosi Y, Iorpagher K, Calabrese E, Aboian M, Linguraru M, Albrecht J, Wiestler B, Kofler F, Janas A, LaBella D, Kzerooni AF, Li HB, Iglesias JE, Farahani K, Eddy J, Bergquist T, Chung V, Shinohara RT, Wiggins W, Reitman Z, Wang C, Liu X, Jiang Z, Familiar A, Van Leemput K, Bukas C, Piraud M, Conte GM, Johansson E, Meier Z, Menze BH, Baid U, Bakas S, Dako F, Fatade A, Anazodo UC. The Brain Tumor Segmentation (BraTS) Challenge 2023: Glioma Segmentation in Sub-Saharan Africa Patient Population (BraTS-Africa). ARXIV 2023:arXiv:2305.19369v1. [PMID: 37396608 PMCID: PMC10312814] [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
Gliomas are the most common type of primary brain tumors. Although gliomas are relatively rare, they are among the deadliest types of cancer, with a survival rate of less than 2 years after diagnosis. Gliomas are challenging to diagnose, hard to treat and inherently resistant to conventional therapy. Years of extensive research to improve diagnosis and treatment of gliomas have decreased mortality rates across the Global North, while chances of survival among individuals in low- and middle-income countries (LMICs) remain unchanged and are significantly worse in Sub-Saharan Africa (SSA) populations. Long-term survival with glioma is associated with the identification of appropriate pathological features on brain MRI and confirmation by histopathology. Since 2012, the Brain Tumor Segmentation (BraTS) Challenge have evaluated state-of-the-art machine learning methods to detect, characterize, and classify gliomas. However, it is unclear if the state-of-the-art methods can be widely implemented in SSA given the extensive use of lower-quality MRI technology, which produces poor image contrast and resolution and more importantly, the propensity for late presentation of disease at advanced stages as well as the unique characteristics of gliomas in SSA (i.e., suspected higher rates of gliomatosis cerebri). Thus, the BraTS-Africa Challenge provides a unique opportunity to include brain MRI glioma cases from SSA in global efforts through the BraTS Challenge to develop and evaluate computer-aided-diagnostic (CAD) methods for the detection and characterization of glioma in resource-limited settings, where the potential for CAD tools to transform healthcare are more likely.
Collapse
Affiliation(s)
- Maruf Adewole
- Medical Artificial Intelligence Laboratory (MAI Lab), Lagos, Nigeria
- Department of Radiation Biology, Radiotherapy and Radiodiagnosis, University of Lagos, Lagos, Nigeria
| | - Jeffrey D Rudie
- Department of Radiology, University of California, San Diego
| | - Anu Gbdamosi
- Medical Artificial Intelligence Laboratory (MAI Lab), Lagos, Nigeria
- Crestview Radiology Limited, Lagos, Nigeria
| | - Oluyemisi Toyobo
- Medical Artificial Intelligence Laboratory (MAI Lab), Lagos, Nigeria
- Crestview Radiology Limited, Lagos, Nigeria
| | | | - Dong Zhang
- Medical Artificial Intelligence Laboratory (MAI Lab), Lagos, Nigeria
| | - Olubukola Omidiji
- Medical Artificial Intelligence Laboratory (MAI Lab), Lagos, Nigeria
- Lagos University Teaching Hospital, Lagos, Nigeria
| | - Rachel Akinola
- Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | | | - Adaobi Emegoakor
- Nnamdi Azikiwe University Teaching Hospital, Nnewi, Anambra State, Nigeria
| | - Nancy Ojo
- Federal Medical Centre, Abeokuta, Ogun State, Nigeria
| | - Kenneth Aguh
- Federal Medical Centre, Umahia, Abia State, Nigeria
| | | | | | | | | | - Kator Iorpagher
- Benue State University Teaching Hospital, Markurdi, Benue State, Nigeria
| | - Evan Calabrese
- Duke University Medical Center, Department of Radiology, USA
- University of California San Francisco, CA, USA
| | | | - Marius Linguraru
- Children's National Hospital, Washington DC, USA
- George Washington University, Washington DC, USA
| | | | - Benedikt Wiestler
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
| | - Florian Kofler
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
- Helmholtz Research Center, Munich, Germany
| | | | - Dominic LaBella
- Duke University Medical Center, Department of Radiation Oncology, USA
| | - Anahita Fathi Kzerooni
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
- Center for AI and Data Science for Integrated Diagnostics (AI2D) & Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Hongwei Bran Li
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
- University of Zurich, Switzerland
| | - Juan Eugenio Iglesias
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, 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) & 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
| | - 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
| | - Koen Van Leemput
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Denmark
| | | | | | | | - Elaine Johansson
- Precision FDA, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | | | - Bjoern H Menze
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
- University of Zurich, Switzerland
| | - Ujjwal Baid
- Center for AI and Data Science for Integrated Diagnostics (AI2D) & 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) & 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
| | - Farouk Dako
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Abiodun Fatade
- Medical Artificial Intelligence Laboratory (MAI Lab), Lagos, Nigeria
- Crestview Radiology Limited, Lagos, Nigeria
| | - Udunna C Anazodo
- Medical Artificial Intelligence Laboratory (MAI Lab), Lagos, Nigeria
- Montreal Neurological Institute, McGill University, Montreal, Canada
- Department of Medicine, University of Cape Town, South Africa
- Department of Radiation Medicine, University of Cape Town, South Africa
| |
Collapse
|
40
|
Pati S, Thakur SP, Hamamcı İE, Baid U, Baheti B, Bhalerao M, Güley O, Mouchtaris S, Lang D, Thermos S, Gotkowski K, González C, Grenko C, Getka A, Edwards B, Sheller M, Wu J, Karkada D, Panchumarthy R, Ahluwalia V, Zou C, Bashyam V, Li Y, Haghighi B, Chitalia R, Abousamra S, Kurc TM, Gastounioti A, Er S, Bergman M, Saltz JH, Fan Y, Shah P, Mukhopadhyay A, Tsaftaris SA, Menze B, Davatzikos C, Kontos D, Karargyris A, Umeton R, Mattson P, Bakas S. GaNDLF: the generally nuanced deep learning framework for scalable end-to-end clinical workflows. COMMUNICATIONS ENGINEERING 2023; 2:23. [PMCID: PMC10956028 DOI: 10.1038/s44172-023-00066-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 03/27/2023] [Indexed: 01/06/2025]
Abstract
Deep Learning (DL) has the potential to optimize machine learning in both the scientific and clinical communities. However, greater expertise is required to develop DL algorithms, and the variability of implementations hinders their reproducibility, translation, and deployment. Here we present the community-driven Generally Nuanced Deep Learning Framework (GaNDLF), with the goal of lowering these barriers. GaNDLF makes the mechanism of DL development, training, and inference more stable, reproducible, interpretable, and scalable, without requiring an extensive technical background. GaNDLF aims to provide an end-to-end solution for all DL-related tasks in computational precision medicine. We demonstrate the ability of GaNDLF to analyze both radiology and histology images, with built-in support for k -fold cross-validation, data augmentation, multiple modalities and output classes. Our quantitative performance evaluation on numerous use cases, anatomies, and computational tasks supports GaNDLF as a robust application framework for deployment in clinical workflows. The increasing complexity of the implementation and operation of deep learning techniques hinders their reproducibility and deployment at scale, especially in healthcare. Pati and colleagues introduce a deep learning framework to analyse healthcare data without requiring extensive computational experience, facilitating the integration of artificial intelligence in clinical workflows.
Collapse
Affiliation(s)
- Sarthak Pati
- MLCommons, Medical Working Group, San Francisco, CA USA
- Center For Artificial Intelligence And Data Science For Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
- Department of Informatics, Technical University of Munich, Munich, Bavaria Germany
| | - Siddhesh P. Thakur
- Center For Artificial Intelligence And Data Science For Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - İbrahim Ethem Hamamcı
- Center For Artificial Intelligence And Data Science For Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
- International School of Medicine, Istanbul Medipol University, Istanbul, Marmara Turkey
| | - Ujjwal Baid
- Center For Artificial Intelligence And Data Science For Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Bhakti Baheti
- Center For Artificial Intelligence And Data Science For Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Megh Bhalerao
- Center For Artificial Intelligence And 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
| | - Orhun Güley
- Department of Informatics, Technical University of Munich, Munich, Bavaria Germany
| | - Sofia Mouchtaris
- Center For Artificial Intelligence And Data Science For Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA USA
| | - David Lang
- Center For Artificial Intelligence And Data Science For Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA USA
- Department of Mathematics, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA USA
| | - Spyridon Thermos
- Institute for Digital Communications, School of Engineering, The University of Edinburgh, Scotland, UK
| | - Karol Gotkowski
- Department of Computer Science, Technical University of Darmstadt, Darmstadt, Hesse Germany
| | - Camila González
- Department of Computer Science, Technical University of Darmstadt, Darmstadt, Hesse Germany
| | - Caleb Grenko
- Center For Artificial Intelligence And Data Science For Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Alexander Getka
- Center For Artificial Intelligence And 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
| | | | - Micah Sheller
- MLCommons, Medical Working Group, San Francisco, CA USA
- Intel Corporation, Santa Clara, CA USA
| | - Junwen Wu
- Intel Corporation, Santa Clara, CA USA
| | | | | | - Vinayak Ahluwalia
- Center For Artificial Intelligence And 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
| | - Chunrui Zou
- Center For Artificial Intelligence And 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
| | - Vishnu Bashyam
- Center For Artificial Intelligence And 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
| | - Yuemeng Li
- Center For Artificial Intelligence And 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
| | - Babak Haghighi
- Center For Artificial Intelligence And 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
| | - Rhea Chitalia
- Center For Artificial Intelligence And 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
| | - Shahira Abousamra
- Department of Computer Science, Stony Brook University, Stony Brook, New York, NY USA
| | - Tahsin M. Kurc
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York, NY USA
| | - Aimilia Gastounioti
- Center For Artificial Intelligence And 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
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO USA
| | - Sezgin Er
- International School of Medicine, Istanbul Medipol University, Istanbul, Marmara Turkey
| | - Mark Bergman
- Center For Artificial Intelligence And 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
| | - Joel H. Saltz
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York, NY USA
| | - Yong Fan
- Center For Artificial Intelligence And 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
| | | | - Anirban Mukhopadhyay
- Department of Computer Science, Technical University of Darmstadt, Darmstadt, Hesse Germany
| | - Sotirios A. Tsaftaris
- Institute for Digital Communications, School of Engineering, The University of Edinburgh, Scotland, UK
| | - Bjoern Menze
- Department of Informatics, Technical University of Munich, Munich, Bavaria Germany
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Christos Davatzikos
- Center For Artificial Intelligence And 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
| | - Despina Kontos
- Center For Artificial Intelligence And 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
| | - Alexandros Karargyris
- MLCommons, Medical Working Group, San Francisco, CA USA
- Institute of Image-Guided Surgery of Strasbourg, Strasbourg, France
| | - Renato Umeton
- MLCommons, Medical Working Group, San Francisco, CA USA
- Department of Informatics & Analytics, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
- Department of Biological Engineering, Department of Mechanical Engineering, Massachusetts Institute of Technology, Boston, MA USA
| | - Peter Mattson
- MLCommons, Medical Working Group, San Francisco, CA USA
- Google, Menlo Park, CA USA
| | - Spyridon Bakas
- MLCommons, Medical Working Group, San Francisco, CA USA
- Center For Artificial Intelligence And Data Science For Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|
41
|
Chakrabarty S, Abidi SA, Mousa M, Mokkarala M, Hren I, Yadav D, Kelsey M, LaMontagne P, Wood J, Adams M, Su Y, Thorpe S, Chung C, Sotiras A, Marcus DS. Integrative Imaging Informatics for Cancer Research: Workflow Automation for Neuro-Oncology (I3CR-WANO). JCO Clin Cancer Inform 2023; 7:e2200177. [PMID: 37146265 PMCID: PMC10281444 DOI: 10.1200/cci.22.00177] [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: 11/21/2022] [Revised: 01/25/2023] [Accepted: 03/06/2023] [Indexed: 05/07/2023] Open
Abstract
PURPOSE Efforts to use growing volumes of clinical imaging data to generate tumor evaluations continue to require significant manual data wrangling, owing to data heterogeneity. Here, we propose an artificial intelligence-based solution for the aggregation and processing of multisequence neuro-oncology MRI data to extract quantitative tumor measurements. MATERIALS AND METHODS Our end-to-end framework (1) classifies MRI sequences using an ensemble classifier, (2) preprocesses the data in a reproducible manner, (3) delineates tumor tissue subtypes using convolutional neural networks, and (4) extracts diverse radiomic features. Moreover, it is robust to missing sequences and adopts an expert-in-the-loop approach in which the segmentation results may be manually refined by radiologists. After the implementation of the framework in Docker containers, it was applied to two retrospective glioma data sets collected from the Washington University School of Medicine (WUSM; n = 384) and The University of Texas MD Anderson Cancer Center (MDA; n = 30), comprising preoperative MRI scans from patients with pathologically confirmed gliomas. RESULTS The scan-type classifier yielded an accuracy of >99%, correctly identifying sequences from 380 of 384 and 30 of 30 sessions from the WUSM and MDA data sets, respectively. Segmentation performance was quantified using the Dice Similarity Coefficient between the predicted and expert-refined tumor masks. The mean Dice scores were 0.882 (±0.244) and 0.977 (±0.04) for whole-tumor segmentation for WUSM and MDA, respectively. CONCLUSION This streamlined framework automatically curated, processed, and segmented raw MRI data of patients with varying grades of gliomas, enabling the curation of large-scale neuro-oncology data sets and demonstrating high potential for integration as an assistive tool in clinical practice.
Collapse
Affiliation(s)
- Satrajit Chakrabarty
- Department of Electrical and Systems Engineering, Washington University in St Louis, St Louis, MO
| | - Syed Amaan Abidi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO
| | - Mina Mousa
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO
| | - Mahati Mokkarala
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO
| | - Isabelle Hren
- Department of Computer Science & Engineering, Washington University in St Louis, St Louis, MO
| | - Divya Yadav
- Division of Radiation Oncology, Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Matthew Kelsey
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO
| | - John Wood
- Division of Radiation Oncology, Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael Adams
- Division of Radiation Oncology, Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yuzhuo Su
- Division of Radiation Oncology, Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sherry Thorpe
- Division of Radiation Oncology, Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Caroline Chung
- Division of Radiation Oncology, Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Aristeidis Sotiras
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO
- Institute for Informatics, Washington University School of Medicine, St Louis, MO
| | - Daniel S. Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
42
|
Robinet L, Siegfried A, Roques M, Berjaoui A, Cohen-Jonathan Moyal E. MRI-Based Deep Learning Tools for MGMT Promoter Methylation Detection: A Thorough Evaluation. Cancers (Basel) 2023; 15:cancers15082253. [PMID: 37190181 DOI: 10.3390/cancers15082253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/29/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Glioblastoma is the most aggressive primary brain tumor, which almost systematically relapses despite surgery (when possible) followed by radio-chemotherapy temozolomide-based treatment. Upon relapse, one option for treatment is another chemotherapy, lomustine. The efficacy of these chemotherapy regimens depends on the methylation of a specific gene promoter known as MGMT, which is the main prognosis factor for glioblastoma. Knowing this biomarker is a key issue for the clinician to personalize and adapt treatment to the patient at primary diagnosis for elderly patients, in particular, and also upon relapse. The association between MRI-derived information and the prediction of MGMT promoter status has been discussed in many studies, and some, more recently, have proposed the use of deep learning algorithms on multimodal scans to extract this information, but they have failed to reach a consensus. Therefore, in this work, beyond the classical performance figures usually displayed, we seek to compute confidence scores to see if a clinical application of such methods can be seriously considered. The systematic approach carried out, using different input configurations and algorithms as well as the exact methylation percentage, led to the following conclusion: current deep learning methods are unable to determine MGMT promoter methylation from MRI data.
Collapse
Affiliation(s)
- Lucas Robinet
- IRT Saint-Exupéry, 31400 Toulouse, France
- IUCT-Oncopole-Institut Claudius Regaud, 31100 Toulouse, France
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), University Paul Sabatier Toulouse III, 31100 Toulouse, France
| | - Aurore Siegfried
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), University Paul Sabatier Toulouse III, 31100 Toulouse, France
- Pathology and Cytology Department, CHU Toulouse, IUCT Oncopole, 31100 Toulouse, France
| | - Margaux Roques
- Department of Neuroradiology, Hopital Pierre Paul Riquet, CHU Purpan, 31300 Toulouse, France
| | | | - Elizabeth Cohen-Jonathan Moyal
- IUCT-Oncopole-Institut Claudius Regaud, 31100 Toulouse, France
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), University Paul Sabatier Toulouse III, 31100 Toulouse, France
| |
Collapse
|
43
|
Pacheco BM, Cassia GDSE, Silva D. Towards fully automated deep-learning-based brain tumor segmentation: Is brain extraction still necessary? Biomed Signal Process Control 2023. [DOI: 10.1016/j.bspc.2022.104514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
44
|
Bornes R, Montero J, Correia A, Marques T, Rosa N. Peri-implant diseases diagnosis, prognosis and dental implant monitoring: a narrative review of novel strategies and clinical impact. BMC Oral Health 2023; 23:183. [PMID: 36997949 PMCID: PMC10061972 DOI: 10.1186/s12903-023-02896-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/17/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND The diagnosis of peri-implantar and periodontal relies mainly on a set of clinical measures and the evaluation of radiographic images. However, these clinical settings alone are not sufficient to determine, much less predict, periimplant bone loss or future implant failure. Early diagnosis of periimplant diseases and its rate of progress may be possible through biomarkers assessment. Once identified, biomarkers of peri-implant and periodontal tissue destruction may alert the clinicians before clinical signs show up. Therefore, it is important to consider developing chair-side diagnostic tests with specificity for a particular biomarker, indicating the current activity of the disease. METHODS A search strategy was created at Pubmed and Web of Science to answer the question: "How the molecular point-of-care tests currently available can help in the early detection of peri-implant diseases and throws light on improvements in point of care diagnostics devices?" RESULTS The PerioSafe® PRO DRS (dentognostics GmbH, Jena) and ImplantSafe® DR (dentognostics GmbH, Jena ORALyzer® test kits, already used clinically, can be a helpful adjunct tool in enhancing the diagnosis and prognosis of periodontal/peri-implantar diseases. With the advances of sensor technology, the biosensors can perform daily monitoring of dental implants or periodontal diseases, making contributions to personal healthcare and improve the current status quo of health management and human health. CONCLUSIONS Based on the findings, more emphasis is given to the role of biomarkers in diagnosing and monitoring periodontal and peri-implant diseases. By combining these strategies with traditional protocols, professionals could increase the accuracy of early detection of peri-implant and periodontal diseases, predicting disease progression, and monitoring of treatment outcomes.
Collapse
Affiliation(s)
- Rita Bornes
- Faculty of Dental Medicine (FMD), Center for Interdisciplinary Research in Health (CIIS), Universidade Católica Portuguesa, Viseu, Portugal.
| | - Javier Montero
- Department of Surgery, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - André Correia
- Faculty of Dental Medicine (FMD), Center for Interdisciplinary Research in Health (CIIS), Universidade Católica Portuguesa, Viseu, Portugal
| | - Tiago Marques
- Faculty of Dental Medicine (FMD), Center for Interdisciplinary Research in Health (CIIS), Universidade Católica Portuguesa, Viseu, Portugal
| | - Nuno Rosa
- Faculty of Dental Medicine (FMD), Center for Interdisciplinary Research in Health (CIIS), Universidade Católica Portuguesa, Viseu, Portugal
| |
Collapse
|
45
|
Chitalia R, Miliotis M, Jahani N, Tastsoglou S, McDonald ES, Belenky V, Cohen EA, Newitt D, Van't Veer LJ, Esserman L, Hylton N, DeMichele A, Hatzigeorgiou A, Kontos D. Radiomic tumor phenotypes augment molecular profiling in predicting recurrence free survival after breast neoadjuvant chemotherapy. COMMUNICATIONS MEDICINE 2023; 3:46. [PMID: 36997615 PMCID: PMC10063641 DOI: 10.1038/s43856-023-00273-1] [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: 06/09/2022] [Accepted: 03/10/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND Early changes in breast intratumor heterogeneity during neoadjuvant chemotherapy may reflect the tumor's ability to adapt and evade treatment. We investigated the combination of precision medicine predictors of genomic and MRI data towards improved prediction of recurrence free survival (RFS). METHODS A total of 100 women from the ACRIN 6657/I-SPY 1 trial were retrospectively analyzed. We estimated MammaPrint, PAM50 ROR-S, and p53 mutation scores from publicly available gene expression data and generated four, voxel-wise 3-D radiomic kinetic maps from DCE-MR images at both pre- and early-treatment time points. Within the primary lesion from each kinetic map, features of change in radiomic heterogeneity were summarized into 6 principal components. RESULTS We identify two imaging phenotypes of change in intratumor heterogeneity (p < 0.01) demonstrating significant Kaplan-Meier curve separation (p < 0.001). Adding phenotypes to established prognostic factors, functional tumor volume (FTV), MammaPrint, PAM50, and p53 scores in a Cox regression model improves the concordance statistic for predicting RFS from 0.73 to 0.79 (p = 0.002). CONCLUSIONS These results demonstrate an important step in combining personalized molecular signatures and longitudinal imaging data towards improved prognosis.
Collapse
Affiliation(s)
- Rhea Chitalia
- Department of Bioengineering, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Marios Miliotis
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- DIANA-Lab, Hellenic Pasteur Institute, Athens, Greece
| | - Nariman Jahani
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Spyros Tastsoglou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- DIANA-Lab, Hellenic Pasteur Institute, Athens, Greece
| | - Elizabeth S McDonald
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Vivian Belenky
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Eric A Cohen
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - David Newitt
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA
| | - Laura J Van't Veer
- Department of Surgery and Oncology, University of California, San Francisco, USA
| | - Laura Esserman
- Department of Surgery and Oncology, University of California, San Francisco, USA
| | - Nola Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA
| | - Angela DeMichele
- Department of Medicine, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Artemis Hatzigeorgiou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- DIANA-Lab, Hellenic Pasteur Institute, Athens, Greece
| | - Despina Kontos
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
46
|
Cepeda S, Luppino LT, Pérez-Núñez A, Solheim O, García-García S, Velasco-Casares M, Karlberg A, Eikenes L, Sarabia R, Arrese I, Zamora T, Gonzalez P, Jiménez-Roldán L, Kuttner S. Predicting Regions of Local Recurrence in Glioblastomas Using Voxel-Based Radiomic Features of Multiparametric Postoperative MRI. Cancers (Basel) 2023; 15:1894. [PMID: 36980783 PMCID: PMC10047582 DOI: 10.3390/cancers15061894] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
The globally accepted surgical strategy in glioblastomas is removing the enhancing tumor. However, the peritumoral region harbors infiltration areas responsible for future tumor recurrence. This study aimed to evaluate a predictive model that identifies areas of future recurrence using a voxel-based radiomics analysis of magnetic resonance imaging (MRI) data. This multi-institutional study included a retrospective analysis of patients diagnosed with glioblastoma who underwent surgery with complete resection of the enhancing tumor. Fifty-five patients met the selection criteria. The study sample was split into training (N = 40) and testing (N = 15) datasets. Follow-up MRI was used for ground truth definition, and postoperative structural multiparametric MRI was used to extract voxel-based radiomic features. Deformable coregistration was used to register the MRI sequences for each patient, followed by segmentation of the peritumoral region in the postoperative scan and the enhancing tumor in the follow-up scan. Peritumoral voxels overlapping with enhancing tumor voxels were labeled as recurrence, while non-overlapping voxels were labeled as nonrecurrence. Voxel-based radiomic features were extracted from the peritumoral region. Four machine learning-based classifiers were trained for recurrence prediction. A region-based evaluation approach was used for model evaluation. The Categorical Boosting (CatBoost) classifier obtained the best performance on the testing dataset with an average area under the curve (AUC) of 0.81 ± 0.09 and an accuracy of 0.84 ± 0.06, using region-based evaluation. There was a clear visual correspondence between predicted and actual recurrence regions. We have developed a method that accurately predicts the region of future tumor recurrence in MRI scans of glioblastoma patients. This could enable the adaptation of surgical and radiotherapy treatment to these areas to potentially prolong the survival of these patients.
Collapse
Affiliation(s)
- Santiago Cepeda
- Department of Neurosurgery, Río Hortega University Hospital, 47014 Valladolid, Spain
| | - Luigi Tommaso Luppino
- Department of Physics and Technology, UiT The Arctic University of Norway, 9019 Tromsø, Norway
| | - Angel Pérez-Núñez
- Department of Neurosurgery, 12 de Octubre University Hospital (i+12), 28041 Madrid, Spain
- Department of Surgery, School of Medicine, Complutense University, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria, 12 de Octubre University Hospital (i+12), 28041 Madrid, Spain
| | - Ole Solheim
- Department of Neurosurgery, St. Olavs University Hospital, 7030 Trondheim, Norway
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, 7034 Trondheim, Norway
| | - Sergio García-García
- Department of Neurosurgery, Río Hortega University Hospital, 47014 Valladolid, Spain
| | | | - Anna Karlberg
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
| | - Live Eikenes
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Rosario Sarabia
- Department of Neurosurgery, Río Hortega University Hospital, 47014 Valladolid, Spain
| | - Ignacio Arrese
- Department of Neurosurgery, Río Hortega University Hospital, 47014 Valladolid, Spain
| | - Tomás Zamora
- Department of Pathology, Río Hortega University Hospital, 47014 Valladolid, Spain
| | - Pedro Gonzalez
- Department of Neurosurgery, 12 de Octubre University Hospital (i+12), 28041 Madrid, Spain
| | - Luis Jiménez-Roldán
- Department of Neurosurgery, 12 de Octubre University Hospital (i+12), 28041 Madrid, Spain
- Department of Surgery, School of Medicine, Complutense University, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria, 12 de Octubre University Hospital (i+12), 28041 Madrid, Spain
| | - Samuel Kuttner
- Department of Physics and Technology, UiT The Arctic University of Norway, 9019 Tromsø, Norway
- The PET Imaging Center, University Hospital of North Norway, 9019 Tromsø, Norway
| |
Collapse
|
47
|
Qureshi SA, Hussain L, Ibrar U, Alabdulkreem E, Nour MK, Alqahtani MS, Nafie FM, Mohamed A, Mohammed GP, Duong TQ. Radiogenomic classification for MGMT promoter methylation status using multi-omics fused feature space for least invasive diagnosis through mpMRI scans. Sci Rep 2023; 13:3291. [PMID: 36841898 PMCID: PMC9961309 DOI: 10.1038/s41598-023-30309-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/21/2023] [Indexed: 02/27/2023] Open
Abstract
Accurate radiogenomic classification of brain tumors is important to improve the standard of diagnosis, prognosis, and treatment planning for patients with glioblastoma. In this study, we propose a novel two-stage MGMT Promoter Methylation Prediction (MGMT-PMP) system that extracts latent features fused with radiomic features predicting the genetic subtype of glioblastoma. A novel fine-tuned deep learning architecture, namely Deep Learning Radiomic Feature Extraction (DLRFE) module, is proposed for latent feature extraction that fuses the quantitative knowledge to the spatial distribution and the size of tumorous structure through radiomic features: (GLCM, HOG, and LBP). The application of the novice rejection algorithm has been found significantly effective in selecting and isolating the negative training instances out of the original dataset. The fused feature vectors are then used for training and testing by k-NN and SVM classifiers. The 2021 RSNA Brain Tumor challenge dataset (BraTS-2021) consists of four structural mpMRIs, viz. fluid-attenuated inversion-recovery, T1-weighted, T1-weighted contrast enhancement, and T2-weighted. We evaluated the classification performance, for the very first time in published form, in terms of measures like accuracy, F1-score, and Matthews correlation coefficient. The Jackknife tenfold cross-validation was used for training and testing BraTS-2021 dataset validation. The highest classification performance is (96.84 ± 0.09)%, (96.08 ± 0.10)%, and (97.44 ± 0.14)% as accuracy, sensitivity, and specificity respectively to detect MGMT methylation status for patients suffering from glioblastoma. Deep learning feature extraction with radiogenomic features, fusing imaging phenotypes and molecular structure, using rejection algorithm has been found to perform outclass capable of detecting MGMT methylation status of glioblastoma patients. The approach relates the genomic variation with radiomic features forming a bridge between two areas of research that may prove useful for clinical treatment planning leading to better outcomes.
Collapse
Affiliation(s)
- Shahzad Ahmad Qureshi
- Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Islamabad, Pakistan.
| | - Lal Hussain
- Department of Computer Science and IT, Neelum Campus, The University of Azad Jammu and Kashmir, Muzaffarabad, Azad Kashmir, Pakistan. .,Department of Computer Science and IT, King Abdullah Campus, The University of Azad Jammu and Kashmir, Muzaffarabad, Azad Kashmir, Pakistan. .,Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 111 East 210th Street, Bronx, NY, 10467, USA.
| | - Usama Ibrar
- grid.461150.7Farooq Hospital, Lahore, Pakistan
| | - Eatedal Alabdulkreem
- grid.449346.80000 0004 0501 7602Department of Computer Sciences, College of Computer and Information Sciences, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671 Saudi Arabia
| | - Mohamed K. Nour
- grid.412832.e0000 0000 9137 6644Department of Computer Sciences, College of Computing and Information System, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Mohammed S. Alqahtani
- grid.412144.60000 0004 1790 7100Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, 61421 Saudi Arabia
| | - Faisal Mohammed Nafie
- grid.449051.d0000 0004 0441 5633Department of Computer Science, College of Science and Humanities at Alghat, Majmaah University, Al-Majmaah, 11952 Saudi Arabia
| | - Abdullah Mohamed
- grid.440865.b0000 0004 0377 3762Research Centre, Future University in Egypt, New Cairo, 11845 Egypt
| | - Gouse Pasha Mohammed
- grid.449553.a0000 0004 0441 5588Department of Computer and Self Development, Preparatory Year Deanship, Prince Sattam Bin Abdulaziz University, AlKharj, Saudi Arabia
| | - Tim Q. Duong
- grid.240283.f0000 0001 2152 0791Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467 USA
| |
Collapse
|
48
|
Chakrabarty S, Abidi SA, Mousa M, Mokkarala M, Kelsey M, LaMontagne P, Sotiras A, Marcus DS. Deep learning-based end-to-end scan-type classification, pre-processing, and segmentation of clinical neuro-oncology studies. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2023; 12469:124690N. [PMID: 39263425 PMCID: PMC11389857 DOI: 10.1117/12.2647656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Modern neuro-oncology workflows are driven by large collections of high-dimensional MRI data obtained using varying acquisition protocols. The concomitant heterogeneity of this data makes extensive manual curation and pre-processing imperative prior to algorithmic use. The limited efforts invested towards automating this curation and processing are fragmented, do not encompass the entire workflow, or still require significant manual intervention. In this work, we propose an artificial intelligence-driven solution for transforming multi-modal raw neuro-oncology MRI Digital Imaging and Communications in Medicine (DICOM) data into quantitative tumor measurements. Our end-to-end framework classifies MRI scans into different structural sequence types, preprocesses the data, and uses convolutional neural networks to segment tumor tissue subtypes. Moreover, it adopts an expert-in-the-loop approach, where segmentation results may be manually refined by radiologists. This framework was implemented as Docker Containers (for command line usage and within the eXtensible Neuroimaging Archive Toolkit [XNAT]) and validated on a retrospective glioma dataset (n = 155) collected from the Washington University School of Medicine, comprising preoperative MRI scans from patients with histopathologically confirmed gliomas. Segmentation results were refined by a neuroradiologist, and performance was quantified using Dice Similarity Coefficient to compare predicted and expert-refined tumor masks. The scan-type classifier yielded a 99.71% accuracy across all sequence types. The segmentation model achieved mean Dice scores of 0.894 (± 0.225) for whole tumor segmentation. The proposed framework can automate tumor segmentation and characterization - thus streamlining workflows in a clinical setting as well as expediting standardized curation of large-scale neuro-oncology datasets in a research setting.
Collapse
Affiliation(s)
- Satrajit Chakrabarty
- Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Syed Amaan Abidi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mina Mousa
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mahati Mokkarala
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew Kelsey
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aristeidis Sotiras
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Institute for Informatics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel S Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
49
|
Haldar D, Kazerooni AF, Arif S, Familiar A, Madhogarhia R, Khalili N, Bagheri S, Anderson H, Shaikh IS, Mahtabfar A, Kim MC, Tu W, Ware J, Vossough A, Davatzikos C, Storm PB, Resnick A, Nabavizadeh A. Unsupervised machine learning using K-means identifies radiomic subgroups of pediatric low-grade gliomas that correlate with key molecular markers. Neoplasia 2023; 36:100869. [PMID: 36566592 PMCID: PMC9803939 DOI: 10.1016/j.neo.2022.100869] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/21/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Despite advancements in molecular and histopathologic characterization of pediatric low-grade gliomas (pLGGs), there remains significant phenotypic heterogeneity among tumors with similar categorizations. We hypothesized that an unsupervised machine learning approach based on radiomic features may reveal distinct pLGG imaging subtypes. METHODS Multi-parametric MR images (T1 pre- and post-contrast, T2, and T2 FLAIR) from 157 patients with pLGGs were collected and 881 quantitative radiomic features were extracted from tumorous region. Clustering was performed using K-means after applying principal component analysis (PCA) for feature dimensionality reduction. Molecular and demographic data was obtained from the PedCBioportal and compared between imaging subtypes. RESULTS K-means identified three distinct imaging-based subtypes. Subtypes differed in mutational frequencies of BRAF (p < 0.05) as well as the gene expression of BRAF (p<0.05). It was also found that age (p < 0.05), tumor location (p < 0.01), and tumor histology (p < 0.0001) differed significantly between the imaging subtypes. CONCLUSION In this exploratory work, it was found that clustering of pLGGs based on radiomic features identifies distinct, imaging-based subtypes that correlate with important molecular markers and demographic details. This finding supports the notion that incorporation of radiomic data could augment our ability to better characterize pLGGs.
Collapse
Affiliation(s)
- Debanjan Haldar
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sherjeel Arif
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ariana Familiar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Rachel Madhogarhia
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nastaran Khalili
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sina Bagheri
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hannah Anderson
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Aria Mahtabfar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Neurological Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Meen Chul Kim
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Wenxin Tu
- College of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jefferey Ware
- Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Arastoo Vossough
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christos Davatzikos
- Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Phillip B Storm
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Division of Neurological Surgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Adam Resnick
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Ali Nabavizadeh
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
50
|
Raunig DL, Pennello GA, Delfino JG, Buckler AJ, Hall TJ, Guimaraes AR, Wang X, Huang EP, Barnhart HX, deSouza N, Obuchowski N. Multiparametric Quantitative Imaging Biomarker as a Multivariate Descriptor of Health: A Roadmap. Acad Radiol 2023; 30:159-182. [PMID: 36464548 PMCID: PMC9825667 DOI: 10.1016/j.acra.2022.10.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/24/2022] [Accepted: 10/29/2022] [Indexed: 12/02/2022]
Abstract
Multiparametric quantitative imaging biomarkers (QIBs) offer distinct advantages over single, univariate descriptors because they provide a more complete measure of complex, multidimensional biological systems. In disease, where structural and functional disturbances occur across a multitude of subsystems, multivariate QIBs are needed to measure the extent of system malfunction. This paper, the first Use Case in a series of articles on multiparameter imaging biomarkers, considers multiple QIBs as a multidimensional vector to represent all relevant disease constructs more completely. The approach proposed offers several advantages over QIBs as multiple endpoints and avoids combining them into a single composite that obscures the medical meaning of the individual measurements. We focus on establishing statistically rigorous methods to create a single, simultaneous measure from multiple QIBs that preserves the sensitivity of each univariate QIB while incorporating the correlation among QIBs. Details are provided for metrological methods to quantify the technical performance. Methods to reduce the set of QIBs, test the superiority of the mp-QIB model to any univariate QIB model, and design study strategies for generating precision and validity claims are also provided. QIBs of Alzheimer's Disease from the ADNI merge data set are used as a case study to illustrate the methods described.
Collapse
Affiliation(s)
- David L Raunig
- Department of Statistical and Quantitative Sciences, Data Science Institute, Takeda Pharmaceuticals, Cambridge, Massachusetts.
| | - Gene A Pennello
- Center for Devices and Radiological Health, US Food and Drug Administration Division of Imaging, Diagnostic and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland
| | - Jana G Delfino
- Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland
| | | | - Timothy J Hall
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin
| | - Alexander R Guimaraes
- Department of Diagnostic Radiology, Oregon Health & Sciences University, Portland, Oregon
| | - Xiaofeng Wang
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland, Ohio
| | - Erich P Huang
- Biometric Research Program, Division of Cancer Treatment and Diagnosis - National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Huiman X Barnhart
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Nandita deSouza
- Division of Radiotherapy and Imaging, the Insitute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Nancy Obuchowski
- Department of Quantitative Health Sciences, Lerner Research Institute Cleveland Clinic Foundation, Cleveland, Ohio
| |
Collapse
|