51
|
de Godoy LL, Chawla S, Brem S, Mohan S. Taming Glioblastoma in "Real Time": Integrating Multimodal Advanced Neuroimaging/AI Tools Towards Creating a Robust and Therapy Agnostic Model for Response Assessment in Neuro-Oncology. Clin Cancer Res 2023; 29:2588-2592. [PMID: 37227179 DOI: 10.1158/1078-0432.ccr-23-0009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/20/2023] [Accepted: 05/04/2023] [Indexed: 05/10/2023]
Abstract
The highly aggressive nature of glioblastoma carries a dismal prognosis despite aggressive multimodal therapy. Alternative treatment regimens, such as immunotherapies, are known to intensify the inflammatory response in the treatment field. Follow-up imaging in these scenarios often mimics disease progression on conventional MRI, making accurate evaluation extremely challenging. To this end, revised criteria for assessment of treatment response in high-grade gliomas were successfully proposed by the RANO Working Group to distinguish pseudoprogression from true progression, with intrinsic constraints related to the postcontrast T1-weighted MRI sequence. To address these existing limitations, our group proposes a more objective and quantifiable "treatment agnostic" model, integrating into the RANO criteria advanced multimodal neuroimaging techniques, such as diffusion tensor imaging (DTI), dynamic susceptibility contrast-perfusion weighted imaging (DSC-PWI), dynamic contrast enhanced (DCE)-MRI, MR spectroscopy, and amino acid-based positron emission tomography (PET) imaging tracers, along with artificial intelligence (AI) tools (radiomics, radiogenomics, and radiopathomics) and molecular information to address this complex issue of treatment-related changes versus tumor progression in "real-time", particularly in the early posttreatment window. Our perspective delineates the potential of incorporating multimodal neuroimaging techniques to improve consistency and automation for the assessment of early treatment response in neuro-oncology.
Collapse
Affiliation(s)
- Laiz Laura de Godoy
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Glioblastoma Translational Center of Excellence, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
52
|
Avellar L, Frizera A, Leal-Junior A. POF Smart Pants: a fully portable optical fiber-integrated smart textile for remote monitoring of lower limb biomechanics. BIOMEDICAL OPTICS EXPRESS 2023; 14:3689-3704. [PMID: 37497490 PMCID: PMC10368064 DOI: 10.1364/boe.492796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 07/28/2023]
Abstract
This paper presents the development of an optical fiber-integrated smart textile used as an instrumented pants for biomechanical and activity recognition. The optical fiber sensor is based on the multiplexed intensity variation technique in which a side coupling between a polymer optical fiber (POF) and light sources with controlled modulation is developed. In addition, the sensor system is integrated into pants, where two POFs with 30 sensors each are placed on the left and right legs of the proposed POF Smart Pants. After the device's fabrication and assembly, the 60 optical fiber sensors are characterized as a function of the transverse displacement on the sensor's region. In this case, each sensor presented its sensitivities (108.03 ± 100 mV/mm), which are used on the sensor normalization prior to the data analysis. Then, the tests with volunteer performing different daily activities indicated the suitability of the proposed device on the assessment of biomechanics of human movement in different activities as well as the spatio-temporal parameters of the gait in different velocity conditions. For activity recognition, a neural network is applied and presented 100% accuracy on the activity recognition. Then, to provide an optimization of the number of sensors, the principal components analysis is applied and indicated a threefold reduction of the number of sensors with an accuracy of 99%. Thus, the proposed POF Smart Pants is a feasible alternative for a low-cost and highly reliable sensor system for remote monitoring of different patients, with the possibility of customizing the device for different users.
Collapse
|
53
|
Sun W, Song C, Tang C, Pan C, Xue P, Fan J, Qiao Y. Performance of deep learning algorithms to distinguish high-grade glioma from low-grade glioma: A systematic review and meta-analysis. iScience 2023; 26:106815. [PMID: 37250800 PMCID: PMC10209541 DOI: 10.1016/j.isci.2023.106815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/23/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
This study aims to evaluate deep learning (DL) performance in differentiating low- and high-grade glioma. Search online database for studies continuously published from 1st January 2015 until 16th August 2022. The random-effects model was used for synthesis, based on pooled sensitivity (SE), specificity (SP), and area under the curve (AUC). Heterogeneity was estimated using the Higgins inconsistency index (I2). 33 were ultimately included in the meta-analysis. The overall pooled SE and SP were 94% and 93%, with an AUC of 0.98. There was great heterogeneity in this field. Our evidence-based study shows DL achieves high accuracy in glioma grading. Subgroup analysis reveals several limitations in this field: 1) Diagnostic trials require standard method for data merging for AI; 2) small sample size; 3) poor-quality image preprocessing; 4) not standard algorithm development; 5) not standard data report; 6) different definition of HGG and LGG; and 7) poor extrapolation.
Collapse
Affiliation(s)
- Wanyi Sun
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng Song
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Tang
- Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Chenghao Pan
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Xue
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinhu Fan
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Youlin Qiao
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
54
|
Noor J, Chaudhry A, Batool S. Microfluidic Technology, Artificial Intelligence, and Biosensors As Advanced Technologies in Cancer Screening: A Review Article. Cureus 2023; 15:e39634. [PMID: 37388583 PMCID: PMC10305590 DOI: 10.7759/cureus.39634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2023] [Indexed: 07/01/2023] Open
Abstract
Cancer screening techniques aim to detect premalignant lesions and enable early intervention to delay the onset of cancer while keeping incidence constant. Technology advancements have led to the development of powerful tools such as microfluidic technology, artificial intelligence, machine learning algorithms, and electrochemical biosensors to aid in early cancer detection. Non-invasive cancer screening methods like virtual colonoscopy and endoscopic ultrasonography have also been developed to provide comprehensive pictures of organs and detect cancer early. This review article provides an overview of recent advances in cancer screening in microfluidic technology, artificial intelligence, and biomarkers through a narrative literature search. Microfluidic devices enable easy handling of sub-microliter volumes and have become a promising tool for cancer detection, drug screening, and modeling angiogenesis and metastasis in cancer research. Machine learning and artificial intelligence have shown high accuracy in oncology-related diagnostic imaging, reducing the manual steps in lesion detection and providing standardized and accurate results, with potential for global standardization in areas like colon polyps, breast cancer, and primary and metastatic brain cancer. A biomarker-based cancer diagnosis is promising for early detection and effective therapy, and electrochemical biosensors integrated with nanoparticles offer multiplexing and amplification capabilities. Understanding these advanced technologies' basics, achievements, and challenges is crucial for advancing their use in oncology.
Collapse
Affiliation(s)
- Jawad Noor
- Internal Medicine, St. Dominic Hospital, Jackson, USA
| | | | - Saima Batool
- Pathology, Nishtar Medical University, Multan, PAK
| |
Collapse
|
55
|
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
|
56
|
Du P, Liu X, Shen L, Wu X, Chen J, Chen L, Cao A, Geng D. Prediction of treatment response in patients with brain metastasis receiving stereotactic radiosurgery based on pre-treatment multimodal MRI radiomics and clinical risk factors: A machine learning model. Front Oncol 2023; 13:1114194. [PMID: 36994193 PMCID: PMC10040663 DOI: 10.3389/fonc.2023.1114194] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
ObjectivesStereotactic radiosurgery (SRS), a therapy that uses radiation to treat brain tumors, has become a significant treatment procedure for patients with brain metastasis (BM). However, a proportion of patients have been found to be at risk of local failure (LF) after treatment. Hence, accurately identifying patients with LF risk after SRS treatment is critical to the development of successful treatment plans and the prognoses of patients. To accurately predict BM patients with the occurrence of LF after SRS therapy, we develop and validate a machine learning (ML) model based on pre-treatment multimodal magnetic resonance imaging (MRI) radiomics and clinical risk factors.Patients and methodsIn this study, 337 BM patients were included (247, 60, and 30 in the training set, internal validation set, and external validation set, respectively). Four clinical features and 223 radiomics features were selected using least absolute shrinkage and selection operator (LASSO) and Max-Relevance and Min-Redundancy (mRMR) filters. We establish the ML model using the selected features and the support vector machine (SVM) classifier to predict the treatment response of BM patients to SRS therapy.ResultsIn the training set, the SVM classifier that uses a combination of clinical and radiomics features demonstrates outstanding discriminative performance (AUC=0.95, 95% CI: 0.93-0.97). Moreover, this model also achieves satisfactory results in the validation sets (AUC=0.95 in the internal validation set and AUC=0.93 in the external validation set), demonstrating excellent generalizability.ConclusionsThis ML model enables a non-invasive prediction of the treatment response of BM patients receiving SRS therapy, which can in turn assist neurologist and radiation oncologists in the development of more precise and individualized treatment plans for BM patients.
Collapse
Affiliation(s)
- Peng Du
- The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Huashan Hospital, Fudan University, Shanghai, China
| | - Xiao Liu
- School of Computer and Information Technology, Beijing, China
| | - Li Shen
- Department of Radiology, Jiahui International Hospital, Shanghai, China
| | - Xuefan Wu
- Department of Radiology, Shanghai Gamma Hospital, Shanghai, China
| | - Jiawei Chen
- Huashan Hospital, Fudan University, Shanghai, China
| | - Lang Chen
- The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Aihong Cao
- The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- *Correspondence: Aihong Cao, ; Daoying Geng,
| | - Daoying Geng
- Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Aihong Cao, ; Daoying Geng,
| |
Collapse
|
57
|
Surianarayanan C, Lawrence JJ, Chelliah PR, Prakash E, Hewage C. Convergence of Artificial Intelligence and Neuroscience towards the Diagnosis of Neurological Disorders-A Scoping Review. SENSORS (BASEL, SWITZERLAND) 2023; 23:3062. [PMID: 36991773 PMCID: PMC10053494 DOI: 10.3390/s23063062] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 06/19/2023]
Abstract
Artificial intelligence (AI) is a field of computer science that deals with the simulation of human intelligence using machines so that such machines gain problem-solving and decision-making capabilities similar to that of the human brain. Neuroscience is the scientific study of the struczture and cognitive functions of the brain. Neuroscience and AI are mutually interrelated. These two fields help each other in their advancements. The theory of neuroscience has brought many distinct improvisations into the AI field. The biological neural network has led to the realization of complex deep neural network architectures that are used to develop versatile applications, such as text processing, speech recognition, object detection, etc. Additionally, neuroscience helps to validate the existing AI-based models. Reinforcement learning in humans and animals has inspired computer scientists to develop algorithms for reinforcement learning in artificial systems, which enables those systems to learn complex strategies without explicit instruction. Such learning helps in building complex applications, like robot-based surgery, autonomous vehicles, gaming applications, etc. In turn, with its ability to intelligently analyze complex data and extract hidden patterns, AI fits as a perfect choice for analyzing neuroscience data that are very complex. Large-scale AI-based simulations help neuroscientists test their hypotheses. Through an interface with the brain, an AI-based system can extract the brain signals and commands that are generated according to the signals. These commands are fed into devices, such as a robotic arm, which helps in the movement of paralyzed muscles or other human parts. AI has several use cases in analyzing neuroimaging data and reducing the workload of radiologists. The study of neuroscience helps in the early detection and diagnosis of neurological disorders. In the same way, AI can effectively be applied to the prediction and detection of neurological disorders. Thus, in this paper, a scoping review has been carried out on the mutual relationship between AI and neuroscience, emphasizing the convergence between AI and neuroscience in order to detect and predict various neurological disorders.
Collapse
Affiliation(s)
| | | | | | - Edmond Prakash
- Research Center for Creative Arts, University for the Creative Arts (UCA), Farnham GU9 7DS, UK
| | - Chaminda Hewage
- Cardiff School of Technologies, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| |
Collapse
|
58
|
Predicting gene mutation status via artificial intelligence technologies based on multimodal integration (MMI) to advance precision oncology. Semin Cancer Biol 2023; 91:1-15. [PMID: 36801447 DOI: 10.1016/j.semcancer.2023.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/30/2023] [Accepted: 02/15/2023] [Indexed: 02/21/2023]
Abstract
Personalized treatment strategies for cancer frequently rely on the detection of genetic alterations which are determined by molecular biology assays. Historically, these processes typically required single-gene sequencing, next-generation sequencing, or visual inspection of histopathology slides by experienced pathologists in a clinical context. In the past decade, advances in artificial intelligence (AI) technologies have demonstrated remarkable potential in assisting physicians with accurate diagnosis of oncology image-recognition tasks. Meanwhile, AI techniques make it possible to integrate multimodal data such as radiology, histology, and genomics, providing critical guidance for the stratification of patients in the context of precision therapy. Given that the mutation detection is unaffordable and time-consuming for a considerable number of patients, predicting gene mutations based on routine clinical radiological scans or whole-slide images of tissue with AI-based methods has become a hot issue in actual clinical practice. In this review, we synthesized the general framework of multimodal integration (MMI) for molecular intelligent diagnostics beyond standard techniques. Then we summarized the emerging applications of AI in the prediction of mutational and molecular profiles of common cancers (lung, brain, breast, and other tumor types) pertaining to radiology and histology imaging. Furthermore, we concluded that there truly exist multiple challenges of AI techniques in the way of its real-world application in the medical field, including data curation, feature fusion, model interpretability, and practice regulations. Despite these challenges, we still prospect the clinical implementation of AI as a highly potential decision-support tool to aid oncologists in future cancer treatment management.
Collapse
|
59
|
Duong MT, Rudie JD, Mohan S. Neuroimaging Patterns of Intracranial Infections: Meningitis, Cerebritis, and Their Complications. Neuroimaging Clin N Am 2023; 33:11-41. [PMID: 36404039 PMCID: PMC10904173 DOI: 10.1016/j.nic.2022.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Neuroimaging provides rapid, noninvasive visualization of central nervous system infections for optimal diagnosis and management. Generalizable and characteristic imaging patterns help radiologists distinguish different types of intracranial infections including meningitis and cerebritis from a variety of bacterial, viral, fungal, and/or parasitic causes. Here, we describe key radiologic patterns of meningeal enhancement and diffusion restriction through profiles of meningitis, cerebritis, abscess, and ventriculitis. We discuss various imaging modalities and recent diagnostic advances such as deep learning through a survey of intracranial pathogens and their radiographic findings. Moreover, we explore critical complications and differential diagnoses of intracranial infections.
Collapse
Affiliation(s)
- Michael Tran Duong
- Division of Neuroradiology, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Jeffrey D Rudie
- Department of Radiology, Scripps Clinic and University of California San Diego, 10666 Torrey Pines Road, La Jolla, CA 92037, USA
| | - Suyash Mohan
- Division of Neuroradiology, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
60
|
Assessing Metabolic Markers in Glioblastoma Using Machine Learning: A Systematic Review. Metabolites 2023; 13:metabo13020161. [PMID: 36837779 PMCID: PMC9958885 DOI: 10.3390/metabo13020161] [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: 12/26/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Glioblastoma (GBM) is a common and deadly brain tumor with late diagnoses and poor prognoses. Machine learning (ML) is an emerging tool that can create highly accurate diagnostic and prognostic prediction models. This paper aimed to systematically search the literature on ML for GBM metabolism and assess recent advancements. A literature search was performed using predetermined search terms. Articles describing the use of an ML algorithm for GBM metabolism were included. Ten studies met the inclusion criteria for analysis: diagnostic (n = 3, 30%), prognostic (n = 6, 60%), or both (n = 1, 10%). Most studies analyzed data from multiple databases, while 50% (n = 5) included additional original samples. At least 2536 data samples were run through an ML algorithm. Twenty-seven ML algorithms were recorded with a mean of 2.8 algorithms per study. Algorithms were supervised (n = 24, 89%), unsupervised (n = 3, 11%), continuous (n = 19, 70%), or categorical (n = 8, 30%). The mean reported accuracy and AUC of ROC were 95.63% and 0.779, respectively. One hundred six metabolic markers were identified, but only EMP3 was reported in multiple studies. Many studies have identified potential biomarkers for GBM diagnosis and prognostication. These algorithms show promise; however, a consensus on even a handful of biomarkers has not yet been made.
Collapse
|
61
|
Luvhengo T, Molefi T, Demetriou D, Hull R, Dlamini Z. Use of Artificial Intelligence in Implementing Mainstream Precision Medicine to Improve Traditional Symptom-driven Practice of Medicine: Allowing Early Interventions and Tailoring better-personalised Cancer Treatments. ARTIFICIAL INTELLIGENCE AND PRECISION ONCOLOGY 2023:49-72. [DOI: 10.1007/978-3-031-21506-3_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
62
|
Jiao Y, Zhang J, Yang X, Zhan T, Wu Z, Li Y, Zhao S, Li H, Weng J, Huo R, Wang J, Xu H, Sun Y, Wang S, Cao Y. Artificial Intelligence-Assisted Evaluation of the Spatial Relationship between Brain Arteriovenous Malformations and the Corticospinal Tract to Predict Postsurgical Motor Defects. AJNR Am J Neuroradiol 2023; 44:17-25. [PMID: 36549849 PMCID: PMC9835926 DOI: 10.3174/ajnr.a7735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 11/07/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE Preoperative evaluation of brain AVMs is crucial for the selection of surgical candidates. Our goal was to use artificial intelligence to predict postsurgical motor defects in patients with brain AVMs involving motor-related areas. MATERIALS AND METHODS Eighty-three patients who underwent microsurgical resection of brain AVMs involving motor-related areas were retrospectively reviewed. Four artificial intelligence-based indicators were calculated with artificial intelligence on TOF-MRA and DTI, including FN5mm/50mm (the proportion of fiber numbers within 5-50mm from the lesion border), FN10mm/50mm (the same but within 10-50mm), FP5mm/50mm (the proportion of fiber voxel points within 5-50mm from the lesion border), and FP10mm/50mm (the same but within 10-50mm). The association between the variables and long-term postsurgical motor defects was analyzed using univariate and multivariate analyses. Least absolute shrinkage and selection operator regression with the Pearson correlation coefficient was used to select the optimal features to develop the machine learning model to predict postsurgical motor defects. The area under the curve was calculated to evaluate the predictive performance. RESULTS In patients with and without postsurgical motor defects, the mean FN5mm/50mm, FN10mm/50mm, FP5mm/50mm, and FP10mm/50mm were 0.24 (SD, 0.24) and 0.03 (SD, 0.06), 0.37 (SD, 0.27) and 0.06 (SD, 0.08), 0.06 (SD, 0.10) and 0.01 (SD, 0.02), and 0.10 (SD, 0.12) and 0.02 (SD, 0.05), respectively. Univariate and multivariate logistic analyses identified FN10mm/50mm as an independent risk factor for long-term postsurgical motor defects (P = .002). FN10mm/50mm achieved a mean area under the curve of 0.86 (SD, 0.08). The mean area under the curve of the machine learning model consisting of FN10mm/50mm, diffuseness, and the Spetzler-Martin score was 0.88 (SD, 0.07). CONCLUSIONS The artificial intelligence-based indicator, FN10mm/50mm, can reflect the lesion-fiber spatial relationship and act as a dominant predictor for postsurgical motor defects in patients with brain AVMs involving motor-related areas.
Collapse
Affiliation(s)
- Y Jiao
- From the Department of Neurosurgery (Y.J., J.Z., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - J Zhang
- From the Department of Neurosurgery (Y.J., J.Z., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - X Yang
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - T Zhan
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - Z Wu
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - Y Li
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - S Zhao
- From the Department of Neurosurgery (Y.J., J.Z., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - H Li
- From the Department of Neurosurgery (Y.J., J.Z., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - J Weng
- From the Department of Neurosurgery (Y.J., J.Z., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - R Huo
- From the Department of Neurosurgery (Y.J., J.Z., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - J Wang
- From the Department of Neurosurgery (Y.J., J.Z., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - H Xu
- From the Department of Neurosurgery (Y.J., J.Z., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - Y Sun
- From the Department of Neurosurgery (Y.J., J.Z., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - S Wang
- From the Department of Neurosurgery (Y.J., J.Z., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| | - Y Cao
- From the Department of Neurosurgery (Y.J., J.Z., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (Y.J., J.Z., X.Y., T.Z., Z.W., Y.L., S.Z., H.L., J. Weng, R.H., J. Wang, H.X., Y.S., S.W., Y.C.), Beijing, China
| |
Collapse
|
63
|
Usmani UA, Happonen A, Watada J, Khakurel J. Artificial Intelligence Applications in Healthcare. LECTURE NOTES IN NETWORKS AND SYSTEMS 2023:1085-1104. [DOI: 10.1007/978-981-99-3091-3_89] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
64
|
Khanna NN, Maindarkar MA, Viswanathan V, Fernandes JFE, Paul S, Bhagawati M, Ahluwalia P, Ruzsa Z, Sharma A, Kolluri R, Singh IM, Laird JR, Fatemi M, Alizad A, Saba L, Agarwal V, Sharma A, Teji JS, Al-Maini M, Rathore V, Naidu S, Liblik K, Johri AM, Turk M, Mohanty L, Sobel DW, Miner M, Viskovic K, Tsoulfas G, Protogerou AD, Kitas GD, Fouda MM, Chaturvedi S, Kalra MK, Suri JS. Economics of Artificial Intelligence in Healthcare: Diagnosis vs. Treatment. Healthcare (Basel) 2022; 10:2493. [PMID: 36554017 PMCID: PMC9777836 DOI: 10.3390/healthcare10122493] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Motivation: The price of medical treatment continues to rise due to (i) an increasing population; (ii) an aging human growth; (iii) disease prevalence; (iv) a rise in the frequency of patients that utilize health care services; and (v) increase in the price. Objective: Artificial Intelligence (AI) is already well-known for its superiority in various healthcare applications, including the segmentation of lesions in images, speech recognition, smartphone personal assistants, navigation, ride-sharing apps, and many more. Our study is based on two hypotheses: (i) AI offers more economic solutions compared to conventional methods; (ii) AI treatment offers stronger economics compared to AI diagnosis. This novel study aims to evaluate AI technology in the context of healthcare costs, namely in the areas of diagnosis and treatment, and then compare it to the traditional or non-AI-based approaches. Methodology: PRISMA was used to select the best 200 studies for AI in healthcare with a primary focus on cost reduction, especially towards diagnosis and treatment. We defined the diagnosis and treatment architectures, investigated their characteristics, and categorized the roles that AI plays in the diagnostic and therapeutic paradigms. We experimented with various combinations of different assumptions by integrating AI and then comparing it against conventional costs. Lastly, we dwell on three powerful future concepts of AI, namely, pruning, bias, explainability, and regulatory approvals of AI systems. Conclusions: The model shows tremendous cost savings using AI tools in diagnosis and treatment. The economics of AI can be improved by incorporating pruning, reduction in AI bias, explainability, and regulatory approvals.
Collapse
Affiliation(s)
- Narendra N. Khanna
- Department of Cardiology, Indraprastha APOLLO Hospitals, New Delhi 110001, India
| | - Mahesh A. Maindarkar
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India
| | | | | | - Sudip Paul
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India
| | - Mrinalini Bhagawati
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India
| | - Puneet Ahluwalia
- Max Institute of Cancer Care, Max Super Specialty Hospital, New Delhi 110017, India
| | - Zoltan Ruzsa
- Invasive Cardiology Division, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Aditya Sharma
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA 22904, USA
| | - Raghu Kolluri
- Ohio Health Heart and Vascular, Columbus, OH 43214, USA
| | - Inder M. Singh
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
| | - John R. Laird
- Heart and Vascular Institute, Adventist Health St. Helena, St. Helena, CA 94574, USA
| | - Mostafa Fatemi
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Azra Alizad
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, 40138 Cagliari, Italy
| | - Vikas Agarwal
- Department of Immunology, SGPGIMS, Lucknow 226014, India
| | - Aman Sharma
- Department of Immunology, SGPGIMS, Lucknow 226014, India
| | - Jagjit S. Teji
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Mustafa Al-Maini
- Allergy, Clinical Immunology and Rheumatology Institute, Toronto, ON L4Z 4C4, Canada
| | | | - Subbaram Naidu
- Electrical Engineering Department, University of Minnesota, Duluth, MN 55812, USA
| | - Kiera Liblik
- Department of Medicine, Division of Cardiology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Amer M. Johri
- Department of Medicine, Division of Cardiology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Monika Turk
- The Hanse-Wissenschaftskolleg Institute for Advanced Study, 27753 Delmenhorst, Germany
| | - Lopamudra Mohanty
- Department of Computer Science, ABES Engineering College, Ghaziabad 201009, India
| | - David W. Sobel
- Rheumatology Unit, National Kapodistrian University of Athens, 15772 Athens, Greece
| | - Martin Miner
- Men’s Health Centre, Miriam Hospital Providence, Providence, RI 02906, USA
| | - Klaudija Viskovic
- Department of Radiology and Ultrasound, University Hospital for Infectious Diseases, 10000 Zagreb, Croatia
| | - George Tsoulfas
- Department of Surgery, Aristoteleion University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Athanasios D. Protogerou
- Cardiovascular Prevention and Research Unit, Department of Pathophysiology, National & Kapodistrian University of Athens, 15772 Athens, Greece
| | - George D. Kitas
- Academic Affairs, Dudley Group NHS Foundation Trust, Dudley DY1 2HQ, UK
- Arthritis Research UK Epidemiology Unit, Manchester University, Manchester M13 9PL, UK
| | - Mostafa M. Fouda
- Department of Electrical and Computer Engineering, Idaho State University, Pocatello, ID 83209, USA
| | - Seemant Chaturvedi
- Department of Neurology & Stroke Program, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Jasjit S. Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
| |
Collapse
|
65
|
García-García S, García-Galindo M, Arrese I, Sarabia R, Cepeda S. Current Evidence, Limitations and Future Challenges of Survival Prediction for Glioblastoma Based on Advanced Noninvasive Methods: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58121746. [PMID: 36556948 PMCID: PMC9786785 DOI: 10.3390/medicina58121746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/16/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
Background and Objectives: Survival estimation for patients diagnosed with Glioblastoma (GBM) is an important information to consider in patient management and communication. Despite some known risk factors, survival estimation remains a major challenge. Novel non-invasive technologies such as radiomics and artificial intelligence (AI) have been implemented to increase the accuracy of these predictions. In this article, we reviewed and discussed the most significant available research on survival estimation for GBM through advanced non-invasive methods. Materials and Methods: PubMed database was queried for articles reporting on survival prognosis for GBM through advanced image and data management methods. Articles including in their title or abstract the following terms were initially screened: ((glioma) AND (survival)) AND ((artificial intelligence) OR (radiomics)). Exclusively English full-text articles, reporting on humans, published as of 1 September 2022 were considered. Articles not reporting on overall survival, evaluating the effects of new therapies or including other tumors were excluded. Research with a radiomics-based methodology were evaluated using the radiomics quality score (RQS). Results: 382 articles were identified. After applying the inclusion criteria, 46 articles remained for further analysis. These articles were thoroughly assessed, summarized and discussed. The results of the RQS revealed some of the limitations of current radiomics investigation on this field. Limitations of analyzed studies included data availability, patient selection and heterogeneity of methodologies. Future challenges on this field are increasing data availability, improving the general understanding of how AI handles data and establishing solid correlations between image features and tumor's biology. Conclusions: Radiomics and AI methods of data processing offer a new paradigm of possibilities to tackle the question of survival prognosis in GBM.
Collapse
Affiliation(s)
- Sergio García-García
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
- Correspondence:
| | - Manuel García-Galindo
- Faculty of Medicine, University of Valladolid, Avenida Ramón y Cajal 7, 47003 Valladolid, Spain
| | - Ignacio Arrese
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
| | - Rosario Sarabia
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
| | - Santiago Cepeda
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
| |
Collapse
|
66
|
Zhou P, Sun Q, Song G, Liu Z, Qi J, Yuan X, Wang X, Yan S, Du J, Dai Z, Wang J, Hu S. Radiomics features from perihematomal edema for prediction of prognosis in the patients with basal ganglia hemorrhage. Front Neurol 2022; 13:982928. [PMID: 36425801 PMCID: PMC9680901 DOI: 10.3389/fneur.2022.982928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/24/2022] [Indexed: 09/18/2024] Open
Abstract
OBJECTIVE We developed and validated a clinical-radiomics nomogram to predict the prognosis of basal ganglia hemorrhage patients. METHODS Retrospective analyses were conducted in 197 patients with basal ganglia hemorrhage (training cohort: n = 136, test cohort: n = 61) who were admitted to The First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital) and underwent computed tomography (CT) scan. According to different prognoses, patients with basal ganglia hemorrhage were divided into two groups. Independent clinical risk factors were derived with univariate and multivariate regression analysis. Radiomics signatures were obtained using least absolute shrinkage and selection operator. A radiomics score (Rad-score) was generated by 12 radiomics signatures of perihematomal edema (PHE) from CT images that were correlated with the prognosis of basal ganglia hemorrhage patients. A clinical-radiomics nomogram was conducted by combing the Rad-score and clinical risk factors using logistic regression analysis. The prediction performance of the nomogram was tested in the training cohort and verified in the test cohort. RESULTS The clinical model conducted by four clinical risk factors and 12 radiomcis features were used to establish the Rad-score. The clinical-radiomics nomogram outperformed the clinical model in the training cohort [area under the curve (AUC), 0.92 vs. 0.85] and the test cohort (AUC, 0.91 vs 0.85). The clinical-radiomics nomogram showed good calibration and clinical benefit in both the training and test cohorts. CONCLUSION Radiomics features of PHE in patients with basal ganglia hemorrhage could contribute to the outcome prediction. The clinical-radiomics nomogram may help first-line clinicians to make individual clinical treatment decisions for patients with basal ganglia hemorrhage.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Quanye Sun
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Gesheng Song
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zexiang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jianfeng Qi
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xuhui Yuan
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Shaofeng Yan
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jianyang Du
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhengjun Dai
- Scientific Research Department, Huiying Medical Technology Co., Ltd, Beijing, China
| | - Jianjun Wang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Shaoshan Hu
- Department of Neurosurgery, Emergency Medicine Center, Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
67
|
Jang EB, Kim HS, Park JE, Park SY, Nam YK, Nam SJ, Kim YH, Kim JH. Diffuse glioma, not otherwise specified: imaging-based risk stratification achieves histomolecular-level prognostication. Eur Radiol 2022; 32:7780-7788. [PMID: 35587830 DOI: 10.1007/s00330-022-08850-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVES To determine whether imaging-based risk stratification enables prognostication in diffuse glioma, NOS (not otherwise specified). METHODS Data from 220 patients classified as diffuse glioma, NOS, between January 2011 and December 2020 were retrospectively included. Two neuroradiologists analyzed pre-surgical CT and MRI to assign gliomas to the three imaging-based risk types considering well-known imaging phenotypes (e.g., T2/FLAIR mismatch). According to the 2021 World Health Organization classification, the three risk types included (1) low-risk, expecting oligodendroglioma, isocitrate dehydrogenase (IDH)-mutant, and 1p/19q-codeleted; (2) intermediate-risk, expecting astrocytoma, IDH-mutant; and (3) high-risk, expecting glioblastoma, IDH-wildtype. Progression-free survival (PFS) and overall survival (OS) were estimated for each risk type. Time-dependent receiver operating characteristic analysis using 10-fold cross-validation with 100-fold bootstrapping was used to compare the performance of an imaging-based survival model with that of a historical molecular-based survival model published in 2015, created using The Cancer Genome Archive data. RESULTS Prognostication according to the three imaging-based risk types was achieved for both PFS and OS (log-rank test, p < 0.001). The imaging-based survival model showed high prognostic value, with areas under the curves (AUCs) of 0.772 and 0.650 for 1-year PFS and OS, respectively, similar to the historical molecular-based survival model (AUC = 0.74 for PFS and 0.87 for OS). The imaging-based survival model achieved high long-term performance in both 3-year PFS (AUC = 0.806) and 5-year OS (AUC = 0.812). CONCLUSION Imaging-based risk stratification achieved histomolecular-level prognostication in diffuse glioma, NOS, and could aid in guiding patient referral for insufficient or unsuccessful molecular diagnosis. KEY POINTS • Three imaging-based risk types enable distinct prognostication in diffuse glioma, NOS (not otherwise specified). • The imaging-based survival model achieved similar prognostic performance as a historical molecular-based survival model. • For long-term prognostication of 3 and 5 years, the imaging-based survival model showed high performance.
Collapse
Affiliation(s)
- Eun Bee Jang
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Seo Young Park
- Department of Statistics and Data Science, Korea National Open University, Seoul, Republic of Korea
| | - Yeo Kyung Nam
- Department of Radiology, Shinchon Yonsei Hospital, Seoul, Republic of Korea
| | - Soo Jung Nam
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Young-Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jeong Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
68
|
Aboian M, Bousabarah K, Kazarian E, Zeevi T, Holler W, Merkaj S, Cassinelli Petersen G, Bahar R, Subramanian H, Sunku P, Schrickel E, Bhawnani J, Zawalich M, Mahajan A, Malhotra A, Payabvash S, Tocino I, Lin M, Westerhoff M. Clinical implementation of artificial intelligence in neuroradiology with development of a novel workflow-efficient picture archiving and communication system-based automated brain tumor segmentation and radiomic feature extraction. Front Neurosci 2022; 16:860208. [PMID: 36312024 PMCID: PMC9606757 DOI: 10.3389/fnins.2022.860208] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 07/13/2022] [Indexed: 11/18/2022] Open
Abstract
Purpose Personalized interpretation of medical images is critical for optimum patient care, but current tools available to physicians to perform quantitative analysis of patient’s medical images in real time are significantly limited. In this work, we describe a novel platform within PACS for volumetric analysis of images and thus development of large expert annotated datasets in parallel with radiologist performing the reading that are critically needed for development of clinically meaningful AI algorithms. Specifically, we implemented a deep learning-based algorithm for automated brain tumor segmentation and radiomics extraction, and embedded it into PACS to accelerate a supervised, end-to- end workflow for image annotation and radiomic feature extraction. Materials and methods An algorithm was trained to segment whole primary brain tumors on FLAIR images from multi-institutional glioma BraTS 2021 dataset. Algorithm was validated using internal dataset from Yale New Haven Health (YHHH) and compared (by Dice similarity coefficient [DSC]) to radiologist manual segmentation. A UNETR deep-learning was embedded into Visage 7 (Visage Imaging, Inc., San Diego, CA, United States) diagnostic workstation. The automatically segmented brain tumor was pliable for manual modification. PyRadiomics (Harvard Medical School, Boston, MA) was natively embedded into Visage 7 for feature extraction from the brain tumor segmentations. Results UNETR brain tumor segmentation took on average 4 s and the median DSC was 86%, which is similar to published literature but lower than the RSNA ASNR MICCAI BRATS challenge 2021. Finally, extraction of 106 radiomic features within PACS took on average 5.8 ± 0.01 s. The extracted radiomic features did not vary over time of extraction or whether they were extracted within PACS or outside of PACS. The ability to perform segmentation and feature extraction before radiologist opens the study was made available in the workflow. Opening the study in PACS, allows the radiologists to verify the segmentation and thus annotate the study. Conclusion Integration of image processing algorithms for tumor auto-segmentation and feature extraction into PACS allows curation of large datasets of annotated medical images and can accelerate translation of research into development of personalized medicine applications in the clinic. The ability to use familiar clinical tools to revise the AI segmentations and natively embedding the segmentation and radiomic feature extraction tools on the diagnostic workstation accelerates the process to generate ground-truth data.
Collapse
Affiliation(s)
- Mariam Aboian
- Department of Radiology and Biomedical Imaging, Brain Tumor Research Group, Yale School of Medicine, Yale University, New Haven, CT, United States
- *Correspondence: Mariam Aboian,
| | | | - Eve Kazarian
- Department of Radiology and Biomedical Imaging, Brain Tumor Research Group, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Tal Zeevi
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT, United States
| | | | - Sara Merkaj
- Department of Radiology and Biomedical Imaging, Brain Tumor Research Group, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Gabriel Cassinelli Petersen
- Department of Radiology and Biomedical Imaging, Brain Tumor Research Group, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Ryan Bahar
- Department of Radiology and Biomedical Imaging, Brain Tumor Research Group, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Harry Subramanian
- Department of Radiology and Biomedical Imaging, Brain Tumor Research Group, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Pranay Sunku
- Department of Radiology and Biomedical Imaging, Brain Tumor Research Group, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Elizabeth Schrickel
- Department of Radiology and Biomedical Imaging, Brain Tumor Research Group, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Jitendra Bhawnani
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Mathew Zawalich
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Amit Mahajan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Ajay Malhotra
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Sam Payabvash
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Irena Tocino
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - MingDe Lin
- Department of Radiology, Yale University and Visage Imaging, New Haven, CT, United States
| | | |
Collapse
|
69
|
Validation of MRI-Based Models to Predict MGMT Promoter Methylation in Gliomas: BraTS 2021 Radiogenomics Challenge. Cancers (Basel) 2022; 14:cancers14194827. [PMID: 36230750 PMCID: PMC9562637 DOI: 10.3390/cancers14194827] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/19/2022] [Accepted: 09/30/2022] [Indexed: 12/05/2022] Open
Abstract
Simple Summary O6-methylguanine-DNA methyl transferase (MGMT) methylation in glioblastoma is an important prognostic and predictive factor that requires an invasive surgical procedure for identification. In several recent studies, MGMT methylation prediction models were developed using MR images, and good diagnostic performance was achieved, which seems to indicate a promising future for radiogenomics. However, the diagnostic performance was not reproducible for numerous research teams when using a larger dataset in the RSNA-MICCAI Brain Tumor Radiogenomic Classification 2021 challenge. To our knowledge, there has been no study regarding the external validation of MGMT prediction models using large-scale multicenter datasets. We tested recent CNN architectures via extensive experiments to investigate whether MGMT methylation in gliomas can be predicted using MRI. With unexpected negative results, approximately 80% of the developed models showed no significant difference with the chance level of 50% in terms of external validation accuracy. In conclusion, MGMT methylation status of gliomas may not be predictable with preoperative MRI, even using deep learning. Abstract O6-methylguanine-DNA methyl transferase (MGMT) methylation prediction models were developed using only small datasets without proper external validation and achieved good diagnostic performance, which seems to indicate a promising future for radiogenomics. However, the diagnostic performance was not reproducible for numerous research teams when using a larger dataset in the RSNA-MICCAI Brain Tumor Radiogenomic Classification 2021 challenge. To our knowledge, there has been no study regarding the external validation of MGMT prediction models using large-scale multicenter datasets. We tested recent CNN architectures via extensive experiments to investigate whether MGMT methylation in gliomas can be predicted using MR images. Specifically, prediction models were developed and validated with different training datasets: (1) the merged (SNUH + BraTS) (n = 985); (2) SNUH (n = 400); and (3) BraTS datasets (n = 585). A total of 420 training and validation experiments were performed on combinations of datasets, convolutional neural network (CNN) architectures, MRI sequences, and random seed numbers. The first-place solution of the RSNA-MICCAI radiogenomic challenge was also validated using the external test set (SNUH). For model evaluation, the area under the receiver operating characteristic curve (AUROC), accuracy, precision, and recall were obtained. With unexpected negative results, 80.2% (337/420) and 60.0% (252/420) of the 420 developed models showed no significant difference with a chance level of 50% in terms of test accuracy and test AUROC, respectively. The test AUROC and accuracy of the first-place solution of the BraTS 2021 challenge were 56.2% and 54.8%, respectively, as validated on the SNUH dataset. In conclusion, MGMT methylation status of gliomas may not be predictable with preoperative MR images even using deep learning.
Collapse
|
70
|
Yin X, Liao H, Yun H, Lin N, Li S, Xiang Y, Ma X. Artificial intelligence-based prediction of clinical outcome in immunotherapy and targeted therapy of lung cancer. Semin Cancer Biol 2022; 86:146-159. [PMID: 35963564 DOI: 10.1016/j.semcancer.2022.08.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022]
Abstract
Lung cancer accounts for the main proportion of malignancy-related deaths and most patients are diagnosed at an advanced stage. Immunotherapy and targeted therapy have great advances in application in clinics to treat lung cancer patients, yet the efficacy is unstable. The response rate of these therapies varies among patients. Some biomarkers have been proposed to predict the outcomes of immunotherapy and targeted therapy, including programmed cell death-ligand 1 (PD-L1) expression and oncogene mutations. Nevertheless, the detection tests are invasive, time-consuming, and have high demands on tumor tissue. The predictive performance of conventional biomarkers is also unsatisfactory. Therefore, novel biomarkers are needed to effectively predict the outcomes of immunotherapy and targeted therapy. The application of artificial intelligence (AI) can be a possible solution, as it has several advantages. AI can help identify features that are unable to be used by humans and perform repetitive tasks. By combining AI methods with radiomics, pathology, genomics, transcriptomics, proteomics, and clinical data, the integrated model has shown predictive value in immunotherapy and targeted therapy, which significantly improves the precision treatment of lung cancer patients. Herein, we reviewed the application of AI in predicting the outcomes of immunotherapy and targeted therapy in lung cancer patients, and discussed the challenges and future directions in this field.
Collapse
Affiliation(s)
- Xiaomeng Yin
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Hu Liao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Hong Yun
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Nan Lin
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Shen Li
- West China School of Medicine, West China Hospital, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Yu Xiang
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Xuelei Ma
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China.
| |
Collapse
|
71
|
Swin Transformer Improves the IDH Mutation Status Prediction of Gliomas Free of MRI-Based Tumor Segmentation. J Clin Med 2022; 11:jcm11154625. [PMID: 35956236 PMCID: PMC9369996 DOI: 10.3390/jcm11154625] [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: 07/11/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Deep learning (DL) could predict isocitrate dehydrogenase (IDH) mutation status from MRIs. Yet, previous work focused on CNNs with refined tumor segmentation. To bridge the gap, this study aimed to evaluate the feasibility of developing a Transformer-based network to predict the IDH mutation status free of refined tumor segmentation. Methods: A total of 493 glioma patients were recruited from two independent institutions for model development (TCIA; N = 259) and external test (AHXZ; N = 234). IDH mutation status was predicted directly from T2 images with a Swin Transformer and conventional ResNet. Furthermore, to investigate the necessity of refined tumor segmentation, seven strategies for the model input image were explored: (i) whole tumor slice; (ii-iii) tumor mask and/or not edema; (iv-vii) tumor bounding box of 0.8, 1.0, 1.2, 1.5 times. Performance comparison was made among the networks of different architectures along with different image input strategies, using area under the curve (AUC) and accuracy (ACC). Finally, to further boost the performance, a hybrid model was built by incorporating the images with clinical features. Results: With the seven proposed input strategies, seven Swin Transformer models and seven ResNet models were built, respectively. Based on the seven Swin Transformer models, an averaged AUC of 0.965 (internal test) and 0.842 (external test) were achieved, outperforming 0.922 and 0.805 resulting from the seven ResNet models, respectively. When a bounding box of 1.0 times was used, Swin Transformer (AUC = 0.868, ACC = 80.7%), achieved the best results against the one that used tumor segmentation (Tumor + Edema, AUC = 0.862, ACC = 78.5%). The hybrid model that integrated age and location features into images yielded improved performance (AUC = 0.878, Accuracy = 82.0%) over the model that used images only. Conclusions: Swin Transformer outperforms the CNN-based ResNet in IDH prediction. Using bounding box input images benefits the DL networks in IDH prediction and makes the IDH prediction free of refined glioma segmentation feasible.
Collapse
|
72
|
Rudie JD, Calabrese E, Saluja R, Weiss D, Colby JB, Cha S, Hess CP, Rauschecker AM, Sugrue LP, Villanueva-Meyer JE. Longitudinal Assessment of Posttreatment Diffuse Glioma Tissue Volumes with Three-dimensional Convolutional Neural Networks. Radiol Artif Intell 2022; 4:e210243. [PMID: 36204543 PMCID: PMC9530762 DOI: 10.1148/ryai.210243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 05/17/2022] [Accepted: 07/15/2022] [Indexed: 12/30/2022]
Abstract
Neural networks were trained for segmentation and longitudinal assessment of posttreatment diffuse glioma. A retrospective cohort (from January 2018 to December 2019) of 298 patients with diffuse glioma (mean age, 52 years ± 14 [SD]; 177 men; 152 patients with glioblastoma, 72 patients with astrocytoma, and 74 patients with oligodendroglioma) who underwent two consecutive multimodal MRI examinations were randomly selected into training (n = 198) and testing (n = 100) samples. A posttreatment tumor segmentation three-dimensional nnU-Net convolutional neural network with multichannel inputs (T1, T2, and T1 postcontrast and fluid-attenuated inversion recovery [FLAIR]) was trained to segment three multiclass tissue types (peritumoral edematous, infiltrated, or treatment-changed tissue [ED]; active tumor or enhancing tissue [AT]; and necrotic core). Separate longitudinal change nnU-Nets were trained on registered and subtracted FLAIR and T1 postlongitudinal images to localize and better quantify and classify changes in ED and AT. Segmentation Dice scores, volume similarities, and 95th percentile Hausdorff distances ranged from 0.72 to 0.89, 0.90 to 0.96, and 2.5 to 3.6 mm, respectively. Accuracy rates of the posttreatment tumor segmentation and longitudinal change networks being able to classify longitudinal changes in ED and AT as increased, decreased, or unchanged were 76%-79% and 90%-91%, respectively. The accuracy levels of the longitudinal change networks were not significantly different from those of three neuroradiologists (accuracy, 90%-92%; κ, 0.58-0.63; P > .05). The results of this study support the potential clinical value of artificial intelligence-based automated longitudinal assessment of posttreatment diffuse glioma. Keywords: MR Imaging, Neuro-Oncology, Neural Networks, CNS, Brain/Brain Stem, Segmentation, Quantification, Convolutional Neural Network (CNN) Supplemental material is available for this article. © RSNA, 2022.
Collapse
|
73
|
Qin J, Tang Y, Wang B. Regional 18F-fluoromisonidazole PET images generated from multiple advanced MR images using neural networks in glioblastoma. Medicine (Baltimore) 2022; 101:e29572. [PMID: 35905276 PMCID: PMC9333488 DOI: 10.1097/md.0000000000029572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Generated 18F-fluoromisonidazole (18F-FMISO) positron emission tomography (PET) images for glioblastoma are highly sought after because 18F-FMISO can be radioactive, and the imaging procedure is not easy. This study aimed to explore the feasibility of using advanced magnetic resonance (MR) images to generate regional 18F-FMISO PET images and its predictive value for survival. Twelve kinds of advanced MR images of 28 patients from The Cancer Imaging Archive were processed. Voxel-by-voxel correlation analysis between 18F-FMISO images and advanced MR images was performed to select the MR images for generating regional 18F-FMISO images. Neural network algorithms provided by the MATLAB toolbox were used to generate regional 18F-FMISO images. The mean square error (MSE) was used to evaluate the regression effect. The prognostic value of generated 18F-FMISO images was evaluated by the Mantel-Cox test. A total of 299 831 voxels were extracted from the segmented regions of all patients. Eleven kinds of advanced MR images were selected to generate 18F-FMISO images. The best neural network algorithm was Bayesian regularization. The MSEs of the training, validation, and testing groups were 2.92E-2, 2.9E-2, and 2.92E-2, respectively. Both the maximum Tissue/Blood ratio (P = .017) and hypoxic volume (P = .023) of the generated images were predictive factors of overall survival, but only hypoxic volume (P = .029) was a predictive factor of progression-free survival. Multiple advanced MR images are feasible to generate qualified regional 18F-FMISO PET images using neural networks. The generated images also have predictive value in the prognostic evaluation of glioblastoma.
Collapse
Affiliation(s)
- Jianhua Qin
- School of Medicine, Qingdao University, Qingdao, P. R. China
- Department of Radiology, Rizhao Central Hospital, Rizhao, P. R. China
| | - Yu Tang
- Department of Radiology, Rizhao Central Hospital, Rizhao, P. R. China
| | - Bao Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, P. R. China
- *Correspondence: Bao Wang, Department of Radiology, Qilu Hospital of Shandong University, Jinan, P. R. China, 250012 (e-mail: )
| |
Collapse
|
74
|
Liu D, Chen J, Ge H, Hu X, Yang K, Liu Y, Hu G, Luo B, Yan Z, Song K, Xiao C, Zou Y, Zhang W, Liu H. Differentiation of malignant brain tumor types using intratumoral and peritumoral radiomic features. Front Oncol 2022; 12:848846. [PMID: 35965511 PMCID: PMC9366472 DOI: 10.3389/fonc.2022.848846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/28/2022] [Indexed: 12/14/2022] Open
Abstract
Tumor infiltration of central nervous system (CNS) malignant tumors may extend beyond visible contrast enhancement. This study explored tumor habitat characteristics in the intratumoral and peritumoral regions to distinguish common malignant brain tumors such as glioblastoma, primary central nervous system lymphoma, and brain metastases. The preoperative MRI data of 200 patients with solitary malignant brain tumors were included from two datasets for training. Quantitative radiomic features from the intratumoral and peritumoral regions were extracted for model training. The performance of the model was evaluated using data (n = 50) from the third clinical center. When combining the intratumoral and peritumoral features, the Adaboost model achieved the best area under the curve (AUC) of 0.91 and accuracy of 76.9% in the test cohort. Based on the optimal features and classifier, the model in the binary classification diagnosis achieves AUC of 0.98 (glioblastoma and lymphoma), 0.86 (lymphoma and metastases), and 0.70 (glioblastoma and metastases) in the test cohort, respectively. In conclusion, quantitative features from non-enhanced peritumoral regions (especially features from the 10-mm margin around the tumor) can provide additional information for the characterization of regional tumoral heterogeneity, which may offer potential value for future individualized assessment of patients with CNS tumors.
Collapse
Affiliation(s)
- Dongming Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jiu Chen
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Institute of Brain Sciences, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Honglin Ge
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Xinhua Hu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Institute of Brain Sciences, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Kun Yang
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Guanjie Hu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Bei Luo
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Zhen Yan
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Kun Song
- Department of Pathology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Chaoyong Xiao
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yuanjie Zou
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wenbin Zhang
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Institute of Brain Sciences, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Wenbin Zhang, ; Hongyi Liu,
| | - Hongyi Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Institute of Brain Sciences, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Wenbin Zhang, ; Hongyi Liu,
| |
Collapse
|
75
|
Xu J, Meng Y, Qiu K, Topatana W, Li S, Wei C, Chen T, Chen M, Ding Z, Niu G. Applications of Artificial Intelligence Based on Medical Imaging in Glioma: Current State and Future Challenges. Front Oncol 2022; 12:892056. [PMID: 35965542 PMCID: PMC9363668 DOI: 10.3389/fonc.2022.892056] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/22/2022] [Indexed: 12/24/2022] Open
Abstract
Glioma is one of the most fatal primary brain tumors, and it is well-known for its difficulty in diagnosis and management. Medical imaging techniques such as magnetic resonance imaging (MRI), positron emission tomography (PET), and spectral imaging can efficiently aid physicians in diagnosing, treating, and evaluating patients with gliomas. With the increasing clinical records and digital images, the application of artificial intelligence (AI) based on medical imaging has reduced the burden on physicians treating gliomas even further. This review will classify AI technologies and procedures used in medical imaging analysis. Additionally, we will discuss the applications of AI in glioma, including tumor segmentation and classification, prediction of genetic markers, and prediction of treatment response and prognosis, using MRI, PET, and spectral imaging. Despite the benefits of AI in clinical applications, several issues such as data management, incomprehension, safety, clinical efficacy evaluation, and ethical or legal considerations, remain to be solved. In the future, doctors and researchers should collaborate to solve these issues, with a particular emphasis on interdisciplinary teamwork.
Collapse
Affiliation(s)
- Jiaona Xu
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuting Meng
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kefan Qiu
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Win Topatana
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shijie Li
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Wei
- Department of Neurology, Affiliated Ningbo First Hospital, Ningbo, China
| | - Tianwen Chen
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingyu Chen
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| | - Guozhong Niu
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| |
Collapse
|
76
|
Liu D, Chen J, Ge H, Yan Z, Luo B, Hu X, Yang K, Liu Y, Liu H, Zhang W. Radiogenomics to characterize the immune-related prognostic signature associated with biological functions in glioblastoma. Eur Radiol 2022; 33:209-220. [PMID: 35881182 DOI: 10.1007/s00330-022-09012-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The tumor microenvironment and immune cell infiltration (ICI) associated with glioblastoma (GBM) play a vital role in cancer development, progression, and prognosis. This study aimed to establish an ICI-related prognostic biomarker and explore the associations between ICI signatures and radiomic features in patients with GBM. METHODS The gene expression and survival data of patients with GBM were obtained from three databases. Based on the ICI pattern, an individualized ICI score for each GBM patient was developed in the discovery set (n = 400) and independently verified in the validation set (n = 374). A total of 5915 radiomic features were extracted from the intratumoral and peritumoral regions. Recursive feature elimination and support vector machine methods were performed to select the key features and generate a model predictive of low- or high- ICI scores. The prognostic value of the identified radio genomic model was examined in an independent dataset (n = 149) using imaging and survival data. RESULTS We found that higher ICI scores often indicated worse patient prognosis (multivariable hazard ratio: 0.48 and 0.63 in discovery and validation set, respectively) and higher expression levels of immune checkpoint-related genes. A model that combined 11 radiomic features could well distinguish tumors with different ICI scores (AUC = 0.96, accuracy = 94%). This model was proven to be helpful for noninvasive prognostic stratification in an independent validation cohort. CONCLUSIONS ICI scores may serve as an effective prognostic biomarker to characterize potential biological processes in patients with GBM. This ICI signature can be evaluated noninvasively through radiogenomic analysis. KEY POINTS • Immune cell infiltration (ICI) scores can serve as an effective prognostic biomarker in patients with glioblastoma. • The ICI signature can be evaluated noninvasively through radiomic features derived from the intratumoral and peritumoral regions. • The prognostic value of the radiogenomic model can be verified by independent survival and MRI data.
Collapse
Affiliation(s)
- Dongming Liu
- Department of Neurosurgery, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jiu Chen
- Institute of Neuropsychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.,Institute of Brain Sciences, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Honglin Ge
- Department of Neurosurgery, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Zhen Yan
- Department of Neurosurgery, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Bei Luo
- Department of Neurosurgery, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Xinhua Hu
- Department of Neurosurgery, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.,Institute of Brain Sciences, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Kun Yang
- Department of Neurosurgery, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yong Liu
- Department of Neurosurgery, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Hongyi Liu
- Department of Neurosurgery, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China. .,Institute of Brain Sciences, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Wenbin Zhang
- Department of Neurosurgery, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China. .,Institute of Brain Sciences, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
77
|
Efficient Prediction of Ki-67 Proliferation Index in Meningiomas on MRI: From Traditional Radiological Findings to a Machine Learning Approach. Cancers (Basel) 2022; 14:cancers14153637. [PMID: 35892896 PMCID: PMC9330288 DOI: 10.3390/cancers14153637] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 02/05/2023] Open
Abstract
Background/aim This study aimed to explore the value of radiological and radiomic features retrieved from magnetic resonance imaging in the prediction of a Ki-67 proliferative index in meningioma patients using a machine learning model. Methods This multicenter, retrospective study included 371 patients collected from two centers. The Ki-67 expression was classified into low-expressed and high-expressed groups with a threshold of 5%. Clinical features and radiological features were collected and analyzed by using univariate and multivariate statistical analyses. Radiomic features were extracted from contrast-enhanced images, followed by three independent feature selections. Six predictive models were constructed with different combinations of features by using linear discriminant analysis (LDA) classifier. Results The multivariate analysis suggested that the presence of intratumoral necrosis (p = 0.032) and maximum diameter (p < 0.001) were independently correlated with a high Ki-67 status. The predictive models showed good performance with AUC of 0.837, accuracy of 0.810, sensitivity of 0.857, and specificity of 0.771 in the internal test and with AUC of 0.700, accuracy of 0.557, sensitivity of 0.314, and specificity of 0.885 in the external test. Conclusion The results of this study suggest that the predictive model can efficiently predict the Ki-67 index of meningioma patients to facilitate the therapeutic management.
Collapse
|
78
|
Blokker M, Hamer PCDW, Wesseling P, Groot ML, Veta M. Fast intraoperative histology-based diagnosis of gliomas with third harmonic generation microscopy and deep learning. Sci Rep 2022; 12:11334. [PMID: 35790792 PMCID: PMC9256596 DOI: 10.1038/s41598-022-15423-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 06/23/2022] [Indexed: 11/09/2022] Open
Abstract
Management of gliomas requires an invasive treatment strategy, including extensive surgical resection. The objective of the neurosurgeon is to maximize tumor removal while preserving healthy brain tissue. However, the lack of a clear tumor boundary hampers the neurosurgeon's ability to accurately detect and resect infiltrating tumor tissue. Nonlinear multiphoton microscopy, in particular higher harmonic generation, enables label-free imaging of excised brain tissue, revealing histological hallmarks within seconds. Here, we demonstrate a real-time deep learning-based pipeline for automated glioma image analysis, matching video-rate image acquisition. We used a custom noise detection scheme, and a fully-convolutional classification network, to achieve on average 79% binary accuracy, 0.77 AUC and 0.83 mean average precision compared to the consensus of three pathologists, on a preliminary dataset. We conclude that the combination of real-time imaging and image analysis shows great potential for intraoperative assessment of brain tissue during tumor surgery.
Collapse
Affiliation(s)
- Max Blokker
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - Philip C de Witt Hamer
- Department of Neurosurgery, Amsterdam UMC location VU University Medical Center, Amsterdam, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, Amsterdam UMC location VU University Medical Center, Amsterdam, The Netherlands
| | - Marie Louise Groot
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mitko Veta
- Medical Image Analysis Group (IMAG/e), Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
79
|
Role of Artificial Intelligence in Radiogenomics for Cancers in the Era of Precision Medicine. Cancers (Basel) 2022; 14:cancers14122860. [PMID: 35740526 PMCID: PMC9220825 DOI: 10.3390/cancers14122860] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Recently, radiogenomics has played a significant role and offered a new understanding of cancer’s biology and behavior in response to standard therapy. It also provides a more precise prognosis, investigation, and analysis of the patient’s cancer. Over the years, Artificial Intelligence (AI) has provided a significant strength in radiogenomics. In this paper, we offer computational and oncological prospects of the role of AI in radiogenomics, as well as its offers, achievements, opportunities, and limitations in the current clinical practices. Abstract Radiogenomics, a combination of “Radiomics” and “Genomics,” using Artificial Intelligence (AI) has recently emerged as the state-of-the-art science in precision medicine, especially in oncology care. Radiogenomics syndicates large-scale quantifiable data extracted from radiological medical images enveloped with personalized genomic phenotypes. It fabricates a prediction model through various AI methods to stratify the risk of patients, monitor therapeutic approaches, and assess clinical outcomes. It has recently shown tremendous achievements in prognosis, treatment planning, survival prediction, heterogeneity analysis, reoccurrence, and progression-free survival for human cancer study. Although AI has shown immense performance in oncology care in various clinical aspects, it has several challenges and limitations. The proposed review provides an overview of radiogenomics with the viewpoints on the role of AI in terms of its promises for computational as well as oncological aspects and offers achievements and opportunities in the era of precision medicine. The review also presents various recommendations to diminish these obstacles.
Collapse
|
80
|
Yoshida K, Kawashima H, Kannon T, Tajima A, Ohno N, Terada K, Takamatsu A, Adachi H, Ohno M, Miyati T, Ishikawa S, Ikeda H, Gabata T. Prediction of pathological complete response to neoadjuvant chemotherapy in breast cancer using radiomics of pretreatment dynamic contrast-enhanced MRI. Magn Reson Imaging 2022; 92:19-25. [PMID: 35636571 DOI: 10.1016/j.mri.2022.05.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 12/29/2022]
Abstract
PURPOSE To investigate if the pretreatment dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI)-based radiomics machine learning predicts the pathological complete response (pCR) to neoadjuvant chemotherapy (NAC) in breast cancer patients. METHODS Seventy-eight breast cancer patients who underwent DCE-MRI before NAC and confirmed as pCR or non-pCR were enrolled. Early enhancement mapping images of pretreatment DCE-MRI were created using subtraction formula as follows: Early enhancement mapping = (Signal 1 min - Signal pre)/Signal pre. Images of the whole tumors were manually segmented and radiomics features extracted. Five prediction models were built using five scenarios that included clinical information, subjective radiological findings, first order texture features, second order texture features, and their combinations. In texture analysis workflow, the corresponding variables were identified by mutual information for feature selection and random forest was used for model prediction. In five models, the area under the receiver operating characteristic curves (AUC) to predict the pCR and several metrics for model evaluation were analyzed. RESULTS The best diagnostic performance based on F-score was achieved when both first and second order texture features with clinical information and subjective radiological findings were used (AUC = 0.77). The second best diagnostic performance was achieved with an AUC of 0.76 for first order texture features followed by an AUC of 0.76 for first and second order texture features. CONCLUSIONS Pretreatment DCE-MRI can improve the prediction of pCR in breast cancer patients when all texture features with clinical information and subjective radiological findings are input to build the prediction model.
Collapse
Affiliation(s)
- Kotaro Yoshida
- Department of Radiology, Kanazawa University Graduate School of Medical Sciences, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| | - Hiroko Kawashima
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| | - Takayuki Kannon
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| | - Naoki Ohno
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| | - Kanako Terada
- Department of Radiology, Kanazawa University Graduate School of Medical Sciences, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan
| | - Atsushi Takamatsu
- Department of Radiology, Kanazawa University Graduate School of Medical Sciences, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan
| | - Hayato Adachi
- Division of Radiology, Kanazawa University Hospital, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan
| | - Masako Ohno
- Division of Radiology, Kanazawa University Hospital, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| | - Tosiaki Miyati
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| | - Satoko Ishikawa
- Department of Breast Surgery, Kanazawa University Hospital, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| | - Hiroko Ikeda
- Diagnostic Pathology, Kanazawa University Hospital, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| | - Toshifumi Gabata
- Department of Radiology, Kanazawa University Graduate School of Medical Sciences, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| |
Collapse
|
81
|
Automatic Classification of Hospital Settings through Artificial Intelligence. ELECTRONICS 2022. [DOI: 10.3390/electronics11111697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Modern hospitals have to meet requirements from national and international institutions in order to ensure hygiene, quality and organisational standards. Moreover, a hospital must be flexible and adaptable to new delivery models for healthcare services. Various hospital monitoring tools have been developed over the years, which allow for a detailed picture of the effectiveness and efficiency of the hospital itself. Many of these systems are based on database management systems (DBMSs), building information modelling (BIM) or geographic information systems (GISs). This work presents an automatic recognition system for hospital settings that integrates these tools. Three alternative proposals were analysed in terms of the construction of the system: the first was based on the use of general models that are present on the cloud for the classification of images; the second consisted of the creation of a customised model and referred to the Clarifai Custom Model service; the third used an object recognition software that was developed by Facebook AI Research combined with a random forest classifier. The obtained results were promising. The customised model almost always classified the photos according to the correct intended use, resulting in a high percentage of confidence of up to 96%. Classification using the third tool was excellent when considering a limited number of hospital settings, with a peak accuracy of higher than 99% and an area under the ROC curve (AUC) of one for specific classes. As expected, increasing the number of room typologies to be discerned negatively affected performance.
Collapse
|
82
|
Park JE. Artificial Intelligence in Neuro-Oncologic Imaging: A Brief Review for Clinical Use Cases and Future Perspectives. Brain Tumor Res Treat 2022; 10:69-75. [PMID: 35545825 PMCID: PMC9098975 DOI: 10.14791/btrt.2021.0031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/24/2022] [Accepted: 04/18/2022] [Indexed: 11/20/2022] Open
Abstract
The artificial intelligence (AI) techniques, both deep learning end-to-end approaches and radiomics with machine learning, have been developed for various imaging-based tasks in neuro-oncology. In this brief review, use cases of AI in neuro-oncologic imaging are summarized: image quality improvement, metastasis detection, radiogenomics, and treatment response monitoring. We then give a brief overview of generative adversarial network and potential utility of synthetic images for various deep learning algorithms of imaging-based tasks and image translation tasks as becoming new data input. Lastly, we highlight the importance of cohorts and clinical trial as a true validation for clinical utility of AI in neuro-oncologic imaging.
Collapse
Affiliation(s)
- Ji Eun Park
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
83
|
Liao G. Artificial Intelligence-Based MRI in Diagnosis of Injury of Cranial Nerves of Premature Infant and Its Correlation with Inflammation of Placenta. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:4550079. [PMID: 35414800 PMCID: PMC8977307 DOI: 10.1155/2022/4550079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 12/14/2022]
Abstract
The study focused on the effects of artificial intelligence algorithms in magnetic resonance imaging (MRI) for diagnosing cranial nerve inflammation of placenta and the correlation between cranial nerve injury with placental inflammation was explored. The subjects were selected from 132 premature infants in the hospital. According to the pathological examination of placenta, 81 cases with chorioamnionitis were taken as the experimental group and 51 cases without chorioamnionitis were taken as the control group. The incidence of cranial nerve injury in different groups of premature infants was analyzed by MRI diagnosis based on the principal component analysis (PCA) artificial intelligence algorithm, so as to analyze the correlation between cranial nerve injury and placental inflammation in premature infants. It was found that when the PCA artificial intelligence algorithm was incorporated into MRI examination of cranial nerve injury of premature infant, the A (accuracy), P (precision), R (recall), and F1 values under the PCA algorithm were 92%, 93.75%, 90%, and 92.87%, respectively. The A, P, R, and F1 of the control group were 54%, 54.1%, 52%, and 53.03%, respectively; there were statistically significant differences between the two groups, P < 0.05. As for the correlation of placental inflammation and cranial nerve injury, the positive detection rate of the experimental group was 53.09%, and the positive detection rate of the control group was 15.69%, and the difference was statistically significant, P < 0.05. In conclusion, the PCA artificial intelligence algorithm has high effectiveness and high accuracy in auxiliary diagnosis of premature brain nerve injury, and placental inflammation greatly increases the chance of premature infant suffering from brain nerve injury.
Collapse
Affiliation(s)
- Gui Liao
- Department of Pediatrics, The Third People's Hospital of Yunnan Province, Kunming 650011, Yunnan, China
| |
Collapse
|
84
|
Avellar L, Stefano Filho C, Delgado G, Frizera A, Rocon E, Leal-Junior A. AI-enabled photonic smart garment for movement analysis. Sci Rep 2022; 12:4067. [PMID: 35260746 PMCID: PMC8904460 DOI: 10.1038/s41598-022-08048-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/24/2022] [Indexed: 02/04/2023] Open
Abstract
Smart textiles are novel solutions for remote healthcare monitoring which involve non-invasive sensors-integrated clothing. Polymer optical fiber (POF) sensors have attractive features for smart textile technology, and combined with Artificial Intelligence (AI) algorithms increase the potential of intelligent decision-making. This paper presents the development of a fully portable photonic smart garment with 30 multiplexed POF sensors combined with AI algorithms to evaluate the system ability on the activity classification of multiple subjects. Six daily activities are evaluated: standing, sitting, squatting, up-and-down arms, walking and running. A k-nearest neighbors classifier is employed and results from 10 trials of all volunteers presented an accuracy of 94.00 (0.14)%. To achieve an optimal amount of sensors, the principal component analysis is used for one volunteer and results showed an accuracy of 98.14 (0.31)% using 10 sensors, 1.82% lower than using 30 sensors. Cadence and breathing rate were estimated and compared to the data from an inertial measurement unit located on the garment back and the highest error was 2.22%. Shoulder flexion/extension was also evaluated. The proposed approach presented feasibility for activity recognition and movement-related parameters extraction, leading to a system fully optimized, including the number of sensors and wireless communication, for Healthcare 4.0.
Collapse
Affiliation(s)
- Leticia Avellar
- Graduate Program in Electrical Engineering, Federal University of Espírito Santo (UFES), Fernando Ferrari Avenue, Vitória, 29075-910, Brazil.
| | - Carlos Stefano Filho
- Neurophysics Group, "Gleb Wataghin" Institute of Physics, University of Campinas, Campinas, Brazil
| | - Gabriel Delgado
- Centro de Automática y Robótica, Ctra. Campo Real, 28500, Arganda del Rey, Madrid, Spain
| | - Anselmo Frizera
- Graduate Program in Electrical Engineering, Federal University of Espírito Santo (UFES), Fernando Ferrari Avenue, Vitória, 29075-910, Brazil
| | - Eduardo Rocon
- Centro de Automática y Robótica, Ctra. Campo Real, 28500, Arganda del Rey, Madrid, Spain
| | - Arnaldo Leal-Junior
- Graduate Program in Electrical Engineering, Federal University of Espírito Santo (UFES), Fernando Ferrari Avenue, Vitória, 29075-910, Brazil
| |
Collapse
|
85
|
Vobugari N, Raja V, Sethi U, Gandhi K, Raja K, Surani SR. Advancements in Oncology with Artificial Intelligence-A Review Article. Cancers (Basel) 2022; 14:1349. [PMID: 35267657 PMCID: PMC8909088 DOI: 10.3390/cancers14051349] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
Well-trained machine learning (ML) and artificial intelligence (AI) systems can provide clinicians with therapeutic assistance, potentially increasing efficiency and improving efficacy. ML has demonstrated high accuracy in oncology-related diagnostic imaging, including screening mammography interpretation, colon polyp detection, glioma classification, and grading. By utilizing ML techniques, the manual steps of detecting and segmenting lesions are greatly reduced. ML-based tumor imaging analysis is independent of the experience level of evaluating physicians, and the results are expected to be more standardized and accurate. One of the biggest challenges is its generalizability worldwide. The current detection and screening methods for colon polyps and breast cancer have a vast amount of data, so they are ideal areas for studying the global standardization of artificial intelligence. Central nervous system cancers are rare and have poor prognoses based on current management standards. ML offers the prospect of unraveling undiscovered features from routinely acquired neuroimaging for improving treatment planning, prognostication, monitoring, and response assessment of CNS tumors such as gliomas. By studying AI in such rare cancer types, standard management methods may be improved by augmenting personalized/precision medicine. This review aims to provide clinicians and medical researchers with a basic understanding of how ML works and its role in oncology, especially in breast cancer, colorectal cancer, and primary and metastatic brain cancer. Understanding AI basics, current achievements, and future challenges are crucial in advancing the use of AI in oncology.
Collapse
Affiliation(s)
- Nikitha Vobugari
- Department of Internal Medicine, Medstar Washington Hospital Center, Washington, DC 20010, USA; (N.V.); (K.G.)
| | - Vikranth Raja
- Department of Medicine, P.S.G Institute of Medical Sciences and Research, Coimbatore 641004, Tamil Nadu, India;
| | - Udhav Sethi
- School of Computer Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Kejal Gandhi
- Department of Internal Medicine, Medstar Washington Hospital Center, Washington, DC 20010, USA; (N.V.); (K.G.)
| | - Kishore Raja
- Department of Pediatric Cardiology, University of Minnesota, Minneapolis, MN 55454, USA;
| | - Salim R. Surani
- Department of Pulmonary and Critical Care, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
86
|
T2-Fluid-Attenuated Inversion Recovery Mismatch Sign in Grade II and III Gliomas: Is There a Coexisting T2-Diffusion-Weighted Imaging Mismatch? J Comput Assist Tomogr 2022; 46:251-256. [PMID: 35297581 DOI: 10.1097/rct.0000000000001267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To determine whether the T2 fluid-attenuated inversion recovery (T2-FLAIR) mismatch sign in diffuse gliomas is associated with an equivalent pattern of disparity in signal intensities when comparing T2- and diffusion-weighted imaging (DWI). METHODS The level of correspondence between T2-FLAIR and T2-DWI evaluations in 34 World Health Organization grade II/III gliomas and interreader agreement among 3 neuroradiologists were assessed by calculating intraclass correlation coefficient and κ statistics, respectively. Tumoral apparent diffusion coefficient values were compared using t test. RESULTS There was an almost perfect correspondence between the 2 mismatch signs (intraclass correlation coefficient = 0.824 [95% confidence interval, 0.68-0.91]) that were associated with higher mean tumoral apparent diffusion coefficient (P < 0.01). Interreader agreement was substantial for T2-FLAIR (Fleiss κ = 0.724) and moderate for T2-DWI comparisons (Fleiss κ = 0.589) (P < 0.001). CONCLUSIONS The T2-FLAIR mismatch sign is usually reflected by a distinct microstructural pattern on DWI. The management of this tumor subtype may benefit from specifically tailored imaging assessments.
Collapse
|
87
|
Booth TC, Wiegers EC, Warnert EAH, Schmainda KM, Riemer F, Nechifor RE, Keil VC, Hangel G, Figueiredo P, Álvarez-Torres MDM, Henriksen OM. High-Grade Glioma Treatment Response Monitoring Biomarkers: A Position Statement on the Evidence Supporting the Use of Advanced MRI Techniques in the Clinic, and the Latest Bench-to-Bedside Developments. Part 2: Spectroscopy, Chemical Exchange Saturation, Multiparametric Imaging, and Radiomics. Front Oncol 2022; 11:811425. [PMID: 35340697 PMCID: PMC8948428 DOI: 10.3389/fonc.2021.811425] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/28/2021] [Indexed: 01/16/2023] Open
Abstract
Objective To summarize evidence for use of advanced MRI techniques as monitoring biomarkers in the clinic, and to highlight the latest bench-to-bedside developments. Methods The current evidence regarding the potential for monitoring biomarkers was reviewed and individual modalities of metabolism and/or chemical composition imaging discussed. Perfusion, permeability, and microstructure imaging were similarly analyzed in Part 1 of this two-part review article and are valuable reading as background to this article. We appraise the clinic readiness of all the individual modalities and consider methodologies involving machine learning (radiomics) and the combination of MRI approaches (multiparametric imaging). Results The biochemical composition of high-grade gliomas is markedly different from healthy brain tissue. Magnetic resonance spectroscopy allows the simultaneous acquisition of an array of metabolic alterations, with choline-based ratios appearing to be consistently discriminatory in treatment response assessment, although challenges remain despite this being a mature technique. Promising directions relate to ultra-high field strengths, 2-hydroxyglutarate analysis, and the use of non-proton nuclei. Labile protons on endogenous proteins can be selectively targeted with chemical exchange saturation transfer to give high resolution images. The body of evidence for clinical application of amide proton transfer imaging has been building for a decade, but more evidence is required to confirm chemical exchange saturation transfer use as a monitoring biomarker. Multiparametric methodologies, including the incorporation of nuclear medicine techniques, combine probes measuring different tumor properties. Although potentially synergistic, the limitations of each individual modality also can be compounded, particularly in the absence of standardization. Machine learning requires large datasets with high-quality annotation; there is currently low-level evidence for monitoring biomarker clinical application. Conclusion Advanced MRI techniques show huge promise in treatment response assessment. The clinical readiness analysis highlights that most monitoring biomarkers require standardized international consensus guidelines, with more facilitation regarding technique implementation and reporting in the clinic.
Collapse
Affiliation(s)
- Thomas C. Booth
- School of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London, United Kingdom
- Department of Neuroradiology, King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | - Evita C. Wiegers
- Department of Radiology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Kathleen M. Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Frank Riemer
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Ruben E. Nechifor
- Department of Clinical Psychology and Psychotherapy International Institute for the Advanced Studies of Psychotherapy and Applied Mental Health, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Vera C. Keil
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, Netherlands
| | - Gilbert Hangel
- Department of Neurosurgery & High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University Vienna, Vienna, Austria
| | - Patrícia Figueiredo
- Department of Bioengineering and Institute for Systems and Robotics - Lisboa, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | | | - Otto M. Henriksen
- Department of Clinical Physiology, Nuclear medicine and PET, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
88
|
Hwang I, Choi SH, Kim JW, Yeon EK, Lee JY, Yoo RE, Kang KM, Yun TJ, Kim JH, Sohn CH. Response prediction of vestibular schwannoma after gamma-knife radiosurgery using pretreatment dynamic contrast-enhanced MRI: a prospective study. Eur Radiol 2022; 32:3734-3743. [PMID: 35084518 DOI: 10.1007/s00330-021-08517-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/09/2021] [Accepted: 12/10/2021] [Indexed: 11/04/2022]
Abstract
OBJECTIVES There are few known predictive factors for response to gamma-knife radiosurgery (GKRS) in vestibular schwannoma (VS). We investigated the predictive role of pretreatment dynamic contrast-enhanced (DCE)-MRI parameters regarding the tumor response after GKRS in sporadic VS. METHODS This single-center prospective study enrolled participants between April 2017 and February 2019. We performed a volumetric measurement of DCE-MRI-derived parameters before GKRS. The tumor volume was measured in a follow-up MRI. The pharmacokinetic parameters were compared between responders and nonresponders according to 20% or more tumor volume reduction. Stepwise multivariable logistic regression analyses were performed, and the diagnostic performance of DCE-MRI parameters for the prediction of tumor response was evaluated by receiver operating characteristic curve analysis. RESULTS Ultimately, 35 participants (21 women, 52 ± 12 years) were included. There were 22 (62.9%) responders with a mean follow-up interval of 30.2 ± 5.7 months. Ktrans (0.036 min-1 vs. 0.057 min-1, p = .008) and initial area under the time-concentration curve within 90 s (IAUC90) (84.4 vs. 143.6, p = .003) showed significant differences between responders and nonresponders. Ktrans (OR = 0.96, p = .021) and IAUC90 (OR = 0.97, p = .004) were significant differentiating variables in each multivariable model with clinical variables for tumor response prediction. Ktrans showed a sensitivity of 81.8% and a specificity of 69.2%, and IAUC90 showed a sensitivity of 100% and a specificity of 53.8% for tumor response prediction. CONCLUSION DCE-MRI (particularly Ktrans and IAUC90) has the potential to be a predictive factor for tumor response in VS after GKRS. KEY POINTS •Pretreatment prediction of gamma-knife radiosurgery response in vestibular schwannoma is still challenging. •Dynamic contrast-enhanced MRI could have predictive value for the response of vestibular schwannoma after gamma-knife radiosurgery.
Collapse
Affiliation(s)
- Inpyeong Hwang
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea. .,Institute of Radiation Medicine, Seoul National University Medical Research Center, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea.
| | - Jin Wook Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eung Koo Yeon
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Ji Ye Lee
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Roh-Eul Yoo
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Institute of Radiation Medicine, Seoul National University Medical Research Center, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tae Jin Yun
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji-Hoon Kim
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Institute of Radiation Medicine, Seoul National University Medical Research Center, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| |
Collapse
|
89
|
Rai S, Raeesa F, Kamath M, Rai S, Pai M, Prabhu S. Multiparametric differentiation of intracranial central nervous system lymphoma and high-grade glioma using diffusion-, perfusion-, susceptibility-weighted magnetic resonance imaging, and spectroscopy. WEST AFRICAN JOURNAL OF RADIOLOGY 2022. [DOI: 10.4103/wajr.wajr_16_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
90
|
Machine Learning-Based Radiomics in Neuro-Oncology. ACTA NEUROCHIRURGICA. SUPPLEMENT 2021; 134:139-151. [PMID: 34862538 DOI: 10.1007/978-3-030-85292-4_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the last decades, modern medicine has evolved into a data-centered discipline, generating massive amounts of granular high-dimensional data exceeding human comprehension. With improved computational methods, machine learning and artificial intelligence (AI) as tools for data processing and analysis are becoming more and more important. At the forefront of neuro-oncology and AI-research, the field of radiomics has emerged. Non-invasive assessments of quantitative radiological biomarkers mined from complex imaging characteristics across various applications are used to predict survival, discriminate between primary and secondary tumors, as well as between progression and pseudo-progression. In particular, the application of molecular phenotyping, envisioned in the field of radiogenomics, has gained popularity for both primary and secondary brain tumors. Although promising results have been obtained thus far, the lack of workflow standardization and availability of multicenter data remains challenging. The objective of this review is to provide an overview of novel applications of machine learning- and deep learning-based radiomics in primary and secondary brain tumors and their implications for future research in the field.
Collapse
|
91
|
Application of artificial intelligence and radiomics in pituitary neuroendocrine and sellar tumors: a quantitative and qualitative synthesis. Neuroradiology 2021; 64:647-668. [PMID: 34839380 DOI: 10.1007/s00234-021-02845-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/21/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE To systematically review the literature regarding the application of machine learning (ML) of magnetic resonance imaging (MRI) radiomics in common sellar tumors. To identify future directions for application of ML in sellar tumor MRI. METHODS PubMed, Medline, Embase, Google Scholar, Scopus, ArxIV, and bioRxiv were searched to identify relevant studies published between 2010 and September 2021. Studies were included if they specifically involved ML of MRI radiomics in the analysis of sellar masses. Risk of bias assessment was performed using the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) Tool. RESULTS Fifty-eight articles were identified for review. All papers utilized retrospective data, and a quantitative systematic review was performed for thirty-one studies utilizing a public dataset which compared pituitary adenomas, meningiomas, and gliomas. One of the analyzed architectures yielded the highest classification accuracy of 0.996. The remaining twenty-seven articles were qualitatively reviewed and showed promising findings in predicting specific tumor characteristics such as tumor consistency, Ki-67 proliferative index, and post-surgical recurrence. CONCLUSION This review highlights the potential clinical application of ML using MRI radiomic data of the sellar region in diagnosis and predicting treatment outcomes. We describe future directions for practical application in the clinical care of patients with pituitary neuroendocrine and other sellar tumors.
Collapse
|
92
|
Pasquini L, Napolitano A, Lucignani M, Tagliente E, Dellepiane F, Rossi-Espagnet MC, Ritrovato M, Vidiri A, Villani V, Ranazzi G, Stoppacciaro A, Romano A, Di Napoli A, Bozzao A. AI and High-Grade Glioma for Diagnosis and Outcome Prediction: Do All Machine Learning Models Perform Equally Well? Front Oncol 2021; 11:601425. [PMID: 34888226 PMCID: PMC8649764 DOI: 10.3389/fonc.2021.601425] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Radiomic models outperform clinical data for outcome prediction in high-grade gliomas (HGG). However, lack of parameter standardization limits clinical applications. Many machine learning (ML) radiomic models employ single classifiers rather than ensemble learning, which is known to boost performance, and comparative analyses are lacking in the literature. We aimed to compare ML classifiers to predict clinically relevant tasks for HGG: overall survival (OS), isocitrate dehydrogenase (IDH) mutation, O-6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation, epidermal growth factor receptor vIII (EGFR) amplification, and Ki-67 expression, based on radiomic features from conventional and advanced magnetic resonance imaging (MRI). Our objective was to identify the best algorithm for each task. One hundred fifty-six adult patients with pathologic diagnosis of HGG were included. Three tumoral regions were manually segmented: contrast-enhancing tumor, necrosis, and non-enhancing tumor. Radiomic features were extracted with a custom version of Pyradiomics and selected through Boruta algorithm. A Grid Search algorithm was applied when computing ten times K-fold cross-validation (K=10) to get the highest mean and lowest spread of accuracy. Model performance was assessed as AUC-ROC curve mean values with 95% confidence intervals (CI). Extreme Gradient Boosting (xGB) obtained highest accuracy for OS (74,5%), Adaboost (AB) for IDH mutation (87.5%), MGMT methylation (70,8%), Ki-67 expression (86%), and EGFR amplification (81%). Ensemble classifiers showed the best performance across tasks. High-scoring radiomic features shed light on possible correlations between MRI and tumor histology.
Collapse
Affiliation(s)
- Luca Pasquini
- Neuroradiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| | - Antonio Napolitano
- Medical Physics Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Martina Lucignani
- Medical Physics Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Emanuela Tagliente
- Medical Physics Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Francesco Dellepiane
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| | - Maria Camilla Rossi-Espagnet
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Matteo Ritrovato
- Unit of Health Technology Assessment (HTA), Biomedical Technology Risk Manager, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Antonello Vidiri
- Radiology and Diagnostic Imaging Department, Regina Elena National Cancer Institute, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Veronica Villani
- Neuro-Oncology Unit, Regina Elena National Cancer Institute, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Giulio Ranazzi
- Department of Clinical and Molecular Medicine, Surgical Pathology Units, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| | - Antonella Stoppacciaro
- Department of Clinical and Molecular Medicine, Surgical Pathology Units, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| | - Andrea Romano
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| | - Alberto Di Napoli
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
- Radiology Department, Castelli Romani Hospital, Rome, Italy
| | - Alessandro Bozzao
- Neuroradiology Unit, Neuroscience, Mental Health and Sensory Organs (NESMOS) Department, Sant’Andrea Hospital, La Sapienza University, Rome, Italy
| |
Collapse
|
93
|
Abstract
Innovations and advances in technologies over the past few years have yielded faster and wider diagnostic applications to patients with neurologic diseases. This article focuses on the foreseeable developments of the diagnostic tools available to the neurologist in the next 15 years. Clinical judgment is and will remain the cornerstone of the diagnostic process, assisted by novel technologies, such as artificial intelligence and machine learning. Future neurologists must be educated to develop, cultivate, and rely on their clinical skills, while becoming familiar with novel, often complex, assistive technologies.
Collapse
|
94
|
Alhasan AS. Clinical Applications of Artificial Intelligence, Machine Learning, and Deep Learning in the Imaging of Gliomas: A Systematic Review. Cureus 2021; 13:e19580. [PMID: 34926051 PMCID: PMC8671075 DOI: 10.7759/cureus.19580] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2021] [Indexed: 02/02/2023] Open
Abstract
In neuro-oncology, magnetic resonance imaging (MRI) is a critically important, non-invasive radiologic assessment technique for brain tumor diagnosis, especially glioma. Deep learning improves MRI image characterization and interpretation through the utilization of raw imaging data and provides unprecedented enhancement of images and representation for detection and classification through deep neural networks. This systematic review and quality appraisal method aim to summarize deep learning approaches used in neuro-oncology imaging to aid healthcare professionals. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, a total of 20 low-risk studies on the established use of deep learning models to identify glioma genetic mutations and grading were selected, based on a Quality Assessment of Diagnostic Accuracy Studies 2 score of ≥9. The included studies provided the deep learning models used alongside their outcome measures, the number of patients, and the molecular markers for brain glioma classification. In 19 studies, the researchers determined that the deep learning model improved the clinical outcome and treatment protocol in patients with a brain tumor. In five studies, the authors determined the sensitivity of the deep learning model used, and in four studies, the authors determined the specificity of the models. Convolutional neural network models were used in 16 studies. In eight studies, the researchers examined glioma grading by using different deep learning models compared with other models. In this review, we found that deep learning models significantly improve the diagnostic and classification accuracy of brain tumors, particularly gliomas without the need for invasive methods. Most studies have presented validated results and can be used in clinical practice to improve patient care and prognosis.
Collapse
|
95
|
Xu Z, Wang X, Zeng S, Ren X, Yan Y, Gong Z. Applying artificial intelligence for cancer immunotherapy. Acta Pharm Sin B 2021; 11:3393-3405. [PMID: 34900525 PMCID: PMC8642413 DOI: 10.1016/j.apsb.2021.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/07/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
Artificial intelligence (AI) is a general term that refers to the use of a machine to imitate intelligent behavior for performing complex tasks with minimal human intervention, such as machine learning; this technology is revolutionizing and reshaping medicine. AI has considerable potential to perfect health-care systems in areas such as diagnostics, risk analysis, health information administration, lifestyle supervision, and virtual health assistance. In terms of immunotherapy, AI has been applied to the prediction of immunotherapy responses based on immune signatures, medical imaging and histological analysis. These features could also be highly useful in the management of cancer immunotherapy given their ever-increasing performance in improving diagnostic accuracy, optimizing treatment planning, predicting outcomes of care and reducing human resource costs. In this review, we present the details of AI and the current progression and state of the art in employing AI for cancer immunotherapy. Furthermore, we discuss the challenges, opportunities and corresponding strategies in applying the technology for widespread clinical deployment. Finally, we summarize the impact of AI on cancer immunotherapy and provide our perspectives about underlying applications of AI in the future.
Collapse
Key Words
- AI, artificial intelligence
- Artificial intelligence
- CT, computed tomography
- CTLA-4, cytotoxic T lymphocyte-associated antigen 4
- Cancer immunotherapy
- DL, deep learning
- Diagnostics
- ICB, immune checkpoint blockade
- MHC-I, major histocompatibility complex class I
- ML, machine learning
- MMR, mismatch repair
- MRI, magnetic resonance imaging
- Machine learning
- PD-1, programmed cell death protein 1
- PD-L1, PD-1 ligand1
- TNBC, triple-negative breast cancer
- US, ultrasonography
- irAEs, immune-related adverse events
Collapse
Affiliation(s)
- Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiang Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xinxin Ren
- Center for Molecular Medicine, Xiangya Hospital, Key Laboratory of Molecular Radiation Oncology of Hunan Province, Central South University, Changsha 410008, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
96
|
Kitamura FC, Pan I, Ferraciolli SF, Yeom KW, Abdala N. Clinical Artificial Intelligence Applications in Radiology: Neuro. Radiol Clin North Am 2021; 59:1003-1012. [PMID: 34689869 DOI: 10.1016/j.rcl.2021.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Radiologists have been at the forefront of the digitization process in medicine. Artificial intelligence (AI) is a promising area of innovation, particularly in medical imaging. The number of applications of AI in neuroradiology has also grown. This article illustrates some of these applications. This article reviews machine learning challenges related to neuroradiology. The first approval of reimbursement for an AI algorithm by the Centers for Medicare and Medicaid Services, covering a stroke software for early detection of large vessel occlusion, is also discussed.
Collapse
Affiliation(s)
- Felipe Campos Kitamura
- DasaInova, Diagnósticos da América SA (Dasa), São Paulo, São Paulo, Brazil; Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil.
| | - Ian Pan
- DasaInova, Diagnósticos da América SA (Dasa), São Paulo, São Paulo, Brazil; Brigham and Woman's Hospital, Boston, MA, USA
| | | | | | - Nitamar Abdala
- Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
97
|
Abstract
The petrous apex may be affected by a range of lesions, commonly encountered as incidental and asymptomatic findings on imaging performed for other clinical reasons. Symptoms associated with petrous apex lesions commonly relate to mass effect and/or direct involvement of closely adjacent structures. Petrous apex lesions are optimally assessed using a combination of high-resolution CT and MRI of the skull base. Management of petrous apex lesions varies widely, reflecting the range of possible pathologies, with imaging playing a key role, including lesion characterization, surveillance, surgical planning, and oncological contouring.
Collapse
Affiliation(s)
- Gillian M Potter
- Department of Neuroradiology, Manchester Centre for Clinical Neurosciences, Salford NHS Foundation Trust, Greater Manchester, England M6 8HD, UK.
| | - Rekha Siripurapu
- Department of Neuroradiology, Manchester Centre for Clinical Neurosciences, Salford NHS Foundation Trust, Greater Manchester, England M6 8HD, UK
| |
Collapse
|
98
|
Abdel Razek AAK, Alksas A, Shehata M, AbdelKhalek A, Abdel Baky K, El-Baz A, Helmy E. Clinical applications of artificial intelligence and radiomics in neuro-oncology imaging. Insights Imaging 2021; 12:152. [PMID: 34676470 PMCID: PMC8531173 DOI: 10.1186/s13244-021-01102-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/26/2021] [Indexed: 12/15/2022] Open
Abstract
This article is a comprehensive review of the basic background, technique, and clinical applications of artificial intelligence (AI) and radiomics in the field of neuro-oncology. A variety of AI and radiomics utilized conventional and advanced techniques to differentiate brain tumors from non-neoplastic lesions such as inflammatory and demyelinating brain lesions. It is used in the diagnosis of gliomas and discrimination of gliomas from lymphomas and metastasis. Also, semiautomated and automated tumor segmentation has been developed for radiotherapy planning and follow-up. It has a role in the grading, prediction of treatment response, and prognosis of gliomas. Radiogenomics allowed the connection of the imaging phenotype of the tumor to its molecular environment. In addition, AI is applied for the assessment of extra-axial brain tumors and pediatric tumors with high performance in tumor detection, classification, and stratification of patient's prognoses.
Collapse
Affiliation(s)
| | - Ahmed Alksas
- Biomaging Lab, Department of Bioengineering, University of Louisville, Louisville, KY, 40292, USA
| | - Mohamed Shehata
- Biomaging Lab, Department of Bioengineering, University of Louisville, Louisville, KY, 40292, USA
| | - Amr AbdelKhalek
- Internship at Mansoura University Hospital, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Khaled Abdel Baky
- Department of Diagnostic Radiology, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Ayman El-Baz
- Biomaging Lab, Department of Bioengineering, University of Louisville, Louisville, KY, 40292, USA
| | - Eman Helmy
- Department of Diagnostic Radiology, Faculty of Medicine, Mansoura University, Elgomheryia Street, Mansoura, 3512, Egypt.
| |
Collapse
|
99
|
Huang J, Shlobin NA, Lam SK, DeCuypere M. Artificial Intelligence Applications in Pediatric Brain Tumor Imaging: A Systematic Review. World Neurosurg 2021; 157:99-105. [PMID: 34648981 DOI: 10.1016/j.wneu.2021.10.068] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/04/2021] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Artificial intelligence (AI) has facilitated the analysis of medical imaging given increased computational capacity and medical data availability in recent years. Although many applications for AI in the imaging of brain tumors have been proposed, their potential clinical impact remains to be explored. A systematic review was performed to examine the role of AI in the analysis of pediatric brain tumor imaging. METHODS PubMed, Embase, and Scopus were searched for relevant articles up to January 27, 2021. RESULTS Literature search identified 298 records, of which 22 studies were included. The most commonly studied tumors were posterior fossa tumors including brainstem glioma, ependymoma, medulloblastoma, and pilocytic astrocytoma (15, 68%). Tumor diagnosis was the most frequently performed task (14, 64%), followed by tumor segmentation (3, 14%) and tumor detection (3, 14%). Of the 6 studies comparing AI to clinical experts, 5 demonstrated superiority of AI for tumor diagnosis. Other tasks including tumor segmentation, attenuation correction of positron emission tomography scans, image registration for patient positioning, and dose calculation for radiotherapy were performed with high accuracy comparable with clinical experts. No studies described use of the AI tool in routine clinical practice. CONCLUSIONS AI methods for analysis of pediatric brain tumor imaging have increased exponentially in recent years. However, adoption of these methods in clinical practice requires further characterization of validity and utility. Implementation of these methods may streamline clinical workflows by improving diagnostic accuracy and automating basic imaging analysis tasks.
Collapse
Affiliation(s)
- Jonathan Huang
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois, USA
| | - Nathan A Shlobin
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois, USA
| | - Sandi K Lam
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois, USA
| | - Michael DeCuypere
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois, USA.
| |
Collapse
|
100
|
Fiani B, Pasko KBD, Sarhadi K, Covarrubias C. Current uses, emerging applications, and clinical integration of artificial intelligence in neuroradiology. Rev Neurosci 2021; 33:383-395. [PMID: 34506699 DOI: 10.1515/revneuro-2021-0101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 08/18/2021] [Indexed: 11/15/2022]
Abstract
Artificial intelligence (AI) is a branch of computer science with a variety of subfields and techniques, exploited to serve as a deductive tool that performs tasks originally requiring human cognition. AI tools and its subdomains are being incorporated into healthcare delivery for the improvement of medical data interpretation encompassing clinical management, diagnostics, and prognostic outcomes. In the field of neuroradiology, AI manifested through deep machine learning and connected neural networks (CNNs) has demonstrated incredible accuracy in identifying pathology and aiding in diagnosis and prognostication in several areas of neurology and neurosurgery. In this literature review, we survey the available clinical data highlighting the utilization of AI in the field of neuroradiology across multiple neurological and neurosurgical subspecialties. In addition, we discuss the emerging role of AI in neuroradiology, its strengths and limitations, as well as future needs in strengthening its role in clinical practice. Our review evaluated data across several subspecialties of neurology and neurosurgery including vascular neurology, spinal pathology, traumatic brain injury (TBI), neuro-oncology, multiple sclerosis, Alzheimer's disease, and epilepsy. AI has established a strong presence within the realm of neuroradiology as a successful and largely supportive technology aiding in the interpretation, diagnosis, and even prognostication of various pathologies. More research is warranted to establish its full scientific validity and determine its maximum potential to aid in optimizing and providing the most accurate imaging interpretation.
Collapse
Affiliation(s)
- Brian Fiani
- Department of Neurosurgery, Desert Regional Medical Center, 1150 N Indian Canyon Dr, Palm Springs, CA, 92262, USA
| | - Kory B Dylan Pasko
- School of Medicine, Georgetown University, 3900 Reservoir Rd NW, Washington, DC, 20007, USA
| | - Kasra Sarhadi
- Department of Neurology, University of Washington, Main Hospital, 325 9th Ave, Seattle, WA, 98104, USA
| | - Claudia Covarrubias
- School of Medicine, Universidad Anáhuac Querétaro, Cto. Universidades I, Fracción 2, 76246 Qro., Querétaro, Mexico
| |
Collapse
|