351
|
Kim JN, Gomez-Perez L, Zimin VN, Makhlouf MHE, Al-Kindi S, Wilson DL, Lee J. Pericoronary adipose tissue radiomics from coronary CT angiography identifies vulnerable plaques characteristics in intravascular OCT. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.09.23284346. [PMID: 36711678 PMCID: PMC9882469 DOI: 10.1101/2023.01.09.23284346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Pericoronary adipose tissue (PCAT) features on CT have been shown to reflect local inflammation, and signals increased cardiovascular risk. Our goal was to determine if PCAT radiomics extracted from coronary CT angiography (CCTA) images are associated with intravascular optical coherence tomography (IVOCT)-identified vulnerable plaque characteristics (e.g., microchannels [MC] and thin-cap fibroatheroma [TCFA]). CCTA and IVOCT images of 30 lesions from 25 patients were registered. Vessels with vulnerable plaques were identified from the registered IVOCT images. PCAT radiomics features were extracted from CCTA images for the lesion region of interest (PCAT-LOI) and the entire vessel (PCAT-Vessel). We extracted 1356 radiomics features, including intensity (first-order), shape, and texture features. Features were reduced using standard approaches (e.g., high feature correlation). Using stratified three-fold cross-validation with 1000 repeats, we determined the ability of PCAT radiomics features from CCTA to predict IVOCT vulnerable plaque characteristics. In identification of TCFA lesions, PCAT-LOI and PCAT-Vessel radiomics models performed comparably (AUC±standard deviation 0.78±0.13, 0.77±0.14). For identification of MC lesions, PCAT-Vessel radiomics model (0.89±0.09) was moderately better associated than that of PCAT-LOI model (0.83±0.12). Both PCAT-LOI and PCAT-Vessel radiomics models also similarly identified coronary vessels thought to be highly vulnerable (i.e., both TCFA and MC) (0.88±0.10, 0.91±0.09). Favorable radiomics features tended to be those describing texture and size of PCAT. PCAT radiomics can identify coronary vessels with TCFA or MC, consistent with IVOCT. CCTA radiomics may improve risk stratification by noninvasively detecting vulnerable plaque characteristics that are only visible with IVOCT.
Collapse
|
352
|
Sansone M, Fusco R, Grassi F, Gatta G, Belfiore MP, Angelone F, Ricciardi C, Ponsiglione AM, Amato F, Galdiero R, Grassi R, Granata V, Grassi R. Machine Learning Approaches with Textural Features to Calculate Breast Density on Mammography. Curr Oncol 2023; 30:839-853. [PMID: 36661713 PMCID: PMC9858566 DOI: 10.3390/curroncol30010064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND breast cancer (BC) is the world's most prevalent cancer in the female population, with 2.3 million new cases diagnosed worldwide in 2020. The great efforts made to set screening campaigns, early detection programs, and increasingly targeted treatments led to significant improvement in patients' survival. The Full-Field Digital Mammograph (FFDM) is considered the gold standard method for the early diagnosis of BC. From several previous studies, it has emerged that breast density (BD) is a risk factor in the development of BC, affecting the periodicity of screening plans present today at an international level. OBJECTIVE in this study, the focus is the development of mammographic image processing techniques that allow the extraction of indicators derived from textural patterns of the mammary parenchyma indicative of BD risk factors. METHODS a total of 168 patients were enrolled in the internal training and test set while a total of 51 patients were enrolled to compose the external validation cohort. Different Machine Learning (ML) techniques have been employed to classify breasts based on the values of the tissue density. Textural features were extracted only from breast parenchyma with which to train classifiers, thanks to the aid of ML algorithms. RESULTS the accuracy of different tested classifiers varied between 74.15% and 93.55%. The best results were reached by a Support Vector Machine (accuracy of 93.55% and a percentage of true positives and negatives equal to TPP = 94.44% and TNP = 92.31%). The best accuracy was not influenced by the choice of the features selection approach. Considering the external validation cohort, the SVM, as the best classifier with the 7 features selected by a wrapper method, showed an accuracy of 0.95, a sensitivity of 0.96, and a specificity of 0.90. CONCLUSIONS our preliminary results showed that the Radiomics analysis and ML approach allow us to objectively identify BD.
Collapse
Affiliation(s)
- Mario Sansone
- Department of Electrical Engineering Information Technology, University of Naples Federico II, 80125 Naples, Italy
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 80013 Napoli, Italy
| | - Francesca Grassi
- Department of Precision Medicine, Division of Radiology, University of Campania Luigi Vanvitelli, 80127 Naples, Italy
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, 20122 Milan, Italy
| | - Gianluca Gatta
- Department of Precision Medicine, Division of Radiology, University of Campania Luigi Vanvitelli, 80127 Naples, Italy
| | - Maria Paola Belfiore
- Department of Precision Medicine, Division of Radiology, University of Campania Luigi Vanvitelli, 80127 Naples, Italy
| | - Francesca Angelone
- Department of Electrical Engineering Information Technology, University of Naples Federico II, 80125 Naples, Italy
| | - Carlo Ricciardi
- Department of Electrical Engineering Information Technology, University of Naples Federico II, 80125 Naples, Italy
| | - Alfonso Maria Ponsiglione
- Department of Electrical Engineering Information Technology, University of Naples Federico II, 80125 Naples, Italy
| | - Francesco Amato
- Department of Electrical Engineering Information Technology, University of Naples Federico II, 80125 Naples, Italy
| | - Roberta Galdiero
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Grassi
- Department of Precision Medicine, Division of Radiology, University of Campania Luigi Vanvitelli, 80127 Naples, Italy
| | - Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberto Grassi
- Department of Precision Medicine, Division of Radiology, University of Campania Luigi Vanvitelli, 80127 Naples, Italy
| |
Collapse
|
353
|
Posa A, Barbieri P, Mazza G, Tanzilli A, Natale L, Sala E, Iezzi R. Technological Advancements in Interventional Oncology. Diagnostics (Basel) 2023; 13:228. [PMID: 36673038 PMCID: PMC9857620 DOI: 10.3390/diagnostics13020228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023] Open
Abstract
Interventional radiology, and particularly interventional oncology, represents one of the medical subspecialties in which technological advancements and innovations play an utterly fundamental role. Artificial intelligence, consisting of big data analysis and feature extrapolation through computational algorithms for disease diagnosis and treatment response evaluation, is nowadays playing an increasingly important role in various healthcare fields and applications, from diagnosis to treatment response prediction. One of the fields which greatly benefits from artificial intelligence is interventional oncology. In addition, digital health, consisting of practical technological applications, can assist healthcare practitioners in their daily activities. This review aims to cover the most useful, established, and interesting artificial intelligence and digital health innovations and updates, to help physicians become more and more involved in their use in clinical practice, particularly in the field of interventional oncology.
Collapse
Affiliation(s)
- Alessandro Posa
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology—A. Gemelli University Hospital Foundation IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Pierluigi Barbieri
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology—A. Gemelli University Hospital Foundation IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Giulia Mazza
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology—A. Gemelli University Hospital Foundation IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Alessandro Tanzilli
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology—A. Gemelli University Hospital Foundation IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Luigi Natale
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology—A. Gemelli University Hospital Foundation IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Istituto di Radiodiagnostica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Evis Sala
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology—A. Gemelli University Hospital Foundation IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Istituto di Radiodiagnostica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Roberto Iezzi
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology—A. Gemelli University Hospital Foundation IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Istituto di Radiodiagnostica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
354
|
Miranda J, Horvat N, Fonseca GM, Araujo-Filho JDAB, Fernandes MC, Charbel C, Chakraborty J, Coelho FF, Nomura CH, Herman P. Current status and future perspectives of radiomics in hepatocellular carcinoma. World J Gastroenterol 2023; 29:43-60. [PMID: 36683711 PMCID: PMC9850949 DOI: 10.3748/wjg.v29.i1.43] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/27/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2023] Open
Abstract
Given the frequent co-existence of an aggressive tumor and underlying chronic liver disease, the management of hepatocellular carcinoma (HCC) patients requires experienced multidisciplinary team discussion. Moreover, imaging plays a key role in the diagnosis, staging, restaging, and surveillance of HCC. Currently, imaging assessment of HCC entails the assessment of qualitative characteristics which are prone to inter-reader variability. Radiomics is an emerging field that extracts high-dimensional mineable quantitative features that cannot be assessed visually with the naked eye from medical imaging. The main potential applications of radiomic models in HCC are to predict histology, response to treatment, genetic signature, recurrence, and survival. Despite the encouraging results to date, there are challenges and limitations that need to be overcome before radiomics implementation in clinical practice. The purpose of this article is to review the main concepts and challenges pertaining to radiomics, and to review recent studies and potential applications of radiomics in HCC.
Collapse
Affiliation(s)
- Joao Miranda
- Department of Radiology, University of Sao Paulo, Sao Paulo 05403-010, Brazil
| | - Natally Horvat
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | | | | | - Maria Clara Fernandes
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | - Charlotte Charbel
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | - Jayasree Chakraborty
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | | | - Cesar Higa Nomura
- Department of Radiology, University of Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Paulo Herman
- Department of Gastroenterology, University of Sao Paulo, Sao Paulo 05403-000, Brazil
| |
Collapse
|
355
|
Mahdavi MMB, Arabfard M, Rafati M, Ghanei M. A Computer-based Analysis for Identification and Quantification of Small Airway Disease in Lung Computed Tomography Images: A Comprehensive Review for Radiologists. J Thorac Imaging 2023; 38:W1-W18. [PMID: 36206107 DOI: 10.1097/rti.0000000000000683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Computed tomography (CT) imaging is being increasingly used in clinical practice for detailed characterization of lung diseases. Respiratory diseases involve various components of the lung, including the small airways. Evaluation of small airway disease on CT images is challenging as the airways cannot be visualized directly by a CT scanner. Small airway disease can manifest as pulmonary air trapping (AT). Although AT may be sometimes seen as mosaic attenuation on expiratory CT images, it is difficult to identify diffuse AT visually. Computer technology advances over the past decades have provided methods for objective quantification of small airway disease on CT images. Quantitative CT (QCT) methods are being rapidly developed to quantify underlying lung diseases with greater precision than subjective visual assessment of CT images. A growing body of evidence suggests that QCT methods can be practical tools in the clinical setting to identify and quantify abnormal regions of the lung accurately and reproducibly. This review aimed to describe the available methods for the identification and quantification of small airway disease on CT images and to discuss the challenges of implementing QCT metrics in clinical care for patients with small airway disease.
Collapse
Affiliation(s)
- Mohammad Mehdi Baradaran Mahdavi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran
| | - Masoud Arabfard
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran
| | - Mehravar Rafati
- Department of Medical Physics and Radiology, Faculty of paramedicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran
| |
Collapse
|
356
|
Ponsiglione A, Stanzione A, Spadarella G, Baran A, Cappellini LA, Lipman KG, Van Ooijen P, Cuocolo R. Ovarian imaging radiomics quality score assessment: an EuSoMII radiomics auditing group initiative. Eur Radiol 2023; 33:2239-2247. [PMID: 36303093 PMCID: PMC9935717 DOI: 10.1007/s00330-022-09180-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/26/2022] [Accepted: 09/18/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To evaluate the methodological rigor of radiomics-based studies using noninvasive imaging in ovarian setting. METHODS Multiple medical literature archives (PubMed, Web of Science, and Scopus) were searched to retrieve original studies focused on computed tomography (CT), magnetic resonance imaging (MRI), ultrasound (US), or positron emission tomography (PET) radiomics for ovarian disorders' assessment. Two researchers in consensus evaluated each investigation using the radiomics quality score (RQS). Subgroup analyses were performed to assess whether the total RQS varied according to first author category, study aim and topic, imaging modality, and journal quartile. RESULTS From a total of 531 items, 63 investigations were finally included in the analysis. The studies were greatly focused (94%) on the field of oncology, with CT representing the most used imaging technique (41%). Overall, the papers achieved a median total RQS 6 (IQR, -0.5 to 11), corresponding to a percentage of 16.7% of the maximum score (IQR, 0-30.6%). The scoring was low especially due to the lack of prospective design and formal validation of the results. At subgroup analysis, the 4 studies not focused on oncological topic showed significantly lower quality scores than the others. CONCLUSIONS The overall methodological rigor of radiomics studies in the ovarian field is still not ideal, limiting the reproducibility of results and potential translation to clinical setting. More efforts towards a standardized methodology in the workflow are needed to allow radiomics to become a viable tool for clinical decision-making. KEY POINTS • The 63 included studies using noninvasive imaging for ovarian applications were mostly focused on oncologic topic (94%). • The included investigations achieved a median total RQS 6 (IQR, -0.5 to 11), indicating poor methodological rigor. • The RQS was low especially due to the lack of prospective design and formal validation of the results.
Collapse
Affiliation(s)
- Andrea Ponsiglione
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy.
| | - Gaia Spadarella
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Agah Baran
- Department of Diagnostic and Interventional Radiology, University of Cologne, Cologne, Germany
| | | | - Kevin Groot Lipman
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Peter Van Ooijen
- Department of Radiation Oncology, University Medical Center Groningen, Groningen, The Netherlands
- Machine Learning Lab, Data Science Center in Health, University Medical Center Groningen, Groningen, the Netherlands
| | - Renato Cuocolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
- Augmented Reality for Health Monitoring Laboratory (ARHeMLab), Department of Electrical Engineering and Information Technology, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
357
|
Multiparameter single-cell proteomic technologies give new insights into the biology of ovarian tumors. Semin Immunopathol 2023; 45:43-59. [PMID: 36635516 PMCID: PMC9974728 DOI: 10.1007/s00281-022-00979-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/11/2022] [Indexed: 01/13/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most lethal gynecological malignancy. Its diagnosis at advanced stage compounded with its excessive genomic and cellular heterogeneity make curative treatment challenging. Two critical therapeutic challenges to overcome are carboplatin resistance and lack of response to immunotherapy. Carboplatin resistance results from diverse cell autonomous mechanisms which operate in different combinations within and across tumors. The lack of response to immunotherapy is highly likely to be related to an immunosuppressive HGSOC tumor microenvironment which overrides any clinical benefit. Results from a number of studies, mainly using transcriptomics, indicate that the immune tumor microenvironment (iTME) plays a role in carboplatin response. However, in patients receiving treatment, the exact mechanistic details are unclear. During the past decade, multiplex single-cell proteomic technologies have come to the forefront of biomedical research. Mass cytometry or cytometry by time-of-flight, measures up to 60 parameters in single cells that are in suspension. Multiplex cellular imaging technologies allow simultaneous measurement of up to 60 proteins in single cells with spatial resolution and interrogation of cell-cell interactions. This review suggests that functional interplay between cell autonomous responses to carboplatin and the HGSOC immune tumor microenvironment could be clarified through the application of multiplex single-cell proteomic technologies. We conclude that for better clinical care, multiplex single-cell proteomic technologies could be an integral component of multimodal biomarker development that also includes genomics and radiomics. Collection of matched samples from patients before and on treatment will be critical to the success of these efforts.
Collapse
|
358
|
Khalid F, Goya-Outi J, Escobar T, Dangouloff-Ros V, Grigis A, Philippe C, Boddaert N, Grill J, Frouin V, Frouin F. Multimodal MRI radiomic models to predict genomic mutations in diffuse intrinsic pontine glioma with missing imaging modalities. Front Med (Lausanne) 2023; 10:1071447. [PMID: 36910474 PMCID: PMC9995801 DOI: 10.3389/fmed.2023.1071447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Purpose Predicting H3.1, TP53, and ACVR1 mutations in DIPG could aid in the selection of therapeutic options. The contribution of clinical data and multi-modal MRI were studied for these three predictive tasks. To keep the maximum number of subjects, which is essential for a rare disease, missing data were considered. A multi-modal model was proposed, collecting all available data for each patient, without performing any imputation. Methods A retrospective cohort of 80 patients with confirmed DIPG and at least one of the four MR modalities (T1w, T1c, T2w, and FLAIR), acquired with two different MR scanners was built. A pipeline including standardization of MR data and extraction of radiomic features within the tumor was applied. The values of radiomic features between the two MR scanners were realigned using the ComBat method. For each prediction task, the most robust features were selected based on a recursive feature elimination with cross-validation. Five different models, one based on clinical data and one per MR modality, were developed using logistic regression classifiers. The prediction of the multi-modal model was defined as the average of all possible prediction results among five for each patient. The performances of the models were compared using a leave-one-out approach. Results The percentage of missing modalities ranged from 6 to 11% across modalities and tasks. The performance of each individual model was dependent on each specific task, with an AUC of the ROC curve ranging from 0.63 to 0.80. The multi-modal model outperformed the clinical model for each prediction tasks, thus demonstrating the added value of MRI. Furthermore, regardless of performance criteria, the multi-modal model came in the first place or second place (very close to first). In the leave-one-out approach, the prediction of H3.1 (resp. ACVR1 and TP53) mutations achieved a balanced accuracy of 87.8% (resp. 82.1 and 78.3%). Conclusion Compared with a single modality approach, the multi-modal model combining multiple MRI modalities and clinical features was the most powerful to predict H3.1, ACVR1, and TP53 mutations and provided prediction, even in the case of missing modality. It could be proposed in the absence of a conclusive biopsy.
Collapse
Affiliation(s)
- Fahad Khalid
- Laboratoire d'Imagerie Translationnelle en Oncologie (LITO)-U1288, Institut Curie, Inserm, Université Paris-Saclay, Orsay, France
| | - Jessica Goya-Outi
- Laboratoire d'Imagerie Translationnelle en Oncologie (LITO)-U1288, Institut Curie, Inserm, Université Paris-Saclay, Orsay, France
| | - Thibault Escobar
- Laboratoire d'Imagerie Translationnelle en Oncologie (LITO)-U1288, Institut Curie, Inserm, Université Paris-Saclay, Orsay, France.,DOSIsoft SA, Cachan, France
| | - Volodia Dangouloff-Ros
- Department of Paediatric Radiology, Hôpital Universitaire Necker Enfants Malades, Paris, France.,Institut Imagine, Inserm U1163 and U1299, Université Paris Cité, Paris, France
| | | | | | - Nathalie Boddaert
- Department of Paediatric Radiology, Hôpital Universitaire Necker Enfants Malades, Paris, France.,Institut Imagine, Inserm U1163 and U1299, Université Paris Cité, Paris, France
| | - Jacques Grill
- Département Cancérologie de l'enfant et de l'adolescent, Gustave-Roussy, Villejuif, France.,Prédicteurs moléculaires et nouvelles cibles en oncologie-U981, Inserm, Université Paris-Saclay, Villejuif, France
| | | | - Frédérique Frouin
- Laboratoire d'Imagerie Translationnelle en Oncologie (LITO)-U1288, Institut Curie, Inserm, Université Paris-Saclay, Orsay, France
| |
Collapse
|
359
|
Niu J, Tan Q, Zou X, Jin S. Accurate prediction of glioma grades from radiomics using a multi-filter and multi-objective-based method. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:2890-2907. [PMID: 36899563 DOI: 10.3934/mbe.2023136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Radiomics, providing quantitative data extracted from medical images, has emerged as a critical role in diagnosis and classification of diseases such as glioma. One main challenge is how to uncover key disease-relevant features from the large amount of extracted quantitative features. Many existing methods suffer from low accuracy or overfitting. We propose a new method, Multiple-Filter and Multi-Objective-based method (MFMO), to identify predictive and robust biomarkers for disease diagnosis and classification. This method combines a multi-filter feature extraction with a multi-objective optimization-based feature selection model, which identifies a small set of predictive radiomic biomarkers with less redundancy. Taking magnetic resonance imaging (MRI) images-based glioma grading as a case study, we identify 10 key radiomic biomarkers that can accurately distinguish low-grade glioma (LGG) from high-grade glioma (HGG) on both training and test datasets. Using these 10 signature features, the classification model reaches training Area Under the receiving operating characteristic Curve (AUC) of 0.96 and test AUC of 0.95, which shows superior performance over existing methods and previously identified biomarkers.
Collapse
Affiliation(s)
- Jingren Niu
- School of Mathematics and Statistics, Wuhan University, Wuhan 430072, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan 430072, China
| | - Qing Tan
- School of Mathematics and Statistics, Wuhan University, Wuhan 430072, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan 430072, China
| | - Xiufen Zou
- School of Mathematics and Statistics, Wuhan University, Wuhan 430072, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan 430072, China
| | - Suoqin Jin
- School of Mathematics and Statistics, Wuhan University, Wuhan 430072, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan 430072, China
| |
Collapse
|
360
|
Sminia P, Guipaud O, Viktorsson K, Ahire V, Baatout S, Boterberg T, Cizkova J, Dostál M, Fernandez-Palomo C, Filipova A, François A, Geiger M, Hunter A, Jassim H, Edin NFJ, Jordan K, Koniarová I, Selvaraj VK, Meade AD, Milliat F, Montoro A, Politis C, Savu D, Sémont A, Tichy A, Válek V, Vogin G. Clinical Radiobiology for Radiation Oncology. RADIOBIOLOGY TEXTBOOK 2023:237-309. [DOI: 10.1007/978-3-031-18810-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
AbstractThis chapter is focused on radiobiological aspects at the molecular, cellular, and tissue level which are relevant for the clinical use of ionizing radiation (IR) in cancer therapy. For radiation oncology, it is critical to find a balance, i.e., the therapeutic window, between the probability of tumor control and the probability of side effects caused by radiation injury to the healthy tissues and organs. An overview is given about modern precision radiotherapy (RT) techniques, which allow optimal sparing of healthy tissues. Biological factors determining the width of the therapeutic window are explained. The role of the six typical radiobiological phenomena determining the response of both malignant and normal tissues in the clinic, the 6R’s, which are Reoxygenation, Redistribution, Repopulation, Repair, Radiosensitivity, and Reactivation of the immune system, is discussed. Information is provided on tumor characteristics, for example, tumor type, growth kinetics, hypoxia, aberrant molecular signaling pathways, cancer stem cells and their impact on the response to RT. The role of the tumor microenvironment and microbiota is described and the effects of radiation on the immune system including the abscopal effect phenomenon are outlined. A summary is given on tumor diagnosis, response prediction via biomarkers, genetics, and radiomics, and ways to selectively enhance the RT response in tumors. Furthermore, we describe acute and late normal tissue reactions following exposure to radiation: cellular aspects, tissue kinetics, latency periods, permanent or transient injury, and histopathology. Details are also given on the differential effect on tumor and late responding healthy tissues following fractionated and low dose rate irradiation as well as the effect of whole-body exposure.
Collapse
|
361
|
Yu K, Ying J, Zhao T, Lei L, Zhong L, Hu J, Zhou JW, Huang C, Zhang X. Prediction model for knee osteoarthritis using magnetic resonance-based radiomic features from the infrapatellar fat pad: data from the osteoarthritis initiative. Quant Imaging Med Surg 2023; 13:352-369. [PMID: 36620171 PMCID: PMC9816749 DOI: 10.21037/qims-22-368] [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: 04/16/2022] [Accepted: 10/31/2022] [Indexed: 11/21/2022]
Abstract
Background The infrapatellar fat pad (IPFP) plays an important role in the incidence of knee osteoarthritis (OA). Magnetic resonance (MR) signal heterogeneity of the IPFP is related to pathologic changes. In this study, we aimed to investigate whether the IPFP radiomic features have predictive value for incident radiographic knee OA (iROA) 1 year prior to iROA diagnosis. Methods Data used in this work were obtained from the osteoarthritis initiative (OAI). In this study, iROA was defined as a knee with a baseline Kellgren-Lawrence grade (KLG) of 0 or 1 that further progressed to KLG ≥2 during the follow-up visit. Intermediate-weighted turbo spin-echo knee MR images at the time of iROA diagnosis and 1 year prior were obtained. Five clinical characteristics-age, sex, body mass index, knee injury history, and knee surgery history-were obtained. A total of 604 knees were selected and matched (302 cases and 302 controls). A U-Net segmentation model was independently trained to automatically segment the IPFP. The prediction models were established in the training set (60%). Three main models were generated using (I) clinical characteristics; (II) radiomic features; (III) combined (clinical plus radiomic) features. Model performance was evaluated in an independent testing set (remaining 40%) using the area under the curve (AUC). Two secondary models were also generated using Hoffa-synovitis scores and clinical characteristics. Results The comparison between the automated and manual segmentations of the IPFP achieved a Dice coefficient of 0.900 (95% CI: 0.891-0.908), which was comparable to that of experienced radiologists. The radiomic features model and the combined model yielded superior AUCs of 0.700 (95% CI: 0.630-0.763) and 0.702 (95% CI: 0.635-0.763), respectively. The DeLong test found no statistically significant difference between the receiver operating curves of the radiomic and combined models (P=0.831); however, both models outperformed the clinical model (P=0.014 and 0.004, respectively). Conclusions Our results demonstrated that radiomic features of the IPFP are predictive of iROA 1 year prior to the diagnosis, suggesting that IPFP radiomic features can serve as an early quantitative prediction biomarker of iROA.
Collapse
Affiliation(s)
- Keyan Yu
- Department of Medical Imaging, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Guangzhou, China;,Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jia Ying
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Tianyun Zhao
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Lan Lei
- Program in Public Health, Stony Brook Medicine, Stony Brook, NY, USA;,Department of Medicine, Northside Hospital Gwinnett, Lawrenceville, GA, USA
| | - Lijie Zhong
- Department of Medical Imaging, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Guangzhou, China
| | - Jiaping Hu
- Department of Medical Imaging, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Guangzhou, China
| | - Juin W. Zhou
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Chuan Huang
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA;,Department of Radiology, Stony Brook Medicine, Stony Brook, NY, USA;,Department of Psychiatry, Stony Brook Medicine, Stony Brook, NY, USA
| | - Xiaodong Zhang
- Department of Medical Imaging, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Guangzhou, China
| |
Collapse
|
362
|
Quan MY, Huang YX, Wang CY, Zhang Q, Chang C, Zhou SC. Deep learning radiomics model based on breast ultrasound video to predict HER2 expression status. Front Endocrinol (Lausanne) 2023; 14:1144812. [PMID: 37143737 PMCID: PMC10153672 DOI: 10.3389/fendo.2023.1144812] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Purpose The detection of human epidermal growth factor receptor 2 (HER2) expression status is essential to determining the chemotherapy regimen for breast cancer patients and to improving their prognosis. We developed a deep learning radiomics (DLR) model combining time-frequency domain features of ultrasound (US) video of breast lesions with clinical parameters for predicting HER2 expression status. Patients and Methods Data for this research was obtained from 807 breast cancer patients who visited from February 2019 to July 2020. Ultimately, 445 patients were included in the study. Pre-operative breast ultrasound examination videos were collected and split into a training set and a test set. Building a training set of DLR models combining time-frequency domain features and clinical features of ultrasound video of breast lesions based on the training set data to predict HER2 expression status. Test the performance of the model using test set data. The final models integrated with different classifiers are compared, and the best performing model is finally selected. Results The best diagnostic performance in predicting HER2 expression status is provided by an Extreme Gradient Boosting (XGBoost)-based time-frequency domain feature classifier combined with a logistic regression (LR)-based clinical parameter classifier of clinical parameters combined DLR, particularly with a high specificity of 0.917. The area under the receiver operating characteristic curve (AUC) for the test cohort was 0.810. Conclusion Our study provides a non-invasive imaging biomarker to predict HER2 expression status in breast cancer patients.
Collapse
Affiliation(s)
- Meng-Yao Quan
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun-Xia Huang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chang-Yan Wang
- Laboratory of The Smart Medicine and AI-based Radiology Technology (SMART), School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Qi Zhang
- Laboratory of The Smart Medicine and AI-based Radiology Technology (SMART), School of Communication and Information Engineering, Shanghai University, Shanghai, China
- *Correspondence: Shi-Chong Zhou, ; Qi Zhang,
| | - Cai Chang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Chong Zhou
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Shi-Chong Zhou, ; Qi Zhang,
| |
Collapse
|
363
|
Colelli G, Barzaghi L, Paoletti M, Monforte M, Bergsland N, Manco G, Deligianni X, Santini F, Ricci E, Tasca G, Mira A, Figini S, Pichiecchio A. Radiomics and machine learning applied to STIR sequence for prediction of quantitative parameters in facioscapulohumeral disease. Front Neurol 2023; 14:1105276. [PMID: 36908599 PMCID: PMC9999017 DOI: 10.3389/fneur.2023.1105276] [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: 11/22/2022] [Accepted: 01/30/2023] [Indexed: 03/14/2023] Open
Abstract
Purpose Quantitative Muscle MRI (qMRI) is a valuable and non-invasive tool to assess disease involvement and progression in neuromuscular disorders being able to detect even subtle changes in muscle pathology. The aim of this study is to evaluate the feasibility of using a conventional short-tau inversion recovery (STIR) sequence to predict fat fraction (FF) and water T2 (wT2) in skeletal muscle introducing a radiomic workflow with standardized feature extraction combined with machine learning algorithms. Methods Twenty-five patients with facioscapulohumeral muscular dystrophy (FSHD) were scanned at calf level using conventional STIR sequence and qMRI techniques. We applied and compared three different radiomics workflows (WF1, WF2, WF3), combined with seven Machine Learning regression algorithms (linear, ridge and lasso regression, tree, random forest, k-nearest neighbor and support vector machine), on conventional STIR images to predict FF and wT2 for six calf muscles. Results The combination of WF3 and K-nearest neighbor resulted to be the best predictor model of qMRI parameters with a mean absolute error about ± 5 pp for FF and ± 1.8 ms for wT2. Conclusion This pilot study demonstrated the possibility to predict qMRI parameters in a cohort of FSHD subjects starting from conventional STIR sequence.
Collapse
Affiliation(s)
- Giulia Colelli
- Department of Mathematics, University of Pavia, Pavia, Italy.,Neuroradiology Department, Advanced Imaging and Radiomics Center, IRCCS Mondino Foundation, Pavia, Italy.,INFN, Group of Pavia, Pavia, Italy
| | - Leonardo Barzaghi
- Department of Mathematics, University of Pavia, Pavia, Italy.,Neuroradiology Department, Advanced Imaging and Radiomics Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Matteo Paoletti
- Neuroradiology Department, Advanced Imaging and Radiomics Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Mauro Monforte
- UOC di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Niels Bergsland
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo Neuroimaging Analysis Center, University of Buffalo, The State University of New York, Buffalo, NY, United States.,IRCCS, Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Giulia Manco
- Neuroradiology Department, Advanced Imaging and Radiomics Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Xeni Deligianni
- Department of Radiology, University Hospital Basel, Basel, Switzerland.,Basel Muscle MRI, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Francesco Santini
- Department of Radiology, University Hospital Basel, Basel, Switzerland.,Basel Muscle MRI, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Enzo Ricci
- UOC di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giorgio Tasca
- UOC di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle upon Tyne, United Kingdom
| | - Antonietta Mira
- Data Science Lab, Università della Svizzera italiana, Lugano, Switzerland.,Department of Science and High Technology, University of Insubria, Como, Italy
| | - Silvia Figini
- Department of Political and Social Sciences, University of Pavia, Pavia, Italy.,BioData Science Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Anna Pichiecchio
- Neuroradiology Department, Advanced Imaging and Radiomics Center, IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
364
|
Kotowski K, Kucharski D, Machura B, Adamski S, Gutierrez Becker B, Krason A, Zarudzki L, Tessier J, Nalepa J. Detecting liver cirrhosis in computed tomography scans using clinically-inspired and radiomic features. Comput Biol Med 2023; 152:106378. [PMID: 36512877 DOI: 10.1016/j.compbiomed.2022.106378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 11/30/2022]
Abstract
Hepatic cirrhosis is an increasing cause of mortality in developed countries-it is the pathological sequela of chronic liver diseases, and the final liver fibrosis stage. Since cirrhosis evolves from the asymptomatic phase, it is of paramount importance to detect it as quickly as possible, because entering the symptomatic phase commonly leads to hospitalization and can be fatal. Understanding the state of the liver based on the abdominal computed tomography (CT) scans is tedious, user-dependent and lacks reproducibility. We tackle these issues and propose an end-to-end and reproducible approach for detecting cirrhosis from CT. It benefits from the introduced clinically-inspired features that reflect the patient's characteristics which are often investigated by experienced radiologists during the screening process. Such features are coupled with the radiomic ones extracted from the liver, and from the suggested region of interest which captures the liver's boundary. The rigorous experiments, performed over two heterogeneous clinical datasets (two cohorts of 241 and 32 patients) revealed that extracting radiomic features from the liver's rectified contour is pivotal to enhance the classification abilities of the supervised learners. Also, capturing clinically-inspired image features significantly improved the performance of such models, and the proposed features were consistently selected as the important ones. Finally, we showed that selecting the most discriminative features leads to the Pareto-optimal models with enhanced feature-level interpretability, as the number of features was dramatically reduced (280×) from thousands to tens.
Collapse
Affiliation(s)
| | | | | | | | - Benjamín Gutierrez Becker
- Roche Pharma Research and Early Development, Informatics, Roche Innovation Center Basel, Basel, Switzerland
| | - Agata Krason
- Roche Pharmaceutical Research and Early Development, Early Clinical Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | - Lukasz Zarudzki
- Department of Radiology and Diagnostic Imaging, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Jean Tessier
- Roche Pharmaceutical Research and Early Development, Early Clinical Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | - Jakub Nalepa
- Graylight Imaging, Gliwice, Poland; Department of Algorithmics and Software, Silesian University of Technology, Gliwice, Poland.
| |
Collapse
|
365
|
Hegazi TM, AlSharydah AM, Alfawaz I, Al-Muhanna AF, Faisal SY. The Impact of Data Management on the Achievable Dose and Efficiency of Mammography and Radiography During the COVID-19 Era: A Facility-Based Cohort Study. Risk Manag Healthc Policy 2023; 16:401-414. [PMID: 36941927 PMCID: PMC10024472 DOI: 10.2147/rmhp.s389960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/05/2023] [Indexed: 03/15/2023] Open
Abstract
Purpose To evaluate the impact of using computational data management resources and analytical software on radiation doses in mammography and radiography during the COVID-19 pandemic, develop departmental diagnostic reference levels (DRLs), and describe achievable doses (ADs) for mammography and radiography based on measured dose parameters. Patients and Methods This ambispective cohort study enrolled 795 and 12,115 patients who underwent mammography and radiography, respectively, at the King Fahd Hospital of the University, Al-Khobar City, Saudi Arabia between May 25 and November 4, 2021. Demographic data were acquired from patients' electronic medical charts. Data on mammographic and radiographic dose determinants were acquired from the data management software. Based on the time when the data management software was operational in the institute, the study was divided into the pre-implementation and post-implementation phases. Continuous and categorical variables were compared between the two phases using an unpaired t-test and the chi-square test. Results The median accumulated average glandular dose (AGD; a mammographic dose determinant) in the post-implementation phase was three-fold higher than that in the pre-implementation phase. The average mammographic exposure time in the post-implementation phase was 16.3 ms shorter than that in the pre-implementation phase. Furthermore, the median values of the dose area product ([DAP], a radiographic dose determinant) were 9.72 and 19.4 cGycm2 in the pre-implementation and post-implementation phases, respectively. Conclusion Although the data management software used in this study helped reduce the radiation exposure time by 16.3 ms in mammography, its impact on the mean accumulated AGD was unfavorable. Similarly, radiographic exposure indices, including DAP, tube voltage, tube current, and exposure time, were not significantly different after the data management software was implemented. Close monitoring of patient radiation doses in mammography and radiography, and dose reduction will become possible if imaging facilities use DRLs and ADs via automated systems.
Collapse
Affiliation(s)
- Tarek Mohammed Hegazi
- Diagnostic and Interventional Radiology Department, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Al-Khobar City, Eastern Province, Saudi Arabia
- Correspondence: Tarek Mohammed Hegazi, Chairperson of the Radiology Department, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Khobar City, Eastern Province, Saudi Arabia, Tel +966-0138966877 (EXT: 2007), Email
| | - Abdulaziz Mohammad AlSharydah
- Diagnostic and Interventional Radiology Department, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Al-Khobar City, Eastern Province, Saudi Arabia
| | - Iba Alfawaz
- Diagnostic and Interventional Radiology Department, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Al-Khobar City, Eastern Province, Saudi Arabia
| | - Afnan Fahad Al-Muhanna
- Diagnostic and Interventional Radiology Department, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Al-Khobar City, Eastern Province, Saudi Arabia
| | - Sarah Yousef Faisal
- Diagnostic and Interventional Radiology Department, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Al-Khobar City, Eastern Province, Saudi Arabia
| |
Collapse
|
366
|
Santos GNM, da Silva HEC, Ossege FEL, Figueiredo PTDS, Melo NDS, Stefani CM, Leite AF. Radiomics in bone pathology of the jaws. Dentomaxillofac Radiol 2023; 52:20220225. [PMID: 36416666 PMCID: PMC9793454 DOI: 10.1259/dmfr.20220225] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/02/2022] [Accepted: 10/02/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE To define which are and how the radiomics features of jawbone pathologies are extracted for diagnosis, predicting prognosis and therapeutic response. METHODS A comprehensive literature search was conducted using eight databases and gray literature. Two independent observers rated these articles according to exclusion and inclusion criteria. 23 papers were included to assess the radiomics features related to jawbone pathologies. Included studies were evaluated by using JBI Critical Appraisal Checklist for Analytical Cross-Sectional Studies. RESULTS Agnostic features were mined from periapical, dental panoramic radiographs, cone beam CT, CT and MRI images of six different jawbone alterations. The most frequent features mined were texture-, shape- and intensity-based features. Only 13 studies described the machine learning step, and the best results were obtained with Support Vector Machine and random forest classifier. For osteoporosis diagnosis and classification, filtering, shape-based and Tamura texture features showed the best performance. For temporomandibular joint pathology, gray-level co-occurrence matrix (GLCM), gray level run length matrix (GLRLM), Gray Level Size Zone Matrix (GLSZM), first-order statistics analysis and shape-based analysis showed the best results. Considering odontogenic and non-odontogenic cysts and tumors, contourlet and SPHARM features, first-order statistical features, GLRLM, GLCM had better indexes. For odontogenic cysts and granulomas, first-order statistical analysis showed better classification results. CONCLUSIONS GLCM was the most frequent feature, followed by first-order statistics, and GLRLM features. No study reported predicting response, prognosis or therapeutic response, but instead diseases diagnosis or classification. Although the lack of standardization in the radiomics workflow of the included studies, texture analysis showed potential to contribute to radiologists' reports, decreasing the subjectivity and leading to personalized healthcare.
Collapse
Affiliation(s)
| | | | | | | | - Nilce de Santos Melo
- Dentistry Department, Faculty of Health Science, University of Brasília, Brasilia, Brazil
| | - Cristine Miron Stefani
- Dentistry Department, Faculty of Health Science, University of Brasília, Brasilia, Brazil
| | - André Ferreira Leite
- Dentistry Department, Faculty of Health Science, University of Brasília, Brasilia, Brazil
| |
Collapse
|
367
|
Radiomics and Deep Learning for Disease Detection in Musculoskeletal Radiology: An Overview of Novel MRI- and CT-Based Approaches. Invest Radiol 2023; 58:3-13. [PMID: 36070548 DOI: 10.1097/rli.0000000000000907] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
ABSTRACT Radiomics and machine learning-based methods offer exciting opportunities for improving diagnostic performance and efficiency in musculoskeletal radiology for various tasks, including acute injuries, chronic conditions, spinal abnormalities, and neoplasms. While early radiomics-based methods were often limited to a smaller number of higher-order image feature extractions, applying machine learning-based analytic models, multifactorial correlations, and classifiers now permits big data processing and testing thousands of features to identify relevant markers. A growing number of novel deep learning-based methods describe magnetic resonance imaging- and computed tomography-based algorithms for diagnosing anterior cruciate ligament tears, meniscus tears, articular cartilage defects, rotator cuff tears, fractures, metastatic skeletal disease, and soft tissue tumors. Initial radiomics and deep learning techniques have focused on binary detection tasks, such as determining the presence or absence of a single abnormality and differentiation of benign versus malignant. Newer-generation algorithms aim to include practically relevant multiclass characterization of detected abnormalities, such as typing and malignancy grading of neoplasms. So-called delta-radiomics assess tumor features before and after treatment, with temporal changes of radiomics features serving as surrogate markers for tumor responses to treatment. New approaches also predict treatment success rates, surgical resection completeness, and recurrence risk. Practice-relevant goals for the next generation of algorithms include diagnostic whole-organ and advanced classification capabilities. Important research objectives to fill current knowledge gaps include well-designed research studies to understand how diagnostic performances and suggested efficiency gains of isolated research settings translate into routine daily clinical practice. This article summarizes current radiomics- and machine learning-based magnetic resonance imaging and computed tomography approaches for musculoskeletal disease detection and offers a perspective on future goals and objectives.
Collapse
|
368
|
Georgiadou E, Bougias H, Leandrou S, Stogiannos N. Radiomics for Alzheimer's Disease: Fundamental Principles and Clinical Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1424:297-311. [PMID: 37486507 DOI: 10.1007/978-3-031-31982-2_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Alzheimer's disease is a neurodegenerative disease with a huge impact on people's quality of life, life expectancy, and morbidity. The ongoing prevalence of the disease, in conjunction with an increased financial burden to healthcare services, necessitates the development of new technologies to be employed in this field. Hence, advanced computational methods have been developed to facilitate early and accurate diagnosis of the disease and improve all health outcomes. Artificial intelligence is now deeply involved in the fight against this disease, with many clinical applications in the field of medical imaging. Deep learning approaches have been tested for use in this domain, while radiomics, an emerging quantitative method, are already being evaluated to be used in various medical imaging modalities. This chapter aims to provide an insight into the fundamental principles behind radiomics, discuss the most common techniques alongside their strengths and weaknesses, and suggest ways forward for future research standardization and reproducibility.
Collapse
Affiliation(s)
- Eleni Georgiadou
- Department of Radiology, Metaxa Anticancer Hospital, Piraeus, Greece
| | - Haralabos Bougias
- Department of Clinical Radiology, University Hospital of Ioannina, Ioannina, Greece
| | - Stephanos Leandrou
- Department of Health Sciences, School of Sciences, European University Cyprus, Engomi, Cyprus
| | - Nikolaos Stogiannos
- Discipline of Medical Imaging and Radiation Therapy, University College Cork, Cork, Ireland.
- Division of Midwifery & Radiography, City, University of London, London, UK.
- Medical Imaging Department, Corfu General Hospital, Corfu, Greece.
| |
Collapse
|
369
|
Lin Z, Wang T, Li H, Xiao M, Ma X, Gu Y, Qiang J. Magnetic resonance-based radiomics nomogram for predicting microsatellite instability status in endometrial cancer. Quant Imaging Med Surg 2023; 13:108-120. [PMID: 36620141 PMCID: PMC9816750 DOI: 10.21037/qims-22-255] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 09/05/2022] [Indexed: 11/30/2022]
Abstract
Background Microsatellite instability (MSI) status is an important indicator for screening patients with endometrial cancer (EC) who have potential Lynch syndrome (LS) and may benefit from immunotherapy. This study aimed to develop a magnetic resonance imaging (MRI)-based radiomics nomogram for the prediction of MSI status in EC. Methods A total of 296 patients with histopathologically diagnosed EC were enrolled, and their MSI status was determined using immunohistochemical (IHC) analysis. Patients were randomly divided into the training cohort (n=236) and the validation cohort (n=60) at a ratio of 8:2. To predict the MSI status in EC, the tumor radiomics features were extracted from T2-weighted images and contrast-enhanced T1-weighted images, which in turn were selected using one-way analysis of variance (ANOVA) and the least absolute shrinkage and selection operator (LASSO) algorithm to build the radiomics signature (radiomics score; radscore) model. Five clinicopathologic characteristics were used to construct a clinicopathologic model. Finally, the nomogram model combining radscore and clinicopathologic characteristics was constructed. The performance of the three models was evaluated using receiver operating characteristic (ROC), calibration, and decision curve analyses (DCA). Results Totals of 21 radiomics features and five clinicopathologic characteristics were selected to develop the radscore and clinicopathological models. The radscore and clinicopathologic models achieved an area under the curve (AUC) of 0.752 and 0.600, respectively, in the training cohort; and of 0.723 and 0.615, respectively, in the validation cohort. The radiomics nomogram model showed improved discrimination efficiency compared with the radscore and clinicopathologic models, with an AUC of 0.773 and 0.740 in the training and validation cohorts, respectively. The calibration curve analysis and DCA showed favorable calibration and clinical utility of the nomogram model. Conclusions The nomogram incorporating MRI-based radiomics features and clinicopathologic characteristics could be a potential tool for the prediction of MSI status in EC.
Collapse
Affiliation(s)
- Zijing Lin
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ting Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Haiming Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Meiling Xiao
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiaoliang Ma
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinwei Qiang
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
370
|
Choi YJ, Jeon KJ, Lee A, Han SS, Lee C. Harmonization of robust radiomic features in the submandibular gland using multi-ultrasound systems: a preliminary study. Dentomaxillofac Radiol 2023; 52:20220284. [PMID: 36341993 PMCID: PMC9974233 DOI: 10.1259/dmfr.20220284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE This study aimed to identify robust radiomic features in multiultrasonography of the submandibular gland and normalize the interdevice discrepancies by applying a machine-learning-based harmonization method. METHODS Ultrasonographic images of normal submandibular gland of young healthy adults, aged between 20 and 40 years, were selected from two different devices. In a total of 30 images, the region of interest was determined along the border of gland parenchyma, and 103 radiomic features were extracted using A-VIEW. The coefficient of variation (CV) was obtained for individual features, and the features showing CV less than 10% were selected. For the selected features, the interdevice discrepancy was normalized using machine-learning method, called the ComBat harmonization. Median differences of the features between the two scanners, before and after harmonization, were compared using Mann-Whitney U-test; confidence interval of 95%. RESULTS Among total 103 radiomic features, 17 features were selected as robust, showing CV less than 10% in both scanners. All values of selected features, except two, showed a statistical difference between the two devices. After applying the ComBat harmonization method, the median and distribution of the 16 features were harmonized to show no significant difference between the two scanners (p > 0.05). One feature remained different (p ≤ 0.05). CONCLUSION On ultrasonographic examination, robust radiomic features for normal submandibular gland were obtained and interdevice normalization was efficiently conducted using ComBat harmonization. Our findings would be useful for multidevices or multicenter studies based on clinical ultrasonographic imaging data to improve the accuracy of the overall diagnostic model.
Collapse
Affiliation(s)
- Yoon Joo Choi
- Department of Oral and Maxillofacial Radiology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Kug Jin Jeon
- Department of Oral and Maxillofacial Radiology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Ari Lee
- Department of Oral and Maxillofacial Radiology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Sang-Sun Han
- Department of Oral and Maxillofacial Radiology, Yonsei University College of Dentistry, Seoul, South Korea
| | | |
Collapse
|
371
|
Li S, Zhou B. A review of radiomics and genomics applications in cancers: the way towards precision medicine. Radiat Oncol 2022; 17:217. [PMID: 36585716 PMCID: PMC9801589 DOI: 10.1186/s13014-022-02192-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
The application of radiogenomics in oncology has great prospects in precision medicine. Radiogenomics combines large volumes of radiomic features from medical digital images, genetic data from high-throughput sequencing, and clinical-epidemiological data into mathematical modelling. The amalgamation of radiomics and genomics provides an approach to better study the molecular mechanism of tumour pathogenesis, as well as new evidence-supporting strategies to identify the characteristics of cancer patients, make clinical decisions by predicting prognosis, and improve the development of individualized treatment guidance. In this review, we summarized recent research on radiogenomics applications in solid cancers and presented the challenges impeding the adoption of radiomics in clinical practice. More standard guidelines are required to normalize radiomics into reproducible and convincible analyses and develop it as a mature field.
Collapse
Affiliation(s)
- Simin Li
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Hospital of China Medical University, Shenyang, 110001 Liaoning People’s Republic of China
| | - Baosen Zhou
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Hospital of China Medical University, Shenyang, 110001 Liaoning People’s Republic of China
| |
Collapse
|
372
|
Hung KF, Ai QYH, Wong LM, Yeung AWK, Li DTS, Leung YY. Current Applications of Deep Learning and Radiomics on CT and CBCT for Maxillofacial Diseases. Diagnostics (Basel) 2022; 13:110. [PMID: 36611402 PMCID: PMC9818323 DOI: 10.3390/diagnostics13010110] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
The increasing use of computed tomography (CT) and cone beam computed tomography (CBCT) in oral and maxillofacial imaging has driven the development of deep learning and radiomics applications to assist clinicians in early diagnosis, accurate prognosis prediction, and efficient treatment planning of maxillofacial diseases. This narrative review aimed to provide an up-to-date overview of the current applications of deep learning and radiomics on CT and CBCT for the diagnosis and management of maxillofacial diseases. Based on current evidence, a wide range of deep learning models on CT/CBCT images have been developed for automatic diagnosis, segmentation, and classification of jaw cysts and tumors, cervical lymph node metastasis, salivary gland diseases, temporomandibular (TMJ) disorders, maxillary sinus pathologies, mandibular fractures, and dentomaxillofacial deformities, while CT-/CBCT-derived radiomics applications mainly focused on occult lymph node metastasis in patients with oral cancer, malignant salivary gland tumors, and TMJ osteoarthritis. Most of these models showed high performance, and some of them even outperformed human experts. The models with performance on par with human experts have the potential to serve as clinically practicable tools to achieve the earliest possible diagnosis and treatment, leading to a more precise and personalized approach for the management of maxillofacial diseases. Challenges and issues, including the lack of the generalizability and explainability of deep learning models and the uncertainty in the reproducibility and stability of radiomic features, should be overcome to gain the trust of patients, providers, and healthcare organizers for daily clinical use of these models.
Collapse
Affiliation(s)
- Kuo Feng Hung
- Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Qi Yong H. Ai
- Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Lun M. Wong
- Imaging and Interventional Radiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Andy Wai Kan Yeung
- Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Dion Tik Shun Li
- Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Yiu Yan Leung
- Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
373
|
Updates on Quantitative MRI of Diffuse Liver Disease: A Narrative Review. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1147111. [PMID: 36619303 PMCID: PMC9812615 DOI: 10.1155/2022/1147111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/29/2022]
Abstract
Diffuse liver diseases are highly prevalent conditions around the world, including pathological liver changes that occur when hepatocytes are damaged and liver function declines, often leading to a chronic condition. In the last years, Magnetic Resonance Imaging (MRI) is reaching an important role in the study of diffuse liver diseases moving from qualitative to quantitative assessment of liver parenchyma. In fact, this can allow noninvasive accurate and standardized assessment of diffuse liver diseases and can represent a concrete alternative to biopsy which represents the current reference standard. MRI approach already tested for other pathologies include diffusion-weighted imaging (DWI) and radiomics, able to quantify different aspects of diffuse liver disease. New emerging MRI quantitative methods include MR elastography (MRE) for the quantification of the hepatic stiffness in cirrhotic patients, dedicated gradient multiecho sequences for the assessment of hepatic fat storage, and iron overload. Thus, the aim of this review is to give an overview of the technical principles and clinical application of new quantitative MRI techniques for the evaluation of diffuse liver disease.
Collapse
|
374
|
A Framework of Analysis to Facilitate the Harmonization of Multicenter Radiomic Features in Prostate Cancer. J Clin Med 2022; 12:jcm12010140. [PMID: 36614941 PMCID: PMC9821561 DOI: 10.3390/jcm12010140] [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: 11/08/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Pooling radiomic features coming from different centers in a statistical framework is challenging due to the variability in scanner models, acquisition protocols, and reconstruction settings. To remove technical variability, commonly called batch effects, different statistical harmonization strategies have been widely used in genomics but less considered in radiomics. The aim of this work was to develop a framework of analysis to facilitate the harmonization of multicenter radiomic features extracted from prostate T2-weighted magnetic resonance imaging (MRI) and to improve the power of radiomics for prostate cancer (PCa) management in order to develop robust non-invasive biomarkers translating into clinical practice. To remove technical variability and correct for batch effects, we investigated four different statistical methods (ComBat, SVA, Arsynseq, and mixed effect). The proposed approaches were evaluated using a dataset of 210 prostate cancer (PCa) patients from two centers. The impacts of the different statistical approaches were evaluated by principal component analysis and classification methods (LogitBoost, random forest, K-nearest neighbors, and decision tree). The ComBat method outperformed all other methods by achieving 70% accuracy and 78% AUC with the random forest method to automatically classify patients affected by PCa. The proposed statistical framework enabled us to define and develop a standardized pipeline of analysis to harmonize multicenter T2W radiomic features, yielding great promise to support PCa clinical practice.
Collapse
|
375
|
McAnena P, Moloney BM, Browne R, O’Halloran N, Walsh L, Walsh S, Sheppard D, Sweeney KJ, Kerin MJ, Lowery AJ. A radiomic model to classify response to neoadjuvant chemotherapy in breast cancer. BMC Med Imaging 2022; 22:225. [PMID: 36564734 PMCID: PMC9789647 DOI: 10.1186/s12880-022-00956-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Medical image analysis has evolved to facilitate the development of methods for high-throughput extraction of quantitative features that can potentially contribute to the diagnostic and treatment paradigm of cancer. There is a need for further improvement in the accuracy of predictive markers of response to neo-adjuvant chemotherapy (NAC). The aim of this study was to develop a radiomic classifier to enhance current approaches to predicting the response to NAC breast cancer. METHODS Data on patients treated for breast cancer with NAC prior to surgery who had a pre-NAC dynamic contrast enhanced breast MRI were included. Response to NAC was assessed using the Miller-Payne system on the excised tumor. Tumor segmentation was carried out manually under the supervision of a consultant breast radiologist. Features were selected using least absolute shrinkage selection operator regression. A support vector machine learning model was used to classify response to NAC. RESULTS 74 patients were included. Patients were classified as having a poor response to NAC (reduction in cellularity < 90%, n = 44) and an excellent response (> 90% reduction in cellularity, n = 30). 4 radiomics features (discretized kurtosis, NGDLM contrast, GLZLM_SZE and GLZLM_ZP) were identified as pertinent predictors of response to NAC. A SVM model using these features stratified patients into poor and excellent response groups producing an AUC of 0.75. Addition of estrogen receptor status improved the accuracy of the model with an AUC of 0.811. CONCLUSION This study identified a radiomic classifier incorporating 4 radiomics features to augment subtype based classification of response to NAC in breast cancer.
Collapse
Affiliation(s)
- Peter McAnena
- grid.412440.70000 0004 0617 9371Department of Surgery, Clinical Sciences Institute, University Hospital Galway, Galway, Ireland
| | - Brian M. Moloney
- grid.412440.70000 0004 0617 9371Department of Radiology, University Hospital Galway, Galway, Ireland
| | - Robert Browne
- grid.412440.70000 0004 0617 9371Department of Surgery, Clinical Sciences Institute, University Hospital Galway, Galway, Ireland
| | - Niamh O’Halloran
- grid.412440.70000 0004 0617 9371Department of Radiology, University Hospital Galway, Galway, Ireland
| | - Leon Walsh
- grid.412440.70000 0004 0617 9371Department of Radiology, University Hospital Galway, Galway, Ireland
| | - Sinead Walsh
- grid.412440.70000 0004 0617 9371Department of Radiology, University Hospital Galway, Galway, Ireland
| | - Declan Sheppard
- grid.412440.70000 0004 0617 9371Department of Radiology, University Hospital Galway, Galway, Ireland
| | - Karl J. Sweeney
- grid.412440.70000 0004 0617 9371Department of Surgery, Clinical Sciences Institute, University Hospital Galway, Galway, Ireland
| | - Michael J. Kerin
- grid.412440.70000 0004 0617 9371Department of Surgery, Clinical Sciences Institute, University Hospital Galway, Galway, Ireland ,grid.6142.10000 0004 0488 0789Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
| | - Aoife J. Lowery
- grid.412440.70000 0004 0617 9371Department of Surgery, Clinical Sciences Institute, University Hospital Galway, Galway, Ireland ,grid.6142.10000 0004 0488 0789Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
| |
Collapse
|
376
|
Tabari A, Chan SM, Omar OMF, Iqbal SI, Gee MS, Daye D. Role of Machine Learning in Precision Oncology: Applications in Gastrointestinal Cancers. Cancers (Basel) 2022; 15:cancers15010063. [PMID: 36612061 PMCID: PMC9817513 DOI: 10.3390/cancers15010063] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal (GI) cancers, consisting of a wide spectrum of pathologies, have become a prominent health issue globally. Despite medical imaging playing a crucial role in the clinical workflow of cancers, standard evaluation of different imaging modalities may provide limited information. Accurate tumor detection, characterization, and monitoring remain a challenge. Progress in quantitative imaging analysis techniques resulted in "radiomics", a promising methodical tool that helps to personalize diagnosis and treatment optimization. Radiomics, a sub-field of computer vision analysis, is a bourgeoning area of interest, especially in this era of precision medicine. In the field of oncology, radiomics has been described as a tool to aid in the diagnosis, classification, and categorization of malignancies and to predict outcomes using various endpoints. In addition, machine learning is a technique for analyzing and predicting by learning from sample data, finding patterns in it, and applying it to new data. Machine learning has been increasingly applied in this field, where it is being studied in image diagnosis. This review assesses the current landscape of radiomics and methodological processes in GI cancers (including gastric, colorectal, liver, pancreatic, neuroendocrine, GI stromal, and rectal cancers). We explain in a stepwise fashion the process from data acquisition and curation to segmentation and feature extraction. Furthermore, the applications of radiomics for diagnosis, staging, assessment of tumor prognosis and treatment response according to different GI cancer types are explored. Finally, we discussed the existing challenges and limitations of radiomics in abdominal cancers and investigate future opportunities.
Collapse
Affiliation(s)
- Azadeh Tabari
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| | - Shin Mei Chan
- Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06510, USA
| | - Omar Mustafa Fathy Omar
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Shams I. Iqbal
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Michael S. Gee
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Dania Daye
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
377
|
Artificial Intelligence (AI) in Breast Imaging: A Scientometric Umbrella Review. Diagnostics (Basel) 2022; 12:diagnostics12123111. [PMID: 36553119 PMCID: PMC9777253 DOI: 10.3390/diagnostics12123111] [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: 11/14/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Artificial intelligence (AI), a rousing advancement disrupting a wide spectrum of applications with remarkable betterment, has continued to gain momentum over the past decades. Within breast imaging, AI, especially machine learning and deep learning, honed with unlimited cross-data/case referencing, has found great utility encompassing four facets: screening and detection, diagnosis, disease monitoring, and data management as a whole. Over the years, breast cancer has been the apex of the cancer cumulative risk ranking for women across the six continents, existing in variegated forms and offering a complicated context in medical decisions. Realizing the ever-increasing demand for quality healthcare, contemporary AI has been envisioned to make great strides in clinical data management and perception, with the capability to detect indeterminate significance, predict prognostication, and correlate available data into a meaningful clinical endpoint. Here, the authors captured the review works over the past decades, focusing on AI in breast imaging, and systematized the included works into one usable document, which is termed an umbrella review. The present study aims to provide a panoramic view of how AI is poised to enhance breast imaging procedures. Evidence-based scientometric analysis was performed in accordance with the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guideline, resulting in 71 included review works. This study aims to synthesize, collate, and correlate the included review works, thereby identifying the patterns, trends, quality, and types of the included works, captured by the structured search strategy. The present study is intended to serve as a "one-stop center" synthesis and provide a holistic bird's eye view to readers, ranging from newcomers to existing researchers and relevant stakeholders, on the topic of interest.
Collapse
|
378
|
Beyond Imaging and Genetic Signature in Glioblastoma: Radiogenomic Holistic Approach in Neuro-Oncology. Biomedicines 2022; 10:biomedicines10123205. [PMID: 36551961 PMCID: PMC9775324 DOI: 10.3390/biomedicines10123205] [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: 11/01/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is a malignant brain tumor exhibiting rapid and infiltrative growth, with less than 10% of patients surviving over 5 years, despite aggressive and multimodal treatments. The poor prognosis and the lack of effective pharmacological treatments are imputable to a remarkable histological and molecular heterogeneity of GBM, which has led, to date, to the failure of precision oncology and targeted therapies. Identification of molecular biomarkers is a paradigm for comprehensive and tailored treatments; nevertheless, biopsy sampling has proved to be invasive and limited. Radiogenomics is an emerging translational field of research aiming to study the correlation between radiographic signature and underlying gene expression. Although a research field still under development, not yet incorporated into routine clinical practice, it promises to be a useful non-invasive tool for future personalized/adaptive neuro-oncology. This review provides an up-to-date summary of the recent advancements in the use of magnetic resonance imaging (MRI) radiogenomics for the assessment of molecular markers of interest in GBM regarding prognosis and response to treatments, for monitoring recurrence, also providing insights into the potential efficacy of such an approach for survival prognostication. Despite a high sensitivity and specificity in almost all studies, accuracy, reproducibility and clinical value of radiomic features are the Achilles heel of this newborn tool. Looking into the future, investigators' efforts should be directed towards standardization and a disciplined approach to data collection, algorithms, and statistical analysis.
Collapse
|
379
|
Nicosia L, Bozzini AC, Ballerini D, Palma S, Pesapane F, Raimondi S, Gaeta A, Bellerba F, Origgi D, De Marco P, Castiglione Minischetti G, Sangalli C, Meneghetti L, Curigliano G, Cassano E. Radiomic Features Applied to Contrast Enhancement Spectral Mammography: Possibility to Predict Breast Cancer Molecular Subtypes in a Non-Invasive Manner. Int J Mol Sci 2022; 23:ijms232315322. [PMID: 36499648 PMCID: PMC9740943 DOI: 10.3390/ijms232315322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
We aimed to investigate the association between the radiomic features of contrast-enhanced spectral mammography (CESM) images and a specific receptor pattern of breast neoplasms. In this single-center retrospective study, we selected patients with neoplastic breast lesions who underwent CESM before a biopsy and surgical assessment between January 2013 and February 2022. Radiomic analysis was performed on regions of interest selected from recombined CESM images. The association between the features and each evaluated endpoint (ER, PR, Ki-67, HER2+, triple negative, G2-G3 expressions) was investigated through univariate logistic regression. Among the significant and highly correlated radiomic features, we selected only the one most associated with the endpoint. From a group of 321 patients, we enrolled 205 malignant breast lesions. The median age at the exam was 50 years (interquartile range (IQR) 45-58). NGLDM_Contrast was the only feature that was positively associated with both ER and PR expression (p-values = 0.01). NGLDM_Coarseness was negatively associated with Ki-67 expression (p-value = 0.02). Five features SHAPE Volume(mL), SHAPE_Volume(vx), GLRLM_RLNU, NGLDM_Busyness and GLZLM_GLNU were all positively and significantly associated with HER2+; however, all of them were highly correlated. Radiomic features of CESM images could be helpful to predict particular molecular subtypes before a biopsy.
Collapse
Affiliation(s)
- Luca Nicosia
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
- Correspondence:
| | - Anna Carla Bozzini
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Daniela Ballerini
- Breast Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy
| | - Simone Palma
- Department of Radiological and Hematological Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Filippo Pesapane
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Sara Raimondi
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO IRCCS, 20139 Milan, Italy
| | - Aurora Gaeta
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO IRCCS, 20139 Milan, Italy
| | - Federica Bellerba
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO IRCCS, 20139 Milan, Italy
| | - Daniela Origgi
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Paolo De Marco
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Giuseppe Castiglione Minischetti
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- School of Medical Physics, University of Milan, Via Celoria 16, 20133 Milan, Italy
| | - Claudia Sangalli
- Data Management, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lorenza Meneghetti
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milano, 20122 Milano, Italy
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Enrico Cassano
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| |
Collapse
|
380
|
Bin X, Zhu C, Tang Y, Li R, Ding Q, Xia W, Tang Y, Tang X, Yao D, Tang A. Nomogram Based on Clinical and Radiomics Data for Predicting Radiation-induced Temporal Lobe Injury in Patients with Non-metastatic Stage T4 Nasopharyngeal Carcinoma. Clin Oncol (R Coll Radiol) 2022; 34:e482-e492. [PMID: 36008245 DOI: 10.1016/j.clon.2022.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/19/2022] [Accepted: 07/21/2022] [Indexed: 01/31/2023]
Abstract
AIMS To use pre-treatment magnetic resonance imaging-based radiomics data with clinical data to predict radiation-induced temporal lobe injury (RTLI) in nasopharyngeal carcinoma (NPC) patients with stage T4/N0-3/M0 within 5 years after radiotherapy. MATERIALS AND METHODS This study retrospectively examined 98 patients (198 temporal lobes) with stage T4/N0-3/M0 NPC. Participants were enrolled into a training cohort or a validation cohort in a ratio of 7:3. Radiomics features were extracted from pre-treatment magnetic resonance imaging that were T1-and T2-weighted. Spearman rank correlation, the t-test and the least absolute shrinkage and selection operator (LASSO) algorithm were used to select significant radiomics features; machine-learning models were used to generate radiomics signatures (Rad-Scores). Rad-Scores and clinical factors were integrated into a nomogram for prediction of RTLI. Nomogram discrimination was evaluated using receiver operating characteristic analysis and clinical benefits were evaluated using decision curve analysis. RESULTS Participants were enrolled into a training cohort (n = 139) or a validation cohort (n = 59). In total, 3568 radiomics features were initially extracted from T1-and T2-weighted images. Age, Dmax, D1cc and 16 stable radiomics features (six from T1-weighted and 10 from T2-weighted images) were identified as independent predictive factors. A greater Rad-Score was associated with a greater risk of RTLI. The nomogram showed good discrimination, with a C-index of 0.85 (95% confidence interval 0.79-0.92) in the training cohort and 0.82 (95% confidence interval 0.71-0.92) in the validation cohort. CONCLUSION We developed models for the prediction of RTLI in patients with stage T4/N0-3/M0 NPC using pre-treatment radiomics data and clinical data. Nomograms from these pre-treatment data improved the prediction of RTLI. These results may allow the selection of patients for earlier clinical interventions.
Collapse
Affiliation(s)
- X Bin
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - C Zhu
- Department of Radiation Oncology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Y Tang
- Department of Neurology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - R Li
- Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University Hangzhou, Zhejiang Province, China; Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Q Ding
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - W Xia
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Y Tang
- Department of Radiology, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - X Tang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - D Yao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - A Tang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
381
|
Liu X, Elbanan MG, Luna A, Haider MA, Smith AD, Sabottke CF, Spieler BM, Turkbey B, Fuentes D, Moawad A, Kamel S, Horvat N, Elsayes KM. Radiomics in Abdominopelvic Solid-Organ Oncologic Imaging: Current Status. AJR Am J Roentgenol 2022; 219:985-995. [PMID: 35766531 PMCID: PMC10616929 DOI: 10.2214/ajr.22.27695] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Radiomics is the process of extraction of high-throughput quantitative imaging features from medical images. These features represent noninvasive quantitative biomarkers that go beyond the traditional imaging features visible to the human eye. This article first reviews the steps of the radiomics pipeline, including image acquisition, ROI selection and image segmentation, image preprocessing, feature extraction, feature selection, and model development and application. Current evidence for the application of radiomics in abdominopelvic solid-organ cancers is then reviewed. Applications including diagnosis, subtype determination, treatment response assessment, and outcome prediction are explored within the context of hepatobiliary and pancreatic cancer, renal cell carcinoma, prostate cancer, gynecologic cancer, and adrenal masses. This literature review focuses on the strongest available evidence, including systematic reviews, meta-analyses, and large multicenter studies. Limitations of the available literature are highlighted, including marked heterogeneity in radiomics methodology, frequent use of small sample sizes with high risk of overfitting, and lack of prospective design, external validation, and standardized radiomics workflow. Thus, although studies have laid a foundation that supports continued investigation into radiomics models, stronger evidence is needed before clinical adoption.
Collapse
Affiliation(s)
- Xiaoyang Liu
- Joint Department of Medical Imaging, Division of Abdominal Imaging, University Health Network, University of Toronto, ON, Canada
| | - Mohamed G Elbanan
- Department of Radiology, Yale New Haven Health, Bridgeport Hospital, Bridgeport, CT
| | | | - Masoom A Haider
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Joint Department of Medical Imaging, University Health Network, Sinai Health System and University of Toronto, Toronto, ON, Canada
| | - Andrew D Smith
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Carl F Sabottke
- Department of Medical Imaging, University of Arizona College of Medicine, Tucson, AZ
| | - Bradley M Spieler
- Department of Radiology, University Medical Center, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD
| | - David Fuentes
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ahmed Moawad
- Department of Diagnostic and Interventional Radiology, Mercy Catholic Medical Center, Darby, PA
| | - Serageldin Kamel
- Department of Lymphoma, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Natally Horvat
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Khaled M Elsayes
- Department of Abdominal Imaging, University of Texas MD Anderson Cancer Center, 1400 Pressler St, Houston, TX 77030
| |
Collapse
|
382
|
O'Donnell J, Gasior S, Davey M, O'Malley E, Lowery A, McGarry J, O'Connell A, Kerin M, McCarthy P. The accuracy of breast MRI radiomic methodologies in predicting pathological complete response to neoadjuvant chemotherapy: A systematic review and network meta-analysis. Eur J Radiol 2022; 157:110561. [DOI: 10.1016/j.ejrad.2022.110561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/13/2022] [Accepted: 10/11/2022] [Indexed: 11/03/2022]
|
383
|
Ultrasonic Texture Features for Assessing Cardiac Remodeling and Dysfunction. J Am Coll Cardiol 2022; 80:2187-2201. [DOI: 10.1016/j.jacc.2022.09.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/08/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022]
|
384
|
Basran PS, Porter I. Radiomics in veterinary medicine: Overview, methods, and applications. Vet Radiol Ultrasound 2022; 63 Suppl 1:828-839. [PMID: 36514226 DOI: 10.1111/vru.13156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/24/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
Radiomics, or quantitative image analysis from radiographic image data, borrows the suffix from other emerging -omics fields of study, such as genomics, proteomics, and metabolomics. This report provides an overview of the general principles of how radiomic features are computed, describes major types of morphological, first order, and texture features, and the applications, challenges, and opportunities of radiomics as applied in veterinary medicine. Some advantages radiomics has over traditional semantic radiological features include standardized methodology in computing semantic features, the ability to compute features in multi-dimensional images, their newfound associations with genomic and pathological abnormalities, and the number of perceptible and imperceptible features available for regression or classification modeling. Some challenges in deploying radiomics in a clinical setting include sensitivity to image acquisition settings and image artifacts, pre- and post-image reconstruction and calculation settings, variability in feature estimates stemming from inter- and intra-observer contouring errors, and challenges with software and data harmonization and generalizability of findings given the challenges of small sample size and patient selection bias in veterinary medicine. Despite this, radiomics has enormous potential in patient-centric diagnostics, prognosis, and theragnostics. Fully leveraging the utility of radiomics in veterinary medicine will require inter-institutional collaborations, data harmonization, and data sharing strategies amongst institutions, transparent and robust model development, and multi-disciplinary efforts within and outside the veterinary medical imaging community.
Collapse
Affiliation(s)
- Parminder S Basran
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Ian Porter
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
385
|
Zhao JW, Shu X, Chen XX, Liu JX, Liu MQ, Ye J, Jiang HJ, Wang GS. Prediction of early recurrence of hepatocellular carcinoma after liver transplantation based on computed tomography radiomics nomogram. Hepatobiliary Pancreat Dis Int 2022; 21:543-550. [PMID: 35705443 DOI: 10.1016/j.hbpd.2022.05.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 05/24/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Early recurrence results in poor prognosis of patients with hepatocellular carcinoma (HCC) after liver transplantation (LT). This study aimed to explore the value of computed tomography (CT)-based radiomics nomogram in predicting early recurrence of patients with HCC after LT. METHODS A cohort of 151 patients with HCC who underwent LT between December 2013 and July 2019 were retrospectively enrolled. A total of 1218 features were extracted from enhanced CT images. The least absolute shrinkage and selection operator algorithm (LASSO) logistic regression was used for dimension reduction and radiomics signature building. The clinical model was constructed after the analysis of clinical factors, and the nomogram was constructed by introducing the radiomics signature into the clinical model. The predictive performance and clinical usefulness of the three models were evaluated using receiver operating characteristic (ROC) curve analysis and decision curve analysis (DCA), respectively. Calibration curves were plotted to assess the calibration of the nomogram. RESULTS There were significant differences in radiomics signature among early recurrence patients and non-early recurrence patients in the training cohort (P < 0.001) and validation cohort (P < 0.001). The nomogram showed the best predictive performance, with the largest area under the ROC curve in the training (0.882) and validation (0.917) cohorts. Hosmer-Lemeshow testing confirmed that the nomogram showed good calibration in the training (P = 0.138) and validation (P = 0.396) cohorts. DCA showed if the threshold probability is within 0.06-1, the nomogram had better clinical usefulness than the clinical model. CONCLUSIONS Our CT-based radiomics nomogram can preoperatively predict the risk of early recurrence in patients with HCC after LT.
Collapse
Affiliation(s)
- Jing-Wei Zhao
- Department of Radiology, the Third Medical Center, Chinese PLA General Hospital, The Training Site for Postgraduate of Jinzhou Medical University, Beijing 100039, China; Department of Radiology, the Third Medical Center, Chinese PLA General Hospital, Beijing 100039, China
| | - Xin Shu
- Medical School of Chinese PLA, Beijing 100853, China
| | - Xiao-Xia Chen
- Department of Radiology, the Third Medical Center, Chinese PLA General Hospital, Beijing 100039, China
| | - Jia-Xiong Liu
- Department of Radiology, the Third Medical Center, Chinese PLA General Hospital, Beijing 100039, China
| | - Mu-Qing Liu
- Department of Radiology, the Third Medical Center, Chinese PLA General Hospital, Beijing 100039, China
| | - Ju Ye
- Department of Radiology, the Third Medical Center, Chinese PLA General Hospital, Beijing 100039, China
| | - Hui-Jie Jiang
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Gui-Sheng Wang
- Department of Radiology, the Third Medical Center, Chinese PLA General Hospital, Beijing 100039, China.
| |
Collapse
|
386
|
Cellina M, Cè M, Khenkina N, Sinichich P, Cervelli M, Poggi V, Boemi S, Ierardi AM, Carrafiello G. Artificial Intellgence in the Era of Precision Oncological Imaging. Technol Cancer Res Treat 2022; 21:15330338221141793. [PMID: 36426565 PMCID: PMC9703524 DOI: 10.1177/15330338221141793] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Rapid-paced development and adaptability of artificial intelligence algorithms have secured their almost ubiquitous presence in the field of oncological imaging. Artificial intelligence models have been created for a variety of tasks, including risk stratification, automated detection, and segmentation of lesions, characterization, grading and staging, prediction of prognosis, and treatment response. Soon, artificial intelligence could become an essential part of every step of oncological workup and patient management. Integration of neural networks and deep learning into radiological artificial intelligence algorithms allow for extrapolating imaging features otherwise inaccessible to human operators and pave the way to truly personalized management of oncological patients.Although a significant proportion of currently available artificial intelligence solutions belong to basic and translational cancer imaging research, their progressive transfer to clinical routine is imminent, contributing to the development of a personalized approach in oncology. We thereby review the main applications of artificial intelligence in oncological imaging, describe the example of their successful integration into research and clinical practice, and highlight the challenges and future perspectives that will shape the field of oncological radiology.
Collapse
Affiliation(s)
- Michaela Cellina
- Radiology Department, Fatebenefratelli Hospital, Milano, Italy,Michaela Cellina, MD, Radiology Department, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, Milano, Piazza Principessa Clotilde 3, 20121, Milano, Italy.
| | - Maurizio Cè
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Natallia Khenkina
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Polina Sinichich
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Marco Cervelli
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Vittoria Poggi
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | - Sara Boemi
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy
| | | | - Gianpaolo Carrafiello
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy,Radiology Department, Fondazione IRCCS Cà Granda, Milan, Italy
| |
Collapse
|
387
|
Haidey J, Low G, Wilson MP. Radiomics-based approaches outperform visual analysis for differentiating lipoma from atypical lipomatous tumors: a review. Skeletal Radiol 2022; 52:1089-1100. [PMID: 36385583 DOI: 10.1007/s00256-022-04232-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/02/2022] [Accepted: 11/06/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Differentiating atypical lipomatous tumors (ALTs) and well-differentiated liposarcomas (WDLs) from benign lipomatous lesions is important for guiding clinical management, though conventional visual analysis of these lesions is challenging due to overlap of imaging features. Radiomics-based approaches may serve as a promising alternative and/or supplementary diagnostic approach to conventional imaging. PURPOSE The purpose of this study is to review the practice of radiomics-based imaging and systematically evaluate the literature available for studies evaluating radiomics applied to differentiating ALTs/WDLs from benign lipomas. REVIEW A background review of the radiomic workflow is provided, outlining the steps of image acquisition, segmentation, feature extraction, and model development. Subsequently, a systematic review of MEDLINE, EMBASE, Scopus, the Cochrane Library, and the grey literature was performed from inception to June 2022 to identify size studies using radiomics for differentiating ALTs/WDLs from benign lipomas. Radiomic models were shown to outperform conventional analysis in all but one model with a sensitivity ranging from 68 to 100% and a specificity ranging from 84 to 100%. However, current approaches rely on user input and no studies used a fully automated method for segmentation, contributing to interobserver variability and decreasing time efficiency. CONCLUSION Radiomic models may show improved performance for differentiating ALTs/WDLs from benign lipomas compared to conventional analysis. However, considerable variability between radiomic approaches exists and future studies evaluating a standardized radiomic model with a multi-institutional study design and preferably fully automated segmentation software are needed before clinical application can be more broadly considered.
Collapse
Affiliation(s)
- Jordan Haidey
- Department of Radiology and Diagnostic Imaging, University of Alberta, 2B2.41 WMC, 8440-112 Street NW, Edmonton, Alberta, T6G 2B7, Canada.
| | - Gavin Low
- Department of Radiology and Diagnostic Imaging, University of Alberta, 2B2.41 WMC, 8440-112 Street NW, Edmonton, Alberta, T6G 2B7, Canada
| | - Mitchell P Wilson
- Department of Radiology and Diagnostic Imaging, University of Alberta, 2B2.41 WMC, 8440-112 Street NW, Edmonton, Alberta, T6G 2B7, Canada
| |
Collapse
|
388
|
Identification of texture MRI brain abnormalities on first-episode psychosis and clinical high-risk subjects using explainable artificial intelligence. Transl Psychiatry 2022; 12:481. [PMID: 36385133 PMCID: PMC9668814 DOI: 10.1038/s41398-022-02242-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 11/17/2022] Open
Abstract
Structural MRI studies in first-episode psychosis and the clinical high-risk state have consistently shown volumetric abnormalities. Aim of the present study was to introduce radiomics texture features in identification of psychosis. Radiomics texture features describe the interrelationship between voxel intensities across multiple spatial scales capturing the hidden information of underlying disease dynamics in addition to volumetric changes. Structural MR images were acquired from 77 first-episode psychosis (FEP) patients, 58 clinical high-risk subjects with no later transition to psychosis (CHR_NT), 15 clinical high-risk subjects with later transition (CHR_T), and 44 healthy controls (HC). Radiomics texture features were extracted from non-segmented images, and two-classification schemas were performed for the identification of FEP vs. HC and FEP vs. CHR_NT. The group of CHR_T was used as external validation in both schemas. The classification of a subject's clinical status was predicted by importing separately (a) the difference of entropy feature map and (b) the contrast feature map, resulting in classification balanced accuracy above 72% in both analyses. The proposed framework enhances the classification decision for FEP, CHR_NT, and HC subjects, verifies diagnosis-relevant features and may potentially contribute to identification of structural biomarkers for psychosis, beyond and above volumetric brain changes.
Collapse
|
389
|
Evaluation of radiomics feature stability in abdominal monoenergetic photon counting CT reconstructions. Sci Rep 2022; 12:19594. [PMID: 36379992 PMCID: PMC9665022 DOI: 10.1038/s41598-022-22877-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
Abstract
Feature stability and standardization remain challenges that impede the clinical implementation of radiomics. This study investigates the potential of spectral reconstructions from photon-counting computed tomography (PCCT) regarding organ-specific radiomics feature stability. Abdominal portal-venous phase PCCT scans of 10 patients in virtual monoenergetic (VM) (keV 40-120 in steps of 10), polyenergetic, virtual non-contrast (VNC), and iodine maps were acquired. Two 2D and 3D segmentations measuring 1 and 2 cm in diameter of the liver, lung, spleen, psoas muscle, subcutaneous fat, and air were obtained for spectral reconstructions. Radiomics features were extracted with pyradiomics. The calculation of feature-specific intraclass correlation coefficients (ICC) was performed by comparing all segmentation approaches and organs. Feature-wise and organ-wise correlations were evaluated. Segmentation-resegmentation stability was evaluated by concordance correlation coefficient (CCC). Compared to non-VM, VM-reconstruction features tended to be more stable. For VM reconstructions, 3D 2 cm segmentation showed the highest average ICC with 0.63. Based on a criterion of ≥ 3 stable organs and an ICC of ≥ 0.75, 12-mainly non-first-order features-are shown to be stable between the VM reconstructions. In a segmentation-resegmentation analysis in 3D 2 cm, three features were identified as stable based on a CCC of > 0.6 in ≥ 3 organs in ≥ 6 VM reconstructions. Certain radiomics features vary between monoenergetic reconstructions and depend on the ROI size. Feature stability was also shown to differ between different organs. Yet, glcm_JointEntropy, gldm_GrayLevelNonUniformity, and firstorder_Entropy could be identified as features that could be interpreted as energy-independent and segmentation-resegmentation stable in this PCCT collective. PCCT may support radiomics feature standardization and comparability between sites.
Collapse
|
390
|
Mao Q, Zhou MT, Zhao ZP, Liu N, Yang L, Zhang XM. Role of radiomics in the diagnosis and treatment of gastrointestinal cancer. World J Gastroenterol 2022; 28:6002-6016. [PMID: 36405385 PMCID: PMC9669820 DOI: 10.3748/wjg.v28.i42.6002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/24/2022] [Accepted: 10/27/2022] [Indexed: 11/10/2022] Open
Abstract
Gastrointestinal cancer (GIC) has high morbidity and mortality as one of the main causes of cancer death. Preoperative risk stratification is critical to guide patient management, but traditional imaging studies have difficulty predicting its biological behavior. The emerging field of radiomics allows the conversion of potential pathophysiological information in existing medical images that cannot be visually recognized into high-dimensional quantitative image features. Tumor lesion characterization, therapeutic response evaluation, and survival prediction can be achieved by analyzing the relationships between these features and clinical and genetic data. In recent years, the clinical application of radiomics to GIC has increased dramatically. In this editorial, we describe the latest progress in the application of radiomics to GIC and discuss the value of its potential clinical applications, as well as its limitations and future directions.
Collapse
Affiliation(s)
- Qi Mao
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Mao-Ting Zhou
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Zhang-Ping Zhao
- Department of Radiology, Panzhihua Central Hospital, Panzhihua 617000, Sichuan Province, China
| | - Ning Liu
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Lin Yang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Xiao-Ming Zhang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| |
Collapse
|
391
|
A Review of Radiomics and Artificial Intelligence and Their Application in Veterinary Diagnostic Imaging. Vet Sci 2022; 9:vetsci9110620. [PMID: 36356097 PMCID: PMC9693121 DOI: 10.3390/vetsci9110620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
Simple Summary The goal of this paper is to provide an overview of current radiomic and AI applications in veterinary diagnostic imaging. We discuss the essential elements of AI for veterinary practitioners with the aim of helping them make informed decisions in applying AI technologies to their practices and that veterinarians will play an integral role in ensuring the appropriate uses and suitable curation of data. The expertise of veterinary professionals will be vital to ensuring suitable data and, subsequently, AI that meets the needs of the profession. Abstract Great advances have been made in human health care in the application of radiomics and artificial intelligence (AI) in a variety of areas, ranging from hospital management and virtual assistants to remote patient monitoring and medical diagnostics and imaging. To improve accuracy and reproducibility, there has been a recent move to integrate radiomics and AI as tools to assist clinical decision making and to incorporate it into routine clinical workflows and diagnosis. Although lagging behind human medicine, the use of radiomics and AI in veterinary diagnostic imaging is becoming more frequent with an increasing number of reported applications. The goal of this paper is to provide an overview of current radiomic and AI applications in veterinary diagnostic imaging.
Collapse
|
392
|
Two-Stage Deep Learning Model for Automated Segmentation and Classification of Splenomegaly. Cancers (Basel) 2022; 14:cancers14225476. [PMID: 36428569 PMCID: PMC9688308 DOI: 10.3390/cancers14225476] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/22/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Splenomegaly is a common cross-sectional imaging finding with a variety of differential diagnoses. This study aimed to evaluate whether a deep learning model could automatically segment the spleen and identify the cause of splenomegaly in patients with cirrhotic portal hypertension versus patients with lymphoma disease. This retrospective study included 149 patients with splenomegaly on computed tomography (CT) images (77 patients with cirrhotic portal hypertension, 72 patients with lymphoma) who underwent a CT scan between October 2020 and July 2021. The dataset was divided into a training (n = 99), a validation (n = 25) and a test cohort (n = 25). In the first stage, the spleen was automatically segmented using a modified U-Net architecture. In the second stage, the CT images were classified into two groups using a 3D DenseNet to discriminate between the causes of splenomegaly, first using the whole abdominal CT, and second using only the spleen segmentation mask. The classification performances were evaluated using the area under the receiver operating characteristic curve (AUC), accuracy (ACC), sensitivity (SEN), and specificity (SPE). Occlusion sensitivity maps were applied to the whole abdominal CT images, to illustrate which regions were important for the prediction. When trained on the whole abdominal CT volume, the DenseNet was able to differentiate between the lymphoma and liver cirrhosis in the test cohort with an AUC of 0.88 and an ACC of 0.88. When the model was trained on the spleen segmentation mask, the performance decreased (AUC = 0.81, ACC = 0.76). Our model was able to accurately segment splenomegaly and recognize the underlying cause. Training on whole abdomen scans outperformed training using the segmentation mask. Nonetheless, considering the performance, a broader and more general application to differentiate other causes for splenomegaly is also conceivable.
Collapse
|
393
|
Stamoulou E, Spanakis C, Manikis GC, Karanasiou G, Grigoriadis G, Foukakis T, Tsiknakis M, Fotiadis DI, Marias K. Harmonization Strategies in Multicenter MRI-Based Radiomics. J Imaging 2022; 8:303. [PMID: 36354876 PMCID: PMC9695920 DOI: 10.3390/jimaging8110303] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 08/13/2023] Open
Abstract
Radiomics analysis is a powerful tool aiming to provide diagnostic and prognostic patient information directly from images that are decoded into handcrafted features, comprising descriptors of shape, size and textural patterns. Although radiomics is gaining momentum since it holds great promise for accelerating digital diagnostics, it is susceptible to bias and variation due to numerous inter-patient factors (e.g., patient age and gender) as well as inter-scanner ones (different protocol acquisition depending on the scanner center). A variety of image and feature based harmonization methods has been developed to compensate for these effects; however, to the best of our knowledge, none of these techniques has been established as the most effective in the analysis pipeline so far. To this end, this review provides an overview of the challenges in optimizing radiomics analysis, and a concise summary of the most relevant harmonization techniques, aiming to provide a thorough guide to the radiomics harmonization process.
Collapse
Affiliation(s)
- Elisavet Stamoulou
- Computational BioMedicine Laboratory (CBML), Foundation for Research and Technology—Hellas (FORTH), 700 13 Heraklion, Greece
| | - Constantinos Spanakis
- Computational BioMedicine Laboratory (CBML), Foundation for Research and Technology—Hellas (FORTH), 700 13 Heraklion, Greece
| | - Georgios C. Manikis
- Computational BioMedicine Laboratory (CBML), Foundation for Research and Technology—Hellas (FORTH), 700 13 Heraklion, Greece
- Department of Oncology-Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Georgia Karanasiou
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, 451 10 Ioannina, Greece
| | - Grigoris Grigoriadis
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, 451 10 Ioannina, Greece
| | - Theodoros Foukakis
- Department of Oncology-Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Manolis Tsiknakis
- Computational BioMedicine Laboratory (CBML), Foundation for Research and Technology—Hellas (FORTH), 700 13 Heraklion, Greece
- Department of Electrical & Computer Engineering, Hellenic Mediterranean University, 714 10 Heraklion, Greece
| | - Dimitrios I. Fotiadis
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, 451 10 Ioannina, Greece
- Department of Biomedical Research, Institute of Molecular Biology and Biotechnology—FORTH, University Campus of Ioannina, 451 15 Ioannina, Greece
| | - Kostas Marias
- Computational BioMedicine Laboratory (CBML), Foundation for Research and Technology—Hellas (FORTH), 700 13 Heraklion, Greece
- Department of Electrical & Computer Engineering, Hellenic Mediterranean University, 714 10 Heraklion, Greece
| |
Collapse
|
394
|
Sadler D, Okwuosa T, Teske AJ, Guha A, Collier P, Moudgil R, Sarkar A, Brown SA. Cardio oncology: Digital innovations, precision medicine and health equity. Front Cardiovasc Med 2022; 9:951551. [PMID: 36407451 PMCID: PMC9669068 DOI: 10.3389/fcvm.2022.951551] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
The rapid emergence of cardio-oncology has resulted in a rapid growth of cardio-oncology programs, dedicated professional societies sections and committees, and multiple collaborative networks that emerged to amplify the access to care in this new subspecialty. However, most existing data, position statements and guidelines are limited by the lack of availability of large clinical trials to support these recommendations. Furthermore, there are significant challenges regarding proper access to cardio-oncology care and treatment, particularly in marginalized and minority populations. The emergence and evolution of personalized medicine, artificial intelligence (AI), and machine learning in medicine and in cardio-oncology provides an opportunity for a more targeted, personalized approach to cardiovascular complications of cancer treatment. The proper implementation of these new modalities may facilitate a more equitable approach to adequate and universal access to cardio-oncology care, improve health related outcomes, and enable health care systems to eliminate the digital divide. This article reviews and analyzes the current status on these important issues.
Collapse
Affiliation(s)
- Diego Sadler
- Cardio Oncology Section, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Florida, Weston, FL, United States
- *Correspondence: Diego Sadler
| | - Tochukwu Okwuosa
- Division of Cardiology, Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - A. J. Teske
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Avirup Guha
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Patrick Collier
- Cleveland Clinic, Cardio Oncology, Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland, OH, United States
| | - Rohit Moudgil
- Cleveland Clinic, Cardio Oncology, Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland, OH, United States
| | - Abdullah Sarkar
- Cardio Oncology Section, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Florida, Weston, FL, United States
| | - Sherry-Ann Brown
- Division of Cardiology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
395
|
Anai K, Hayashida Y, Ueda I, Hozuki E, Yoshimatsu Y, Tsukamoto J, Hamamura T, Onari N, Aoki T, Korogi Y. The effect of CT texture-based analysis using machine learning approaches on radiologists' performance in differentiating focal-type autoimmune pancreatitis and pancreatic duct carcinoma. Jpn J Radiol 2022; 40:1156-1165. [PMID: 35727458 PMCID: PMC9616757 DOI: 10.1007/s11604-022-01298-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/28/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE To develop a support vector machine (SVM) classifier using CT texture-based analysis in differentiating focal-type autoimmune pancreatitis (AIP) and pancreatic duct carcinoma (PD), and to assess the radiologists' diagnostic performance with or without SVM. MATERIALS AND METHODS This retrospective study included 50 patients (20 patients with focal-type AIP and 30 patients with PD) who underwent dynamic contrast-enhanced CT. Sixty-two CT texture-based features were extracted from 2D images of the arterial and portal phase CTs. We conducted data compression and feature selections using principal component analysis (PCA) and produced the SVM classifier. Four readers participated in this observer performance study and the statistical significance of differences with and without the SVM was assessed by receiver operating characteristic (ROC) analysis. RESULTS The SVM performance indicated a high performance in differentiating focal-type AIP and PD (AUC = 0.920). The AUC for all 4 readers increased significantly from 0.827 to 0.911 when using the SVM outputs (p = 0.010). The AUC for inexperienced readers increased significantly from 0.781 to 0.905 when using the SVM outputs (p = 0.310). The AUC for experienced readers increased from 0.875 to 0.912 when using the SVM outputs, however, there was no significant difference (p = 0.018). CONCLUSION The use of SVM classifier using CT texture-based features improved the diagnostic performance for differentiating focal-type AIP and PD on CT.
Collapse
Affiliation(s)
- Kenta Anai
- Department of Radiology, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555 Japan
| | - Yoshiko Hayashida
- Department of Radiology, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555 Japan
| | - Issei Ueda
- Department of Radiology, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555 Japan
| | - Eri Hozuki
- Department of Radiology, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555 Japan
| | - Yuuta Yoshimatsu
- Department of Radiology, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555 Japan
| | - Jun Tsukamoto
- Department of Radiology, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555 Japan
| | - Toshihiko Hamamura
- Department of Radiology, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555 Japan
| | - Norihiro Onari
- Department of Radiology, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555 Japan
| | - Takatoshi Aoki
- Department of Radiology, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555 Japan
| | - Yukunori Korogi
- Department of Radiology, Kyushu Rosai Hospital, Moji Medical Center, 3-1, Higashiminatomachi, Moji-ku, Kitakyushu, Fukuoka 801-8502 Japan
| |
Collapse
|
396
|
Luo C, Zhao S, Peng C, Wang C, Hu K, Zhong X, Luo T, Huang J, Lu D. Mammography radiomics features at diagnosis and progression-free survival among patients with breast cancer. Br J Cancer 2022; 127:1886-1892. [PMID: 36050449 PMCID: PMC9643418 DOI: 10.1038/s41416-022-01958-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The associations between mammographic radiomics and breast cancer clinical endpoints are unclear. We aimed to identify mammographic radiomics features associated with breast cancer prognosis. METHODS Nested from a large breast cancer cohort in our institution, we conducted an extreme case-control study consisting of 207 cases with any invasive disease-free survival (iDFS) endpoint <5 years and 207 molecular subtype-matched controls with >5-year iDFS. A total of 632 radiomics features in craniocaudal (CC) and mediolateral oblique (MLO) views were extracted from pre-treatment mammography. Logistic regression was used to identify iDFS-associated features with multiple testing corrections (Benjamini-Hochberg method). In a subsample with RNA-seq data (n = 96), gene set enrichment analysis was employed to identify pathways associated with lead features. RESULTS We identified 15 iDFS-associated features from CC-view yet none from MLO-view. S(1,-1)SumAverg and WavEnLL_s-6 were the lead ones and associated with favourable (OR 0.64, 95% CI 0.42-0.87, P = 0.01) and poor iDFS (OR 1.53, 95% CI 1.31-1.76, P = 0.01), respectively. Both features were associated with eight pathways (primarily involving cell cycle regulation) in tumour but not adjacent normal tissues. CONCLUSION Our findings suggest mammographic radiomics features are associated with breast cancer iDFS, potentially through pathways involving cell cycle regulation.
Collapse
Affiliation(s)
- Chuanxu Luo
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, China
| | - Shuang Zhao
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Cheng Peng
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Chengshi Wang
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, China
- Department of breast surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Kejia Hu
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, China
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Xiaorong Zhong
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, China
- Department of Head, Neck and Mammary Gland Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Luo
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, China
- Department of Head, Neck and Mammary Gland Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Juan Huang
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Donghao Lu
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
397
|
Fasterholdt I, Naghavi-Behzad M, Rasmussen BSB, Kjølhede T, Skjøth MM, Hildebrandt MG, Kidholm K. Value assessment of artificial intelligence in medical imaging: a scoping review. BMC Med Imaging 2022; 22:187. [PMID: 36316665 PMCID: PMC9620604 DOI: 10.1186/s12880-022-00918-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/22/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Artificial intelligence (AI) is seen as one of the major disrupting forces in the future healthcare system. However, the assessment of the value of these new technologies is still unclear, and no agreed international health technology assessment-based guideline exists. This study provides an overview of the available literature in the value assessment of AI in the field of medical imaging. METHODS We performed a systematic scoping review of published studies between January 2016 and September 2020 using 10 databases (Medline, Scopus, ProQuest, Google Scholar, and six related databases of grey literature). Information about the context (country, clinical area, and type of study) and mentioned domains with specific outcomes and items were extracted. An existing domain classification, from a European assessment framework, was used as a point of departure, and extracted data were grouped into domains and content analysis of data was performed covering predetermined themes. RESULTS Seventy-nine studies were included out of 5890 identified articles. An additional seven studies were identified by searching reference lists, and the analysis was performed on 86 included studies. Eleven domains were identified: (1) health problem and current use of technology, (2) technology aspects, (3) safety assessment, (4) clinical effectiveness, (5) economics, (6) ethical analysis, (7) organisational aspects, (8) patients and social aspects, (9) legal aspects, (10) development of AI algorithm, performance metrics and validation, and (11) other aspects. The frequency of mentioning a domain varied from 20 to 78% within the included papers. Only 15/86 studies were actual assessments of AI technologies. The majority of data were statements from reviews or papers voicing future needs or challenges of AI research, i.e. not actual outcomes of evaluations. CONCLUSIONS This review regarding value assessment of AI in medical imaging yielded 86 studies including 11 identified domains. The domain classification based on European assessment framework proved useful and current analysis added one new domain. Included studies had a broad range of essential domains about addressing AI technologies highlighting the importance of domains related to legal and ethical aspects.
Collapse
Affiliation(s)
- Iben Fasterholdt
- CIMT – Centre for Innovative Medical Technology, Odense University Hospital, Sdr. Boulevard 29, Entrance 102, 4rd Floor, 5000 Odense C, Denmark
| | - Mohammad Naghavi-Behzad
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Benjamin S. B. Rasmussen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Radiology, Odense University Hospital, Odense, Denmark
- CAI-X – Centre for Clinical Artificial Intelligence, Odense University Hospital, Odense, Denmark
| | - Tue Kjølhede
- CIMT – Centre for Innovative Medical Technology, Odense University Hospital, Sdr. Boulevard 29, Entrance 102, 4rd Floor, 5000 Odense C, Denmark
| | - Mette Maria Skjøth
- Department of Dermatology and Allergy Centre, Odense University Hospital, Odense, Denmark
| | - Malene Grubbe Hildebrandt
- CIMT – Centre for Innovative Medical Technology, Odense University Hospital, Sdr. Boulevard 29, Entrance 102, 4rd Floor, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Kristian Kidholm
- CIMT – Centre for Innovative Medical Technology, Odense University Hospital, Sdr. Boulevard 29, Entrance 102, 4rd Floor, 5000 Odense C, Denmark
| |
Collapse
|
398
|
Yin F, Zhang H, Qi A, Zhu Z, Yang L, Wen G, Xie W. An exploratory study of CT radiomics using differential network feature selection for WHO/ISUP grading and progression-free survival prediction of clear cell renal cell carcinoma. Front Oncol 2022; 12:979613. [DOI: 10.3389/fonc.2022.979613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesTo explore the feasibility of predicting the World Health Organization/International Society of Urological Pathology (WHO/ISUP) grade and progression-free survival (PFS) of clear cell renal cell cancer (ccRCC) using the radiomics features (RFs) based on the differential network feature selection (FS) method using the maximum-entropy probability model (MEPM).Methods175 ccRCC patients were divided into a training set (125) and a test set (50). The non-contrast phase (NCP), cortico-medullary phase, nephrographic phase, excretory phase phases, and all-phase WHO/ISUP grade prediction models were constructed based on a new differential network FS method using the MEPM. The diagnostic performance of the best phase model was compared with the other state-of-the-art machine learning models and the clinical models. The RFs of the best phase model were used for survival analysis and visualized using risk scores and nomograms. The performance of the above models was tested in both cross-validated and independent validation and checked by the Hosmer-Lemeshow test.ResultsThe NCP RFs model was the best phase model, with an AUC of 0.89 in the test set, and performed superior to other machine learning models and the clinical models (all p <0.05). Kaplan-Meier survival analysis, univariate and multivariate cox regression results, and risk score analyses showed the NCP RFs could predict PFS well (almost all p < 0.05). The nomogram model incorporated the best two RFs and showed good discrimination, a C-index of 0.71 and 0.69 in the training and test set, and good calibration.ConclusionThe NCP CT-based RFs selected by differential network FS could predict the WHO/ISUP grade and PFS of RCC.
Collapse
|
399
|
Kulanthaivelu R, Kohan A, Hinzpeter R, Liu ZA, Hope A, Huang SH, Waldron J, O’Sullivan B, Ortega C, Metser U, Veit-Haibach P. Prognostic value of PET/CT and MR-based baseline radiomics among patients with non-metastatic nasopharyngeal carcinoma. Front Oncol 2022; 12:952763. [PMID: 36353565 PMCID: PMC9638017 DOI: 10.3389/fonc.2022.952763] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/03/2022] [Indexed: 11/09/2023] Open
Abstract
PURPOSE Radiomics is an emerging imaging assessment technique that has shown promise in predicting survival among nasopharyngeal carcinoma (NPC) patients. Studies so far have focused on PET or MR-based radiomics independently. The aim of our study was to evaluate the prognostic value of clinical and radiomic parameters derived from both PET/CT and MR. METHODS Retrospective evaluation of 124 NPC patients with PET/CT and radiotherapy planning MR (RP-MR). Primary tumors were segmented using dedicated software (LIFEx version 6.1) from PET, CT, contrast-enhanced T1-weighted (T1-w), and T2-weighted (T2-w) MR sequences with 376 radiomic features extracted. Summary statistics describe patient, disease, and treatment characteristics. The Kaplan-Meier (KM) method estimates overall survival (OS) and progression-free survival (PFS). Clinical factors selected based on univariable analysis and the multivariable Cox model were subsequently constructed with radiomic features added. RESULTS The final models comparing clinical, clinical + RP-MR, clinical + PET/CT and clinical + RP-MR + PET/CT for OS and PFS demonstrated that combined radiomic signatures were significantly associated with improved survival prognostication (AUC 0.62 vs 0.81 vs 0.75 vs 0.86 at 21 months for PFS and 0.56 vs 0.85 vs 0.79 vs 0.96 at 24 months for OS). Clinical + RP-MR features initially outperform clinical + PET/CT for both OS and PFS (<18 months), and later in the clinical course for PFS (>42 months). CONCLUSION Our study demonstrated that PET/CT-based radiomic features may improve survival prognostication among NPC patients when combined with baseline clinical and MR-based radiomic features.
Collapse
Affiliation(s)
- Roshini Kulanthaivelu
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women’s College Hospital, University of Toronto, Toronto, ON, Canada
| | - Andres Kohan
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women’s College Hospital, University of Toronto, Toronto, ON, Canada
| | - Ricarda Hinzpeter
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women’s College Hospital, University of Toronto, Toronto, ON, Canada
| | - Zhihui Amy Liu
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Andrew Hope
- Department of Radiation Oncology, University Health Network, Mount Sinai Hospital and Women’s College Hospital, University of Toronto, Toronto, ON, Canada
| | - Shao Hui Huang
- Department of Radiation Oncology, University Health Network, Mount Sinai Hospital and Women’s College Hospital, University of Toronto, Toronto, ON, Canada
| | - John Waldron
- Department of Radiation Oncology, University Health Network, Mount Sinai Hospital and Women’s College Hospital, University of Toronto, Toronto, ON, Canada
| | - Brian O’Sullivan
- Department of Radiation Oncology, University Health Network, Mount Sinai Hospital and Women’s College Hospital, University of Toronto, Toronto, ON, Canada
| | - Claudia Ortega
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women’s College Hospital, University of Toronto, Toronto, ON, Canada
| | - Ur Metser
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women’s College Hospital, University of Toronto, Toronto, ON, Canada
| | - Patrick Veit-Haibach
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women’s College Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
400
|
Application of Machine Learning Techniques for Characterization of Ischemic Stroke with MRI Images: A Review. Diagnostics (Basel) 2022; 12:diagnostics12102535. [DOI: 10.3390/diagnostics12102535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Magnetic resonance imaging (MRI) is a standard tool for the diagnosis of stroke, but its manual interpretation by experts is arduous and time-consuming. Thus, there is a need for computer-aided-diagnosis (CAD) models for the automatic segmentation and classification of stroke on brain MRI. The heterogeneity of stroke pathogenesis, morphology, image acquisition modalities, sequences, and intralesional tissue signal intensity, as well as lesion-to-normal tissue contrast, pose significant challenges to the development of such systems. Machine learning (ML) is increasingly being used in predictive neuroimaging diagnosis and prognostication. This paper reviews image processing and machine learning techniques that have been applied to detect ischemic stroke on brain MRI, including details on image acquisition, pre-processing, techniques to segment, extraction of features, and classification into stroke types. The main objective of this work is to find the state-of-art machine learning techniques used to predict the ischemic stroke and their application in clinical set-up. The article selection is performed according to PRISMA guideline. The state-of-the-art on automated MRI stroke diagnosis, with a focus on machine learning, is discussed, along with its advantages and limitations. We found that the various machine learning models discussed in this article are able to detect the infarcts with an acceptable accuracy of 70–90%. However, no one has highlighted the time complexity to predict the stroke in the model developed, which is an important factor. The work concludes with proposals for future recommendations for building efficient and robust deep learning (DL) models for quantitative brain MRI analysis. In recent work, with the application of DL approaches, using large datasets to train the models has improved the detection accuracy and reduced computational complexity. We suggest that the design of a decision support system based on artificial intelligence (AI) and clinical data presenting symptoms is essential to support clinicians to accelerate diagnosis and timeous therapy in the emergency management of stroke.
Collapse
|