1
|
Marfisi D, Giannelli M, Marzi C, Del Meglio J, Barucci A, Masturzo L, Vignali C, Mascalchi M, Traino A, Casolo G, Diciotti S, Tessa C. Test-retest repeatability of myocardial radiomic features from quantitative cardiac magnetic resonance T1 and T2 mapping. Magn Reson Imaging 2024; 113:110217. [PMID: 39067653 DOI: 10.1016/j.mri.2024.110217] [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: 03/23/2024] [Revised: 06/14/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Radiomics of cardiac magnetic resonance (MR) imaging has proved to be potentially useful in the study of various myocardial diseases. Therefore, assessing the repeatability degree in radiomic features measurement is of fundamental importance. The aim of this study was to assess test-retest repeatability of myocardial radiomic features extracted from quantitative T1 and T2 maps. A representative group of 24 subjects (mean age 54 ± 18 years) referred for clinical cardiac MR imaging were enrolled in the study. For each subject, T1 and T2 mapping through MOLLI and T2-prepared TrueFISP acquisition sequences, respectively, were performed at 1.5 T. Then, 98 radiomic features of different classes (shape, first-order, second-order) were extracted from a region of interest encompassing the whole left ventricle myocardium in a short axis slice. The repeatability was assessed performing different and complementary analyses: intraclass correlation coefficient (ICC) and limits of agreement (LOA) (i.e., the interval within which 95% of the percentage differences between two repeated measures are expected to lie). Radiomic features were characterized by a relatively wide range of repeatability degree in terms of both ICC and LOA. Overall, 44.9% and 38.8% of radiomic features showed ICC values > 0.75 for T1 and T2 maps, respectively, while 25.5% and 23.4% of radiomic features showed LOA between ±10%. A subset of radiomic features for T1 (Mean, Median, 10Percentile, 90Percentile, RootMeanSquared, Imc2, RunLengthNonUniformityNormalized, RunPercentage and ShortRunEmphasis) and T2 (MaximumDiameter, RunLengthNonUniformityNormalized, RunPercentage, ShortRunEmphasis) maps presented both ICC > 0.75 and LOA between ±5%. Overall, radiomic features extracted from T1 maps showed better repeatability performance than those extracted from T2 maps, with shape features characterized by better repeatability than first-order and textural features. Moreover, only a limited subset of 9 and 4 radiomic features for T1 and T2 maps, respectively, showed high repeatability degree in terms of both ICC and LOA. These results confirm the importance of assessing test-retest repeatability degree in radiomic feature estimation and might be useful for a more effective/reliable use of myocardial T1 and T2 mapping radiomics in clinical or research studies.
Collapse
Affiliation(s)
- Daniela Marfisi
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", 56126 Pisa, Italy
| | - Marco Giannelli
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", 56126 Pisa, Italy.
| | - Chiara Marzi
- Department of Statistics, Computer Science, Applications "Giuseppe Parenti", University of Florence, 50134 Florence, Italy
| | - Jacopo Del Meglio
- Unit of Cardiology, Azienda USL Toscana Nord Ovest, Versilia Hospital, 55041 Lido di Camaiore, Italy
| | - Andrea Barucci
- Institute of Applied Physics "Nello Carrara" (IFAC), Council of National Research (CNR), 50019 Sesto Fiorentino, Italy
| | - Luigi Masturzo
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", 56126 Pisa, Italy
| | - Claudio Vignali
- Unit of Radiology, Azienda USL Toscana Nord Ovest, Versilia Hospital, 55041 Lido di Camaiore, Italy
| | - Mario Mascalchi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50121 Florence, Italy; Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), 50139 Florence, Italy
| | - Antonio Traino
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", 56126 Pisa, Italy
| | - Giancarlo Casolo
- Unit of Cardiology, Azienda USL Toscana Nord Ovest, Versilia Hospital, 55041 Lido di Camaiore, Italy
| | - Stefano Diciotti
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, 47522 Cesena, Italy
| | - Carlo Tessa
- Unit of Radiology, Azienda USL Toscana Nord Ovest, Apuane Hospital, 54100 Massa, Italy
| |
Collapse
|
2
|
Fok WYR, Fieselmann A, Huemmer C, Biniazan R, Beister M, Geiger B, Kappler S, Saalfeld S. Adversarial robustness improvement for X-ray bone segmentation using synthetic data created from computed tomography scans. Sci Rep 2024; 14:25813. [PMID: 39468116 PMCID: PMC11519576 DOI: 10.1038/s41598-024-73363-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/17/2024] [Indexed: 10/30/2024] Open
Abstract
Deep learning-based image analysis offers great potential in clinical practice. However, it faces mainly two challenges: scarcity of large-scale annotated clinical data for training and susceptibility to adversarial data in inference. As an example, an artificial intelligence (AI) system could check patient positioning, by segmenting and evaluating relative positions of anatomical structures in medical images. Nevertheless, data to train such AI system might be highly imbalanced with mostly well-positioned images being available. Thus, we propose the use of synthetic X-ray images and annotation masks forward projected from 3D photon-counting CT volumes to create realistic non-optimally positioned X-ray images for training. An open-source model (TotalSegmentator) was used to annotate the clavicles in 3D CT volumes. We evaluated model robustness with respect to the internal (simulated) patient rotation α on real-data-trained models and real&synthetic-data-trained models. Our results showed that real&synthetic- data-trained models have Dice score percentage improvements of 3% to 15% across different α groups compared to the real-data-trained model. Therefore, we demonstrated that synthetic data could be supplementary used to train and enrich heavily underrepresented conditions to increase model robustness.
Collapse
Affiliation(s)
- Wai Yan Ryana Fok
- Faculty of Computer Science, Otto-von-Guericke-University Magdeburg, 39106, Magdeburg, Germany.
- X-ray Products, Siemens Healthineers AG, 91301, Forchheim, Germany.
| | | | | | - Ramyar Biniazan
- X-ray Products, Siemens Healthineers AG, 91301, Forchheim, Germany
| | - Marcel Beister
- X-ray Products, Siemens Healthineers AG, 91301, Forchheim, Germany
| | - Bernhard Geiger
- X-ray Products, Siemens Healthineers AG, 91301, Forchheim, Germany
| | - Steffen Kappler
- X-ray Products, Siemens Healthineers AG, 91301, Forchheim, Germany
| | - Sylvia Saalfeld
- Faculty of Computer Science, Otto-von-Guericke-University Magdeburg, 39106, Magdeburg, Germany
- Institute for Medical Informatics and Statistics, University Hospital Schleswig-Holstein Campus Kiel, 24105, Kiel, Germany
| |
Collapse
|
3
|
Kallos-Balogh P, Vas NF, Toth Z, Szakall S, Szabo P, Garai I, Kepes Z, Forgacs A, Szatmáriné Egeresi L, Magnus D, Balkay L. Multicentric study on the reproducibility and robustness of PET-based radiomics features with a realistic activity painting phantom. PLoS One 2024; 19:e0309540. [PMID: 39446842 PMCID: PMC11500893 DOI: 10.1371/journal.pone.0309540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/13/2024] [Indexed: 10/26/2024] Open
Abstract
Previously, we developed an "activity painting" tool for PET image simulation; however, it could simulate heterogeneous patterns only in the air. We aimed to improve this phantom technique to simulate arbitrary lesions in a radioactive background to perform relevant multi-center radiomic analysis. We conducted measurements moving a 22Na point source in a 20-liter background volume filled with 5 kBq/mL activity with an adequately controlled robotic system to prevent the surge of the water. Three different lesion patterns were "activity-painted" in five PET/CT cameras, resulting in 8 different reconstructions. We calculated 46 radiomic indeces (RI) for each lesion and imaging setting, applying absolute and relative discretization. Reproducibility and reliability were determined by the inter-setting coefficient of variation (CV) and the intraclass correlation coefficient (ICC). Hypothesis tests were used to compare RI between lesions. By simulating precisely the same lesions, we confirmed that the reconstructed voxel size and the spatial resolution of different PET cameras were critical for higher order RI. Considering conventional RIs, the SUVpeak and SUVmean proved the most reliable (CV<10%). CVs above 25% are more common for higher order RIs, but we also found that low CVs do not necessarily imply robust parameters but often rather insensitive RIs. Based on the hypothesis test, most RIs could clearly distinguish between the various lesions using absolute resampling. ICC analysis also revealed that most RIs were more reproducible with absolute discretization. The activity painting method in a real radioactive environment proved suitable for precisely detecting the radiomic differences derived from the different camera settings and texture characteristics. We also found that inter-setting CV is not an appropriate metric for analyzing RI parameters' reliability and robustness. Although multicentric cohorts are increasingly common in radiomics analysis, realistic texture phantoms can provide indispensable information on the sensitivity of an RI and how an individual RI parameter measures the texture.
Collapse
Affiliation(s)
- Piroska Kallos-Balogh
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Norman Felix Vas
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltan Toth
- Medicopus Healthcare Provider and Public Nonprofit Ltd., Somogy County Moritz Kaposi Teaching Hospital, Kaposvár, Hungary
| | | | | | - Ildiko Garai
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Scanomed Ltd., Debrecen, Debrecen, Hungary
| | - Zita Kepes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Lilla Szatmáriné Egeresi
- Division of Radiology and Imaging Science, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dahlbom Magnus
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California, United States of America
| | - Laszlo Balkay
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
4
|
Gong J, Wang Q, Li J, Yang Z, Zhang J, Teng X, Sun H, Cai J, Zhao L. Using high-repeatable radiomic features improves the cross-institutional generalization of prognostic model in esophageal squamous cell cancer receiving definitive chemoradiotherapy. Insights Imaging 2024; 15:239. [PMID: 39373828 PMCID: PMC11458848 DOI: 10.1186/s13244-024-01816-3] [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: 08/05/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
OBJECTIVES Repeatability is crucial for ensuring the generalizability and clinical utility of radiomics-based prognostic models. This study aims to investigate the repeatability of radiomic feature (RF) and its impact on the cross-institutional generalizability of the prognostic model for predicting local recurrence-free survival (LRFS) and overall survival (OS) in esophageal squamous cell cancer (ESCC) receiving definitive (chemo) radiotherapy (dCRT). METHODS Nine hundred and twelve patients from two hospitals were included as training and external validation sets, respectively. Image perturbations were applied to contrast-enhanced computed tomography to generate perturbed images. Six thousand five hundred ten RFs from different feature types, bin widths, and filters were extracted from the original and perturbed images separately to evaluate RF repeatability by intraclass correlation coefficient (ICC). The high-repeatable and low-repeatable RF groups grouped by the median ICC were further analyzed separately by feature selection and multivariate Cox proportional hazards regression model for predicting LRFS and OS. RESULTS First-order statistical features were more repeatable than texture features (median ICC: 0.70 vs 0.42-0.62). RFs from LoG had better repeatability than that of wavelet (median ICC: 0.70-0.84 vs 0.14-0.64). Features with smaller bin widths had higher repeatability (median ICC of 8-128: 0.65-0.47). For both LRFS and OS, the performance of the models based on high- and low-repeatable RFs remained stable in the training set with similar C-index (LRFS: 0.65 vs 0.67, p = 0.958; OS: 0.64 vs 0.65, p = 0.651), while the performance of the model based on the low-repeatable group was significantly lower than that based on the high-repeatable group in the external validation set (LRFS: 0.61 vs 0.67, p = 0.013; OS: 0.56 vs 0.63, p = 0.013). CONCLUSIONS Applying high-repeatable RFs in modeling could safeguard the cross-institutional generalizability of the prognostic model in ESCC. CRITICAL RELEVANCE STATEMENT The exploration of repeatable RFs in different diseases and different types of imaging is conducive to promoting the proper use of radiomics in clinical research. KEY POINTS The repeatability of RFs impacts the generalizability of the radiomic model. The high-repeatable RFs safeguard the cross-institutional generalizability of the model. Smaller bin width helps improve the repeatability of RFs.
Collapse
Affiliation(s)
- Jie Gong
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qifeng Wang
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jie Li
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhi Yang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiang Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xinzhi Teng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Hongfei Sun
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Lina Zhao
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
5
|
Shiiba T, Watanabe M. Stability of radiomic features from positron emission tomography images: a phantom study comparing advanced reconstruction algorithms and ordered subset expectation maximization. Phys Eng Sci Med 2024; 47:929-937. [PMID: 38625624 DOI: 10.1007/s13246-024-01416-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/18/2024] [Indexed: 04/17/2024]
Abstract
In this study, we compared the repeatability and reproducibility of radiomic features obtained from positron emission tomography (PET) images according to the reconstruction algorithm used-advanced reconstruction algorithms, such as HYPER iterative (IT), HYPER deep learning reconstruction (DLR), and HYPER deep progressive reconstruction (DPR), or traditional Ordered Subset Expectation Maximization (OSEM)-to understand the potential variations and implications of using advanced reconstruction techniques in PET-based radiomics. We used a heterogeneous phantom with acrylic spherical beads (4- or 8-mm diameter) filled with 18F. PET images were acquired and reconstructed using OSEM, IT, DLR, and DPR. Original and wavelet radiomic features were calculated using SlicerRadiomics. Radiomic feature repeatability was assessed using the Coefficient of Variance (COV) and intraclass correlation coefficient (ICC), and inter-acquisition time reproducibility was assessed using the concordance correlation coefficient (CCC). For the 4- and 8-mm diameter beads phantom, the proportion of radiomic features with a COV < 10% was equivocal or higher for the advanced reconstruction algorithm than for OSEM. ICC indicated that advanced methods generally outperformed OSEM in repeatability, except for the original features of the 8-mm beads phantom. In the inter-acquisition time reproducibility analysis, the combinations of 3 and 5 min exhibited the highest reproducibility in both phantoms, with IT and DPR showing the highest proportion of radiomic features with CCC > 0.8. Advanced reconstruction methods provided enhanced stability of radiomic features compared with OSEM, suggesting their potential for optimal image reconstruction in PET-based radiomics, offering potential benefits in clinical diagnostics and prognostics.
Collapse
Affiliation(s)
- Takuro Shiiba
- Department of Molecular Imaging, Clinical and Educational Collaboration Unit, School of Medical Sciences, Fujita Health University, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| | - Masanori Watanabe
- Department of Radiology, Fujita Health University Hospital, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| |
Collapse
|
6
|
Teng X, Wang Y, Nicol AJ, Ching JCF, Wong EKY, Lam KTC, Zhang J, Lee SWY, Cai J. Enhancing the Clinical Utility of Radiomics: Addressing the Challenges of Repeatability and Reproducibility in CT and MRI. Diagnostics (Basel) 2024; 14:1835. [PMID: 39202322 PMCID: PMC11353986 DOI: 10.3390/diagnostics14161835] [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: 06/26/2024] [Revised: 08/03/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Radiomics, which integrates the comprehensive characterization of imaging phenotypes with machine learning algorithms, is increasingly recognized for its potential in the diagnosis and prognosis of oncological conditions. However, the repeatability and reproducibility of radiomic features are critical challenges that hinder their widespread clinical adoption. This review aims to address the paucity of discussion regarding the factors that influence the reproducibility and repeatability of radiomic features and their subsequent impact on the application of radiomic models. We provide a synthesis of the literature on the repeatability and reproducibility of CT/MR-based radiomic features, examining sources of variation, the number of reproducible features, and the availability of individual feature repeatability indices. We differentiate sources of variation into random effects, which are challenging to control but can be quantified through simulation methods such as perturbation, and biases, which arise from scanner variability and inter-reader differences and can significantly affect the generalizability of radiomic model performance in diverse settings. Four suggestions for repeatability and reproducibility studies are suggested: (1) detailed reporting of variation sources, (2) transparent disclosure of calculation parameters, (3) careful selection of suitable reliability indices, and (4) comprehensive reporting of reliability metrics. This review underscores the importance of random effects in feature selection and harmonizing biases between development and clinical application settings to facilitate the successful translation of radiomic models from research to clinical practice.
Collapse
Affiliation(s)
- Xinzhi Teng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Yongqiang Wang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Alexander James Nicol
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Jerry Chi Fung Ching
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Edwin Ka Yiu Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Kenneth Tsz Chun Lam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Jiang Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Shara Wee-Yee Lee
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
- Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
7
|
Scicolone R, Vacca S, Pisu F, Benson JC, Nardi V, Lanzino G, Suri JS, Saba L. Radiomics and artificial intelligence: General notions and applications in the carotid vulnerable plaque. Eur J Radiol 2024; 176:111497. [PMID: 38749095 DOI: 10.1016/j.ejrad.2024.111497] [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: 03/16/2024] [Revised: 04/14/2024] [Accepted: 05/03/2024] [Indexed: 06/17/2024]
Abstract
Carotid atherosclerosis plays a substantial role in cardiovascular morbidity and mortality. Given the multifaceted impact of this disease, there has been increasing interest in harnessing artificial intelligence (AI) and radiomics as complementary tools for the quantitative analysis of medical imaging data. This integrated approach holds promise not only in refining medical imaging data analysis but also in optimizing the utilization of radiologists' expertise. By automating time consuming tasks, AI allows radiologists to focus on more pertinent responsibilities. Simultaneously, the capacity of AI in radiomics to extract nuanced patterns from raw data enhances the exploration of carotid atherosclerosis, advancing efforts in terms of (1) early detection and diagnosis, (2) risk stratification and predictive modeling, (3) improving workflow efficiency, and (4) contributing to advancements in research. This review provides an overview of general concepts related to radiomics and AI, along with their application in the field of carotid vulnerable plaque. It also offers insights into various research studies conducted on this topic across different imaging techniques.
Collapse
Affiliation(s)
- Roberta Scicolone
- Department of Radiology, Azienda Ospedaliero-Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, Cagliari, Italy
| | - Sebastiano Vacca
- University of Cagliari, School of Medicine and Surgery, Cagliari, Italy
| | - Francesco Pisu
- Department of Radiology, Azienda Ospedaliero-Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, Cagliari, Italy
| | - John C Benson
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Valentina Nardi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, USA
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero-Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, Cagliari, Italy.
| |
Collapse
|
8
|
Brown KH, Kerr BN, Pettigrew M, Connor K, Miller IS, Shiels L, Connolly C, McGarry C, Byrne AT, Butterworth KT. A comparative analysis of preclinical computed tomography radiomics using cone-beam and micro-computed tomography scanners. Phys Imaging Radiat Oncol 2024; 31:100615. [PMID: 39157293 PMCID: PMC11328005 DOI: 10.1016/j.phro.2024.100615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/17/2024] [Accepted: 07/20/2024] [Indexed: 08/20/2024] Open
Abstract
Background and purpose Radiomics analysis extracts quantitative data (features) from medical images. These features could potentially reflect biological characteristics and act as imaging biomarkers within precision medicine. However, there is a lack of cross-comparison and validation of radiomics outputs which is paramount for clinical implementation. In this study, we compared radiomics outputs across two computed tomography (CT)-based preclinical scanners. Materials and methods Cone beam CT (CBCT) and µCT scans were acquired using different preclinical CT imaging platforms. The reproducibility of radiomics features on each scanner was assessed using a phantom across imaging energies (40 & 60 kVp) and segmentation volumes (44-238 mm3). Retrospective mouse scans were used to compare feature reliability across varying tissue densities (lung, heart, bone), scanners and after voxel size harmonisation. Reliable features had an intraclass correlation coefficient (ICC) > 0.8. Results First order and GLCM features were the most reliable on both scanners across different volumes. There was an inverse relationship between tissue density and feature reliability, with the highest number of features in lung (CBCT=580, µCT=734) and lowest in bone (CBCT=110, µCT=560). Comparable features for lung and heart tissues increased when voxel sizes were harmonised. We have identified tissue-specific preclinical radiomics signatures in mice for the lung (133), heart (35), and bone (15). Conclusions Preclinical CBCT and µCT scans can be used for radiomics analysis to support the development of meaningful radiomics signatures. This study demonstrates the importance of standardisation and emphasises the need for multi-centre studies.
Collapse
Affiliation(s)
- Kathryn H Brown
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Brianna N Kerr
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Mihaela Pettigrew
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Kate Connor
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Ian S Miller
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- National Preclinical Imaging Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Liam Shiels
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Colum Connolly
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Conor McGarry
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
- Northern Ireland Cancer Centre, Belfast Health & Social Care Trust, Belfast, United Kingdom
| | - Annette T Byrne
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- National Preclinical Imaging Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Karl T Butterworth
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
9
|
Koçak B, Yüzkan S, Mutlu S, Karagülle M, Kala A, Kadıoğlu M, Solak S, Sunman Ş, Temiz ZH, Ganiyusufoğlu AK. Influence of image preprocessing on the segmentation-based reproducibility of radiomic features: in vivo experiments on discretization and resampling parameters. Diagn Interv Radiol 2024; 30:152-162. [PMID: 38073244 PMCID: PMC11095065 DOI: 10.4274/dir.2023.232543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/14/2023] [Indexed: 05/15/2024]
Abstract
PURPOSE To systematically investigate the impact of image preprocessing parameters on the segmentation-based reproducibility of magnetic resonance imaging (MRI) radiomic features. METHODS The MRI scans of 50 patients were included from the multi-institutional Brain Tumor Segmentation 2021 public glioma dataset. Whole tumor volumes were manually segmented by two independent readers, with the participation of eight readers. Radiomic features were extracted from two sequences: T2-weighted (T2) and contrast-enhanced T1-weighted (T1ce). Two methods were considered for discretization: bin count (i.e., relative discretization) and bin width (i.e., absolute discretization). Ten discretization (five for each method) and five resampling parameters were varied while other parameters were fixed. The intraclass correlation coefficient (ICC) was used for reliability analysis based on two commonly used cut-off values (0.75 and 0.90). RESULTS Image preprocessing parameters had a significant impact on the segmentation-based reproducibility of radiomic features. The bin width method yielded more reproducible features than the bin count method. In discretization experiments using the bin width on both sequences, according to the ICC cut-off values of 0.75 and 0.90, the rate of reproducible features ranged from 70% to 84% and from 35% to 57%, respectively, with an increasing percentage trend as parameter values decreased (from 84 to 5 for T2; 100 to 6 for T1ce). In the resampling experiments, these ranged from 53% to 74% and from 10% to 20%, respectively, with an increasing percentage trend from lower to higher parameter values (physical voxel size; from 1 x 1 x 1 to 2 x 2 x 2 mm3). CONCLUSION The segmentation-based reproducibility of radiomic features appears to be substantially influenced by discretization and resampling parameters. Our findings indicate that the bin width method should be used for discretization and lower bin width and higher resampling values should be used to allow more reproducible features.
Collapse
Affiliation(s)
- Burak Koçak
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Sabahattin Yüzkan
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Samet Mutlu
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Mehmet Karagülle
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Ahmet Kala
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Mehmet Kadıoğlu
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Sıla Solak
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Şeyma Sunman
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Zişan Hayriye Temiz
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| | - Ali Kürşad Ganiyusufoğlu
- University of Health Sciences, Başakşehir Çam and Sakura City Hospital, Clinic of Radiology, İstanbul, Türkiye
| |
Collapse
|
10
|
Watzenboeck ML, Beer L, Kifjak D, Röhrich S, Heidinger BH, Prayer F, Milos RI, Apfaltrer P, Langs G, Baltzer PAT, Prosch H. Contrast Agent Dynamics Determine Radiomics Profiles in Oncologic Imaging. Cancers (Basel) 2024; 16:1519. [PMID: 38672601 PMCID: PMC11049400 DOI: 10.3390/cancers16081519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The reproducibility of radiomics features extracted from CT and MRI examinations depends on several physiological and technical factors. The aim was to evaluate the impact of contrast agent timing on the stability of radiomics features using dynamic contrast-enhanced perfusion CT (dceCT) or MRI (dceMRI) in prostate and lung cancers. METHODS Radiomics features were extracted from dceCT or dceMRI images in patients with biopsy-proven peripheral prostate cancer (pzPC) or biopsy-proven non-small cell lung cancer (NSCLC), respectively. Features that showed significant differences between contrast phases were identified using linear mixed models. An L2-penalized logistic regression classifier was used to predict class labels for pzPC and unaffected prostate regions-of-interest (ROIs). RESULTS Nine pzPC and 28 NSCLC patients, who were imaged with dceCT and/or dceMRI, were included in this study. After normalizing for individual enhancement patterns by defining seven individual phases based on a reference vessel, 19, 467 and 128 out of 1204 CT features showed significant temporal dynamics in healthy prostate parenchyma, prostate tumors and lung tumors, respectively. CT radiomics-based classification accuracy of healthy and tumor ROIs was highly dependent on contrast agent phase. For dceMRI, 899 and 1027 out of 1118 features were significantly dependent on time after contrast agent injection for prostate and lung tumors. CONCLUSIONS CT and MRI radiomics features in both prostate and lung tumors are significantly affected by interindividual differences in contrast agent dynamics.
Collapse
Affiliation(s)
- Martin L. Watzenboeck
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria (G.L.); (P.A.T.B.); (H.P.)
- Computational Imaging Research Lab, Christian Doppler Laboratory for Machine Learning Driven Precision Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Lucian Beer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria (G.L.); (P.A.T.B.); (H.P.)
- Computational Imaging Research Lab, Christian Doppler Laboratory for Machine Learning Driven Precision Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Daria Kifjak
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria (G.L.); (P.A.T.B.); (H.P.)
- Computational Imaging Research Lab, Christian Doppler Laboratory for Machine Learning Driven Precision Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Sebastian Röhrich
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria (G.L.); (P.A.T.B.); (H.P.)
- Computational Imaging Research Lab, Christian Doppler Laboratory for Machine Learning Driven Precision Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Benedikt H. Heidinger
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria (G.L.); (P.A.T.B.); (H.P.)
- Computational Imaging Research Lab, Christian Doppler Laboratory for Machine Learning Driven Precision Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Florian Prayer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria (G.L.); (P.A.T.B.); (H.P.)
- Computational Imaging Research Lab, Christian Doppler Laboratory for Machine Learning Driven Precision Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Ruxandra-Iulia Milos
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria (G.L.); (P.A.T.B.); (H.P.)
- Computational Imaging Research Lab, Christian Doppler Laboratory for Machine Learning Driven Precision Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Paul Apfaltrer
- Zentralröntgeninstitut für Diagnostik, Interventionelle Radiologie und Nuklearmedizin, Landesklinikum Wiener Neustadt, 2700 Wiener Neustadt, Austria
| | - Georg Langs
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria (G.L.); (P.A.T.B.); (H.P.)
- Computational Imaging Research Lab, Christian Doppler Laboratory for Machine Learning Driven Precision Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Pascal A. T. Baltzer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria (G.L.); (P.A.T.B.); (H.P.)
| | - Helmut Prosch
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria (G.L.); (P.A.T.B.); (H.P.)
- Computational Imaging Research Lab, Christian Doppler Laboratory for Machine Learning Driven Precision Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
11
|
van der Reijd DJ, Chupetlovska K, van Dijk E, Westerink B, Monraats MA, Van Griethuysen JJM, Lambregts DMJ, Tissier R, Beets-Tan RGH, Benson S, Maas M. Multi-sequence MRI radiomics of colorectal liver metastases: Which features are reproducible across readers? Eur J Radiol 2024; 172:111346. [PMID: 38309217 DOI: 10.1016/j.ejrad.2024.111346] [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: 11/17/2023] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
PURPOSE To assess the inter-reader reproducibility of radiomics features on multiple MRI sequences after segmentations of colorectal liver metastases (CRLM). METHOD 30 CRLM (in 23 patients) were manually delineated by three readers on MRI before the start of chemotherapy on the contrast enhanced T1-weighted images (CE-T1W) in the portal venous phase, T2-weighted images (T2W) and b800 diffusion weighted images (DWI). DWI delineations were copied to the ADC-maps. 107 radiomics features were extracted per sequence. The intraclass correlation coefficient (ICC) was calculated per feature. Features were considered reproducible if ICC > 0.9. RESULTS 90% of CE-T1W features were reproducible with a median ICC of 0.98 (range 0.76-1.00). 81% of DWI features were robust with median ICC = 0.97 (range 0.38-1.00). The T2W features had a median ICC of 0.96 (range 0.55-0.99) and were reproducible in 80%. ADC showed the lowest number of reproducible features with 58% and median ICC = 0.91 (range 0.38-0.99) When considering the lower bound of the ICC 95% confidence intervals, 58%, 66%, 54% and 29% reached 0.9 for the CE-T1W, DWI, T2W and ADC features, respectively. The feature class with the best reproducibility differed per sequence. CONCLUSIONS The majority of MRI radiomics features from CE-T1W, T2W, DWI and ADC in colorectal liver metastases were robust for segmentation variability between readers. The CE-T1W yielded slightly better reproducibility results compared to DWI and T2W. The ADC features seem more susceptible to reader differences compared to the other three sequences.
Collapse
Affiliation(s)
- Denise J van der Reijd
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Kalina Chupetlovska
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Eleanor van Dijk
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Bram Westerink
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Melanie A Monraats
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Joost J M Van Griethuysen
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Doenja M J Lambregts
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Renaud Tissier
- Biostatistics Center, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - Regina G H Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands; Faculty of Health Sciences, University of Southern Denmark, Campusvej 55, DK 5203 Odense, Denmark
| | - Sean Benson
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands; Department of Cardiology, Amsterdam University Medical Centres, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Monique Maas
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands.
| |
Collapse
|
12
|
Lee DY, Shin J, Kim S, Baek SE, Lee S, Son NH, Park MS. Radiomics model versus 2017 revised international consensus guidelines for predicting malignant intraductal papillary mucinous neoplasms. Eur Radiol 2024; 34:1222-1231. [PMID: 37615762 DOI: 10.1007/s00330-023-10158-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/05/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023]
Abstract
OBJECTIVES To evaluate a CT-based radiomics model for identifying malignant pancreatic intraductal papillary mucinous neoplasms (IPMNs) and compare its performance with the 2017 international consensus guidelines (ICGs). MATERIALS AND METHODS We retrospectively included 194 consecutive patients who underwent surgical resection of pancreatic IPMNs between January 2008 and December 2020. Surgical histopathology was the reference standard for diagnosing malignancy. Using radiomics features from preoperative contrast-enhanced CT, a radiomics model was built with the least absolute shrinkage and selection operator by a five-fold cross-validation. CT and MR images were independently reviewed based on the 2017 ICGs by two abdominal radiologists, and the performances of the 2017 ICGs and radiomics model were compared. The areas under the curve (AUCs) were compared using the DeLong method. RESULTS A total of 194 patients with pancreatic IPMNs (benign, 83 [43%]; malignant, 111 [57%]) were chronologically divided into training (n = 141; age, 65 ± 8.6 years; 88 males) and validation sets (n = 53; age, 66 ± 9.7 years; 31 males). There was no statistically significant difference in the diagnostic performance of the 2017 ICGs between CT and MRI (AUC, 0.71 vs. 0.71; p = 0.93) with excellent intermodality agreement (k = 0.86). In the validation set, the CT radiomics model had higher AUC (0.85 vs. 0.71; p = 0.038), specificity (84.6% vs. 61.5%; p = 0.041), and positive predictive value (84.0% vs. 66.7%; p = 0.044) than the 2017 ICGs. CONCLUSION The CT radiomics model exhibited better diagnostic performance than the 2017 ICGs in classifying malignant IPMNs. CLINICAL RELEVANCE STATEMENT Compared with the radiologists' evaluation based on the 2017 international consensus guidelines, the CT radiomics model exhibited better diagnostic performance in classifying malignant intraductal papillary mucinous neoplasms. KEY POINTS • There is a paucity of comparisons between the 2017 international consensus guidelines (ICGs) and radiomics models for malignant intraductal papillary mucinous neoplasms (IPMNs). • The CT radiomics model developed in this study exhibited better diagnostic performance than the 2017 ICGs in classifying malignant IPMNs. • The radiomics model may serve as a valuable complementary tool to the 2017 ICGs, potentially allowing a more quantitative assessment of IPMNs.
Collapse
Affiliation(s)
- Doo Young Lee
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jaeseung Shin
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University College of Medicine, 81 Irwon-Ro, Kangnam-Gu, Seoul, 06351, Republic of Korea.
| | - Sungwon Kim
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Song-Ee Baek
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Suji Lee
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Nak-Hoon Son
- Department of Statistics, Keimyung University, Daegu, Korea
| | - Mi-Suk Park
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Majumder S, Katz S, Kontos D, Roshkovan L. State of the art: radiomics and radiomics-related artificial intelligence on the road to clinical translation. BJR Open 2024; 6:tzad004. [PMID: 38352179 PMCID: PMC10860524 DOI: 10.1093/bjro/tzad004] [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: 02/15/2023] [Revised: 09/15/2023] [Accepted: 10/30/2023] [Indexed: 02/16/2024] Open
Abstract
Radiomics and artificial intelligence carry the promise of increased precision in oncologic imaging assessments due to the ability of harnessing thousands of occult digital imaging features embedded in conventional medical imaging data. While powerful, these technologies suffer from a number of sources of variability that currently impede clinical translation. In order to overcome this impediment, there is a need to control for these sources of variability through harmonization of imaging data acquisition across institutions, construction of standardized imaging protocols that maximize the acquisition of these features, harmonization of post-processing techniques, and big data resources to properly power studies for hypothesis testing. For this to be accomplished, it will be critical to have multidisciplinary and multi-institutional collaboration.
Collapse
Affiliation(s)
- Shweta Majumder
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| | - Sharyn Katz
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| | - Despina Kontos
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| | - Leonid Roshkovan
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| |
Collapse
|
14
|
Zhang J, Teng X, Zhang X, Lam SK, Lin Z, Liang Y, Yu H, Siu SWK, Chang ATY, Zhang H, Kong FM, Yang R, Cai J. Comparing effectiveness of image perturbation and test retest imaging in improving radiomic model reliability. Sci Rep 2023; 13:18263. [PMID: 37880324 PMCID: PMC10600245 DOI: 10.1038/s41598-023-45477-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023] Open
Abstract
Image perturbation is a promising technique to assess radiomic feature repeatability, but whether it can achieve the same effect as test-retest imaging on model reliability is unknown. This study aimed to compare radiomic model reliability based on repeatable features determined by the two methods using four different classifiers. A 191-patient public breast cancer dataset with 71 test-retest scans was used with pre-determined 117 training and 74 testing samples. We collected apparent diffusion coefficient images and manual tumor segmentations for radiomic feature extraction. Random translations, rotations, and contour randomizations were performed on the training images, and intra-class correlation coefficient (ICC) was used to filter high repeatable features. We evaluated model reliability in both internal generalizability and robustness, which were quantified by training and testing AUC and prediction ICC. Higher testing performance was found at higher feature ICC thresholds, but it dropped significantly at ICC = 0.95 for the test-retest model. Similar optimal reliability can be achieved with testing AUC = 0.7-0.8 and prediction ICC > 0.9 at the ICC threshold of 0.9. It is recommended to include feature repeatability analysis using image perturbation in any radiomic study when test-retest is not feasible, but care should be taken when deciding the optimal feature repeatability criteria.
Collapse
Affiliation(s)
- Jiang Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Y920, Lee Shau Kee Building, Hung Hom, Kowloon, Hong Kong, China
| | - Xinzhi Teng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Y920, Lee Shau Kee Building, Hung Hom, Kowloon, Hong Kong, China
| | - Xinyu Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Y920, Lee Shau Kee Building, Hung Hom, Kowloon, Hong Kong, China
| | - Sai-Kit Lam
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
- Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China
| | - Zhongshi Lin
- Shenzhen Institute for Drug Control (Shenzhen Testing Center of Medical Devices), Shenzhen, China
| | - Yongyi Liang
- Shenzhen Institute for Drug Control (Shenzhen Testing Center of Medical Devices), Shenzhen, China
| | - Hao Yu
- Institute of Biomedical and Health Engineering, Chinese Academy of Sciences Shenzhen Institutes of Advanced Technology, Shenzhen, China
| | - Steven Wai Kwan Siu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Amy Tien Yee Chang
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Hua Zhang
- Beijing Linking Medical Technology Co., Ltd., Beijing, China
| | - Feng-Ming Kong
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ruijie Yang
- Department of Radiation Oncology, Cancer Center, Peking University Third Hospital, Beijing, China
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Y920, Lee Shau Kee Building, Hung Hom, Kowloon, Hong Kong, China.
- Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
15
|
Singh A, Roshkovan L, Thompson JC, Kontos D, Katz SI. Are radiomic signatures ready for incorporation in the clinical pipeline? Transl Lung Cancer Res 2023; 12:1845-1849. [PMID: 37854152 PMCID: PMC10579831 DOI: 10.21037/tlcr-23-502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/01/2023] [Indexed: 10/20/2023]
Affiliation(s)
- Apurva Singh
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Leonid Roshkovan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey C. Thompson
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Thoracic Oncology Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Despina Kontos
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sharyn I. Katz
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
16
|
Tan D, Mohamad Salleh SA, Manan HA, Yahya N. Delta-radiomics-based models for toxicity prediction in radiotherapy: A systematic review and meta-analysis. J Med Imaging Radiat Oncol 2023; 67:564-579. [PMID: 37309680 DOI: 10.1111/1754-9485.13546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/28/2023] [Indexed: 06/14/2023]
Abstract
INTRODUCTION Delta-radiomics models are potentially able to improve the treatment assessment than single-time point features. The purpose of this study is to systematically synthesize the performance of delta-radiomics-based models for radiotherapy (RT)-induced toxicity. METHODS A literature search was performed following the PRISMA guidelines. Systematic searches were performed in PubMed, Scopus, Cochrane and Embase databases in October 2022. Retrospective and prospective studies on the delta-radiomics model for RT-induced toxicity were included based on predefined PICOS criteria. A random-effect meta-analysis of AUC was performed on the performance of delta-radiomics models, and a comparison with non-delta radiomics models was included. RESULTS Of the 563 articles retrieved, 13 selected studies of RT-treated patients on different types of cancer (HNC = 571, NPC = 186, NSCLC = 165, oesophagus = 106, prostate = 33, OPC = 21) were eligible for inclusion in the systematic review. Included studies show that morphological and dosimetric features may improve the predictive model performance for the selected toxicity. Four studies that reported both delta and non-delta radiomics features with AUC were included in the meta-analysis. The AUC random effects estimate for delta and non-delta radiomics models were 0.80 and 0.78 with heterogeneity, I2 of 73% and 27% respectively. CONCLUSION Delta-radiomics-based models were found to be promising predictors of predefined end points. Future studies should consider using standardized methods and radiomics features and external validation to the reviewed delta-radiomics model.
Collapse
Affiliation(s)
- Daryl Tan
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | - Hanani Abdul Manan
- Functional Image Processing Laboratory, Department of Radiology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Noorazrul Yahya
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
17
|
Abe K, Kadoya N, Ito K, Tanaka S, Nakajima Y, Hashimoto S, Suda Y, Uno T, Jingu K. Evaluation of the MVCT-based radiomic features as prognostic factor in patients with head and neck squamous cell carcinoma. BMC Med Imaging 2023; 23:102. [PMID: 37528392 PMCID: PMC10391970 DOI: 10.1186/s12880-023-01055-w] [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: 12/23/2022] [Accepted: 07/18/2023] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND Megavoltage computed tomography (MVCT) images acquired during each radiotherapy session may be useful for delta radiomics. However, no studies have examined whether the MVCT-based radiomics has prognostic power. Therefore, the purpose of this study was to examine the prognostic power of the MVCT-based radiomics for head and neck squamous cell carcinoma (HNSCC) patients. METHODS 100 HNSCC patients who received definitive radiotherapy were analyzed and divided into two groups: training (n = 70) and test (n = 30) sets. MVCT images obtained using TomoTherapy for the first fraction of radiotherapy and planning kilovoltage CT (kVCT) images obtained using Aquilion LB CT scanner were analyzed. Primary gross tumor volume (GTV) was propagated from kVCT to MVCT images using rigid registration, and 107 radiomic features were extracted from the GTV in MVCT and kVCT images. Least absolute shrinkage and selection operator (LASSO) Cox regression model was used to examine the association between overall survival (OS) and rad score calculated for each patient by weighting the feature value through the coefficient when features were selected. Then, the predictive values of MVCT-based and kVCT-based rad score and patient-, treatment-, and tumor-specific factors were evaluated. RESULTS C-indices of the rad score for MVCT- and kVCT-based radiomics were 0.667 and 0.685, respectively. The C-indices of 6 clinical factors were 0.538-0.622. The 3-year OS was significantly different between high- and low-risk groups according to the MVCT-based rad score (50% vs. 83%; p < 0.01). CONCLUSIONS Our results suggested that MVCT-based radiomics had stronger prognostic power than any single clinical factor and was a useful prognostic factor when predicting OS in HNSCC patients.
Collapse
Affiliation(s)
- Kota Abe
- Department of Radiation Oncology, MR Linac ART Division, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
- Department of Radiation Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo- machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Noriyuki Kadoya
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo- machi, Aoba-ku, Sendai, 980-8574, Japan.
| | - Kei Ito
- Department of Radiation Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Shohei Tanaka
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo- machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Yujiro Nakajima
- Department of Radiation Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
- Department of Radiological Sciences, Komazawa University, 1-23-1 komazawa, Setagaya-ku, Tokyo, 154-8525, Japan
| | - Shimpei Hashimoto
- Saitama Prefectural Cancer Center, 780 large section of a town Omuro, Ina-machi, Kitaadachi- gun, Saitama, 362-0806, Japan
| | - Yuhi Suda
- Department of Radiation Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Takashi Uno
- Diagnostic Radiology and Radiation Oncology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Keiichi Jingu
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo- machi, Aoba-ku, Sendai, 980-8574, Japan
| |
Collapse
|
18
|
Chaddad A, Tan G, Liang X, Hassan L, Rathore S, Desrosiers C, Katib Y, Niazi T. Advancements in MRI-Based Radiomics and Artificial Intelligence for Prostate Cancer: A Comprehensive Review and Future Prospects. Cancers (Basel) 2023; 15:3839. [PMID: 37568655 PMCID: PMC10416937 DOI: 10.3390/cancers15153839] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The use of multiparametric magnetic resonance imaging (mpMRI) has become a common technique used in guiding biopsy and developing treatment plans for prostate lesions. While this technique is effective, non-invasive methods such as radiomics have gained popularity for extracting imaging features to develop predictive models for clinical tasks. The aim is to minimize invasive processes for improved management of prostate cancer (PCa). This study reviews recent research progress in MRI-based radiomics for PCa, including the radiomics pipeline and potential factors affecting personalized diagnosis. The integration of artificial intelligence (AI) with medical imaging is also discussed, in line with the development trend of radiogenomics and multi-omics. The survey highlights the need for more data from multiple institutions to avoid bias and generalize the predictive model. The AI-based radiomics model is considered a promising clinical tool with good prospects for application.
Collapse
Affiliation(s)
- Ahmad Chaddad
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
- The Laboratory for Imagery, Vision and Artificial Intelligence, École de Technologie Supérieure (ETS), Montreal, QC H3C 1K3, Canada
| | - Guina Tan
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
| | - Xiaojuan Liang
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
| | - Lama Hassan
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
| | | | - Christian Desrosiers
- The Laboratory for Imagery, Vision and Artificial Intelligence, École de Technologie Supérieure (ETS), Montreal, QC H3C 1K3, Canada
| | - Yousef Katib
- Department of Radiology, Taibah University, Al Madinah 42361, Saudi Arabia
| | - Tamim Niazi
- Lady Davis Institute for Medical Research, McGill University, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
19
|
Zhang YP, Zhang XY, Cheng YT, Li B, Teng XZ, Zhang J, Lam S, Zhou T, Ma ZR, Sheng JB, Tam VCW, Lee SWY, Ge H, Cai J. Artificial intelligence-driven radiomics study in cancer: the role of feature engineering and modeling. Mil Med Res 2023; 10:22. [PMID: 37189155 DOI: 10.1186/s40779-023-00458-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/04/2023] [Indexed: 05/17/2023] Open
Abstract
Modern medicine is reliant on various medical imaging technologies for non-invasively observing patients' anatomy. However, the interpretation of medical images can be highly subjective and dependent on the expertise of clinicians. Moreover, some potentially useful quantitative information in medical images, especially that which is not visible to the naked eye, is often ignored during clinical practice. In contrast, radiomics performs high-throughput feature extraction from medical images, which enables quantitative analysis of medical images and prediction of various clinical endpoints. Studies have reported that radiomics exhibits promising performance in diagnosis and predicting treatment responses and prognosis, demonstrating its potential to be a non-invasive auxiliary tool for personalized medicine. However, radiomics remains in a developmental phase as numerous technical challenges have yet to be solved, especially in feature engineering and statistical modeling. In this review, we introduce the current utility of radiomics by summarizing research on its application in the diagnosis, prognosis, and prediction of treatment responses in patients with cancer. We focus on machine learning approaches, for feature extraction and selection during feature engineering and for imbalanced datasets and multi-modality fusion during statistical modeling. Furthermore, we introduce the stability, reproducibility, and interpretability of features, and the generalizability and interpretability of models. Finally, we offer possible solutions to current challenges in radiomics research.
Collapse
Affiliation(s)
- Yuan-Peng Zhang
- Department of Medical Informatics, Nantong University, Nantong, 226001, Jiangsu, China
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, Guangdong, China
| | - Xin-Yun Zhang
- Department of Medical Informatics, Nantong University, Nantong, 226001, Jiangsu, China
| | - Yu-Ting Cheng
- Department of Medical Informatics, Nantong University, Nantong, 226001, Jiangsu, China
| | - Bing Li
- Department of Radiation Oncology, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, Henan, China
| | - Xin-Zhi Teng
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Jiang Zhang
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Saikit Lam
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Ta Zhou
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Zong-Rui Ma
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Jia-Bao Sheng
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Victor C W Tam
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Shara W Y Lee
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Hong Ge
- Department of Radiation Oncology, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, Henan, China
| | - Jing Cai
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China.
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
20
|
Liu Y, Wei X, Feng X, Liu Y, Feng G, Du Y. Repeatability of radiomics studies in colorectal cancer: a systematic review. BMC Gastroenterol 2023; 23:125. [PMID: 37059990 PMCID: PMC10105401 DOI: 10.1186/s12876-023-02743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 03/22/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Recently, radiomics has been widely used in colorectal cancer, but many variable factors affect the repeatability of radiomics research. This review aims to analyze the repeatability of radiomics studies in colorectal cancer and to evaluate the current status of radiomics in the field of colorectal cancer. METHODS The included studies in this review by searching from the PubMed and Embase databases. Then each study in our review was evaluated using the Radiomics Quality Score (RQS). We analyzed the factors that may affect the repeatability in the radiomics workflow and discussed the repeatability of the included studies. RESULTS A total of 188 studies was included in this review, of which only two (2/188, 1.06%) studies controlled the influence of individual factors. In addition, the median score of RQS was 11 (out of 36), range-1 to 27. CONCLUSIONS The RQS score was moderately low, and most studies did not consider the repeatability of radiomics features, especially in terms of Intra-individual, scanners, and scanning parameters. To improve the generalization of the radiomics model, it is necessary to further control the variable factors of repeatability.
Collapse
Affiliation(s)
- Ying Liu
- School of Medical Imaging, North Sichuan Medical College, Sichuan Province, Nanchong City, 637000, China
| | - Xiaoqin Wei
- School of Medical Imaging, North Sichuan Medical College, Sichuan Province, Nanchong City, 637000, China
| | | | - Yan Liu
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China
| | - Guiling Feng
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China
| | - Yong Du
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China.
| |
Collapse
|
21
|
Brown KH, Payan N, Osman S, Ghita M, Walls GM, Patallo IS, Schettino G, Prise KM, McGarry CK, Butterworth KT. Development and optimisation of a preclinical cone beam computed tomography-based radiomics workflow for radiation oncology research. Phys Imaging Radiat Oncol 2023; 26:100446. [PMID: 37252250 PMCID: PMC10213103 DOI: 10.1016/j.phro.2023.100446] [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: 02/24/2023] [Revised: 05/03/2023] [Accepted: 05/08/2023] [Indexed: 05/31/2023] Open
Abstract
Background and purpose Radiomics features derived from medical images have the potential to act as imaging biomarkers to improve diagnosis and predict treatment response in oncology. However, the complex relationships between radiomics features and the biological characteristics of tumours are yet to be fully determined. In this study, we developed a preclinical cone beam computed tomography (CBCT) radiomics workflow with the aim to use in vivo models to further develop radiomics signatures. Materials and methods CBCT scans of a mouse phantom were acquired using onboard imaging from a small animal radiotherapy research platform (SARRP, Xstrahl). The repeatability and reproducibility of radiomics outputs were compared across different imaging protocols, segmentation sizes, pre-processing parameters and materials. Robust features were identified and used to compare scans of two xenograft mouse tumour models (A549 and H460). Results Changes to the radiomics workflow significantly impact feature robustness. Preclinical CBCT radiomics analysis is feasible with 119 stable features identified from scans imaged at 60 kV, 25 bin width and 0.26 mm slice thickness. Large variation in segmentation volumes reduced the number of reliable radiomics features for analysis. Standardization in imaging and analysis parameters is essential in preclinical radiomics analysis to improve accuracy of outputs, leading to more consistent and reproducible findings. Conclusions We present the first optimised workflow for preclinical CBCT radiomics to identify imaging biomarkers. Preclinical radiomics has the potential to maximise the quantity of data captured in in vivo experiments and could provide key information supporting the wider application of radiomics.
Collapse
Affiliation(s)
- Kathryn H. Brown
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Northern Ireland, UK
| | - Neree Payan
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Northern Ireland, UK
| | - Sarah Osman
- University College London Hospitals NHS Foundation Trust Department of Radiotherapy, London, UK
| | - Mihaela Ghita
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Northern Ireland, UK
| | - Gerard M. Walls
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Northern Ireland, UK
- Cancer Centre, Belfast Health & Social Care Trust, Lisburn Road, Belfast BT9 7AB, Northern Ireland, UK
| | | | | | - Kevin M. Prise
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Northern Ireland, UK
| | - Conor K. McGarry
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Northern Ireland, UK
- Cancer Centre, Belfast Health & Social Care Trust, Lisburn Road, Belfast BT9 7AB, Northern Ireland, UK
| | - Karl T. Butterworth
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Northern Ireland, UK
| |
Collapse
|
22
|
Park S, Kim JH, Kim J, Joseph W, Lee D, Park SJ. Development of a deep learning-based auto-segmentation algorithm for hepatocellular carcinoma (HCC) and application to predict microvascular invasion of HCC using CT texture analysis: preliminary results. Acta Radiol 2023; 64:907-917. [PMID: 35570797 DOI: 10.1177/02841851221100318] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Automatic segmentation has recently been developed to yield objective data. Prediction of microvascular invasion (MVI) of hepatocellular carcinoma (HCC) using radiomics has been reported. PURPOSE To develop a deep learning-based auto-segmentation algorithm (DL-AS) for the detection of HCC and to predict MVI using computed tomography (CT) texture analysis. MATERIAL AND METHODS We retrospectively collected training data from 249 patients with HCC and validation set from 35 patients. Lesions of the training set were manually drawn by radiologist, in the delayed phase. 2D U-Net was selected as the DL architecture. Using the validation set, one radiologist manually drew 2D and 3D regions of interest twice, and the developed DL-AS was performed twice with a one-month time interval. The reproducibility was calculated using intraclass correlation coefficients (ICC). Logistic regression was performed to predict MVI. RESULTS ICC was in the range of 0.190-0.998/0.341-0.997 in the manual 3D/2D segmentation. In contrast, it was perfect in 3D/2D using DL-AS, with a success rate of 88.6% for the detection of HCC. For predicting MVI, sphericity was a significant parameter (odds ratio <0.001; 95% confidence interval <0.001-0.206; P = 0.020) for predicting MVI using 2D DL-AS. However, 3D DL-AS segmentation did not yield a predictive parameter. CONCLUSION The auto-segmentation of HCC using DL-AS provides perfect reproducibility, although it failed to detect 11.4% (4/35). However, the extracted parameters yielded different important predictors of MVI in HCC. Sphericity was a significant predictor in 2D DL-AS and 3D manual segmentation, while discrete compactness was a significant predictor in 2D manual segmentation.
Collapse
Affiliation(s)
- Sungeun Park
- Department of Radiology, 119754Konkuk University Medical Center, Seoul, Republic of Korea
| | - Jung Hoon Kim
- Department of Radiology, 58927Seoul National University Hospital, Seoul, Republic of Korea
- Department of Radiology, 37990Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jieun Kim
- Department of Radiology, 58927Seoul National University Hospital, Seoul, Republic of Korea
| | | | - Doohee Lee
- Medical IP Co., Ltd, Seoul, Republic of Korea
| | | |
Collapse
|
23
|
Zhang J, Lam SK, Teng X, Ma Z, Han X, Zhang Y, Cheung ALY, Chau TC, Ng SCY, Lee FKH, Au KH, Yip CWY, Lee VHF, Han Y, Cai J. Radiomic feature repeatability and its impact on prognostic model generalizability: A multi-institutional study on nasopharyngeal carcinoma patients. Radiother Oncol 2023; 183:109578. [PMID: 36822357 DOI: 10.1016/j.radonc.2023.109578] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND AND PURPOSE To investigate the radiomic feature (RF) repeatability via perturbation and its impact on cross-institutional prognostic model generalizability in Nasopharyngeal Carcinoma (NPC) patients. MATERIALS AND METHODS 286 and 183 NPC patients from two institutions were included for model training and validation. Perturbations with random translations and rotations were applied to contrast-enhanced T1-weighted (CET1-w) MR images. RFs were extracted from primary tumor volume under a wide range of image filtering and discretization settings. RF repeatability was assessed by intraclass correlation coefficient (ICC), which was used to equally separate the RFs into low- and high-repeatable groups by the median value. After feature selection, multivariate Cox regression and Kaplan-Meier analysis were independently employed to develop and analyze prognostic models. Concordance index (C-index) and P-value from log-rank test were used to assess model performance. RESULTS Most textural RFs from high-pass wavelet-filtered images were susceptible to image perturbations. It was more prominent when a smaller discretization bin number was used (e.g., 8, mean ICC = 0.69). Using high-repeatable RFs for model development yielded a significantly higher C-index (0.63) in the validation cohort than when only low-repeatable RFs were used (0.57, P = 0.024), suggesting higher model generalizability. Besides, significant risk stratification in the validation cohort was observed only when high-repeatable RFs were used (P < 0.001). CONCLUSION Repeatability of RFs from high-pass wavelet-filtered CET1-w MR images of primary NPC tumor was poor, particularly when a smaller bin number was used. Exclusive use of high-repeatable RFs is suggested to safeguard model generalizability for wide-spreading clinical utilization.
Collapse
Affiliation(s)
- Jiang Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Sai-Kit Lam
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China; Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xinzhi Teng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Zongrui Ma
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xinyang Han
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yuanpeng Zhang
- Department of Medical Informatics, Nantong University, Nantong, Jiangsu, China
| | - Andy Lai-Yin Cheung
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Tin-Ching Chau
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong, China
| | - Sherry Chor-Yi Ng
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong, China
| | - Francis Kar-Ho Lee
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Kwok-Hung Au
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Celia Wai-Yi Yip
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Victor Ho-Fun Lee
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Ying Han
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
24
|
Huang EP, O'Connor JPB, McShane LM, Giger ML, Lambin P, Kinahan PE, Siegel EL, Shankar LK. Criteria for the translation of radiomics into clinically useful tests. Nat Rev Clin Oncol 2023; 20:69-82. [PMID: 36443594 PMCID: PMC9707172 DOI: 10.1038/s41571-022-00707-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2022] [Indexed: 11/29/2022]
Abstract
Computer-extracted tumour characteristics have been incorporated into medical imaging computer-aided diagnosis (CAD) algorithms for decades. With the advent of radiomics, an extension of CAD involving high-throughput computer-extracted quantitative characterization of healthy or pathological structures and processes as captured by medical imaging, interest in such computer-extracted measurements has increased substantially. However, despite the thousands of radiomic studies, the number of settings in which radiomics has been successfully translated into a clinically useful tool or has obtained FDA clearance is comparatively small. This relative dearth might be attributable to factors such as the varying imaging and radiomic feature extraction protocols used from study to study, the numerous potential pitfalls in the analysis of radiomic data, and the lack of studies showing that acting upon a radiomic-based tool leads to a favourable benefit-risk balance for the patient. Several guidelines on specific aspects of radiomic data acquisition and analysis are already available, although a similar roadmap for the overall process of translating radiomics into tools that can be used in clinical care is needed. Herein, we provide 16 criteria for the effective execution of this process in the hopes that they will guide the development of more clinically useful radiomic tests in the future.
Collapse
Affiliation(s)
- Erich P Huang
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, MD, USA.
| | - James P B O'Connor
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Lisa M McShane
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | | | - Philippe Lambin
- Department of Precision Medicine, Maastricht University, Maastricht, Netherlands
| | - Paul E Kinahan
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Eliot L Siegel
- Department of Diagnostic Radiology, University of Maryland, Baltimore, MD, USA
| | - Lalitha K Shankar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
25
|
Keyl J, Hosch R, Berger A, Ester O, Greiner T, Bogner S, Treckmann J, Ting S, Schumacher B, Albers D, Markus P, Wiesweg M, Forsting M, Nensa F, Schuler M, Kasper S, Kleesiek J. Deep learning-based assessment of body composition and liver tumour burden for survival modelling in advanced colorectal cancer. J Cachexia Sarcopenia Muscle 2023; 14:545-552. [PMID: 36544260 PMCID: PMC9891942 DOI: 10.1002/jcsm.13158] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/16/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Personalized therapy planning remains a significant challenge in advanced colorectal cancer care, despite extensive research on prognostic and predictive markers. A strong correlation of sarcopenia or overall body composition and survival has been described. Here, we explore whether automated assessment of body composition and liver metastases from standard of care CT images can add to clinical parameters in personalized survival risk prognostication. METHODS We retrospectively analysed clinical imaging data from 85 patients (50.6% female, mean age 58.9 SD 12.2 years) with colorectal cancer and synchronous liver metastases. Pretrained deep learning models were used to assess body composition and liver metastasis geometry from abdominal CT images before the initiation of systemic treatment. Abdominal muscle-to-bone ratio (MBR) was calculated by dividing abdominal muscle volume by abdominal bone volume. MBR was compared with body mass index (BMI), abdominal muscle volume, and abdominal muscle volume divided by height squared. Differences in overall survival based on body composition and liver metastasis parameters were compared using Kaplan-Meier survival curves. Results were correlated with clinical and biomarker data to develop a machine learning model for survival risk prognostication. RESULTS The MBR, unlike abdominal muscle volume or BMI, was significantly associated with overall survival (HR 0.39, 95% CI: 0.19-0.80, P = 0.009). The MBR (P = 0.022), liver metastasis surface area (P = 0.01) and primary tumour sidedness (P = 0.007) were independently associated with overall survival in multivariate analysis. Body composition parameters did not correlate with KRAS mutational status or primary tumour sidedness. A prediction model based on MBR, liver metastasis surface area and primary tumour sidedness achieved a concordance index of 0.69. CONCLUSIONS Automated segmentation enables to extract prognostic parameters from routine imaging data for personalized survival modelling in advanced colorectal cancer patients.
Collapse
Affiliation(s)
- Julius Keyl
- Department of Medical Oncology, West German Cancer CenterUniversity Hospital Essen (AöR)EssenGermany
- Institute for Artificial Intelligence in MedicineUniversity Hospital Essen (AöR)EssenGermany
- German Cancer Consortium (DKTK)Partner site University Hospital Essen (AöR)EssenGermany
| | - René Hosch
- Institute for Artificial Intelligence in MedicineUniversity Hospital Essen (AöR)EssenGermany
- Department of Diagnostic and Interventional Radiology and NeuroradiologyUniversity Hospital Essen (AöR)EssenGermany
| | - Aaron Berger
- Institute for Artificial Intelligence in MedicineUniversity Hospital Essen (AöR)EssenGermany
| | - Oliver Ester
- Institute for Artificial Intelligence in MedicineUniversity Hospital Essen (AöR)EssenGermany
| | | | - Simon Bogner
- Department of Medical Oncology, West German Cancer CenterUniversity Hospital Essen (AöR)EssenGermany
| | - Jürgen Treckmann
- Department of General, Visceral and Transplant Surgery, West German Cancer CenterUniversity Hospital Essen (AöR)EssenGermany
| | - Saskia Ting
- Institute of Pathology EssenWest German Cancer Center, University Hospital Essen (AöR)EssenGermany
| | | | - David Albers
- Department of GastroenterologyElisabeth Hospital EssenEssenGermany
| | - Peter Markus
- Department of General Surgery and TraumatologyElisabeth Hospital EssenEssenGermany
| | - Marcel Wiesweg
- Department of Medical Oncology, West German Cancer CenterUniversity Hospital Essen (AöR)EssenGermany
- Medical FacultyUniversity of Duisburg‐EssenEssenGermany
| | - Michael Forsting
- Department of Diagnostic and Interventional Radiology and NeuroradiologyUniversity Hospital Essen (AöR)EssenGermany
| | - Felix Nensa
- Institute for Artificial Intelligence in MedicineUniversity Hospital Essen (AöR)EssenGermany
- Department of Diagnostic and Interventional Radiology and NeuroradiologyUniversity Hospital Essen (AöR)EssenGermany
| | - Martin Schuler
- Department of Medical Oncology, West German Cancer CenterUniversity Hospital Essen (AöR)EssenGermany
- German Cancer Consortium (DKTK)Partner site University Hospital Essen (AöR)EssenGermany
- Medical FacultyUniversity of Duisburg‐EssenEssenGermany
| | - Stefan Kasper
- Department of Medical Oncology, West German Cancer CenterUniversity Hospital Essen (AöR)EssenGermany
- German Cancer Consortium (DKTK)Partner site University Hospital Essen (AöR)EssenGermany
- Medical FacultyUniversity of Duisburg‐EssenEssenGermany
| | - Jens Kleesiek
- Institute for Artificial Intelligence in MedicineUniversity Hospital Essen (AöR)EssenGermany
- Medical FacultyUniversity of Duisburg‐EssenEssenGermany
| |
Collapse
|
26
|
Zhang Y, Hu HH, Zhou SH, Xia WY, Zhang Y, Zhang JP, Fu XL, Yu W. PET-based radiomics visualizes tumor-infiltrating CD8 T cell exhaustion to optimize radiotherapy/immunotherapy combination in mouse models of lung cancer. Biomark Res 2023; 11:10. [PMID: 36694213 PMCID: PMC9875413 DOI: 10.1186/s40364-023-00454-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Cumulative preclinical and clinical evidences showed radiotherapy might augment systemic antitumoral responses to immunotherapy for metastatic non-small cell lung cancer, but the optimal timing of combination is still unclear. The overall infiltration and exhausted subpopulations of tumor-infiltrating CD8+ T cells might be a potential biomarker indicating the response to immune checkpoint inhibitors (ICI), the alteration of which is previously uncharacterized during peri-irradiation period, while dynamic monitoring is unavailable via repeated biopsies in clinical practice. METHODS Basing on tumor-bearing mice model, we investigated the dynamics of overall infiltration and exhausted subpopulations of CD8+ T cells after ablative irradiation. With the understanding of distinct metabolic characteristics accompanied with T cell exhaustion, we developed a PET radiomics approach to identify and visualize T cell exhaustion status. RESULTS CD8+ T cell infiltration increased from 3 to 14 days after ablative irradiation while terminally exhausted populations significantly predominated CD8+ T cells during late course of this infiltrating period, indicating that 3-7 days post-irradiation might be a potential appropriate window for delivering ICI treatment. A PET radiomics approach was established to differentiate T cell exhaustion status, which fitted well in both ICI and irradiation settings. We also visualized the underlying association of more heterogeneous texture on PET images with progressed T cell exhaustion. CONCLUSIONS We proposed a non-invasive imaging predictor which accurately assessed heterogeneous T cell exhaustion status relevant to ICI treatment and irradiation, and might serve as a promising solution to timely estimate immune-responsiveness of tumor microenvironment and the optimal timing of combined therapy.
Collapse
Affiliation(s)
- Ying Zhang
- grid.412524.40000 0004 0632 3994Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200030 China
| | - Hui-Hui Hu
- grid.412524.40000 0004 0632 3994Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200030 China
| | - Shi-Hong Zhou
- grid.412524.40000 0004 0632 3994Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wu-Yan Xia
- grid.412524.40000 0004 0632 3994Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200030 China
| | - Yan Zhang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Ping Zhang
- grid.452404.30000 0004 1808 0942Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiao-Long Fu
- grid.412524.40000 0004 0632 3994Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200030 China
| | - Wen Yu
- grid.412524.40000 0004 0632 3994Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200030 China
| |
Collapse
|
27
|
Koçak B, Cuocolo R, dos Santos DP, Stanzione A, Ugga L. Must-have Qualities of Clinical Research on Artificial Intelligence and Machine Learning. Balkan Med J 2023; 40:3-12. [PMID: 36578657 PMCID: PMC9874249 DOI: 10.4274/balkanmedj.galenos.2022.2022-11-51] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/06/2022] [Indexed: 12/30/2022] Open
Abstract
In the field of computer science, known as artificial intelligence, algorithms imitate reasoning tasks that are typically performed by humans. The techniques that allow machines to learn and get better at tasks such as recognition and prediction, which form the basis of clinical practice, are referred to as machine learning, which is a subfield of artificial intelligence. The number of artificial intelligence-and machine learnings-related publications in clinical journals has grown exponentially, driven by recent developments in computation and the accessibility of simple tools. However, clinicians are often not included in data science teams, which may limit the clinical relevance, explanability, workflow compatibility, and quality improvement of artificial intelligence solutions. Thus, this results in the language barrier between clinicians and artificial intelligence developers. Healthcare practitioners sometimes lack a basic understanding of artificial intelligence research because the approach is difficult for non-specialists to understand. Furthermore, many editors and reviewers of medical publications might not be familiar with the fundamental ideas behind these technologies, which may prevent journals from publishing high-quality artificial intelligence studies or, worse still, could allow for the publication of low-quality works. In this review, we aim to improve readers’ artificial intelligence literacy and critical thinking. As a result, we concentrated on what we consider the 10 most important qualities of artificial intelligence research: valid scientific purpose, high-quality data set, robust reference standard, robust input, no information leakage, optimal bias-variance tradeoff, proper model evaluation, proven clinical utility, transparent reporting, and open science. Before designing a study, one should have defined a sound scientific purpose. Then, it should be backed by a high-quality data set, robust input, and a solid reference standard. The artificial intelligence development pipeline should prevent information leakage. For the models, optimal bias-variance tradeoff should be achieved, and generalizability assessment must be adequately performed. The clinical value of the final models must also be established. After the study, thought should be given to transparency in publishing the process and results as well as open science for sharing data, code, and models. We hope this work may improve the artificial intelligence literacy and mindset of the readers.
Collapse
Affiliation(s)
- Burak Koçak
- Clinic of Radiology, University of Health Sciences Turkey, Başakşehir Çam and Sakura City Hospital, İstanbul, Turkey
| | - Renato Cuocolo
- Department of Medicine, Surgery and Dentistry University of Salerno, Baronissi, Italy
| | - Daniel Pinto dos Santos
- Department of Radiology, University Hospital of Cologne, Cologne, Germany
- Department of Radiology, University Hospital of Frankfurt, Frankfurt, Germany
| | - Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Napoli, Italy
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Napoli, Italy
| |
Collapse
|
28
|
Prayer F, Watzenböck ML, Heidinger BH, Rainer J, Schmidbauer V, Prosch H, Ulm B, Rubesova E, Prayer D, Kasprian G. Fetal MRI radiomics: non-invasive and reproducible quantification of human lung maturity. Eur Radiol 2023; 33:4205-4213. [PMID: 36604329 PMCID: PMC10182107 DOI: 10.1007/s00330-022-09367-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/26/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023]
Abstract
OBJECTIVES To assess the reproducibility of radiomics features extracted from the developing lung in repeated in-vivo fetal MRI acquisitions. METHODS In-vivo MRI (1.5 Tesla) scans of 30 fetuses, each including two axial and one coronal T2-weighted sequences of the whole lung with all other acquisition parameters kept constant, were retrospectively identified. Manual segmentation of the lungs was performed using ITK-Snap. One hundred radiomics features were extracted from fetal lung MRI data using Pyradiomics, resulting in 90 datasets. Intra-class correlation coefficients (ICC) of radiomics features were calculated between baseline and repeat axial acquisitions and between baseline axial and coronal acquisitions. RESULTS MRI data of 30 fetuses (12 [40%] females, 18 [60%] males) at a median gestational age of 24 + 5 gestational weeks plus days (GW) (interquartile range [IQR] 3 + 3 GW, range 21 + 1 to 32 + 6 GW) were included. Median ICC of radiomics features between baseline and repeat axial MR acquisitions was 0.92 (IQR 0.13, range 0.33 to 1), with 60 features exhibiting excellent (ICC > 0.9), 27 good (> 0.75-0.9), twelve moderate (0.5-0.75), and one poor (ICC < 0.5) reproducibility. Median ICC of radiomics features between baseline axial and coronal MR acquisitions was 0.79 (IQR 0.15, range 0.2 to 1), with 20 features exhibiting excellent, 47 good, 29 moderate, and four poor reproducibility. CONCLUSION Standardized in-vivo fetal MRI allows reproducible extraction of lung radiomics features. In the future, radiomics analysis may improve diagnostic and prognostic yield of fetal MRI in normal and pathologic lung development. KEY POINTS • Non-invasive fetal MRI acquired using a standardized protocol allows reproducible extraction of radiomics features from the developing lung for objective tissue characterization. • Alteration of imaging plane between fetal MRI acquisitions has a negative impact on lung radiomics feature reproducibility. • Fetal MRI radiomics features reflecting the microstructure and shape of the fetal lung could complement observed-to-expected lung volume in the prediction of postnatal outcome and optimal treatment of fetuses with abnormal lung development in the future.
Collapse
Affiliation(s)
- Florian Prayer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Martin L Watzenböck
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Benedikt H Heidinger
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Julian Rainer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Victor Schmidbauer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Helmut Prosch
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Barbara Ulm
- Department of Obstetrics and Gynecology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Vienna, Austria
| | - Erika Rubesova
- Department of Pediatric Radiology, Lucile Packard Children's Hospital at Stanford, Stanford University, 725 Welch Road, Stanford, CA, 94305, USA
| | - Daniela Prayer
- Imaging Bellaria, Bellariastrasse 3, 1010, Vienna, Austria
| | - Gregor Kasprian
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria.
| |
Collapse
|
29
|
Singh A, Horng H, Chitalia R, Roshkovan L, Katz SI, Noël P, Shinohara RT, Kontos D. Resampling and harmonization for mitigation of heterogeneity in image parameters of baseline scans. Sci Rep 2022; 12:21505. [PMID: 36513760 PMCID: PMC9747915 DOI: 10.1038/s41598-022-26083-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Our study investigates the effects of heterogeneity in image parameters on the reproducibility of prognostic performance of models built using radiomic biomarkers. We compare the prognostic performance of models derived from the heterogeneity-mitigated features with that of models obtained from raw features, to assess whether reproducibility of prognostic scores improves upon application of our methods. We used two datasets: The Breast I-SPY1 dataset-Baseline DCE-MRI scans of 156 women with locally advanced breast cancer, treated with neoadjuvant chemotherapy, publicly available via The Cancer Imaging Archive (TCIA); The NSCLC IO dataset-Baseline CT scans of 107 patients with stage 4 non-small cell lung cancer (NSCLC), treated with pembrolizumab immunotherapy at our institution. Radiomic features (n = 102) are extracted from the tumor ROIs. We use a variety of resampling and harmonization scenarios to mitigate the heterogeneity in image parameters. The patients were divided into groups based on batch variables. For each group, the radiomic phenotypes are combined with the clinical covariates into a prognostic model. The performance of the groups is assessed using the c-statistic, derived from a Cox proportional hazards model fitted on all patients within a group. The heterogeneity-mitigation scenario (radiomic features, derived from images that have been resampled to minimum voxel spacing, are harmonized using the image acquisition parameters as batch variables) gave models with highest prognostic scores (for e.g., IO dataset; batch variable: high kernel resolution-c-score: 0.66). The prognostic performance of patient groups is not comparable in case of models built using non-heterogeneity mitigated features (for e.g., I-SPY1 dataset; batch variable: small pixel spacing-c-score: 0.54, large pixel spacing-c-score: 0.65). The prognostic performance of patient groups is closer in case of heterogeneity-mitigated scenarios (for e.g., scenario-harmonize by voxel spacing parameters: IO dataset; thin slice-c-score: 0.62, thick slice-c-score: 0.60). Our results indicate that accounting for heterogeneity in image parameters is important to obtain more reproducible prognostic scores, irrespective of image site or modality. For non-heterogeneity mitigated models, the prognostic scores are not comparable across patient groups divided based on batch variables. This study can be a step in the direction of constructing reproducible radiomic biomarkers, thus increasing their application in clinical decision making.
Collapse
Affiliation(s)
- Apurva Singh
- grid.25879.310000 0004 1936 8972Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104 USA ,grid.25879.310000 0004 1936 8972Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Hannah Horng
- grid.25879.310000 0004 1936 8972Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Rhea Chitalia
- grid.25879.310000 0004 1936 8972Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104 USA ,grid.25879.310000 0004 1936 8972Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Leonid Roshkovan
- grid.25879.310000 0004 1936 8972Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Sharyn I. Katz
- grid.25879.310000 0004 1936 8972Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Peter Noël
- grid.25879.310000 0004 1936 8972Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Russell T. Shinohara
- grid.25879.310000 0004 1936 8972Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Despina Kontos
- grid.25879.310000 0004 1936 8972Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104 USA ,grid.25879.310000 0004 1936 8972Department of Radiology, Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Rm D702 Richards Bldg., 3700 Hamilton Walk, Philadelphia, PA 19104 USA
| |
Collapse
|
30
|
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
|
31
|
Christie JR, Daher O, Abdelrazek M, Romine PE, Malthaner RA, Qiabi M, Nayak R, Napel S, Nair VS, Mattonen SA. Predicting recurrence risks in lung cancer patients using multimodal radiomics and random survival forests. J Med Imaging (Bellingham) 2022; 9:066001. [PMID: 36388142 PMCID: PMC9641263 DOI: 10.1117/1.jmi.9.6.066001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022] Open
Abstract
Purpose We developed a model integrating multimodal quantitative imaging features from tumor and nontumor regions, qualitative features, and clinical data to improve the risk stratification of patients with resectable non-small cell lung cancer (NSCLC). Approach We retrospectively analyzed 135 patients [mean age, 69 years (43 to 87, range); 100 male patients and 35 female patients] with NSCLC who underwent upfront surgical resection between 2008 and 2012. The tumor and peritumoral regions on both preoperative CT and FDG PET-CT and the vertebral bodies L3 to L5 on FDG PET were segmented to assess the tumor and bone marrow uptake, respectively. Radiomic features were extracted and combined with clinical and CT qualitative features. A random survival forest model was developed using the top-performing features to predict the time to recurrence/progression in the training cohort ( n = 101 ), validated in the testing cohort ( n = 34 ) using the concordance, and compared with a stage-only model. Patients were stratified into high- and low-risks of recurrence/progression using Kaplan-Meier analysis. Results The model, consisting of stage, three wavelet texture features, and three wavelet first-order features, achieved a concordance of 0.78 and 0.76 in the training and testing cohorts, respectively, significantly outperforming the baseline stage-only model results of 0.67 ( p < 0.005 ) and 0.60 ( p = 0.008 ), respectively. Patients at high- and low-risks of recurrence/progression were significantly stratified in both the training ( p < 0.005 ) and the testing ( p = 0.03 ) cohorts. Conclusions Our radiomic model, consisting of stage and tumor, peritumoral, and bone marrow features from CT and FDG PET-CT significantly stratified patients into low- and high-risk of recurrence/progression.
Collapse
Affiliation(s)
- Jaryd R. Christie
- Western University, Department of Medical Biophysics, London, Ontario, Canada
- London Regional Cancer Program, Baines Imaging Research Laboratory, London, Ontario, Canada
| | - Omar Daher
- Western University, Department of Medical Imaging, London, Ontario, Canada
| | - Mohamed Abdelrazek
- Western University, Department of Medical Imaging, London, Ontario, Canada
| | - Perrin E. Romine
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington, United States
- University of Washington School of Medicine, Division of Medical Oncology, Seattle, Washington, United States
| | - Richard A. Malthaner
- Western University, Division of Thoracic Surgery, Department of Surgery, London, Ontario, Canada
| | - Mehdi Qiabi
- Western University, Division of Thoracic Surgery, Department of Surgery, London, Ontario, Canada
| | - Rahul Nayak
- Western University, Division of Thoracic Surgery, Department of Surgery, London, Ontario, Canada
| | - Sandy Napel
- Stanford University, Department of Radiology, Stanford, California, United States
| | - Viswam S. Nair
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington, United States
- University of Washington School of Medicine, Division of Pulmonary and Critical Care Medicine, Seattle, Washington, United States
| | - Sarah A. Mattonen
- Western University, Department of Medical Biophysics, London, Ontario, Canada
- London Regional Cancer Program, Baines Imaging Research Laboratory, London, Ontario, Canada
- Western University, Department of Oncology, London, Ontario, Canada
| |
Collapse
|
32
|
Ge G, Zhang J. Uniqueness of radiomic features in non-small cell lung cancer. J Appl Clin Med Phys 2022; 23:e13787. [PMID: 36173022 PMCID: PMC9797180 DOI: 10.1002/acm2.13787] [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: 02/08/2022] [Revised: 07/26/2022] [Accepted: 08/24/2022] [Indexed: 01/01/2023] Open
Abstract
PURPOSE The uniqueness of radiomic features, combined with their reproducibility, determines the reliability of radiomic studies. This study is to test the hypothesis that radiomic features extracted from a defined region of interest (ROI) are unique to the underlying structure (e.g., tumor). APPROACH Two cohorts of non-small cell lung cancer (NSCLC) patients were retrospectively retrieved from a GE and a Siemens CT scanner. The lung nodules (ROI) were delineated manually and radiomic features were extracted using IBEX. The same ROI was then translocated randomly to four other tissue regions of the same set of images: adipose, heart, lung beyond nodule, and muscle for radiomic feature extraction. Coefficient of variation (CV) within different ROIs and concordance correlation coefficient (CCC) between lung nodule and a given tissue region were calculated to test to determine feature uniqueness. The radiomic features were considered nonunique when (1) the CV < 10% and CCC > 0.85 for over 50% of patients; and (2) the CCC > 0.85 appeared in ≥2 tissue regions beyond the defined region. RESULTS In total, 14 patients from GE and 18 patients from Siemens are analyzed. The results show that 12 features fall below the 10% CV threshold for over 50% of patients in the GE cohort and 29 features in the Siemens cohort. According to CCC, 18 radiomic features in GE and 16 features in Siemens are identified as nonunique, with 11 overlapping features. Combining CV and CCC, 9 of 123 calculated features (7.3%) are identified as nonunique to a defined ROI. CONCLUSIONS Radiomic feature uniqueness should be considered to improve the reliability of radiomics study.
Collapse
Affiliation(s)
- Gary Ge
- Department of RadiologyUniversity of KentuckyLexingtonKentucky40536USA
| | - Jie Zhang
- Department of RadiologyUniversity of KentuckyLexingtonKentucky40536USA
| |
Collapse
|
33
|
Horvat N, Miranda J, El Homsi M, Peoples JJ, Long NM, Simpson AL, Do RKG. A primer on texture analysis in abdominal radiology. Abdom Radiol (NY) 2022; 47:2972-2985. [PMID: 34825946 DOI: 10.1007/s00261-021-03359-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 01/18/2023]
Abstract
The number of publications on texture analysis (TA), radiomics, and radiogenomics has been growing exponentially, with abdominal radiologists aiming to build new prognostic or predictive biomarkers for a wide range of clinical applications including the use of oncological imaging to advance the field of precision medicine. TA is specifically concerned with the study of the variation of pixel intensity values in radiological images. Radiologists aim to capture pixel variation in radiological images to deliver new insights into tumor biology that cannot be derived from visual inspection alone. TA remains an active area of investigation and requires further standardization prior to its clinical acceptance and applicability. This review is for radiologists interested in this rapidly evolving field, who are thinking of performing research or want to better interpret results in this arena. We will review the main concepts in TA, workflow processes, and existing challenges and steps to overcome them, as well as look at publications in body imaging with external validation.
Collapse
Affiliation(s)
- Natally Horvat
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Joao Miranda
- Department of Radiology, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Maria El Homsi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Jacob J Peoples
- School of Computing, Queen's University, Kingston, ON, Canada
| | - Niamh M Long
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Amber L Simpson
- School of Computing, Queen's University, Kingston, ON, Canada.,Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Richard K G Do
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
| |
Collapse
|
34
|
Ren Q, Xiong F, Zhu P, Chang X, Wang G, He N, Jin Q. Assessing the robustness of radiomics/deep learning approach in the identification of efficacy of anti–PD-1 treatment in advanced or metastatic non-small cell lung carcinoma patients. Front Oncol 2022; 12:952749. [PMID: 35992867 PMCID: PMC9390967 DOI: 10.3389/fonc.2022.952749] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/04/2022] [Indexed: 12/11/2022] Open
Abstract
Administration of anti–PD-1 is now a standard therapy in advanced non-small cell lung carcinoma (NSCLC) patients. The clinical application of biomarkers reflecting tumor immune microenvironment is hurdled by the invasiveness of obtaining tissues despite its importance in immunotherapy. This study aimed to develop a robust and non-invasive radiomics/deep learning machine biomarker for predicting the response to immunotherapy in NSCLC patients. Radiomics/deep learning features were exacted from computed tomography (CT) images of NSCLC patients treated with Nivolumab or Pembrolizumab. The robustness of radiomics/deep learning features was assessed against various perturbations, then robust features were selected based on the Intraclass Correlation Coefficient (ICC). Radiomics/deep learning machine-learning classifiers were constructed by combining seven feature exactors, 13 feature selection methods, and 12 classifiers. The optimal model was selected using the mean area under the curve (AUC) and relative standard deviation (RSD). The consistency of image features against various perturbations was high (the range of median ICC: 0.78–0.97), but the consistency was poor in test–retest testing (the range of median ICC: 0.42–0.67). The optimal model, InceptionV3_RELF_Nearest Neighbors classifiers, had the highest prediction efficacy (AUC: 0.96 and RSD: 0.50) for anti–PD-1/PD-L1 treatment. Accuracy (ACC), sensitivity, specificity, precision, and F1 score were 95.24%, 95.00%, 95.50%, 91.67%, and 95.30%, respectively. For successful model robustification, tailoring perturbations for robustness testing to the target dataset is key. Robust radiomics/deep learning features, when paired with machine-learning methodologies, will work on the exactness and the repeatability of anticipating immunotherapy adequacy.
Collapse
Affiliation(s)
- Qianqian Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fu Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhu
- Department of Hepatobiliary Surgery, Wuhan No.1 Hospital, Wuhan, China
| | - Xiaona Chang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nan He
- Cancer Center, Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Nan He, ; Qianna Jin,
| | - Qianna Jin
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Nan He, ; Qianna Jin,
| |
Collapse
|
35
|
Bao J, Feng X, Ma Y, Wang Y, Qi J, Qin C, Tan X, Tian Y. The latest application progress of radiomics in prediction and diagnosis of liver diseases. Expert Rev Gastroenterol Hepatol 2022; 16:707-719. [PMID: 35880549 DOI: 10.1080/17474124.2022.2104711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Early detection and individualized treatment of patients with liver disease is the key to survival. Radiomics can extract high-throughput quantitative features by multimode imaging, which has good application prospects for the diagnosis, staging and prognosis of benign and malignant liver diseases. Therefore, this paper summarizes the current research status in the field of liver disease, in order to help these patients achieve personalized and precision medical care. AREAS COVERED This paper uses several keywords on the PubMed database to search the references, and reviews the workflow of traditional radiomics, as well as the characteristics and influencing factors of different imaging modes. At the same time, the references on the application of imaging in different benign and malignant liver diseases were also summarized. EXPERT OPINION For patients with liver disease, the traditional imaging evaluation can only provide limited information. Radiomics exploits the characteristics of high-throughput and high-dimensional extraction, enabling liver imaging capabilities far beyond the scope of traditional visual image analysis. Recent studies have demonstrated the prospect of this technology in personalized diagnosis and treatment decision in various fields of the liver. However, further clinical validation is needed in its application and practice.
Collapse
Affiliation(s)
- Jiaying Bao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Xiao Feng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Yan Ma
- Department of Ultrasound, Zibo Central Hospital, Zibo, P.R. China
| | - Yanyan Wang
- Departments of Emergency Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Jianni Qi
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Chengyong Qin
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Xu Tan
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Yongmei Tian
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| |
Collapse
|
36
|
Zhang JZ, Ganesh H, Raslau FD, Nair R, Escott E, Wang C, Wang G, Zhang J. Deep learning versus iterative reconstruction on image quality and dose reduction in abdominal CT: a live animal study. Phys Med Biol 2022; 67. [DOI: 10.1088/1361-6560/ac7999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/16/2022] [Indexed: 11/12/2022]
Abstract
Abstract
Objective. While simulated low-dose CT images and phantom studies cannot fully approximate subjective and objective effects of deep learning (DL) denoising on image quality, live animal models may afford this assessment. This study is to investigate the potential of DL in CT dose reduction on image quality compared to iterative reconstruction (IR). Approach. The upper abdomen of a live 4 year old sheep was scanned on a CT scanner at different exposure levels. Images were reconstructed using FBP and ADMIRE with 5 strengths. A modularized DL network with 5 modules was used for image reconstruction via progressive denoising. Radiomic features were extracted from a region over the liver. Concordance correlation coefficient (CCC) was applied to quantify agreement between any two sets of radiomic features. Coefficient of variation was calculated to measure variation in a radiomic feature series. Structural similarity index (SSIM) was used to measure the similarity between any two images. Diagnostic quality, low-contrast detectability, and image texture were qualitatively evaluated by two radiologists. Pearson correlation coefficient was computed across all dose-reconstruction/denoising combinations. Results. A total of 66 image sets, with 405 radiomic features extracted from each, are analyzed. IR and DL can improve diagnostic quality and low-contrast detectability and similarly modulate image texture features. In terms of SSIM, DL has higher potential in preserving image structure. There is strong correlation between SSIM and radiologists’ evaluations for diagnostic quality (0.559) and low-contrast detectability (0.635) but moderate correlation for texture (0.313). There is moderate correlation between CCC of radiomic features and radiologists’ evaluation for diagnostic quality (0.397), low-contrast detectability (0.417), and texture (0.326), implying that improvement of image features may not relate to improvement of diagnostic quality. Conclusion. DL shows potential to further reduce radiation dose while preserving structural similarity, while IR is favored by radiologists and more predictably alters radiomic features.
Collapse
|
37
|
Thrussell I, Winfield JM, Orton MR, Miah AB, Zaidi SH, Arthur A, Thway K, Strauss DC, Collins DJ, Koh DM, Oelfke U, Huang PH, O’Connor JPB, Messiou C, Blackledge MD. Radiomic Features From Diffusion-Weighted MRI of Retroperitoneal Soft-Tissue Sarcomas Are Repeatable and Exhibit Change After Radiotherapy. Front Oncol 2022; 12:899180. [PMID: 35924167 PMCID: PMC9343063 DOI: 10.3389/fonc.2022.899180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Background Size-based assessments are inaccurate indicators of tumor response in soft-tissue sarcoma (STS), motivating the requirement for new response imaging biomarkers for this rare and heterogeneous disease. In this study, we assess the test-retest repeatability of radiomic features from MR diffusion-weighted imaging (DWI) and derived maps of apparent diffusion coefficient (ADC) in retroperitoneal STS and compare baseline repeatability with changes in radiomic features following radiotherapy (RT). Materials and Methods Thirty patients with retroperitoneal STS received an MR examination prior to treatment, of whom 23/30 were investigated in our repeatability analysis having received repeat baseline examinations and 14/30 patients were investigated in our post-treatment analysis having received an MR examination after completing pre-operative RT. One hundred and seven radiomic features were extracted from the full manually delineated tumor region using PyRadiomics. Test-retest repeatability was assessed using an intraclass correlation coefficient (baseline ICC), and post-radiotherapy variance analysis (post-RT-IMS) was used to compare the change in radiomic feature value to baseline repeatability. Results For the ADC maps and DWI images, 101 and 102 features demonstrated good baseline repeatability (baseline ICC > 0.85), respectively. Forty-three and 2 features demonstrated both good baseline repeatability and a high post-RT-IMS (>0.85), respectively. Pearson correlation between the baseline ICC and post-RT-IMS was weak (0.432 and 0.133, respectively). Conclusions The ADC-based radiomic analysis shows better test-retest repeatability compared with features derived from DWI images in STS, and some of these features are sensitive to post-treatment change. However, good repeatability at baseline does not imply sensitivity to post-treatment change.
Collapse
Affiliation(s)
- Imogen Thrussell
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | - Jessica M. Winfield
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | - Matthew R. Orton
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | - Aisha B. Miah
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Sarcoma Unit, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Shane H. Zaidi
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Sarcoma Unit, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Amani Arthur
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | - Khin Thway
- Sarcoma Unit, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
- Department of Histopathology, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Dirk C. Strauss
- Department of Surgery, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - David J. Collins
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | - Dow-Mu Koh
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | - Uwe Oelfke
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Paul H. Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - James P. B. O’Connor
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Radiology, The Christie Hospital, Manchester, United Kingdom
| | - Christina Messiou
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | - Matthew D. Blackledge
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| |
Collapse
|
38
|
The Potential and Emerging Role of Quantitative Imaging Biomarkers for Cancer Characterization. Cancers (Basel) 2022; 14:cancers14143349. [PMID: 35884409 PMCID: PMC9321521 DOI: 10.3390/cancers14143349] [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: 06/05/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Modern, personalized therapy approaches are increasingly changing advanced cancer into a chronic disease. Compared to imaging, novel omics methodologies in molecular biology have already achieved an individual characterization of cancerous lesions. With quantitative imaging biomarkers, analyzed by radiomics or deep learning, an imaging-based assessment of tumoral biology can be brought into clinical practice. Combining these with other non-invasive methods, e.g., liquid profiling, could allow for more individual decision making regarding therapies and applications. Abstract Similar to the transformation towards personalized oncology treatment, emerging techniques for evaluating oncologic imaging are fostering a transition from traditional response assessment towards more comprehensive cancer characterization via imaging. This development can be seen as key to the achievement of truly personalized and optimized cancer diagnosis and treatment. This review gives a methodological introduction for clinicians interested in the potential of quantitative imaging biomarkers, treating of radiomics models, texture visualization, convolutional neural networks and automated segmentation, in particular. Based on an introduction to these methods, clinical evidence for the corresponding imaging biomarkers—(i) dignity and etiology assessment; (ii) tumoral heterogeneity; (iii) aggressiveness and response; and (iv) targeting for biopsy and therapy—is summarized. Further requirements for the clinical implementation of these imaging biomarkers and the synergistic potential of personalized molecular cancer diagnostics and liquid profiling are discussed.
Collapse
|
39
|
Rai R, Barton MB, Chlap P, Liney G, Brink C, Vinod S, Heinke M, Trada Y, Holloway LC. Repeatability and reproducibility of magnetic resonance imaging-based radiomic features in rectal cancer. J Med Imaging (Bellingham) 2022; 9:044005. [PMID: 35992729 PMCID: PMC9386367 DOI: 10.1117/1.jmi.9.4.044005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 08/09/2022] [Indexed: 08/20/2023] Open
Abstract
Purpose: Radiomics of magnetic resonance images (MRIs) in rectal cancer can non-invasively characterize tumor heterogeneity with potential to discover new imaging biomarkers. However, for radiomics to be reliable, the imaging features measured must be stable and reproducible. The aim of this study is to quantify the repeatability and reproducibility of MRI-based radiomic features in rectal cancer. Approach: An MRI radiomics phantom was used to measure the longitudinal repeatability of radiomic features and the impact of post-processing changes related to image resolution and noise. Repeatability measurements in rectal cancers were also quantified in a cohort of 10 patients with test-retest imaging among two observers. Results: We found that many radiomic features, particularly from texture classes, were highly sensitive to changes in image resolution and noise. About 49% of features had coefficient of variations ≤ 10 % in longitudinal phantom measurements. About 75% of radiomic features in in vivo test-retest measurements had an intraclass correlation coefficient of ≥ 0.8 . We saw excellent interobserver agreement with mean Dice similarity coefficient of 0.95 ± 0.04 for test and retest scans. Conclusions: The results of this study show that even when using a consistent imaging protocol many radiomic features were unstable. Therefore, caution must be taken when selecting features for potential imaging biomarkers.
Collapse
Affiliation(s)
- Robba Rai
- University of New South Wales, South Western Sydney Clinical School, Liverpool, New South Wales, Australia
- Liverpool Hospital, Liverpool and Macarthur Cancer Therapy Centre, Liverpool, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Michael B. Barton
- University of New South Wales, South Western Sydney Clinical School, Liverpool, New South Wales, Australia
- Liverpool Hospital, Liverpool and Macarthur Cancer Therapy Centre, Liverpool, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Phillip Chlap
- University of New South Wales, South Western Sydney Clinical School, Liverpool, New South Wales, Australia
- Liverpool Hospital, Liverpool and Macarthur Cancer Therapy Centre, Liverpool, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Gary Liney
- University of New South Wales, South Western Sydney Clinical School, Liverpool, New South Wales, Australia
- Liverpool Hospital, Liverpool and Macarthur Cancer Therapy Centre, Liverpool, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Carsten Brink
- Odense University Hospital, Laboratory of Radiation Physics, Department of Oncology, Odense, Denmark
- University of Southern Denmark, Department of Clinical Research, Odense, Denmark
| | - Shalini Vinod
- University of New South Wales, South Western Sydney Clinical School, Liverpool, New South Wales, Australia
- Liverpool Hospital, Liverpool and Macarthur Cancer Therapy Centre, Liverpool, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | | | - Yuvnik Trada
- Calvary Mater Newcastle, Department of Radiation Oncology, Newcastle, New South Wales, Australia
| | - Lois C. Holloway
- University of New South Wales, South Western Sydney Clinical School, Liverpool, New South Wales, Australia
- Liverpool Hospital, Liverpool and Macarthur Cancer Therapy Centre, Liverpool, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- University of Wollongong, Centre of Radiation Physics, Wollongong, New South Wales, Australia
- University of Sydney, Institute of Medical Physics, School of Physics, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Teng X, Zhang J, Zwanenburg A, Sun J, Huang Y, Lam S, Zhang Y, Li B, Zhou T, Xiao H, Liu C, Li W, Han X, Ma Z, Li T, Cai J. Building reliable radiomic models using image perturbation. Sci Rep 2022; 12:10035. [PMID: 35710850 PMCID: PMC9203573 DOI: 10.1038/s41598-022-14178-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/16/2022] [Indexed: 02/06/2023] Open
Abstract
Radiomic model reliability is a central premise for its clinical translation. Presently, it is assessed using test–retest or external data, which, unfortunately, is often scarce in reality. Therefore, we aimed to develop a novel image perturbation-based method (IPBM) for the first of its kind toward building a reliable radiomic model. We first developed a radiomic prognostic model for head-and-neck cancer patients on a training (70%) and evaluated on a testing (30%) cohort using C-index. Subsequently, we applied the IPBM to CT images of both cohorts (Perturbed-Train and Perturbed-Test cohort) to generate 60 additional samples for both cohorts. Model reliability was assessed using intra-class correlation coefficient (ICC) to quantify consistency of the C-index among the 60 samples in the Perturbed-Train and Perturbed-Test cohorts. Besides, we re-trained the radiomic model using reliable RFs exclusively (ICC > 0.75) to validate the IPBM. Results showed moderate model reliability in Perturbed-Train (ICC: 0.565, 95%CI 0.518–0.615) and Perturbed-Test (ICC: 0.596, 95%CI 0.527–0.670) cohorts. An enhanced reliability of the re-trained model was observed in Perturbed-Train (ICC: 0.782, 95%CI 0.759–0.815) and Perturbed-Test (ICC: 0.825, 95%CI 0.782–0.867) cohorts, indicating validity of the IPBM. To conclude, we demonstrated capability of the IPBM toward building reliable radiomic models, providing community with a novel model reliability assessment strategy prior to prospective evaluation.
Collapse
Affiliation(s)
- Xinzhi Teng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Jiang Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Alex Zwanenburg
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Jiachen Sun
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Yuhua Huang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Saikit Lam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Yuanpeng Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Bing Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Ta Zhou
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Haonan Xiao
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Chenyang Liu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Wen Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Xinyang Han
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Zongrui Ma
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Tian Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China.
| |
Collapse
|
41
|
Ren Q, Zhu P, Li C, Yan M, Liu S, Zheng C, Xia X. Pretreatment Computed Tomography-Based Machine Learning Models to Predict Outcomes in Hepatocellular Carcinoma Patients who Received Combined Treatment of Trans-Arterial Chemoembolization and Tyrosine Kinase Inhibitor. Front Bioeng Biotechnol 2022; 10:872044. [PMID: 35677305 PMCID: PMC9168370 DOI: 10.3389/fbioe.2022.872044] [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: 02/09/2022] [Accepted: 04/22/2022] [Indexed: 11/15/2022] Open
Abstract
Aim: Trans-arterial chemoembolization (TACE) in combination with tyrosine kinase inhibitor (TKI) has been evidenced to improve outcomes in a portion of patients with hepatocellular carcinoma (HCC). Developing biomarkers to identify patients who might benefit from the combined treatment is needed. This study aims to investigate the efficacy of radiomics/deep learning features-based models in predicting short-term disease control and overall survival (OS) in HCC patients who received the combined treatment. Materials and Methods: A total of 103 HCC patients who received the combined treatment from Sep. 2015 to Dec. 2019 were enrolled in the study. We exacted radiomics features and deep learning features of six pre-trained convolutional neural networks (CNNs) from pretreatment computed tomography (CT) images. The robustness of features was evaluated, and those with excellent stability were used to construct predictive models by combining each of the seven feature exactors, 13 feature selection methods and 12 classifiers. The models were evaluated for predicting short-term disease by using the area under the receiver operating characteristics curve (AUC) and relative standard deviation (RSD). The optimal models were further analyzed for predictive performance on overall survival. Results: A total of the 1,092 models (156 with radiomics features and 936 with deep learning features) were constructed. Radiomics_GINI_Nearest Neighbors (RGNN) and Resnet50_MIM_Nearest Neighbors (RMNN) were identified as optimal models, with the AUC of 0.87 and 0.94, accuracy of 0.89 and 0.92, sensitivity of 0.88 and 0.97, specificity of 0.90 and 0.90, precision of 0.87 and 0.83, F1 score of 0.89 and 0.92, and RSD of 1.30 and 0.26, respectively. Kaplan-Meier survival analysis showed that RGNN and RMNN were associated with better OS (p = 0.006 for RGNN and p = 0.033 for RMNN). Conclusion: Pretreatment CT-based radiomics/deep learning models could non-invasively and efficiently predict outcomes in HCC patients who received combined therapy of TACE and TKI.
Collapse
Affiliation(s)
- Qianqian Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Peng Zhu
- Department of Hepatobiliary Surgery, Wuhan No.1 Hospital, Wuhan, China
| | - Changde Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Meijun Yan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Song Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiangwen Xia
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- *Correspondence: Xiangwen Xia,
| |
Collapse
|
42
|
Deep Learning-Based Image Conversion Improves the Reproducibility of Computed Tomography Radiomics Features: A Phantom Study. Invest Radiol 2022; 57:308-317. [PMID: 34839305 DOI: 10.1097/rli.0000000000000839] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVES This study aimed to evaluate the usefulness of deep learning-based image conversion to improve the reproducibility of computed tomography (CT) radiomics features. MATERIALS AND METHODS This study was conducted using an abdominal phantom with liver nodules. We developed an image conversion algorithm using a residual feature aggregation network to reproduce radiomics features with CT images under various CT protocols and reconstruction kernels. External validation was performed using images from different scanners, consisting of 8 different protocols. To evaluate the variability of radiomics features, regions of interest (ROIs) were drawn by targeting the liver parenchyma, vessels, paraspinal area, and liver nodules. We extracted 18 first-order, 68 second-order, and 688 wavelet radiomics features. Measurement variability was assessed using the concordance correlation coefficient (CCC), compared with the ground-truth image. RESULTS In the ROI-based analysis, there was an 83.3% improvement of CCC (80/96; 4 ROIs with 3 categories of radiomics features and 8 protocols) in synthetic images compared with the original images. Among them, the 56 CCC pairs showed a significant increase after image synthesis. In the radiomics feature-based analysis, 62.0% (3838 of 6192; 774 radiomics features with 8 protocols) features showed increased CCC after image synthesis, and a significant increase was noted in 26.9% (1663 of 6192) features. In particular, the first-order feature (79.9%, 115/144) showed better improvement in terms of the reproducibility of radiomics feature than the second-order (59.9%, 326/544) or wavelet feature (61.7%, 3397/5504). CONCLUSIONS Our study demonstrated that a deep learning model for image conversion can improve the reproducibility of radiomics features across various CT protocols, reconstruction kernels, and CT scanners.
Collapse
|
43
|
Abdali SH, Afzali F, Baseri S, Abdalvand N, Abdollahi H. Bone radiomics reproducibility: a three-centered study on the impacts of image contrast, edge enhancement, and latitude variations. Phys Eng Sci Med 2022; 45:497-511. [PMID: 35389137 DOI: 10.1007/s13246-022-01116-4] [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: 08/02/2021] [Accepted: 03/01/2022] [Indexed: 11/25/2022]
Abstract
This study aims to measure the reproducibility of radiomics features in ankle bone radiography over changes in post-processing parameters including contrast, edge enhancement and latitude. Lateral ankle bone radiographies for sixty patients were obtained from three digital radiology centers. All images were acquired by same image acquisition settings. A two-dimensional region of interest was drawn in any image and 93 features from 6 feature sets including first and second order were extracted. The coefficient of variation (COV) and intraclass correlation coefficient (ICC) were calculated to assess feature reproducibility for each center and among all centers in three scenarios: Adams (Nat Rev Endocrinol 9(1):28, 2013) ten different contrast Brown et al. (J Med Imaging 5(1):011017, 2018) ten different edge enhancement and Hirvasniemi et al. (Osteoarthr Cartilage 27(6):906-914, 2019) ten different image latitude parameters. Based on ICC analysis, it is observed that 46-100-44% of Histogram, 54-72-42% of GLCM, 43-76-36% of GLDM, 60-90-17% of GLRLM, 33-19-21% of GLSZM and 13-20-0% of NGTDM radiomics features had 90% < ICC < 100% over changes in contrast-edge enhancement-latitude changes respectively. Based on COV, GLRLM was only feature set that 100% of their features had COV ≤ 5% over changes in contrast and edge enhancement. The results presented here, indicating that radiomics features extracted are vulnerable over changes in contrast, edge enhancement and latitude. The most reproducible features that introduced in this study could be used for further clinical decision making.
Collapse
Affiliation(s)
- Seyed Hamid Abdali
- Student Research Committee, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Firoozeh Afzali
- Student Research Committee, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeid Baseri
- Student Research Committee, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Neda Abdalvand
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, P.O. Box: 15785 - 6171, Junction of Shahid Hemmat & Shahid Chamran Expressways, 14496, Tehran, Iran.
| | - Hamid Abdollahi
- Student Research Committee, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Department of Radiologic Technology, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
44
|
Tharmaseelan H, Hertel A, Tollens F, Rink J, Woźnicki P, Haselmann V, Ayx I, Nörenberg D, Schoenberg SO, Froelich MF. Identification of CT Imaging Phenotypes of Colorectal Liver Metastases from Radiomics Signatures-Towards Assessment of Interlesional Tumor Heterogeneity. Cancers (Basel) 2022; 14:cancers14071646. [PMID: 35406418 PMCID: PMC8997087 DOI: 10.3390/cancers14071646] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Tumoral heterogeneity (TH) is a major challenge in the treatment of metastatic colorectal cancer (mCRC) and is associated with inferior response. Therefore, the identification of TH would be beneficial for treatment planning. TH can be assessed by identifying genetic alterations. In this work, a radiomics-based approach for assessment of TH in colorectal liver metastases (CRLM) in CT scans is demonstrated. (2) Methods: In this retrospective study, CRLM of mCRC were segmented and radiomics features extracted using pyradiomics. Unsupervised k-means clustering was applied to features and lesions. Feature redundancy was evaluated by principal component analysis and reduced by Pearson correlation coefficient cutoff. Feature selection was conducted by LASSO regression and visual analysis of the clusters by radiologists. (3) Results: A total of 47 patients’ (36% female, median age 64) CTs with 261 lesions were included. Five clusters were identified, and the categories small disseminated (n = 31), heterogeneous (n = 105), homogeneous (n = 64), mixed (n = 59), and very large type (n = 2) were assigned based on visual characteristics. Further statistical analysis showed correlation (p < 0.01) of clusters with sex, primary location, T- and N-status, and mutational status. Feature reduction and selection resulted in the identification of four features as a final set for cluster definition. (4) Conclusions: Radiomics features can characterize TH in liver metastases of mCRC in CT scans, and may be suitable for a better pretherapeutic classification of liver lesion phenotypes.
Collapse
Affiliation(s)
- Hishan Tharmaseelan
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Alexander Hertel
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Fabian Tollens
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Johann Rink
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Piotr Woźnicki
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Verena Haselmann
- Institute of Clinical Chemistry, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany;
| | - Isabelle Ayx
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Dominik Nörenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Stefan O. Schoenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Matthias F. Froelich
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
- Correspondence:
| |
Collapse
|
45
|
Linsalata S, Borgheresi R, Marfisi D, Barca P, Sainato A, Paiar F, Neri E, Traino AC, Giannelli M. Radiomics of Patients with Locally Advanced Rectal Cancer: Effect of Preprocessing on Features Estimation from Computed Tomography Imaging. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2003286. [PMID: 35355820 PMCID: PMC8958068 DOI: 10.1155/2022/2003286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 01/14/2022] [Accepted: 01/30/2022] [Indexed: 12/24/2022]
Abstract
The purpose of this study was to investigate the effect of image preprocessing on radiomic features estimation from computed tomography (CT) imaging of locally advanced rectal cancer (LARC). CT images of 20 patients with LARC were used to estimate 105 radiomic features of 7 classes (shape, first-order, GLCM, GLDM, GLRLM, GLSZM, and NGTDM). Radiomic features were estimated for 6 different isotropic resampling voxel sizes, using 10 interpolation algorithms (at fixed bin width) and 6 different bin widths (at fixed interpolation algorithm). The intraclass correlation coefficient (ICC) and the coefficient of variation (CV) were calculated to assess the variability in radiomic features estimation due to preprocessing. A repeated measures correlation analysis was performed to assess any linear correlation between radiomic feature estimate and resampling voxel size or bin width. Reproducibility of radiomic feature estimate, when assessed through ICC analysis, was nominally excellent (ICC > 0.9) for shape features, good (0.75 < ICC ≤ 0.9) or moderate (0.5 < ICC ≤ 0.75) for first-order features, and moderate or poor (0 ≤ ICC ≤ 0.5) for textural features. A number of radiomic features characterized by good or excellent reproducibility in terms of ICC showed however median CV values greater than 15%. For most textural features, a significant (p < 0.05) correlation between their estimate and resampling voxel size or bin width was found. In CT imaging of patients with LARC, the estimate of textural features, as well as of first-order features to a lesser extent, is appreciably biased by preprocessing. Accordingly, this should be taken into account when planning clinical or research studies, as well as when comparing results from different studies and performing multicenter studies.
Collapse
Affiliation(s)
- Stefania Linsalata
- Unit of Medical Physics, Pisa University Hospital “Azienda Ospedaliero-Universitaria Pisana”, Pisa, Italy
| | - Rita Borgheresi
- Unit of Medical Physics, Pisa University Hospital “Azienda Ospedaliero-Universitaria Pisana”, Pisa, Italy
| | - Daniela Marfisi
- Unit of Medical Physics, Pisa University Hospital “Azienda Ospedaliero-Universitaria Pisana”, Pisa, Italy
| | - Patrizio Barca
- Unit of Medical Physics, Pisa University Hospital “Azienda Ospedaliero-Universitaria Pisana”, Pisa, Italy
| | - Aldo Sainato
- Radiation Oncology Unit, Pisa University Hospital “Azienda Ospedaliero-Universitaria Pisana”, Pisa, Italy
| | - Fabiola Paiar
- Radiation Oncology Unit, Pisa University Hospital “Azienda Ospedaliero-Universitaria Pisana”, Pisa, Italy
| | - Emanuele Neri
- Diagnostic and Interventional Radiology, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Antonio Claudio Traino
- Unit of Medical Physics, Pisa University Hospital “Azienda Ospedaliero-Universitaria Pisana”, Pisa, Italy
| | - Marco Giannelli
- Unit of Medical Physics, Pisa University Hospital “Azienda Ospedaliero-Universitaria Pisana”, Pisa, Italy
| |
Collapse
|
46
|
Assessing radiomics feature stability with simulated CT acquisitions. Sci Rep 2022; 12:4732. [PMID: 35304508 PMCID: PMC8933485 DOI: 10.1038/s41598-022-08301-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 03/03/2022] [Indexed: 11/29/2022] Open
Abstract
Medical imaging quantitative features had once disputable usefulness in clinical studies. Nowadays, advancements in analysis techniques, for instance through machine learning, have enabled quantitative features to be progressively useful in diagnosis and research. Tissue characterisation is improved via the “radiomics” features, whose extraction can be automated. Despite the advances, stability of quantitative features remains an important open problem. As features can be highly sensitive to variations of acquisition details, it is not trivial to quantify stability and efficiently select stable features. In this work, we develop and validate a Computed Tomography (CT) simulator environment based on the publicly available ASTRA toolbox (www.astra-toolbox.com). We show that the variability, stability and discriminative power of the radiomics features extracted from the virtual phantom images generated by the simulator are similar to those observed in a tandem phantom study. Additionally, we show that the variability is matched between a multi-center phantom study and simulated results. Consequently, we demonstrate that the simulator can be utilised to assess radiomics features’ stability and discriminative power.
Collapse
|
47
|
Robustness and reproducibility of radiomics in T2 weighted images from magnetic resonance image guided linear accelerator in a phantom study. Phys Med 2022; 96:130-139. [PMID: 35287100 DOI: 10.1016/j.ejmp.2022.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/07/2022] [Accepted: 03/04/2022] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Quantitative radiomics features extracted from medical images have been shown to provide value in predicting clinical outcomes. The study for robustness and reproducibility of radiomics features obtained with magnetic resonance image guided linear accelerator (MR-Linac) is insufficient. The objective of this work was to investigate the stability of radiomics features extracted from T2-weighted images of MR-Linac for five common effect factors. MATERIALS AND METHOD In this work, ten jellies, five fruits/vegetables, and a dynamic phantom were used to evaluate the impact of test-retest, intraobserver, varied thicknesses, radiation, and motion. These phantoms were scanned on a 1.5 T MRI system of MR-Linac. For test-retest data, the phantoms were scanned twice with repositioning within 15 min. To assess for intraobserver comparison, the segmentation of MR images was repeated by one observer in a double-blind manner. Three slice thicknesses (1.2 mm, 2.4 mm, and 4.8 mm) were used to select robust features that were insensitive to different thicknesses. The effect of radiation on features was studied by acquiring images when the beam was on. Common movement images of patients during radiotherapy were simulated by a dynamic phantom with five motion states to study the motion effect. A total of 1409 radiomics features, including shape features, first-order features, and texture features, were extracted from the original, wavelet, square, logarithmic, exponential and gradient images. The robustness and reproducibility features were evaluated using the concordance correlation coefficient (CCC). RESULT The intraobserver group had the most robust features (936/1079, 86.7%), while the group of motion effects had the lowest robustness (56/936, 6.0%), followed by the group of different thickness cohorts (374/936, 40.0%). The stability of features in the test-retest and radiation groups was 1072 of 1312 (81.7%) and 810 of 936 (86.5%), respectively. Overall, 25 of 1409 (2.4%) radiomics features remained robust in all five tests, mostly focusing on the image type of the wavelet. The number of stable features extracted from when the beam was on was less than that extracted when the beam was off. Shape features were the most robust of all of the features in all of the groups, excluding the motion group. CONCLUSION Compared with other factors fewer features remained robust to the effect of motion. This result emphasizes the need to consider the effect of respiration motion. The study for T2-weighted images from MR-Linac under different conditions will help us to build a robust predictive model applicable for radiotherapy.
Collapse
|
48
|
Hinzpeter R, Baumann L, Guggenberger R, Huellner M, Alkadhi H, Baessler B. Radiomics for detecting prostate cancer bone metastases invisible in CT: a proof-of-concept study. Eur Radiol 2022; 32:1823-1832. [PMID: 34559264 PMCID: PMC8831270 DOI: 10.1007/s00330-021-08245-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/13/2021] [Accepted: 08/03/2021] [Indexed: 01/02/2023]
Abstract
OBJECTIVES To investigate, in patients with metastatic prostate cancer, whether radiomics of computed tomography (CT) image data enables the differentiation of bone metastases not visible on CT from unaffected bone using 68 Ga-PSMA PET imaging as reference standard. METHODS In this IRB-approved retrospective study, 67 patients (mean age 71 ± 7 years; range: 55-84 years) showing a total of 205 68 Ga-PSMA-positive prostate cancer bone metastases in the thoraco-lumbar spine and pelvic bone being invisible in CT were included. Metastases and 86 68 Ga-PSMA-negative bone volumes in the same body region were segmented and further post-processed. Intra- and inter-reader reproducibility was assessed, with ICCs < 0.90 being considered non-reproducible. To account for imbalances in the dataset, data augmentation was performed to achieve improved class balance and to avoid model overfitting. The dataset was split into training, test, and validation set. After a multi-step dimension reduction process and feature selection process, the 11 most important and independent features were selected for statistical analyses. RESULTS A gradient-boosted tree was trained on the selected 11 radiomic features in order to classify patients' bones into bone metastasis and normal bone using the training dataset. This trained model achieved a classification accuracy of 0.85 (95% confidence interval [CI]: 0.76-0.92, p < .001) with 78% sensitivity and 93% specificity. The tuned model was applied on the original, non-augmented dataset resulting in a classification accuracy of 0.90 (95% CI: 0.82-0.98) with 91% sensitivity and 88% specificity. CONCLUSION Our proof-of-concept study indicates that radiomics may accurately differentiate unaffected bone from metastatic bone, being invisible by the human eye on CT. KEY POINTS • This proof-of-concept study showed that radiomics applied on CT images may accurately differentiate between bone metastases and metastatic-free bone in patients with prostate cancer. • Future promising applications include automatic bone segmentation, followed by a radiomics classifier, allowing for a screening-like approach in the detection of bone metastases.
Collapse
Affiliation(s)
- Ricarda Hinzpeter
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Raemistr. 100, CH-8091, Zurich, Switzerland.
| | - Livia Baumann
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Raemistr. 100, CH-8091, Zurich, Switzerland
| | - Roman Guggenberger
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Raemistr. 100, CH-8091, Zurich, Switzerland
| | - Martin Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Hatem Alkadhi
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Raemistr. 100, CH-8091, Zurich, Switzerland
| | - Bettina Baessler
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Raemistr. 100, CH-8091, Zurich, Switzerland
| |
Collapse
|
49
|
Carbonell G, Kennedy P, Bane O, Kirmani A, El Homsi M, Stocker D, Said D, Mukherjee P, Gevaert O, Lewis S, Hectors S, Taouli B. Precision of MRI radiomics features in the liver and hepatocellular carcinoma. Eur Radiol 2022; 32:2030-2040. [PMID: 34564745 DOI: 10.1007/s00330-021-08282-1] [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: 03/16/2021] [Revised: 07/12/2021] [Accepted: 08/17/2021] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To assess the precision of MRI radiomics features in hepatocellular carcinoma (HCC) tumors and liver parenchyma. METHODS The study population consisted of 55 patients, including 16 with untreated HCCs, who underwent two repeat contrast-enhanced abdominal MRI exams within 1 month to evaluate: (1) test-retest repeatability using the same MRI system (n = 28, 10 HCCs); (2) inter-platform reproducibility between different MRI systems (n = 27, 6 HCCs); (3) inter-observer reproducibility (n = 16, 16 HCCs). Shape and 1st- and 2nd-order radiomics features were quantified on pre-contrast T1-weighted imaging (WI), T1WI portal venous phase (pvp), T2WI, and ADC (apparent diffusion coefficient), on liver regions of interest (ROIs) and HCC volumes of interest (VOIs). Precision was assessed by calculating intraclass correlation coefficient (ICC), concordance correlation coefficient (CCC), and coefficient of variation (CV). RESULTS There was moderate to excellent test-retest repeatability of shape and 1st- and 2nd-order features for all sequences in HCCs (ICC: 0.53-0.99; CV: 3-29%), and moderate to good test-retest repeatability of 1st- and 2nd-order features for T1WI sequences, and 2nd-order features for T2WI in the liver (ICC: 0.53-0.73; CV: 12-19%). There was poor inter-platform reproducibility for all features and sequences, except for shape and 1st-order features on T1WI in HCCs (CCC: 0.58-0.99; CV: 3-15%). Good to excellent inter-observer reproducibility was found for all features and sequences in HCCs (CCC: 0.80-0.99; CV: 4-15%) and moderate to good for liver (CCC: 0.45-0.86; CV: 6-25%). CONCLUSIONS MRI radiomics features have acceptable repeatability in the liver and HCC when using the same MRI system and across readers but have low reproducibility across MR systems, except for shape and 1st-order features on T1WI. Data must be interpreted with caution when performing multiplatform radiomics studies. KEY POINTS • MRI radiomics features have acceptable repeatability when using the same MRI system but less reproducible when using different MRI platforms. • MRI radiomics features extracted from T1 weighted-imaging show greater stability across exams than T2 weighted-imaging and ADC. • Inter-observer reproducibility of MRI radiomics features was found to be good in HCC tumors and acceptable in liver parenchyma.
Collapse
Affiliation(s)
- Guillermo Carbonell
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Radiology, University Hospital Virgen de La Arrixaca, Murcia, Spain
| | - Paul Kennedy
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Octavia Bane
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ammar Kirmani
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria El Homsi
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel Stocker
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Daniela Said
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Radiology, Universidad de los Andes, Santiago, Chile
| | | | - Olivier Gevaert
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Sara Lewis
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stefanie Hectors
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bachir Taouli
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
50
|
Li Y, Reyhan M, Zhang Y, Wang X, Zhou J, Zhang Y, Yue NJ, Nie K. The impact of phantom design and material-dependence on repeatability and reproducibility of CT-based radiomics features. Med Phys 2022; 49:1648-1659. [PMID: 35103332 DOI: 10.1002/mp.15491] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/26/2021] [Accepted: 12/29/2021] [Indexed: 12/28/2022] Open
Abstract
PURPOSE To understand the design of radiomics phantom and material-dependence on repeatability and reproducibility of CT radiomics features METHODS: : A radiomics phantom consisting of various materials with uniformity, textural and biological components, was constructed. The phantom was scanned with different manufacturer CT scanners and the scans were repeated multiple times on the same scanner with different acquisition settings as kVp, mAs, orientation, field of view (FOV), slice thickness, pitch, reconstruction kernels and acquisition mode. A total of 72 phantom scans were included. For each scan, 18 different regions of interest (ROI) were contoured and 708 radiomics features were extracted from each ROI via an open source radiomics tool, IBEX. To relate the phantom data to patient data, the radiomics features from different phantom materials were compared with those extracted from 50 patients' images of five disease sites as brain, head-and-neck, breast, liver and lung cases using box-plots comparison and principal component analysis (PCA). The temporal stability of imaging features was then evaluated with respect to a controlled scenario (test-retest) via the intra-class correlation coefficient (ICC). The reproducibility of radiomics features with respect to different scanners or acquisition settings were further evaluated with concordance correlation coefficients (CCC). RESULTS Among all phantom materials, the biological component had feature values closest to human tissues, especially for tumors in brain and liver. The textural component showed similar ranges of variation to lung lesions, particularly for cartridges of rice, cereal, and the 3D-printed textural phantom with fine and rough-grid. It also showed that certain materials, such as polystyrene foam, plaster and peanuts, did not have comparable values to human tissue and could be excluded for future phantom design. High repeatability was observed in the test-retest study as indicated by an ICC value of 0.998 ± 0.020. All materials were used for feature stability analysis. For the inter-scanner study, shape-related features were the most-reliable category with 94% of features having CCC ≥0.9, while GOH were the least-reliable with only 14.6% meeting the criteria. For the intra-scanner study, the reproducibility of CT-based radiomics features showed material-dependence. In general, the instability of radiomics features introduced by kVp, mAs, pitch, acquisition mode and orientation were relatively mild. However, the homogeneous materials were more vulnerable to those changes compared to materials with textural patterns. Regardless of material compositions, resolution parameters like FOV and slice thickness, could have large impact on feature stability. Switching between standard and bone reconstruction kernels could also result significant changes to feature reproducibility. CONCLUSION We have built a radiomics phantom using materials that cover a wide span of tumor textures seen in oncological CT images. The designed phantom presents a preliminary opportunity for investigating reproducibility of radiomics features and the reproducibility can be material dependent. Thus, in the radiomics quality assurance design, it is important to choose appropriate materials that can provide a close range of radiomics features to patients with specific disease sites dependency taken into consideration. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yanjing Li
- Department of Radiation Oncology, Rutgers-Cancer Institute of New Jersey, New Brunswick, NJ, 08901
| | - Meral Reyhan
- Department of Radiation Oncology, Rutgers-Cancer Institute of New Jersey, New Brunswick, NJ, 08901
| | - Yin Zhang
- Department of Radiation Oncology, Rutgers-Cancer Institute of New Jersey, New Brunswick, NJ, 08901
| | - Xiao Wang
- Department of Radiation Oncology, Rutgers-Cancer Institute of New Jersey, New Brunswick, NJ, 08901
| | - Jinghao Zhou
- Department of Radiation Oncology, Rutgers-Cancer Institute of New Jersey, New Brunswick, NJ, 08901
| | - Yang Zhang
- Department of Radiation Oncology, Rutgers-Cancer Institute of New Jersey, New Brunswick, NJ, 08901
| | - Ning J Yue
- Department of Radiation Oncology, Rutgers-Cancer Institute of New Jersey, New Brunswick, NJ, 08901
| | - Ke Nie
- Department of Radiation Oncology, Rutgers-Cancer Institute of New Jersey, New Brunswick, NJ, 08901
| |
Collapse
|