101
|
Priya S, Reutzel A, Ferreira Dalla Pria OA, Goetz S, Pham HT, Alatoum A, Aher PY, Narayanasamy S, Nagpal P, Carter KD. Addressing Inter-reconstruction variability in multi-energy myocardial CT Radiomics: The Benefits of combat harmonization. Eur J Radiol 2024; 183:111891. [PMID: 39708706 DOI: 10.1016/j.ejrad.2024.111891] [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: 06/09/2024] [Revised: 10/18/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
RATIONALE AND OBJECTIVES To investigate the effect of ComBat harmonization on the stability of myocardial radiomic features derived from multi-energy CT reconstructions. MATERIALS AND METHODS A retrospective study was conducted on 205 patients who underwent dual-energy chest CTA at a single center. The data was reconstructed into multiple spectral reconstructions (mixed energy simulating standard 120 Kv acquisition and monoenergetic images ranging from 40 to 190 keV in increments of 10). The left ventricle myocardium was segmented using semiautomated software (Syngo.Via FRONTIER, version 5.0.2; Siemens). Radiomic features were extracted from multiple spectral reconstructions (batches). The consistency of these radiomics features across different batches was evaluated with and without ComBat harmonization using Cohen's d and Principal component analysis (PCA). Both parametric and nonparametric ComBat methods were considered. RESULTS Without any ComBat technique, 43.40% of features remained consistent across all multienergy reconstructions. Applying ComBat harmonization increased this consistency to 98.37% with parametric empirical bayes (EB) ComBat and EB M-ComBat, and to 91.52% and 92.33% with nonparametric EB ComBat and nonparametric EB M-ComBat, respectively. PCA without ComBat revealed noticeable differences in the first two principal components between batches, indicating a batch effect or unstable radiomic features. Following ComBat harmonization, the principal components showed more consistency between batches, demonstrating radiomics feature stability between batches. CONCLUSION ComBat harmonization enhanced the consistency of radiomic features from multi-energy CT data. Integrating ComBat harmonization may lead to more reproducible results in multienergy CT radiomics studies.
Collapse
Affiliation(s)
- Sarv Priya
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa, USA.
| | - Abigail Reutzel
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa, USA
| | | | - Sawyer Goetz
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa, USA
| | - Hanh Td Pham
- Department of Biostatistics, University of Iowa, Iowa City, IA, USA
| | - Aiah Alatoum
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa, USA
| | - Pritish Y Aher
- Department of Radiology, University of Miami, Miller School of Medicine, Miami, FL, United States
| | | | - Prashant Nagpal
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Knute D Carter
- Department of Biostatistics, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
102
|
Wan CF, Jiang ZY, Wang YQ, Wang L, Fang H, Jin Y, Dong Q, Zhang XQ, Jiang LX. Radiomics of multimodal ultrasound for early prediction of pathologic complete response to neoadjuvant chemotherapy in breast cancer. Acad Radiol 2024:S1076-6332(24)00855-9. [PMID: 39690072 DOI: 10.1016/j.acra.2024.11.012] [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: 08/03/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 12/19/2024]
Abstract
RATIONALE AND OBJECTIVES To construct and validate a clinical-radiomics model based on radiomics features extracted from two-stage multimodal ultrasound and clinicopathologic information for early predicting pathologic complete response (pCR) to neoadjuvant chemotherapy (NAC) in breast cancer patients treated with NAC. MATERIALS AND METHODS Consecutive women with biopsy-proven breast cancer undergoing multimodal US pretreatment and after two cycles of NAC and followed by surgery between January 2014 and November 2023 were retrospectively collected for clinical-radiomics model construction (n = 274) and retrospective test (n = 134). The predictive performance of it was further tested in a subsequent prospective internal test set recruited between January 2024 to July 2024 (n = 76). Finally, a total of 484 patients were enrolled. The clinical-radiomics model predictive performance was compared with radiomics model, clinical model and radiologists' visual assessment by area under the receiver operating characteristic curve (AUC) analysis and DeLong test. RESULTS The proposed clinical-radiomics model obtained the AUC values of 0.92 (95%CI: 0.88, 0.94) and 0.85 (95%CI: 0.79, 0.89) in retrospective and prospective test sets, respectively, which were significantly higher than that those of the radiomics model (AUCs: 0.75-0.85), clinical model (AUCs: 0.68-0.72) and radiologists' visual assessments (AUCs:0.59-0.68) (all p < 0.05). In addition, the predictive efficacy of the radiologists was improved under the assistance of the clinical-radiomics model significantly. CONCLUSION The clinical-radiomics model developed in this study, which integrated clinicopathologic information and two-stage multimodal ultrasound features, was able to early predict pCR to NAC in breast cancer patients with favorable predictive effectiveness.
Collapse
Affiliation(s)
- Cai-Feng Wan
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Zhuo-Yun Jiang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China (Z-y.J.)
| | - Yu-Qun Wang
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Lin Wang
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Hua Fang
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Ye Jin
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Qi Dong
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Xue-Qing Zhang
- Department of Pathology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (X-q.Z.)
| | - Li-Xin Jiang
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.).
| |
Collapse
|
103
|
Darbari Kaul R, Sacks PL, Thiel C, Rimmer J, Kalish L, Campbell RG, Sacks R, Di Ieva A, Harvey RJ. Radiomics of the Paranasal Sinuses: A Systematic Review of Computer-Assisted Techniques to Assess Computed Tomography Radiological Data. Am J Rhinol Allergy 2024:19458924241304082. [PMID: 39686586 DOI: 10.1177/19458924241304082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
BACKGROUND Radiomics is a quantitative approach to medical imaging, aimed to extract features into large datasets. By using artificial intelligence (AI) methodologies, large radiomic data can be analysed and translated into meaningful clinical applications. In rhinology, there is heavy reliance on computed tomography (CT) imaging of the paranasal sinus for diagnostics and assessment of treatment outcomes. Currently, there is an emergence of literature detailing radiomics use in rhinology. OBJECTIVE This systematic review aims to assess the current techniques used to analyze radiomic data from paranasal sinus CT imaging. METHODS A systematic search was performed using Ovid MEDLINE and EMBASE databases from January 1, 2019 until March 16, 2024 using the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) checklist and Cochrane Library Systematic Reviews for Diagnostic and Prognostic Studies. The QUADAS-2 and PROBAST tools were utilized to assess risk of bias. RESULTS Our search generated 1456 articles with 10 articles meeting eligibility criteria. Articles were divided into 2 categories, diagnostic (n = 7) and prognostic studies (n = 3). The number of radiomic features extracted ranged 4 to 1409, with analysis including non-AI-based statistical analyses (n = 3) or machine learning algorithms (n = 7). The diagnostic or prognostic utility of radiomics analyses were rated as excellent (n = 3), very good (n = 2), good (n = 2), or not reported (n = 3) based upon area under the curve receiver operating characteristic (AUC-ROC) or accuracy. The average radiomics quality score was 36.95%. CONCLUSION Radiomics is an evolving field which can augment our understanding of rhinology diseases, however there are currently only minimal quality studies with limited clinical utility.
Collapse
Affiliation(s)
- Rhea Darbari Kaul
- Rhinology and Skull Base Research Group, Applied Medical Research Centre, University of New South Wales, Sydney, Australia
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, Australia
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Peta-Lee Sacks
- Rhinology and Skull Base Research Group, Applied Medical Research Centre, University of New South Wales, Sydney, Australia
| | - Cedric Thiel
- Rhinology and Skull Base Research Group, Applied Medical Research Centre, University of New South Wales, Sydney, Australia
| | - Janet Rimmer
- Rhinology and Skull Base Research Group, Applied Medical Research Centre, University of New South Wales, Sydney, Australia
- Woolcock Institute, University of Sydney, Sydney, Australia
- Faculty of Medicine, Notre Dame University, Sydney, Australia
| | - Larry Kalish
- Rhinology and Skull Base Research Group, Applied Medical Research Centre, University of New South Wales, Sydney, Australia
- Department of Otolaryngology, Head and Neck Surgery, Concord General Hospital, University of Sydney, Sydney, Australia
- Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Raewyn Gay Campbell
- Rhinology and Skull Base Research Group, Applied Medical Research Centre, University of New South Wales, Sydney, Australia
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Faculty of Medicine, University of Sydney, Sydney, Australia
- Department of Otolaryngology Head and Neck Surgery, Royal Prince Alfred Hospital, Sydney, Australia
| | - Raymond Sacks
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Department of Otolaryngology, Head and Neck Surgery, Concord General Hospital, University of Sydney, Sydney, Australia
- Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Antonio Di Ieva
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, Australia
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Richard John Harvey
- Rhinology and Skull Base Research Group, Applied Medical Research Centre, University of New South Wales, Sydney, Australia
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW, Sydney, Australia
| |
Collapse
|
104
|
Shahzadi M, Rafique H, Waheed A, Naz H, Waheed A, Zokirova FR, Khan H. Artificial intelligence for chimeric antigen receptor-based therapies: a comprehensive review of current applications and future perspectives. Ther Adv Vaccines Immunother 2024; 12:25151355241305856. [PMID: 39691280 PMCID: PMC11650588 DOI: 10.1177/25151355241305856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Using artificial intelligence (AI) to enhance chimeric antigen receptor (CAR)-based therapies' design, production, and delivery is a novel and promising approach. This review provides an overview of the current applications and challenges of AI for CAR-based therapies and suggests some directions for future research and development. This paper examines some of the recent advances of AI for CAR-based therapies, for example, using deep learning (DL) to design CARs that target multiple antigens and avoid antigen escape; using natural language processing to extract relevant information from clinical reports and literature; using computer vision to analyze the morphology and phenotype of CAR cells; using reinforcement learning to optimize the dose and schedule of CAR infusion; and using AI to predict the efficacy and toxicity of CAR-based therapies. These applications demonstrate the potential of AI to improve the quality and efficiency of CAR-based therapies and to provide personalized and precise treatments for cancer patients. However, there are also some challenges and limitations of using AI for CAR-based therapies, for example, the lack of high-quality and standardized data; the need for validation and verification of AI models; the risk of bias and error in AI outputs; the ethical, legal, and social issues of using AI for health care; and the possible impact of AI on the human role and responsibility in cancer immunotherapy. It is important to establish a multidisciplinary collaboration among researchers, clinicians, regulators, and patients to address these challenges and to ensure the safe and responsible use of AI for CAR-based therapies.
Collapse
Affiliation(s)
- Muqadas Shahzadi
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Hamad Rafique
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Ahmad Waheed
- Department of Zoology, Faculty of Life Sciences, University of Okara, 2 KM Lahore Road, Renala Khurd, Okara 56130, Punjab, Pakistan
| | - Hina Naz
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Atifa Waheed
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | | | - Humera Khan
- Department of Biochemistry, Sahiwal Medical College, Sahiwal, Pakistan
| |
Collapse
|
105
|
Yin Y, Zhang W, Chen Y, Zhang Y, Shen X. Radiomics predicting immunohistochemical markers in primary hepatic carcinoma: Current status and challenges. Heliyon 2024; 10:e40588. [PMID: 39660185 PMCID: PMC11629216 DOI: 10.1016/j.heliyon.2024.e40588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/28/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Primary hepatic carcinoma, comprising hepatocellular carcinoma (HCC), intrahepatic cholangiocarcinoma (ICC), and combined hepatocellular cholangiocarcinoma (cHCC-CCA), ranks among the most common malignancies worldwide. The heterogeneity of tumors is a primary factor impeding the efficacy of treatments for primary hepatic carcinoma. Immunohistochemical markers may play a potential role in characterizing this heterogeneity, providing significant guidance for prognostic analysis and the development of personalized treatment plans for the patients with primary hepatic carcinoma. Currently, primary hepatic carcinoma immunohistochemical analysis primarily relies on invasive techniques such as surgical pathology and tissue biopsy. Consequently, the non-invasive preoperative acquisition of primary hepatic carcinoma immunohistochemistry has emerged as a focal point of research. As an emerging non-invasive diagnostic technique, radiomics possesses the potential to extensively characterize tumor heterogeneity. It can predict immunohistochemical markers associated with hepatocellular carcinoma preoperatively, demonstrating significant auxiliary utility in clinical guidance. This article summarizes the progress in using radiomics to predict immunohistochemical markers in primary hepatic carcinoma, addresses the challenges faced in this field of study, and anticipates its future application prospects.
Collapse
Affiliation(s)
- Yunqing Yin
- The Second Clinical Medical College, Jinan University, China
| | - Wei Zhang
- Department of Intervention, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Yanhui Chen
- Department of Intervention, Shenzhen Bao'an People's Hospital, Shenzhen, 518100, Guangdong, China
| | - Yanfang Zhang
- Department of Intervention, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Xinying Shen
- Department of Intervention, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| |
Collapse
|
106
|
Liu Z, Liu Z, Wan X, Wang Y, Huang X. Predicting high-intensity focused ultrasound efficacy in adenomyosis treatment based on magnetic resonance (MR) radiomics and clinical-imaging features. Clin Radiol 2024; 81:106778. [PMID: 39798274 DOI: 10.1016/j.crad.2024.106778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/09/2024] [Accepted: 12/09/2024] [Indexed: 01/15/2025]
Abstract
AIMS To develop a model predicting high-intensity focused ultrasound (HIFU) efficacy in adenomyosis treatment using enhanced T1WI and T2WI-FS radiomics combined with clinical imaging features. MATERIALS AND METHODS The study included 137 adenomyosis patients treated with HIFU from September 2021 to December 2023. Based on nonperfused volume ratio (NPVR), participants were divided into two groups: NPVR < 50% (n=77) and NPVR ≥ 50% (n=60). Patients were randomly split into training and test sets (7:3 ratio). Radiomics features were extracted from enhanced T1WI and T2WI-FS sequences, while clinical imaging features were selected using univariate analysis and binary logistic regression. Logistic regression models were built for radiomics, clinical imaging, and combined data. Model performance was assessed using ROC curves, Delong's test, and calibration curves. RESULTS AUCs for the radiomics, clinical-imaging, and combined models in the training set were 0.831, 0.664, and 0.845, respectively, and 0.829, 0.597, and 0.831 in the test set. The combined model outperformed the clinical-imaging model (training p=0.001, test p=0.01) and the radiomics model (training p=0.012, test p=0.032). However, no significant difference was found between the combined and radiomics models (p>0.05). Calibration curves and decision curve analysis confirmed the combined model's accuracy and clinical applicability. CONCLUSION A model incorporating clinical-imaging features with T1WI and T2WI-FS radiomics effectively predicts HIFU success in adenomyosis treatment, offering valuable guidance for clinical decision-making.
Collapse
Affiliation(s)
- Z Liu
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Z Liu
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - X Wan
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Y Wang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - X Huang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
107
|
Zeng S, Wang XL, Yang H. Radiomics and radiogenomics: extracting more information from medical images for the diagnosis and prognostic prediction of ovarian cancer. Mil Med Res 2024; 11:77. [PMID: 39673071 PMCID: PMC11645790 DOI: 10.1186/s40779-024-00580-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/07/2024] [Indexed: 12/15/2024] Open
Abstract
Ovarian cancer (OC) remains one of the most lethal gynecological malignancies globally. Despite the implementation of various medical imaging approaches for OC screening, achieving accurate differential diagnosis of ovarian tumors continues to pose significant challenges due to variability in image performance, resulting in a lack of objectivity that relies heavily on the expertise of medical professionals. This challenge can be addressed through the emergence and advancement of radiomics, which enables high-throughput extraction of valuable information from conventional medical images. Furthermore, radiomics can integrate with genomics, a novel approach termed radiogenomics, which allows for a more comprehensive, precise, and personalized assessment of tumor biological features. In this review, we present an extensive overview of the application of radiomics and radiogenomics in diagnosing and predicting ovarian tumors. The findings indicate that artificial intelligence methods based on imaging can accurately differentiate between benign and malignant ovarian tumors, as well as classify their subtypes. Moreover, these methods are effective in forecasting survival rates, treatment outcomes, metastasis risk, and recurrence for patients with OC. It is anticipated that these advancements will function as decision-support tools for managing OC while contributing to the advancement of precision medicine.
Collapse
Affiliation(s)
- Song Zeng
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xin-Lu Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Hua Yang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
108
|
Bhandari A, Johnson K, Oh K, Yu F, Huynh LM, Lei Y, Wisnoskie S, Zhou S, Baine MJ, Lin C, Zhang C, Wang S. Developing a novel dosiomics model to predict treatment failures following lung stereotactic body radiation therapy. Front Oncol 2024; 14:1438861. [PMID: 39726705 PMCID: PMC11669717 DOI: 10.3389/fonc.2024.1438861] [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: 05/26/2024] [Accepted: 11/07/2024] [Indexed: 12/28/2024] Open
Abstract
Purpose The purpose of this study was to investigate the dosiomics features of the interplay between CT density and dose distribution in lung SBRT plans, and to develop a model to predict treatment failure following lung SBRT treatment. Methods A retrospective study was conducted involving 179 lung cancer patients treated with SBRT at the University of Nebraska Medical Center (UNMC) between October 2007 and June 2022. Features from the CT image, Biological Effective Dose (BED) and five interaction matrices between CT and BED were extracted using radiomics mathematics. Our in-house feature selection pipeline was utilized to evaluate and rank features based on their importance and redundancy, with only the selected non-redundant features being used for predictive modeling. We randomly selected 151 cases and 28 cases as training and test datasets. Four different models were trained utilizing the Balanced Random Forest framework on the same training dataset to differentiate between failure and non-failure cases. These four models utilized the same number of selected features extracted from CT-only, BED-only, a combination of CT and BED, and a composite of CT and BED including their interaction matrices, respectively. Results The cohort included 125 non-failure cases and 54 failure cases, with a median follow-up time of 34.4 months. We selected the top 17 important and non-redundant features (with the Pearsons's coefficient < 0.5) in each model. When evaluated on the same independent test set, the four models-CT features-only, BED features-only, a combination of CT and BED features, and a composite model including features from CT and BED that includes their interaction matrices-achieved AUC values of 0.56, 0.75, 0.73, and 0.82, respectively, with corresponding accuracies of 0.61, 0.79, 0.71, and 0.79. The composite model demonstrated the highest AUC and accuracy, indicating that incorporating interactions between CT and BED reveals more predictive capabilities in distinguishing between failure and non-failure cases. Conclusion The dosiomics model integrating the interaction between CT and Dose can effectively predict treatment failure following lung SBRT treatment and may serve as a useful tool to proactively evaluate and select lung SBRT treatment plans to reduce treatment failure in the future.
Collapse
Affiliation(s)
- Ashok Bhandari
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kurtis Johnson
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kyuhak Oh
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Fang Yu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, United States
| | - Linda M. Huynh
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Yu Lei
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sarah Wisnoskie
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Radiation Oncology, Novant Health Cancer Institute, Winston-Salem, NC, United States
| | - Sumin Zhou
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michael James Baine
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Chi Zhang
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shuo Wang
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
109
|
Jia XF, Chen YC, Zheng KK, Zhu DQ, Chen C, Liu J, Yang YJ, Li CT. Clinical-Radiomics Nomogram Model Based on CT Angiography for Prediction of Intracranial Aneurysm Rupture: A Multicenter Study. J Multidiscip Healthc 2024; 17:5917-5926. [PMID: 39678712 PMCID: PMC11645942 DOI: 10.2147/jmdh.s491697] [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: 08/15/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024] Open
Abstract
Objective Risk estimation of intracranial aneurysm rupture is critical in determining treatment strategy. There is a scarcity of multicenter studies on the predictive power of clinical-radiomics models for aneurysm rupture. This study aims to develop a clinical-radiomics model and explore its additional value in the discrimination of aneurysm rupture. Methods A total of 516 aneurysms, including 273 (52.9%) with ruptured aneurysms, were retrospectively enrolled from four hospitals between January 2019 and August 2020. Relevant clinical features were collected, and radiomic characteristics associated with aneurysm were extracted. Subsequently, three models, including a clinical model, a radiomics model, and a clinical-radiomics model were constructed using multivariate logistic regression analysis to effectively classify aneurysm rupture. The performance of models was analyzed through operating characteristic curves, decision curve, and calibration curves analysis. Different models' comparison used DeLong tests. To offer an understandable and intuitive scoring system for assessing rupture risk, we developed a comprehensive nomogram based on the developed model. Results Three clinical risk factors and fourteen radiomics features were explored to establish three models. The area under the receiver operating curve (AUC) for the radiomics model was 0.775 (95% CI,0.719-0.830), 0.752 (95% CI,0.663-0.841), 0.747 (95% CI,0.658-0.835) in the training, internal and external test datasets, respectively. The AUC for clinical model was 0.802 (95% CI, 0.749-0.854), 0.736 (95% CI, 0.644-0.828), 0.789 (95% CI, 0.709-0.870) in these three sets, respectively. The clinical-radiomics model showed an AUC of 0.880 (95% CI,0.840-0.920), 0.807 (95% CI,0.728-0.887), 0.815 (95% CI,0.740-0.891) in three datasets respectively. Compared with the radiomics and clinical models, the clinical-radiomics model demonstrated better diagnostic performance (DeLong' test P < 0.05). Conclusion The clinical-radiomics model represents a promising approach for predicting rupture of intracranial aneurysms.
Collapse
Affiliation(s)
- Xiu-Fen Jia
- Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, People’s Republic of China
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Yong-Chun Chen
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Kui-Kui Zheng
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Dong-Qin Zhu
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Chao Chen
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Jinjin Liu
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Yun-Jun Yang
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Chuan-Ting Li
- Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, People’s Republic of China
| |
Collapse
|
110
|
Wang Q, Brismar TB, Björk D, Baubeta E, Lindell G, Björnsson B, Sparrelid E. Development and External Validation of a Combined Clinical-Radiomic Model for Predicting Insufficient Hypertrophy of the Future Liver Remnant following Portal Vein Embolization. Ann Surg Oncol 2024:10.1245/s10434-024-16592-z. [PMID: 39658716 DOI: 10.1245/s10434-024-16592-z] [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: 08/19/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024]
Abstract
OBJECTIVES This study aimed to develop and externally validate a model for predicting insufficient future liver remnant (FLR) hypertrophy after portal vein embolization (PVE) based on clinical factors and radiomics of pretreatment computed tomography (CT) PATIENTS AND METHODS: Clinical information and CT scans of 241 consecutive patients from three Swedish centers were retrospectively collected. One center (120 patients) was applied for model development, and the other two (59 and 62 patients) as test cohorts. Logistic regression analysis was adopted for clinical model development. A FLR radiomics signature was constructed from the CT images using the support vector machine. A model combining clinical factors and FLR radiomics signature was developed. Area under the curve (AUC) was adopted for predictive performance evaluation RESULTS: Three independent clinical factors were identified for model construction: pretreatment standardized FLR (odds ratio (OR): 1.12, 95% confidence interval (CI): 1.04-1.20), alanine transaminase (ALT) level (OR: 0.98, 95% CI: 0.97-0.99), and PVE material (OR: 0.27, 95% CI: 0.08-0.87). This clinical model showed an AUC of 0.75, 0.71, and 0.68 in the three cohorts, respectively. A total of 833 radiomics features were extracted, and after feature dimension reduction, 16 features were selected for FLR radiomics signature construction. When adding it to the clinical model, the AUC of the combined model increased to 0.80, 0.76, and 0.72, respectively. However, the increase was not significant. CONCLUSIONS Pretreatment CT radiomics showed added value to the clinical model for predicting FLR hypertrophy following PVE. Although not reaching statistically significant, the evolving radiomics holds a potential to supplement traditional predictors of FLR hypertrophy.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden.
- Department of Radiology, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | - Torkel B Brismar
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Dennis Björk
- Department of Surgery, Linköping University Hospital, Linköping, Sweden
| | - Erik Baubeta
- Department of Imaging and Functional Medicine, Skåne University Hospital, Lund, Sweden
- Diagnostic Radiology, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Gert Lindell
- Department of Surgery, Skåne University Hospital Comprehensive Cancer Center, Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | | | - Ernesto Sparrelid
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
111
|
Müller D, Voran JC, Macedo M, Hartmann D, Lind C, Frank D, Schreiweis B, Kramer F, Ulrich H. Assessing Patient Health Dynamics by Comparative CT Analysis: An Automatic Approach to Organ and Body Feature Evaluation. Diagnostics (Basel) 2024; 14:2760. [PMID: 39682668 DOI: 10.3390/diagnostics14232760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/24/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: The integration of machine learning into the domain of radiomics has revolutionized the approach to personalized medicine, particularly in oncology. Our research presents RadTA (RADiomics Trend Analysis), a novel framework developed to facilitate the automatic analysis of quantitative imaging biomarkers (QIBs) from time-series CT volumes. Methods: RadTA is designed to bridge a technical gap for medical experts and enable sophisticated radiomic analyses without deep learning expertise. The core of RadTA includes an automated command line interface, streamlined image segmentation, comprehensive feature extraction, and robust evaluation mechanisms. RadTA utilizes advanced segmentation models, specifically TotalSegmentator and Body Composition Analysis (BCA), to accurately delineate anatomical structures from CT scans. These models enable the extraction of a wide variety of radiomic features, which are subsequently processed and compared to assess health dynamics across timely corresponding CT series. Results: The effectiveness of RadTA was tested using the HNSCC-3DCT-RT dataset, which includes CT scans from oncological patients undergoing radiation therapy. The results demonstrate significant changes in tissue composition and provide insights into the physical effects of the treatment. Conclusions: RadTA demonstrates a step of clinical adoption in the field of radiomics, offering a user-friendly, robust, and effective tool for the analysis of patient health dynamics. It can potentially also be used for other medical specialties.
Collapse
Affiliation(s)
- Dominik Müller
- IT-Infrastructure for Translational Medical Research, University of Augsburg, 86159 Augsburg, Germany
- Institute for Digital Medicine, University Hospital Augsburg, 86156 Augsburg, Germany
- Institute for Medical Informatics and Statistics, Kiel University and University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Jakob Christoph Voran
- Department of Cardiology, Kiel University and University Hospital Schleswig-Holstein, 24105 Kiel, Germany
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 24103 Kiel, Germany
| | - Mário Macedo
- Institute for Medical Informatics and Statistics, Kiel University and University Hospital Schleswig-Holstein, 24105 Kiel, Germany
- Medical Data Integration Center, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Dennis Hartmann
- IT-Infrastructure for Translational Medical Research, University of Augsburg, 86159 Augsburg, Germany
| | - Charlotte Lind
- Department of Cardiology, Kiel University and University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Derk Frank
- Department of Cardiology, Kiel University and University Hospital Schleswig-Holstein, 24105 Kiel, Germany
- German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 24103 Kiel, Germany
| | - Björn Schreiweis
- Institute for Medical Informatics and Statistics, Kiel University and University Hospital Schleswig-Holstein, 24105 Kiel, Germany
- Medical Data Integration Center, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Frank Kramer
- IT-Infrastructure for Translational Medical Research, University of Augsburg, 86159 Augsburg, Germany
| | - Hannes Ulrich
- Institute for Medical Informatics and Statistics, Kiel University and University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| |
Collapse
|
112
|
Sharafi A, Klein AP, Koch KM. Quantitative MRI Assessment of Post-Surgical Spinal Cord Injury Through Radiomic Analysis. J Imaging 2024; 10:312. [PMID: 39728209 DOI: 10.3390/jimaging10120312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024] Open
Abstract
This study investigates radiomic efficacy in post-surgical traumatic spinal cord injury (SCI), overcoming MRI limitations from metal artifacts to enhance diagnosis, severity assessment, and lesion characterization or prognosis and therapy guidance. Traumatic spinal cord injury (SCI) causes severe neurological deficits. While MRI allows qualitative injury evaluation, standard imaging alone has limitations for precise SCI diagnosis, severity stratification, and pathology characterization, which are needed to guide prognosis and therapy. Radiomics enables quantitative tissue phenotyping by extracting a high-dimensional set of descriptive texture features from medical images. However, the efficacy of postoperative radiomic quantification in the presence of metal-induced MRI artifacts from spinal instrumentation has yet to be fully explored. A total of 50 healthy controls and 12 SCI patients post-stabilization surgery underwent 3D multi-spectral MRI. Automated spinal cord segmentation was followed by radiomic feature extraction. Supervised machine learning categorized SCI versus controls, injury severity, and lesion location relative to instrumentation. Radiomics differentiated SCI patients (Matthews correlation coefficient (MCC) 0.97; accuracy 1.0), categorized injury severity (MCC: 0.95; ACC: 0.98), and localized lesions (MCC: 0.85; ACC: 0.90). Combined T1 and T2 features outperformed individual modalities across tasks with gradient boosting models showing the highest efficacy. The radiomic framework achieved excellent performance, differentiating SCI from controls and accurately categorizing injury severity. The ability to reliably quantify SCI severity and localization could potentially inform diagnosis, prognosis, and guide therapy. Further research is warranted to validate radiomic SCI biomarkers and explore clinical integration.
Collapse
Affiliation(s)
- Azadeh Sharafi
- Radiology Department, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Andrew P Klein
- Radiology Department, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kevin M Koch
- Radiology Department, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
113
|
Wang Q, Brismar TB, Björk D, Baubeta E, Lindell G, Björnsson B, Sparrelid E. ASO Author Reflections: Added Value of Pretreatment CT Radiomics for Accurate Prediction of Liver Hypertrophy Following Portal Vein Embolization. Ann Surg Oncol 2024:10.1245/s10434-024-16654-2. [PMID: 39645557 DOI: 10.1245/s10434-024-16654-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/09/2024]
Affiliation(s)
- Qiang Wang
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden.
- Department of Radiology, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | - Torkel B Brismar
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Dennis Björk
- Department of Surgery, Linköping University Hospital, Linköping, Sweden
| | - Erik Baubeta
- Department of Imaging and Functional Medicine, Skåne University Hospital, Lund, Sweden
- Diagnostic Radiology, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Gert Lindell
- Department of Surgery, Skåne University Hospital Comprehensive Cancer Center, Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | | | - Ernesto Sparrelid
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
114
|
Ghezzo S, Bharathi PG, Duan H, Mapelli P, Sorgo P, Davidzon GA, Bezzi C, Chung BI, Samanes Gajate AM, Thong AEC, Russo T, Brembilla G, Loening AM, Ghanouni P, Grattagliano A, Briganti A, De Cobelli F, Sonn G, Chiti A, Iagaru A, Moradi F, Picchio M. The Challenge of External Generalisability: Insights from the Bicentric Validation of a [ 68Ga]Ga-PSMA-11 PET Based Radiomics Signature for Primary Prostate Cancer Characterisation Using Histopathology as Reference. Cancers (Basel) 2024; 16:4103. [PMID: 39682289 DOI: 10.3390/cancers16234103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Background: PSMA PET radiomics is a promising tool for primary prostate cancer (PCa) characterisation. However, small single-centre studies and lack of external validation hinder definitive conclusions on the potential of PSMA PET radiomics in the initial workup of PCa. We aimed to validate a radiomics signature in a larger internal cohort and in an external cohort from a separate centre. Methods: One hundred and twenty-seven PCa patients were retrospectively enrolled across two independent hospitals. The first centre (IRCCS San Raffaele Scientific Institute, Centre 1) contributed 62 [68Ga]Ga-PSMA-11 PET scans, 20 patients classified as low-grade (ISUP grade < 4), and 42 as high-grade (ISUP grade ≥ 4). The second centre (Stanford University Hospital, Centre 2) provided 65 [68Ga]Ga-PSMA-11 PET scans, and 49 low-grade and 16 high-grade patients. A radiomics model previously generated in Centre 1 was tested on the two cohorts separately and afterward on the entire dataset. Then, we evaluated whether the radiomics features selected in the previous investigation could generalise to new data. Several machine learning (ML) models underwent training and testing using 100-fold Monte Carlo cross-validation, independently at both Centre 1 and Centre 2, with a 70-30% train-test split. Additionally, models were trained in one centre and tested in the other, and vice versa. Furthermore, data from both centres were combined for training and testing using Monte Carlo cross-validation. Finally, a new radiomics signature built on this bicentric dataset was proposed. Several performance metrics were computed. Results: The previously generated radiomics signature resulted in an area under the receiver operating characteristic curve (AUC) of 80.4% when tested on Centre 1, while it generalised poorly to Centre 2, where it reached an AUC of 62.7%. When the whole cohort was considered, AUC was 72.5%. Similarly, new ML models trained on the previously selected features yielded, at best, an AUC of 80.9% for Centre 1 and performed at chance for Centre 2 (AUC of 49.3%). A new signature built on this bicentric dataset reached, at best, an average AUC of 91.4% in the test set. Conclusions: The satisfying performance of radiomics models when used in the original development settings, paired with the poor performance otherwise observed, emphasises the need to consider centre-specific factors and dataset characteristics when developing radiomics models. Combining radiomics datasets is a viable strategy to reduce such centre-specific biases, but external validation is still needed.
Collapse
Affiliation(s)
- Samuele Ghezzo
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Praveen Gurunath Bharathi
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Heying Duan
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Paola Mapelli
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Philipp Sorgo
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Guido Alejandro Davidzon
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Carolina Bezzi
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | | | | | - Tommaso Russo
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Department of Radiology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giorgio Brembilla
- Department of Radiology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Andreas Markus Loening
- Division of Body MRI, Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Pejman Ghanouni
- Division of Body MRI, Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Anna Grattagliano
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Alberto Briganti
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Division of Experimental Oncology, Department of Urology, Urological Research Institute (URI), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesco De Cobelli
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Department of Radiology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Geoffrey Sonn
- Department of Urology, Stanford University, Stanford, CA 94305, USA
| | - Arturo Chiti
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Farshad Moradi
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Maria Picchio
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
115
|
Xu ZL, Qian GX, Li YH, Lu JL, Wei MT, Bu XY, Ge YS, Cheng Y, Jia WD. Evaluating microvascular invasion in hepatitis B virus-related hepatocellular carcinoma based on contrast-enhanced computed tomography radiomics and clinicoradiological factors. World J Gastroenterol 2024; 30:4801-4816. [PMID: 39649551 PMCID: PMC11606376 DOI: 10.3748/wjg.v30.i45.4801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/28/2024] [Accepted: 09/23/2024] [Indexed: 11/13/2024] Open
Abstract
BACKGROUND Microvascular invasion (MVI) is a significant indicator of the aggressive behavior of hepatocellular carcinoma (HCC). Expanding the surgical resection margin and performing anatomical liver resection may improve outcomes in patients with MVI. However, no reliable preoperative method currently exists to predict MVI status or to identify patients at high-risk group (M2). AIM To develop and validate models based on contrast-enhanced computed tomography (CECT) radiomics and clinicoradiological factors to predict MVI and identify M2 among patients with hepatitis B virus-related HCC (HBV-HCC). The ultimate goal of the study was to guide surgical decision-making. METHODS A total of 270 patients who underwent surgical resection were retrospectively analyzed. The cohort was divided into a training dataset (189 patients) and a validation dataset (81) with a 7:3 ratio. Radiomics features were selected using intra-class correlation coefficient analysis, Pearson or Spearman's correlation analysis, and the least absolute shrinkage and selection operator algorithm, leading to the construction of radscores from CECT images. Univariate and multivariate analyses identified significant clinicoradiological factors and radscores associated with MVI and M2, which were subsequently incorporated into predictive models. The models' performance was evaluated using calibration, discrimination, and clinical utility analysis. RESULTS Independent risk factors for MVI included non-smooth tumor margins, absence of a peritumoral hypointensity ring, and a high radscore based on delayed-phase CECT images. The MVI prediction model incorporating these factors achieved an area under the curve (AUC) of 0.841 in the training dataset and 0.768 in the validation dataset. The M2 prediction model, which integrated the radscore from the 5 mm peritumoral area in the CECT arterial phase, α-fetoprotein level, enhancing capsule, and aspartate aminotransferase level achieved an AUC of 0.865 in the training dataset and 0.798 in the validation dataset. Calibration and decision curve analyses confirmed the models' good fit and clinical utility. CONCLUSION Multivariable models were constructed by combining clinicoradiological risk factors and radscores to preoperatively predict MVI and identify M2 among patients with HBV-HCC. Further studies are needed to evaluate the practical application of these models in clinical settings.
Collapse
Affiliation(s)
- Zi-Ling Xu
- Department of General Surgery, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, Anhui Province, China
| | - Gui-Xiang Qian
- Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Yong-Hai Li
- Department of Anorectal Surgery, The First People's Hospital of Hefei, Hefei 230001, Anhui Province, China
| | - Jian-Lin Lu
- Department of General Surgery, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, Anhui Province, China
| | - Ming-Tong Wei
- Department of General Surgery, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, Anhui Province, China
| | - Xiang-Yi Bu
- Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Yong-Sheng Ge
- Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Yuan Cheng
- Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Wei-Dong Jia
- Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| |
Collapse
|
116
|
Zhang L, Diao B, Fan Z, Zhan H. Radiomics for differentiating pancreatic Mucinous Cystic Neoplasm from Serous Cystic Neoplasm: Systematic Review and Meta-Analysis. Acad Radiol 2024:S1076-6332(24)00892-4. [PMID: 39648097 DOI: 10.1016/j.acra.2024.11.047] [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: 08/29/2024] [Revised: 11/17/2024] [Accepted: 11/17/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND As pancreatic cystic neoplasms (PCN) differ in current standard of care, and these treatments can affect quality of life to varying degrees, a definitive preoperative diagnosis must be reliable. Current diagnostic approaches, specifically traditional cross-sectional imaging techniques, face certain limitations. But radiomics has been shown to have high diagnostic accuracy across a range of diseases. Objective to conduct a comprehensive review of the literature on the use of radiomics to differentiate Mucinous Cystic Neoplasm (MCN) from Serous Cystic Neoplasm (SCN). METHODS This study was comprehensively searched in Pubmed, Scopus and Web of Science databases for meta-analysis of studies that used radiomics to distinguish MCN from SCN. Risk of bias was assessed using the diagnostic accuracy study quality assessment method and combined with sensitivity, specificity, diagnostic odds ratio, and summary receiver operating characteristic (SROC)curve analysis. RESULTS A total of 884 patients from 8 studies were included in this analysis, including 365 MCN and 519 SCN. The Meta-analysis found that radiomics identified MCN and SCN with high sensitivity and specificity, with combined sensitivity and specificity of 0.84(0.82-0.87) and 0.82(0.79-0.84). The positive likelihood ratio (PLR) and the negative likelihood ratio (NLR) are 5.61(3.72, 8.47) and 0.14(0.09-0.26). In addition, the area under the SROC curve (AUC) was drawn at 0.93. No significant risk of publication bias was detected through the funnel plot analysis. The performances of feature extraction from the volume of interest (VOI) or Using AI classifier in the radiomics models were superior to those of protocols employing region of interest (ROI) or absence of AI classifier. CONCLUSION This meta-analysis demonstrates that radiomics exhibits high sensitivity and specificity in distinguishing between MCN and SCN, and has the potential to become a reliable diagnostic tool for their identification.
Collapse
Affiliation(s)
- Longjia Zhang
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China (L.Z., B.D., Z.F., H.Z.)
| | - Boyu Diao
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China (L.Z., B.D., Z.F., H.Z.)
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China (L.Z., B.D., Z.F., H.Z.)
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China (L.Z., B.D., Z.F., H.Z.).
| |
Collapse
|
117
|
Zheng X, Liu K, Gao Z, Li C, Tong L, Rong C, Li S, Liu Y, Wu X. Predicting overall survival and prophylactic cranial irradiation benefit in small cell lung cancer patients: a multicenter cohort study. BMC Cancer 2024; 24:1507. [PMID: 39643886 PMCID: PMC11622659 DOI: 10.1186/s12885-024-13274-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND To construct a CT-based radiomics nomogram, enabling the estimation of overall survival (OS) in small cell lung cancer (SCLC) patients and facilitating the identification of prophylactic cranial irradiation (PCI) beneficiaries through risk stratification using the radiomics score (RS). METHODS A retrospective recruitment of 375 patients with pathologically confirmed SCLC was conducted across three medical centers, followed by their division into different cohorts. To generate the RS, a series of analyses were performed, including Pearson correlation analysis, univariate Cox analysis, and least absolute shrinkage and selection operator (LASSO) Cox regression analysis. Subsequently, patients were stratified into either the low RS or high RS group, determined by identifying the optimal RS cutoff value. Subsequently, a radiomics nomogram was constructed using the RS, followed by assessments of its discrimination, calibration, clinical utility and reclassification. Moreover, we evaluated the potential benefits of PCI following stratification by RS. RESULTS For the internal and external validation cohorts, the radiomics nomogram (concordance index [C-index]: 0.770, 0.763) outperformed clinical nomogram (C-index: 0.625, 0.570) in predicting OS. Besides, patients with high RS had survival benefit from PCI in both the limited and extensive stage (hazard ratio [HR]: 0.304, 95% confidence interval [CI]: 0.087-1.065, P = 0.003; HR: 0.481, 95% CI: 0.270-0.860, P = 0.019, respectively), while no significant association were observed in patients with low RS. CONCLUSION A radiomics nomogram based on CT shows potential in predicting OS for individuals with SCLC. The RS could assist in tailoring treatment plans to identify patients likely to benefit from PCI.
Collapse
Affiliation(s)
- Xiaomin Zheng
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230031, China
| | - Kaicai Liu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230031, China
| | - Zhao Gao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230031, China
| | - Cuiping Li
- Department of Radiology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China
- Department of Radiology, Anhui Provincial Cancer Hospital, Hefei, 230031, China
| | - Li Tong
- Department of Radiology, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
| | - Chang Rong
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230031, China
| | - Shuai Li
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230031, China
| | - Yichao Liu
- Department of Radiology, The Affiliated Bozhou Hospital of Anhui Medical University, No. 616 Duzhong Road, Qiaocheng District, Bozhou, Anhui Province, 236000, China.
| | - Xingwang Wu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230031, China.
| |
Collapse
|
118
|
Hong Y, Jeong S, Park MJ, Song W, Lee N. Application of Pathomic Features for Differentiating Dysplastic Cells in Patients with Myelodysplastic Syndrome. Bioengineering (Basel) 2024; 11:1230. [PMID: 39768048 PMCID: PMC11673167 DOI: 10.3390/bioengineering11121230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Myelodysplastic syndromes (MDSs) are a group of hematologic neoplasms accompanied by dysplasia of bone marrow (BM) hematopoietic cells with cytopenia. Recently, digitalized pathology and pathomics using computerized feature analysis have been actively researched for classifying and predicting prognosis in various tumors of hematopoietic tissues. This study analyzed the pathomic features of hematopoietic cells in BM aspiration smears of patients with MDS according to each hematopoietic cell lineage and dysplasia. We included 24 patients with an MDS and 21 with normal BM. The 12,360 hematopoietic cells utilized were to be classified into seven types: normal erythrocytes, normal granulocytes, normal megakaryocytes, dysplastic erythrocytes, dysplastic granulocytes, dysplastic megakaryocytes, and others. Four hundred seventy-six pathomic features quantifying cell intensity, shape, and texture were extracted from each segmented cell. After comparing the combination of feature selection and machine learning classifier methods using 5-fold cross-validation area under the receiver operating characteristic curve (AUROC), the quadratic discriminant analysis (QDA) with gradient boosting decision tree (AUROC = 0.63) and QDA with eXtreme gradient boosting (XGB) (AUROC = 0.64) showed a high AUROC combination. Through a feature selection process, 30 characteristics were further analyzed. Dysplastic erythrocytes and granulocytes showed lower median values on heatmap analysis compared to that of normal erythrocytes and granulocytes. The data suggest that pathomic features could be applied to cell differentiation in hematologic malignancies. It could be used as a new biomarker with an auxiliary role for more accurate diagnosis. Further studies including prediction survival and prognosis with larger cohort of patients are needed.
Collapse
Affiliation(s)
- Youngtaek Hong
- CONNECT-AI Research Center, Yonsei University College of Medicine, Seoul 03764, Republic of Korea;
| | - Seri Jeong
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07440, Republic of Korea; (S.J.); (M.-J.P.); (W.S.)
| | - Min-Jeong Park
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07440, Republic of Korea; (S.J.); (M.-J.P.); (W.S.)
| | - Wonkeun Song
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07440, Republic of Korea; (S.J.); (M.-J.P.); (W.S.)
| | - Nuri Lee
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07440, Republic of Korea; (S.J.); (M.-J.P.); (W.S.)
| |
Collapse
|
119
|
Begal J, Sabo E, Goldberg N, Bitterman A, Khoury W. Wavelets-Based Texture Analysis of Post Neoadjuvant Chemoradiotherapy Magnetic Resonance Imaging as a Tool for Recognition of Pathological Complete Response in Rectal Cancer, a Retrospective Study. J Clin Med 2024; 13:7383. [PMID: 39685841 DOI: 10.3390/jcm13237383] [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: 08/07/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Patients with locally advanced rectal cancer (LARC) treated by neoadjuvant chemoradiotherapy (nCRT) may experience pathological complete response (pCR). Tools that can identify pCR are required to define candidates suitable for the watch and wait (WW) strategy. Automated image analysis is used for predicting clinical aspects of diseases. Texture analysis of magnetic resonance imaging (MRI) wavelets algorithms provides a novel way to identify pCR. We aimed to evaluate wavelets-based image analysis of MRI for predicting pCR. Methods: MRI images of rectal cancer from 22 patients who underwent nCRT were captured at best representative views of the tumor. The MRI images were digitized and their texture was analyzed using different mother wavelets. Each mother wavelet was used to scan the image repeatedly at different frequencies. Based on these analyses, coefficients of similarity were calculated providing a variety of textural variables that were subsequently correlated with histopathology in each case. This allowed for proper identification of the best mother wavelets able to predict pCR. The predictive formula of complete response was computed using the independent statistical variables that were singled out by the multivariate regression model. Results: The statistical model used four wavelet variables to predict pCR with an accuracy of 100%, sensitivity of 100%, specificity of 100%, and PPV and NPV of 100%. Conclusions: Wavelet-transformed texture analysis of radiomic MRI can predict pCR in patients with LARC. It may provide a potential accurate surrogate method for the prediction of clinical outcomes of nCRT, resulting in an effective selection of patients amenable to WW.
Collapse
Affiliation(s)
- Julia Begal
- Department of General Surgery, Carmel Medical Center, Haifa 3436212, Israel
| | - Edmond Sabo
- Department of Human Pathology, Carmel Medical Center, Haifa 3436212, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Natalia Goldberg
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
- Department of Radiology, Carmel Medical Center, Haifa 3436212, Israel
| | - Arie Bitterman
- Department of General Surgery, Carmel Medical Center, Haifa 3436212, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Wissam Khoury
- Department of General Surgery, Carmel Medical Center, Haifa 3436212, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
120
|
Zheng Y, Mei P, Wang M, Luo Q, Li H, Ding C, Zhang K, Chen L, Gu J, Li Y, Guo T, Zhang C, Yao W, Wei L, Liao Y, Han X, Shi H. CT-based delta-radiomics for predicting pathological response to neoadjuvant immunochemotherapy in esophageal squamous cell carcinoma: a multicenter study. BMC Med Imaging 2024; 24:329. [PMID: 39627736 PMCID: PMC11616236 DOI: 10.1186/s12880-024-01503-1] [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: 07/13/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND The study aimed to investigate the predictive value of delta-radiomics derived from computed tomography (CT) for pathological complete response (pCR) to neoadjuvant immunochemotherapy (NICT) among patients with esophageal squamous cell carcinoma (ESCC), helping clinicians determine whether to modify the neoadjuvant treatment strategy, proceed to surgery, or forgo surgery altogether. METHODS A total of 140 ESCC patients from two institutions (Database 1 = 93; Database 2 = 47) who underwent NICT and surgery were retrospectively included in the study. The training set consisted of patients from Database 1, while the testing set included patients from Database 2. All patients underwent contrast-enhanced CT scans before the start of the treatment and prior to the operation. The delta-radiomics features were calculated as the relative net change of radiomics features between the two-time points. Feature selection was conducted using Pearson correlation analysis, intraclass correlation coefficients, and the fivefold cross-validation with least absolute shrinkage and selection analysis. Four models were established, comprising a clinical model, a pre-treatment radiomics model, a delta-radiomics model, and a mixed model. Area under the curve (AUC) and decision curve analysis were used to assess the performance and the clinical value of the models. RESULTS Less than half of the tumors (40/140, 28.6%) showed pCR following NICT. The delta-radiomics model displayed AUC of 0.827 and 0.790 in the training and testing set for predicting pCR, which was superior to the clinical model based on age and clinical tumor node metastasis (cTNM) stage (0.758 and 0.615) and the pre-treatment radiomics model (0.787 and 0.621). Furthermore, the delta-radiomics model demonstrated a more excellent AUC value in the testing set than the mixed model (0.847 and 0.719), which integrated clinical and delta-radiomics features. CONCLUSIONS The delta-radiomics model exhibited better diagnostic performance in preoperative prediction of pCR for NICT in ESCC patients compared to the clinical, pre-treatment radiomics, and mixed models.
Collapse
Affiliation(s)
- Yuting Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Peiyuan Mei
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mingliang Wang
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, 450003, China
| | - Qinyue Luo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Hanting Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chengyu Ding
- Bayer Healthcare, No. 399, West Haiyang Road, Shanghai, 200126, China
| | - Kailu Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Leqing Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jin Gu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yumin Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Tingting Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chi Zhang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenjian Yao
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, 450003, China
| | - Li Wei
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, 450003, China.
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xiaoyu Han
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Heshui Shi
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
121
|
Migliorelli A, Manuelli M, Ciorba A, Stomeo F, Pelucchi S, Bianchini C. Role of Artificial Intelligence in Human Papillomavirus Status Prediction for Oropharyngeal Cancer: A Scoping Review. Cancers (Basel) 2024; 16:4040. [PMID: 39682226 PMCID: PMC11640028 DOI: 10.3390/cancers16234040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/21/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Human papillomavirus (HPV) infection is sexually transmitted and commonly widespread in the head and neck region; however, its role in tumor development and prognosis has only been demonstrated for oropharyngeal squamous cell carcinoma (HPV-OPSCC). The aim of this review is to analyze the results of the most recent literature that has investigated the use of artificial intelligence (AI) as a method for discerning HPV-positive from HPV-negative OPSCC tumors. A review of the literature was performed using PubMed/MEDLINE, EMBASE, and Cochrane Library databases, according to PRISMA for scoping review criteria (from 2017 to July 2024). A total of 15 articles and 4063 patients have been included. Eleven studies analyzed the role of radiomics, and four analyzed the role of AI in determining HPV histological positivity. The results of this scoping review indicate that AI has the potential to play a role in predicting HPV positivity or negativity in OPSCC. Further studies are required to confirm these results.
Collapse
Affiliation(s)
- Andrea Migliorelli
- ENT & Audiology Unit, Department of Neurosciences, University Hospital of Ferrara, 44100 Ferrara, Italy
| | | | | | | | | | | |
Collapse
|
122
|
Pattathil N, Lee TSJ, Huang RS, Lena ER, Felfeli T. Adherence of studies involving artificial intelligence in the analysis of ophthalmology electronic medical records to AI-specific items from the CONSORT-AI guideline: a systematic review. Graefes Arch Clin Exp Ophthalmol 2024; 262:3741-3748. [PMID: 38953984 DOI: 10.1007/s00417-024-06553-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 06/09/2024] [Accepted: 06/12/2024] [Indexed: 07/04/2024] Open
Abstract
PURPOSE In the context of ophthalmologic practice, there has been a rapid increase in the amount of data collected using electronic health records (EHR). Artificial intelligence (AI) offers a promising means of centralizing data collection and analysis, but to date, most AI algorithms have only been applied to analyzing image data in ophthalmologic practice. In this review we aimed to characterize the use of AI in the analysis of EHR, and to critically appraise the adherence of each included study to the CONSORT-AI reporting guideline. METHODS A comprehensive search of three relevant databases (MEDLINE, EMBASE, and Cochrane Library) from January 2010 to February 2023 was conducted. The included studies were evaluated for reporting quality based on the AI-specific items from the CONSORT-AI reporting guideline. RESULTS Of the 4,968 articles identified by our search, 89 studies met all inclusion criteria and were included in this review. Most of the studies utilized AI for ocular disease prediction (n = 41, 46.1%), and diabetic retinopathy was the most studied ocular pathology (n = 19, 21.3%). The overall mean CONSORT-AI score across the 14 measured items was 12.1 (range 8-14, median 12). Categories with the lowest adherence rates were: describing handling of poor quality data (48.3%), specifying participant inclusion and exclusion criteria (56.2%), and detailing access to the AI intervention or its code, including any restrictions (62.9%). CONCLUSIONS In conclusion, we have identified that AI is prominently being used for disease prediction in ophthalmology clinics, however these algorithms are limited by their lack of generalizability and cross-center reproducibility. A standardized framework for AI reporting should be developed, to improve AI applications in the management of ocular disease and ophthalmology decision making.
Collapse
Affiliation(s)
| | - Tin-Suet Joan Lee
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ryan S Huang
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Eleanor R Lena
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tina Felfeli
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada.
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
123
|
Tavakkoli MB, Abedi I, Abdollahi H, Amouheidari A, Azmoonfar R, Saber K, Hassaninejad H. Comparison prediction models of bladder toxicity based on radiomic features of CT and MRI in patients with prostate cancer undergoing radiotherapy. J Med Imaging Radiat Sci 2024; 55:101765. [PMID: 39306942 DOI: 10.1016/j.jmir.2024.101765] [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: 06/30/2024] [Revised: 08/19/2024] [Accepted: 09/04/2024] [Indexed: 12/02/2024]
Abstract
PURPOSE This study aimed to assess the radiomic features of computed tomography (CT) and magnetic resonance imaging (MRI) of the bladder wall before radiotherapy using machine learning (ML) methods to predict bladder radiotoxicity in patients with prostate cancer. METHODS This study enrolled 70 patients with pathologically confirmed prostate cancer who were candidates for radiation therapy (RT). CT and MRI of the bladder wall before radiotherapy were used to extract radiomic features. The least absolute shrinkage and selection operator (LASSO) was used for feature selection. Algorithms such as Random Forest (RF), Decision Tree (DT), Logistic Regression (LR), and K-Nearest Neighbors (KNN) have been used to develop models based on radiomic, dosimetry, and clinical parameters. The area under the curve (AUC) of the receiver operating characteristic (ROC) curve and accuracy were used to analyze the predictive power of all models. RESULTS The RF and LR models based on the radiomic features of MRI and clinical/dosimetry parameters with an AUC of 0.95 and 0.93, and an accuracy of 86% and 86%, respectively, had the highest performance in the prediction of bladder radiation toxicity. CONCLUSIONS This study showed that, firstly, CT and MRI radiomic features of the bladder wall before treatment could be used to predict bladder radiotoxicity. Second, MRI is better than CT in predicting bladder toxicity caused by radiation. And thirdly, the performance of the predictive models based on the combination of radiomic, clinical, and dosimetry characteristics was improved.
Collapse
Affiliation(s)
- Mohammad Bagher Tavakkoli
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Iraj Abedi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Abdollahi
- Department of Radiology Technology, Faculty of Allied Medical Sciences, Kerman University of Medical Sciences, Kerman, Iran; Department of Radiology, University of British Columbia, Vancouver, BC, Canada; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | | | - Rasool Azmoonfar
- Department of Radiology, School of Allied Medical Sciences, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Korosh Saber
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Hassaninejad
- Department of Radiology, Faculty of Paramedical, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
124
|
Sun C, Jiang C, Wang X, Ma S, Zhang D, Jia W. MR-Based Radiomics Predicts CDK6 Expression and Prognostic Value in High-grade Glioma. Acad Radiol 2024; 31:5141-5153. [PMID: 38964985 DOI: 10.1016/j.acra.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024]
Abstract
RATIONALE AND OBJECTIVES This study aims to assess the prognostic value of Cyclin-dependent kinases 6 (CDK6) expression levels and establish a machine learning-based radiomics model for predicting the expression levels of CDK6 in high-grade gliomas (HGG). MATERIALS AND METHODS Clinical parameters and genomic data were extracted from 310 HGG patients in the Cancer Genome Atlas (TCGA) database and 27 patients in the Repository of Molecular Brain Neoplasia Data (REMBRANDT) database. Univariate and multivariate Cox regression, as well as Kaplan-Meier analysis, were performed for prognosis analysis. The correlation between immune cell Infiltration with CDK6 was assessed using spearman correlation analysis. Radiomic features were extracted from contrast-enhanced magnetic resonance imaging (CE-MRI) in the Cancer Imaging Archive (TCIA) database (n = 82) and REMBRANDT database (n = 27). Logistic regression (LR) and support vector machine (SVM) were employed to establish the radiomics model for predicting CDK6 expression. Receiver operating characteristic curve (ROC), calibration curve, and decision curve analysis (DCA) were utilized to assess the predictive performance of the radiomics model. Generate radiomic scores (RS) based on the LR model. An RS-based nomogram was constructed to predict the prognosis of HGG. RESULTS CDK6 was significantly overexpressed in HGG tissues and was related to lower overall survival. A significant elevation in infiltrating M0 macrophages was observed in the CDK6 high group (P < 0.001). The LR radiomics model for the prediction of CDK6 expression levels (AUC=0.810 in the training cohort, AUC = 0.784 after cross-validation, AUC=0.750 in the testing cohort) was established utilizing three radiomic features. The predictive efficiencies of the RS-based nomogram, as measured by AUC, were 0.769 for 1-year, 0.815 for 3-year, and 0.780 for 5-year, respectively. CONCLUSION The expression level of CDK6 can impact the prognosis of patients with HGG. The expression level of HGG can be noninvasively prognosticated utilizing a radiomics model.
Collapse
Affiliation(s)
- Chen Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Chenggang Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Xi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Dainan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China.
| |
Collapse
|
125
|
Benz L, Heck K, Hevisov D, Kugelmann D, Tseng PC, Sreij Z, Litzenburger F, Waschke J, Schwendicke F, Kienle A, Hickel R, Kunzelmann KH, Walter E. Visualization of Pulpal Structures by SWIR in Endodontic Access Preparation. J Dent Res 2024; 103:1375-1383. [PMID: 39101558 PMCID: PMC11633072 DOI: 10.1177/00220345241262949] [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] [Indexed: 08/06/2024] Open
Abstract
Endodontic access preparation is one of the initial steps in root canal treatments and can be hindered by the obliteration of pulp canals and formation of tertiary dentin. Until now, methods for direct intraoperative visualization of the 3-dimensional anatomy of teeth have been missing. Here, we evaluate the use of shortwave infrared radiation (SWIR) for navigation during stepwise access preparation. Nine teeth (3 anteriors, 3 premolars, and 3 molars) were explanted en bloc with intact periodontium including alveolar bone and mucosa from the upper or lower jaw of human body donors. Analysis was performed at baseline as well as at preparation depths of 5 mm, 7 mm, and 9 mm, respectively. For reflection, SWIR was used at a wavelength of 1,550 nm from the occlusal direction, whereas for transillumination, SWIR was passed through each sample at the marginal gingiva from the buccal as well as oral side at a wavelength of 1,300 nm. Pulpal structures could be identified as darker areas approximately 2 mm before reaching the pulp chamber using SWIR transillumination, although they were indistinguishable under normal circumstances. Furcation areas in molars appeared with higher intensity than areas with canals. The location of pulpal structures was confirmed by superimposition of segmented micro-computed tomography (µCT) images. By radiomic analysis, significant differences between pulpal and parapulpal areas could be detected in image features. With hierarchical cluster analysis, both segments could be confirmed and associated with specific clusters. The local thickness of µCTs was calculated and correlated with SWIR transillumination images, by which a linear dependency of thickness and intensity could be demonstrated. Lastly, by in silico simulations of light propagation, dentin tubules were shown to be a crucial factor for understanding the visibility of the pulp. In conclusion, SWIR transillumination may allow direct clinical live navigation during endodontic access preparation.
Collapse
Affiliation(s)
- L. Benz
- Department of Conservative Dentistry and Periodontology, LMU Hospital, LMU, Munich, Germany
| | - K. Heck
- Department of Conservative Dentistry and Periodontology, LMU Hospital, LMU, Munich, Germany
| | - D. Hevisov
- Institut für Lasertechnologien in der Medizin und Meßtechnik an der Universität Ulm, Ulm, Germany
| | - D. Kugelmann
- Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - P.-C. Tseng
- Department of Conservative Dentistry and Periodontology, LMU Hospital, LMU, Munich, Germany
| | - Z. Sreij
- Department of Conservative Dentistry and Periodontology, LMU Hospital, LMU, Munich, Germany
| | - F. Litzenburger
- Department of Conservative Dentistry and Periodontology, LMU Hospital, LMU, Munich, Germany
| | - J. Waschke
- Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - F. Schwendicke
- Department of Conservative Dentistry and Periodontology, LMU Hospital, LMU, Munich, Germany
| | - A. Kienle
- Institut für Lasertechnologien in der Medizin und Meßtechnik an der Universität Ulm, Ulm, Germany
| | - R. Hickel
- Department of Conservative Dentistry and Periodontology, LMU Hospital, LMU, Munich, Germany
| | - K.-H. Kunzelmann
- Department of Conservative Dentistry and Periodontology, LMU Hospital, LMU, Munich, Germany
| | - E. Walter
- Department of Conservative Dentistry and Periodontology, LMU Hospital, LMU, Munich, Germany
| |
Collapse
|
126
|
Wang W, Sheng R, Liao S, Wu Z, Wang L, Liu C, Yang C, Jiang R. LightGBM is an Effective Predictive Model for Postoperative Complications in Gastric Cancer: A Study Integrating Radiomics with Ensemble Learning. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:3034-3048. [PMID: 38940888 PMCID: PMC11612084 DOI: 10.1007/s10278-024-01172-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/29/2024]
Abstract
Postoperative complications of radical gastrectomy seriously affect postoperative recovery and require accurate risk prediction. Therefore, this study aimed to develop a prediction model specifically tailored to guide perioperative clinical decision-making for postoperative complications in patients with gastric cancer. A retrospective analysis was conducted on patients who underwent radical gastrectomy at the First Affiliated Hospital of Nanjing Medical University between April 2022 and June 2023. A total of 166 patients were enrolled. Patient demographic characteristics, laboratory examination results, and surgical pathological features were recorded. Preoperative abdominal CT scans were used to segment the visceral fat region of the patients through 3Dslicer, a 3D Convolutional Neural Network (3D-CNN) to extract image features and the LASSO regression model was employed for feature selection. Moreover, an ensemble learning strategy was adopted to train the features and predict postoperative complications of gastric cancer. The prediction performance of the LGBM (Light Gradient Boosting Machine), XGB (XGBoost), RF (Random Forest), and GBDT (Gradient Boosting Decision Tree) models was evaluated through fivefold cross-validation. This study successfully constructed a model for predicting early complications following radical gastrectomy based on the optimal algorithm, LGBM. The LGBM model yielded an AUC value of 0.9232 and an accuracy of 87.28% (95% CI, 75.61-98.95%), surpassing the performance of other models. Through ensemble learning and integration of perioperative clinical data and visceral fat radiomics, a predictive LGBM model was established. This model has the potential to facilitate individualized clinical decision-making and the early recovery of patients with gastric cancer post-surgery.
Collapse
Affiliation(s)
- Wenli Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Rongrong Sheng
- Information Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Shumei Liao
- Information Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Linjun Wang
- Department of Gastric Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Riyue Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
127
|
Tan HQ, Cai J, Tay SH, Sim AY, Huang L, Chua ML, Tang Y. Cluster-based radiomics reveal spatial heterogeneity of bevacizumab response for treatment of radiotherapy-induced cerebral necrosis. Comput Struct Biotechnol J 2024; 23:43-51. [PMID: 38125298 PMCID: PMC10730953 DOI: 10.1016/j.csbj.2023.11.040] [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: 08/02/2023] [Revised: 11/21/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Background Bevacizumab is used in the treatment of radiation necrosis (RN), which is a debilitating toxicity following head and neck radiotherapy. However, there is no biomarker to predict if a patient would respond to bevacizumab. Purpose We aimed to develop a cluster-based radiomics approach to characterize the spatial heterogeneity of RN and map their responses to bevacizumab. Methods 118 consecutive nasopharyngeal carcinoma patients diagnosed with RN were enrolled. We divided 152 lesions from the patients into 101 for training, and 51 for validation. We extracted voxel-level radiomics features from each lesion segmented on T1-weighted+contrast and T2 FLAIR sequences of pre- and post-bevacizumab magnetic resonance images, followed by a three-step analysis involving individual- and population-level clustering, before delta-radiomics to derive five radiomics clusters within the lesions. We tested the association of each cluster with response to bevacizumab and developed a clinico-radiomics model using clinical predictors and cluster-specific features. Results 71 (70.3%) and 34 (66.7%) lesions had responded to bevacizumab in the training and validation datasets, respectively. Two radiomics clusters were spatially mapped to the edema region, and the volume changes were significantly associated with bevacizumab response (OR:11.12 [95% CI: 2.54-73.47], P = 0.004; and 1.63[1.07-2.78], P = 0.042). The combined clinico-radiomics model based on textural features extracted from the most significant cluster improved the prediction of bevacizumab response, compared with a clinical-only model (AUC:0.755 [0.645-0.865] to 0.852 [0.764-0.940], training; 0.708 [0.554-0.861] to 0.816 [0.699-0.933], validation). Conclusion Our radiomics approach yielded intralesional resolution, enabling a more refined feature selection for predicting bevacizumab efficacy in the treatment of RN.
Collapse
Affiliation(s)
- Hong Qi Tan
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore
| | - Jinhua Cai
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shi Hui Tay
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore
| | - Adelene Y.L. Sim
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore
| | - Luo Huang
- Department of Radiation Oncology, Chongqing University Cancer Hospital, People's Republic of China
| | - Melvin L.K. Chua
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore
- Oncology Academic Programme, Duke-NUS Medical School, Singapore
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
128
|
Xu C, Wang Z, Wang A, Zheng Y, Song Y, Wang C, Yang G, Ma M, He M. Breast Cancer: Multi-b-Value Diffusion Weighted Habitat Imaging in Predicting Pathologic Complete Response to Neoadjuvant Chemotherapy. Acad Radiol 2024; 31:4733-4742. [PMID: 38890032 DOI: 10.1016/j.acra.2024.06.004] [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: 04/19/2024] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 06/20/2024]
Abstract
RATIONALE AND OBJECTIVES The aim of this study was to ascertain whether the utilization of multiple b-value diffusion-weighted habitat imaging, a technique that depicts tumor heterogeneity, could aid in identifying breast cancer patients who would derive substantial benefit from neoadjuvant chemotherapy (NAC). MATERIALS AND METHODS This prospective study enrolled 143 women (II-III breast cancer), who underwent multi-b-value diffusion-weighted imaging (DWI) in 3-T magnetic resonance (MR) before NAC. The patient cohort was partitioned into a training set (consisting of 100 patients, of which 36 demonstrated a pathologic complete response [pCR]) and a test set (featuring 43 patients, 16 of whom exhibited pCR). Utilizing the training set, predictive models for pCR, were constructed using different parameters: whole-tumor radiomics (ModelWH), diffusion-weighted habitat-imaging (ModelHabitats), conventional MRI features (ModelCF), along with combined models ModelHabitats+CF. The performance of these models was assessed based on the area under the receiver operating characteristic curve (AUC) and calibration slope. RESULTS In the prediction of pCR, ModelWH, ModelHabitats, ModelCF, and ModelHabitats+CF achieved AUCs of 0.733, 0.722, 0.705, and 0.756 respectively, within the training set. These scores corresponded to AUCs of 0.625, 0.801, 0.700, and 0.824 respectively in the test set. The DeLong test revealed no significant difference between ModelWH and ModelHabitats (P = 0.182), between ModelHabitats and ModelHabitats+CF (P = 0.113). CONCLUSION The habitat model we developed, incorporating first-order features along with conventional MRI features, has demonstrated accurate predication of pCR prior to NAC. This model holds the potential to augment decision-making processes in personalized treatment strategies for breast cancer.
Collapse
Affiliation(s)
- Chao Xu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China (C.X., Z.W., Y.Z., M.M., M.H.); Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, China (C.X.)
| | - Zhihong Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China (C.X., Z.W., Y.Z., M.M., M.H.); Department of Hematology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, China (Z.W.)
| | - Ailing Wang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, China (A.W., C.W., G.Y.)
| | - Yunyan Zheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China (C.X., Z.W., Y.Z., M.M., M.H.); Department of Radiology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, China (Y.Z., M.M., M.H.)
| | - Yang Song
- MR Scientific Marketing, Siemens Healthineers Ltd., Shanghai, China (Y.S.)
| | - Chenglong Wang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, China (A.W., C.W., G.Y.)
| | - Guang Yang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, China (A.W., C.W., G.Y.)
| | - Mingping Ma
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China (C.X., Z.W., Y.Z., M.M., M.H.); Department of Radiology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, China (Y.Z., M.M., M.H.)
| | - Muzhen He
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China (C.X., Z.W., Y.Z., M.M., M.H.); Department of Radiology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, China (Y.Z., M.M., M.H.).
| |
Collapse
|
129
|
Alikhani R, Horbal SR, Rothberg AE, Pai MP. Radiomic-based biomarkers: Transforming age and body composition metrics into personalized age-informed indices. Clin Transl Sci 2024; 17:e70062. [PMID: 39644153 PMCID: PMC11624483 DOI: 10.1111/cts.70062] [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: 07/25/2024] [Revised: 09/03/2024] [Accepted: 10/16/2024] [Indexed: 12/09/2024] Open
Abstract
Chronological age has been the standard for quantifying the aging process. While it is simple to quantify it cannot fully discern the biological variability of aging between individuals. The growing body of interest in this variability of human aging has led to the introduction of new biomarkers to operationalize biological age. The inclusion of body composition may provide additional value to biological aging as a prediction and estimation factor of individual health outcomes. Diagnostic images based on radiomic techniques such as Computed Tomography contain an untapped wealth of patient-specific data that remain inaccessible to healthcare providers. These images are beneficial for collecting information from body composition that adds precision and granularity when compared to traditional measures. This information can subsequently be aggregated to construct models for changes in the human body associated with aging. In addition, aging leads to a natural decline in the best parameter of drug dosing in older adults, glomerular filtration rate. Since the conventional models of kidney function are correlated with age and body composition, the radiomic biomarkers representing age-related changes in body composition may also serve as potential new imaging biomarkers of kidney function for personalized dosing. Our review introduces potential radiomic biomarkers as measures of body composition change targeting the aging processes. As a functional example, we have hypothesized an age-related model of radiomics as a covariate of kidney function to improve personalized dosing. Future research focusing on evaluating this hypothesis in human subject studies is acknowledged.
Collapse
Affiliation(s)
- Radin Alikhani
- Department of Clinical Pharmacy, College of PharmacyUniversity of MichiganAnn ArborMichiganUSA
| | | | - Amy E. Rothberg
- Department of Internal Medicine – Metabolism, Endocrinology, and DiabetesUniversity of MichiganAnn ArborMichiganUSA
| | - Manjunath P. Pai
- Department of Clinical Pharmacy, College of PharmacyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
130
|
Huang W, Pan Y, Wang H, Jiang L, Liu Y, Wang S, Dai H, Ye R, Yan C, Li Y. Delta-radiomics Analysis Based on Multi-phase Contrast-enhanced MRI to Predict Early Recurrence in Hepatocellular Carcinoma After Percutaneous Thermal Ablation. Acad Radiol 2024; 31:4934-4945. [PMID: 38902111 DOI: 10.1016/j.acra.2024.06.002] [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: 04/26/2024] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 06/22/2024]
Abstract
RATIONALE AND OBJECTIVES It is critical to predict early recurrence (ER) after percutaneous thermal ablation (PTA) for hepatocellular carcinoma (HCC). We aimed to develop and validate a delta-radiomics nomogram based on multi-phase contrast-enhanced magnetic resonance imaging (MRI) to preoperatively predict ER of HCC after PTA. MATERIALS AND METHODS We retrospectively enrolled 164 patients with HCC and divided them into training, temporal validation, and other-scanner validation cohorts (n = 110, 29, and 25, respectively). The volumes of interest of the intratumoral and/or peritumoral regions were delineated on preoperative multi-phase MR images. Original radiomics features were extracted from each phase, and delta-radiomics features were calculated. Logistic regression was used to train the corresponding radiomics models. The clinical and radiological characteristics were evaluated and combined to establish a clinical-radiological model. A fusion model comprising the best radiomics scores and clinical-radiological risk factors was constructed and presented as a nomogram. The performance of each model was evaluated and recurrence-free survival (RFS) was assessed. RESULTS Child-Pugh grade B, high-risk tumor location, and an incomplete/absent tumor capsule were independent predictors of ER. The optimal radiomics model comprised 12 delta-radiomics features with areas under the curve (AUCs) of 0.834, 0.795, and 0.769 in the training, temporal validation, and other-scanner validation cohorts, respectively. The nomogram showed the best predictive performance with AUCs as 0.893, 0.854, and 0.827 in the three datasets. There was a statistically significant difference in RFS between the risk groups calculated using the delta-radiomics model and nomogram. CONCLUSIONS The nomogram combined with the delta-radiomic score and clinical-radiological risk factors could non-invasively predict ER of HCC after PTA.
Collapse
Affiliation(s)
- Wanrong Huang
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Yifan Pan
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Huifang Wang
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Lu Jiang
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Yamei Liu
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Shunli Wang
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Hanting Dai
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Rongping Ye
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China; Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Chuan Yan
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China; Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yueming Li
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China; Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China; Key Laboratory of Radiation Biology of Fujian higher education institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China.
| |
Collapse
|
131
|
Wu C, Chen Q, Wang H, Guan Y, Mian Z, Huang C, Ruan C, Song Q, Jiang H, Pan J, Li X. A review of deep learning approaches for multimodal image segmentation of liver cancer. J Appl Clin Med Phys 2024; 25:e14540. [PMID: 39374312 PMCID: PMC11633801 DOI: 10.1002/acm2.14540] [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: 03/27/2024] [Revised: 05/30/2024] [Accepted: 08/13/2024] [Indexed: 10/09/2024] Open
Abstract
This review examines the recent developments in deep learning (DL) techniques applied to multimodal fusion image segmentation for liver cancer. Hepatocellular carcinoma is a highly dangerous malignant tumor that requires accurate image segmentation for effective treatment and disease monitoring. Multimodal image fusion has the potential to offer more comprehensive information and more precise segmentation, and DL techniques have achieved remarkable progress in this domain. This paper starts with an introduction to liver cancer, then explains the preprocessing and fusion methods for multimodal images, then explores the application of DL methods in this area. Various DL architectures such as convolutional neural networks (CNN) and U-Net are discussed and their benefits in multimodal image fusion segmentation. Furthermore, various evaluation metrics and datasets currently used to measure the performance of segmentation models are reviewed. While reviewing the progress, the challenges of current research, such as data imbalance, model generalization, and model interpretability, are emphasized and future research directions are suggested. The application of DL in multimodal image segmentation for liver cancer is transforming the field of medical imaging and is expected to further enhance the accuracy and efficiency of clinical decision making. This review provides useful insights and guidance for medical practitioners.
Collapse
Affiliation(s)
- Chaopeng Wu
- Department of Radiation OncologyRenmin HospitalWuhan UniversityWuhanHubeiChina
| | - Qiyao Chen
- Department of Radiation OncologyRenmin HospitalWuhan UniversityWuhanHubeiChina
| | - Haoyu Wang
- Department of Radiation OncologyRenmin HospitalWuhan UniversityWuhanHubeiChina
| | - Yu Guan
- Department of Radiation OncologyRenmin HospitalWuhan UniversityWuhanHubeiChina
| | - Zhangyang Mian
- Department of Radiation OncologyRenmin HospitalWuhan UniversityWuhanHubeiChina
| | - Cong Huang
- Department of Radiation OncologyRenmin HospitalWuhan UniversityWuhanHubeiChina
| | - Changli Ruan
- Department of Radiation OncologyRenmin HospitalWuhan UniversityWuhanHubeiChina
| | - Qibin Song
- Department of Radiation OncologyRenmin HospitalWuhan UniversityWuhanHubeiChina
| | - Hao Jiang
- School of Electronic InformationWuhan UniversityWuhanHubeiChina
| | - Jinghui Pan
- Department of Radiation OncologyRenmin HospitalWuhan UniversityWuhanHubeiChina
- School of Electronic InformationWuhan UniversityWuhanHubeiChina
| | - Xiangpan Li
- Department of Radiation OncologyRenmin HospitalWuhan UniversityWuhanHubeiChina
| |
Collapse
|
132
|
Li H, Liu Z, Li F, Xia Y, Zhang T, Shi F, Zeng Q. Identification of Prolactinoma in Pituitary Neuroendocrine Tumors Using Radiomics Analysis Based on Multiparameter MRI. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:2865-2873. [PMID: 38844718 PMCID: PMC11612092 DOI: 10.1007/s10278-024-01153-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 12/05/2024]
Abstract
This study aims to investigate the feasibility of preoperatively predicting histological subtypes of pituitary neuroendocrine tumors (PitNETs) using machine learning and radiomics based on multiparameter MRI. Patients with PitNETs from January 2016 to May 2022 were retrospectively enrolled from four medical centers. A cfVB-Net network was used to automatically segment PitNET multiparameter MRI. Radiomics features were extracted from the MRI, and the radiomics score (Radscore) of each patient was calculated. To predict histological subtypes, the Gaussian process (GP) machine learning classifier based on radiomics features was performed. Multi-classification (six-class histological subtype) and binary classification (PRL vs. non-PRL) GP model was constructed. Then, a clinical-radiomics nomogram combining clinical factors and Radscores was constructed using the multivariate logistic regression analysis. The performance of the models was evaluated using receiver operating characteristic (ROC) curves. The PitNET auto-segmentation model eventually achieved the mean Dice similarity coefficient of 0.888 in 1206 patients (mean age 49.3 ± SD years, 52% female). In the multi-classification model, the GP of T2WI got the best area under the ROC curve (AUC), with 0.791, 0.801, and 0.711 in the training, validation, and external testing set, respectively. In the binary classification model, the GP of T2WI combined with CE T1WI demonstrated good performance, with AUC of 0.936, 0.882, and 0.791 in training, validation, and external testing sets, respectively. In the clinical-radiomics nomogram, Radscores and Hardy' grade were identified as predictors for PRL expression. Machine learning and radiomics analysis based on multiparameter MRI exhibited high efficiency and clinical application value in predicting the PitNET histological subtypes.
Collapse
Affiliation(s)
- Hongxia Li
- Department of Radiology, The Second Hospital of Shandong University, No.247 Beiyuan Road, Jinan, 250033, China
| | - Zhiling Liu
- Department of Radiology, Shandong Provincial Hospital, Jinan, 250098, China
| | - Fuyan Li
- Department of Radiology, Shandong Medical Imaging Research Institute, Jinan, 250021, China
| | - Yuwei Xia
- Shanghai United Imaging Intelligence, Co., Ltd, 701 Yunjin Road, Xuhui District, Shanghai, 200030, China
| | - Tong Zhang
- Department of Radiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin City, 150001, China
| | - Feng Shi
- Shanghai United Imaging Intelligence, Co., Ltd, 701 Yunjin Road, Xuhui District, Shanghai, 200030, China
| | - Qingshi Zeng
- Department of Radiology, Shandong Provincial Qianfoshan Hospital, No.16766 Jingshi Road, Jinan, 250013, China.
| |
Collapse
|
133
|
Chen Y, Du P, Zhang Y, Guo X, Song Y, Wang J, Yang LL, He W. Image-based multi-omics analysis for oral science: Recent progress and perspectives. J Dent 2024; 151:105425. [PMID: 39427959 DOI: 10.1016/j.jdent.2024.105425] [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: 06/30/2024] [Revised: 10/01/2024] [Accepted: 10/18/2024] [Indexed: 10/22/2024] Open
Abstract
OBJECTIVES The diagnosis and treatment of oral and dental diseases rely heavily on various types of medical imaging. Deep learning-mediated multi-omics analysis can extract more representative features than those identified through traditional diagnostic methods. This review aims to discuss the applications and recent advances in image-based multi-omics analysis in oral science and to highlight its potential to enhance traditional diagnostic approaches for oral diseases. STUDY SELECTION, DATA, AND SOURCES A systematic search was conducted in the PubMed, Web of Science, and Google Scholar databases, covering all available records. This search thoroughly examined and summarized advances in image-based multi-omics analysis in oral and maxillofacial medicine. CONCLUSIONS This review comprehensively summarizes recent advancements in image-based multi-omics analysis for oral science, including radiomics, pathomics, and photographic-based omics analysis. It also discusses the ongoing challenges and future perspectives that could provide new insights into exploiting the potential of image-based omics analysis in the field of oral science. CLINICAL SIGNIFICANCE This review article presents the state of image-based multi-omics analysis in stomatology, aiming to help oral clinicians recognize the utility of combining omics analyses with imaging during diagnosis and treatment, which can improve diagnostic accuracy, shorten times to diagnosis, save medical resources, and reduce disparity in professional knowledge among clinicians.
Collapse
Affiliation(s)
- Yizhuo Chen
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Pengxi Du
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yinyin Zhang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
134
|
Ebrahimpour L, Lemaréchal Y, Yolchuyeva S, Orain M, Lamaze F, Driussi A, Coulombe F, Joubert P, Després P, Manem VSK. Sensitivity of CT-derived radiomic features to extraction libraries and gray-level discretization in the context of immune biomarker discovery. Br J Radiol 2024; 97:1982-1991. [PMID: 39287013 DOI: 10.1093/bjr/tqae187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 05/05/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVES Radiomics can predict patient outcomes by automatically extracting a large number of features from medical images. This study is aimed to investigate the sensitivity of radiomics features extracted from 2 different pipelines, namely, Pyradiomics and RaCat, as well as the impact of gray-level discretization on the discovery of immune checkpoint inhibitors (ICIs) biomarkers. METHODS A retrospective cohort of 164 non-small cell lung cancer patients administered with ICIs was used in this study. Radiomic features were extracted from the pre-treatment CT scans. Univariate models were used to assess the association of common radiomics features between 2 libraries with progression-free survival (PFS), programmed death ligand 1 (PD-L1), and tumour infiltrating lymphocytes (CD8 counts). In addition, we also examined the impact of gray-level discretization incorporated in Pyradiomics on the robustness of features across various clinical endpoints. RESULTS We extracted 1224, 441 radiomic features using Pyradiomics and RaCat, respectively. Among these, 75 features were found to be common between the 2 libraries. Our analysis revealed that the directionality of association between radiomic features and clinical endpoints is highly dependent on the library. Notably, a larger number of Pyradiomics features were statistically associated with PFS, whereas RaCat features showed a stronger association with PD-L1 expression. Furthermore, intensity-based features were found to have a consistent association with clinical endpoints regardless of the gray-level discretization parameters in Pyradiomics-extracted features. CONCLUSIONS This study highlights the heterogeneity of radiomics libraries and the gray-level discretization parameters that will impact the feature selection and predictive model development for biomarkers. Importantly, our work highlights the significance of standardizing radiomic features to facilitate translational studies that use imaging as an endpoint. ADVANCES IN KNOWLEDGE Our study emphasizes the need to select stable CT-derived handcrafted features to build immunotherapy biomarkers, which is a necessary precursor for multi-institutional validation of imaging biomarkers.
Collapse
Affiliation(s)
- Leyla Ebrahimpour
- Department of Physics, Engineering Physics and Optics, Université Laval, Quebec City, G1V 0A6, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, G1V 0A6, Canada
- Centre de Recherche du CHU de Québec - Université Laval, 6 McMahon, Quebec, Quebec G1R3S3, Canada
| | - Yannick Lemaréchal
- Department of Physics, Engineering Physics and Optics, Université Laval, Quebec City, G1V 0A6, Canada
| | - Sevinj Yolchuyeva
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, G1V 0A6, Canada
- Centre de Recherche du CHU de Québec - Université Laval, 6 McMahon, Quebec, Quebec G1R3S3, Canada
- Cancer Research Center, Université Laval, 2325 Rue de l'Université, Quebec, Quebe G1V0A6, Canada
- Big Data Research Center, Université Laval, 2325 Rue de l'Université, Quebec, Quebec G1V0A6, Canada
| | - Michèle Orain
- Quebec Heart & Lung Institute Research Center, Quebec City, G1V 4G5, Canada
| | - Fabien Lamaze
- Quebec Heart & Lung Institute Research Center, Quebec City, G1V 4G5, Canada
| | - Arnaud Driussi
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, G1V 0A6, Canada
- Quebec Heart & Lung Institute Research Center, Quebec City, G1V 4G5, Canada
| | - François Coulombe
- Quebec Heart & Lung Institute Research Center, Quebec City, G1V 4G5, Canada
| | - Philippe Joubert
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, G1V 0A6, Canada
- Quebec Heart & Lung Institute Research Center, Quebec City, G1V 4G5, Canada
| | - Philippe Després
- Department of Physics, Engineering Physics and Optics, Université Laval, Quebec City, G1V 0A6, Canada
- Quebec Heart & Lung Institute Research Center, Quebec City, G1V 4G5, Canada
| | - Venkata S K Manem
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, G1V 0A6, Canada
- Centre de Recherche du CHU de Québec - Université Laval, 6 McMahon, Quebec, Quebec G1R3S3, Canada
- Cancer Research Center, Université Laval, 2325 Rue de l'Université, Quebec, Quebe G1V0A6, Canada
- Big Data Research Center, Université Laval, 2325 Rue de l'Université, Quebec, Quebec G1V0A6, Canada
| |
Collapse
|
135
|
Liu M, Duan R, Xu Z, Fu Z, Li Z, Pan A, Lin Y. CT-based radiomics combined with clinical features for invasiveness prediction and pathological subtypes classification of subsolid pulmonary nodules. Eur J Radiol Open 2024; 13:100584. [PMID: 39041055 PMCID: PMC11260948 DOI: 10.1016/j.ejro.2024.100584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/29/2024] [Accepted: 06/18/2024] [Indexed: 07/24/2024] Open
Abstract
Purpose To construct optimal models for predicting the invasiveness and pathological subtypes of subsolid nodules (SSNs) based on CT radiomics and clinical features. Materials and Methods This study was a retrospective study involving two centers. A total of 316 patients with 353 SSNs confirmed as atypical adenomatous hyperplasia (AAH), adenocarcinoma in situ (AIS), minimally invasive adenocarcinoma (MIA) and invasive adenocarcinoma (IAC) were included from January 2019 to February 2023. Models based on CT radiomics and clinical features were constructed for classification of AAH/AIS and MIA, MIA and IAC, as well as lepidic-predominant adenocarcinoma (LPA) and acinar-predominant adenocarcinoma (APA). Receiver operating characteristic (ROC) curve was used to evaluate the model performance. Finally, the nomograms based on the optimal models were established. Results The nomogram based on the combined model (AAH/AIS versus MIA) consisting of lobulation, the GGN-vessel relationship, diameter, CT value, consolidation tumor ratio (CTR) and rad-score performed the best (AUC=0.841), while age, CT value, CTR and rad-score were the significant features for distinguishing MIA from IAC, the nomogram based on these features performed the best (AUC=0.878). There were no significant differences in clinical features between LPA and APA, while the radiomics model based on rad-score showed good performance for distinguishing LPA from APA (AUC=0.926). Conclusions The nomograms based on radiomics and clinical features could predict the invasiveness of SSNs accurately. Moreover, radiomics models showed good performance in distinguishing LPA from APA.
Collapse
Affiliation(s)
- Miaozhi Liu
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province 515041, PR China
| | - Rui Duan
- Department of Radiology, First People's Hospital of Foshan, Foshan, Guangdong Province 528000, PR China
| | - Zhifeng Xu
- Department of Radiology, First People's Hospital of Foshan, Foshan, Guangdong Province 528000, PR China
| | - Zijie Fu
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province 515041, PR China
| | - Zhiheng Li
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province 515041, PR China
| | - Aizhen Pan
- Department of Radiology, First People's Hospital of Foshan, Foshan, Guangdong Province 528000, PR China
| | - Yan Lin
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province 515041, PR China
| |
Collapse
|
136
|
Hatamikia S, George G, Schwarzhans F, Mahbod A, Woitek R. Breast MRI radiomics and machine learning-based predictions of response to neoadjuvant chemotherapy - How are they affected by variations in tumor delineation? Comput Struct Biotechnol J 2024; 23:52-63. [PMID: 38125296 PMCID: PMC10730996 DOI: 10.1016/j.csbj.2023.11.016] [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: 05/14/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 12/23/2023] Open
Abstract
Manual delineation of volumes of interest (VOIs) by experts is considered the gold-standard method in radiomics analysis. However, it suffers from inter- and intra-operator variability. A quantitative assessment of the impact of variations in these delineations on the performance of the radiomics predictors is required to develop robust radiomics based prediction models. In this study, we developed radiomics models for the prediction of pathological complete response to neoadjuvant chemotherapy in patients with two different breast cancer subtypes based on contrast-enhanced magnetic resonance imaging acquired prior to treatment (baseline MRI scans). Different mathematical operations such as erosion, smoothing, dilation, randomization, and ellipse fitting were applied to the original VOIs delineated by experts to simulate variations of segmentation masks. The effects of such VOI modifications on various steps of the radiomics workflow, including feature extraction, feature selection, and prediction performance, were evaluated. Using manual tumor VOIs and radiomics features extracted from baseline MRI scans, an AUC of up to 0.96 and 0.89 was achieved for human epidermal growth receptor 2 positive and triple-negative breast cancer, respectively. For smoothing and erosion, VOIs yielded the highest number of robust features and the best prediction performance, while ellipse fitting and dilation lead to the lowest robustness and prediction performance for both breast cancer subtypes. At most 28% of the selected features were similar to manual VOIs when different VOI delineation data were used. Differences in VOI delineation affect different steps of radiomics analysis, and their quantification is therefore important for development of standardized radiomics research.
Collapse
Affiliation(s)
- Sepideh Hatamikia
- Danube Private University, Krems, Rathausplatz 1, Krems-Stein, AT-3500, Austria
- Austrian Center for Medical Innovation and Technology (ACMIT), Viktor Kaplan-Straße 2/1, Wiener Neustadt 2700, Austria
| | - Geevarghese George
- Danube Private University, Krems, Rathausplatz 1, Krems-Stein, AT-3500, Austria
| | - Florian Schwarzhans
- Danube Private University, Krems, Rathausplatz 1, Krems-Stein, AT-3500, Austria
| | - Amirreza Mahbod
- Danube Private University, Krems, Rathausplatz 1, Krems-Stein, AT-3500, Austria
| | - Ramona Woitek
- Danube Private University, Krems, Rathausplatz 1, Krems-Stein, AT-3500, Austria
| |
Collapse
|
137
|
Liu WQ, Xue YT, Huang XY, Lin B, Li XD, Ke ZB, Chen DN, Chen JY, Wei Y, Zheng QS, Xue XY, Xu N. Development and Validation of an MRI-Based Radiomics Nomogram to Predict the Prognosis of De Novo Oligometastatic Prostate Cancer Patients. Cancer Med 2024; 13:e70481. [PMID: 39704412 DOI: 10.1002/cam4.70481] [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: 01/06/2024] [Revised: 11/16/2024] [Accepted: 11/28/2024] [Indexed: 12/21/2024] Open
Abstract
OBJECTIVE We aimed to develop and validate a nomogram based on MRI radiomics to predict overall survival (OS) for patients with de novo oligometastatic prostate cancer (PCa). METHODS A total of 165 patients with de novo oligometastatic PCa were included in the study (training cohort, n = 115; validating cohort, n = 50). Among them, MRI scans were conducted and T2-weighted imaging (T2WI) and apparent diffusion coefficient (ADC) sequences were collected for radiomics features along with their clinicopathological features. Radiological features were extracted from T2WI and ADC sequences for prostate tumors. Univariate Cox regression analysis and the least absolute shrinkage and selection operator (LASSO) combined with 10-fold cross-validation were used to select the optimal features on each sequence. Then, a weighted radiomics score (Rad-score) was generated and independent risk factors were obtained from univariate and multivariate Cox regressions to build the nomogram. Model performance was assessed using receiver operating characteristic (ROC) curves, calibration, and decision curve analysis (DCA). RESULTS Eastern Cooperative Oncology Group (ECOG) score, absolute neutrophil count (ANC) and Rad-score were included in the nomogram as independent risk factors for OS in de novo oligometastatic PCa patients. We found that the areas under the curves (AUCs) in the training cohort were 0.734, 0.851, and 0.773 for predicting OS at 1, 2, and 3 years, respectively. In the validating cohort, the AUCs were 0.703, 0.799, and 0.833 for predicting OS at 1, 2, and 3 years, respectively. Furthermore, the clinical relevance of the predictive nomogram was confirmed through the analysis of DCA and calibration curve analysis. CONCLUSION The MRI-based nomogram incorporating Rad-score and clinical data was developed to guide the OS assessment of oligometastatic PCa. This helps in understanding the prognosis and improves the shared decision-making process.
Collapse
Affiliation(s)
- Wen-Qi Liu
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yu-Ting Xue
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xu-Yun Huang
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Bin Lin
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiao-Dong Li
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zhi-Bin Ke
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Dong-Ning Chen
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jia-Yin Chen
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yong Wei
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qing-Shui Zheng
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xue-Yi Xue
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ning Xu
- Department of Urology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
138
|
Lyu W, Gong J, Zhu L, Xu T, Huang S, Shen C, Wang C, He X, Ying H, Hu C, Wang Y, Ji Q, Gu Y, Zhou X, Lu X. MR radiomics unveils neoadjuvant chemo-responsiveness with insights into selective treatment de-intensification in HPV-positive oropharyngeal carcinoma. Oral Oncol 2024; 159:107049. [PMID: 39341091 DOI: 10.1016/j.oraloncology.2024.107049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/26/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Accurate prediction of neoadjuvant chemotherapy (NAC) response allows for NAC-guided personalized treatment de-intensification in HPV-positive oropharyngeal squamous cell carcinoma (OPSCC). In this study, we aimed to apply baseline MR radiomic features to predict NAC response to help select NAC-guided de-intensification candidates, and to explore biological underpinnings of response-oriented radiomics. METHODS Pre-treatment MR images and clinical data of 131 patients with HPV-positive OPSCC were retrieved from Fudan University Shanghai Cancer Center. Patients were divided into training cohort (n = 47), validation cohort 1 (n = 49) from NAC response-adapted de-intensification trial (IChoice-01, NCT04012502) and real-world validation cohort 2 (n = 35). NAC prediction model using linear support vector machine (SVM) was built and validated. Subsequent nomograms combined radiomics and clinical characteristics were established to predict survival outcomes. RNA-seq and proteomic data were compared to interpret the molecular features underlying radiomic signatures with differential NAC response. FINDINGS For NAC response prediction, the fusion model with both oropharyngeal and nodal signatures achieved encouraging performance to predict good responders in the training cohort (AUC 0·89, 95% CI, 0·79-0·95) and validation cohort 1 (AUC 0·71, 95% CI, 0·59-0·83). For prognosis prediction, radiomics-based nomograms exhibited satisfactory discriminative ability between low-risk and high-risk patients (PFS, C-index 0·85, 0·76 and 0·83; OS, C-index 0·79, 0·76 and 0·87, respectively) in three cohorts. Expression analysis unveiled NAC poor responders had predominantly enhanced keratinization while good responders were featured by upregulated immune response and oxidative stress. INTERPRETATION The MR-based radiomic models and prognostic models efficiently discriminate among patients with different NAC response and survival risk, which help candidate selection in HPV-positive OPSCC with regard to personalized treatment de-intensification.
Collapse
Affiliation(s)
- Wenjiao Lyu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Jing Gong
- Department of Radiology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Lin Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Tingting Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Shenglin Huang
- The Shanghai Key Laboratory of Medical Epigenetics, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Chunying Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Cuihong Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Xiayun He
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Hongmei Ying
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Chaosu Hu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Qinghai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China.
| | - Xin Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China.
| | - Xueguan Lu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China.
| |
Collapse
|
139
|
Elahi R, Nazari M. An updated overview of radiomics-based artificial intelligence (AI) methods in breast cancer screening and diagnosis. Radiol Phys Technol 2024; 17:795-818. [PMID: 39285146 DOI: 10.1007/s12194-024-00842-6] [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: 06/19/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 11/21/2024]
Abstract
Current imaging methods for diagnosing breast cancer (BC) are associated with limited sensitivity and specificity and modest positive predictive power. The recent progress in image analysis using artificial intelligence (AI) has created great promise to improve BC diagnosis and subtype differentiation. In this case, novel quantitative computational methods, such as radiomics, have been developed to enhance the sensitivity and specificity of early BC diagnosis and classification. The potential of radiomics in improving the diagnostic efficacy of imaging studies has been shown in several studies. In this review article, we discuss the radiomics workflow and current handcrafted radiomics methods in the diagnosis and classification of BC based on the most recent studies on different imaging modalities, e.g., MRI, mammography, contrast-enhanced spectral mammography (CESM), ultrasound imaging, and digital breast tumosynthesis (DBT). We also discuss current challenges and potential strategies to improve the specificity and sensitivity of radiomics in breast cancer to help achieve a higher level of BC classification and diagnosis in the clinical setting. The growing field of AI incorporation with imaging information has opened a great opportunity to provide a higher level of care for BC patients.
Collapse
Affiliation(s)
- Reza Elahi
- Department of Radiology, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Mahdis Nazari
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
140
|
Ferrari R, Trinci M, Casinelli A, Treballi F, Leone E, Caruso D, Polici M, Faggioni L, Neri E, Galluzzo M. Radiomics in radiology: What the radiologist needs to know about technical aspects and clinical impact. LA RADIOLOGIA MEDICA 2024; 129:1751-1765. [PMID: 39472389 DOI: 10.1007/s11547-024-01904-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/16/2024] [Indexed: 12/17/2024]
Abstract
Radiomics represents the science of extracting and analyzing a multitude of quantitative features from medical imaging, revealing the quantitative potential of radiologic images. This scientific review aims to provide radiologists with a comprehensive understanding of radiomics, emphasizing its principles, applications, challenges, limits, and prospects. The limitations of standardization in current scientific production are analyzed, along with possible solutions proposed by some of the referenced papers. As the continuous evolution of medical imaging is ongoing, radiologists must be aware of new perspectives to play a central role in patient management.
Collapse
Affiliation(s)
- Riccardo Ferrari
- Emergency Radiology Department, San Camillo-Forlanini Hospital, Rome, Italy.
| | - Margherita Trinci
- Dipartimento Di Radiologia, P.O. Colline Dell'Albegna, Orbetello, Grosseto, Italy
| | - Alice Casinelli
- Diagnostic Imaging Department, Sandro Pertini Hospital, Rome, Italy
| | | | - Edoardo Leone
- Emergency Radiology Department, San Camillo-Forlanini Hospital, Rome, Italy
| | - Damiano Caruso
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant'Andrea University Hospital, Rome, Italy
| | - Michela Polici
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome-Sant'Andrea University Hospital, Rome, Italy
| | - Lorenzo Faggioni
- Department of Translational Research on New Technologies in Medicine e Surgery, Pisa University, Pisa, Italy
| | - Emanuele Neri
- Department of Translational Research on New Technologies in Medicine e Surgery, Pisa University, Pisa, Italy
| | - Michele Galluzzo
- Emergency Radiology Department, San Camillo-Forlanini Hospital, Rome, Italy
| |
Collapse
|
141
|
Le EPV, Wong MYZ, Rundo L, Tarkin JM, Evans NR, Weir-McCall JR, Chowdhury MM, Coughlin PA, Pavey H, Zaccagna F, Wall C, Sriranjan R, Corovic A, Huang Y, Warburton EA, Sala E, Roberts M, Schönlieb CB, Rudd JHF. Using machine learning to predict carotid artery symptoms from CT angiography: A radiomics and deep learning approach. Eur J Radiol Open 2024; 13:100594. [PMID: 39280120 PMCID: PMC11402422 DOI: 10.1016/j.ejro.2024.100594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 09/18/2024] Open
Abstract
Purpose To assess radiomics and deep learning (DL) methods in identifying symptomatic Carotid Artery Disease (CAD) from carotid CT angiography (CTA) images. We further compare the performance of these novel methods to the conventional calcium score. Methods Carotid CT angiography (CTA) images from symptomatic patients (ischaemic stroke/transient ischaemic attack within the last 3 months) and asymptomatic patients were analysed. Carotid arteries were classified into culprit, non-culprit and asymptomatic. The calcium score was assessed using the Agatston method. 93 radiomic features were extracted from regions-of-interest drawn on 14 consecutive CTA slices. For DL, convolutional neural networks (CNNs) with and without transfer learning were trained directly on CTA slices. Predictive performance was assessed over 5-fold cross validated AUC scores. SHAP and GRAD-CAM algorithms were used for explainability. Results 132 carotid arteries were analysed (41 culprit, 41 non-culprit, and 50 asymptomatic). For asymptomatic vs symptomatic arteries, radiomics attained a mean AUC of 0.96(± 0.02), followed by DL 0.86(± 0.06) and then calcium 0.79(± 0.08). For culprit vs non-culprit arteries, radiomics achieved a mean AUC of 0.75(± 0.09), followed by DL 0.67(± 0.10) and then calcium 0.60(± 0.02). For multi-class classification, the mean AUCs were 0.95(± 0.07), 0.79(± 0.05), and 0.71(± 0.07) for radiomics, DL and calcium, respectively. Explainability revealed consistent patterns in the most important radiomic features. Conclusions Our study highlights the potential of novel image analysis techniques in extracting quantitative information beyond calcification in the identification of CAD. Though further work is required, the transition of these novel techniques into clinical practice may eventually facilitate better stroke risk stratification.
Collapse
Affiliation(s)
| | - Mark Y Z Wong
- Department of Medicine, University of Cambridge, United Kingdom
| | - Leonardo Rundo
- Department of Radiology, University of Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, United Kingdom
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, Italy
| | - Jason M Tarkin
- Department of Medicine, University of Cambridge, United Kingdom
| | - Nicholas R Evans
- Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Jonathan R Weir-McCall
- Department of Radiology, University of Cambridge, United Kingdom
- Department of Radiology, Royal Papworth Hospital, Cambridge, UK
| | - Mohammed M Chowdhury
- Division of Vascular Surgery, Department of Surgery, University of Cambridge, United Kingdom
| | | | - Holly Pavey
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, United Kingdom
| | - Fulvio Zaccagna
- Department of Radiology, University of Cambridge, United Kingdom
- Department of Imaging, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Investigative Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Chris Wall
- Department of Medicine, University of Cambridge, United Kingdom
| | | | - Andrej Corovic
- Department of Medicine, University of Cambridge, United Kingdom
| | - Yuan Huang
- Department of Medicine, University of Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, United Kingdom
- EPSRC Centre for Mathematical Imaging in Healthcare, University of Cambridge, United Kingdom
| | | | - Evis Sala
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Michael Roberts
- Department of Medicine, University of Cambridge, United Kingdom
- EPSRC Centre for Mathematical Imaging in Healthcare, University of Cambridge, United Kingdom
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, United Kingdom
| | | | - James H F Rudd
- Department of Medicine, University of Cambridge, United Kingdom
- EPSRC Centre for Mathematical Imaging in Healthcare, University of Cambridge, United Kingdom
| |
Collapse
|
142
|
Ma Y, Yue P, Zhang J, Yuan J, Liu Z, Chen Z, Zhang H, Zhang C, Zhang Y, Dong C, Lin Y, Liu Y, Li S, Meng W. Early prediction of acute gallstone pancreatitis severity: a novel machine learning model based on CT features and open access online prediction platform. Ann Med 2024; 56:2357354. [PMID: 38813815 PMCID: PMC11141304 DOI: 10.1080/07853890.2024.2357354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 04/26/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Early diagnosis of acute gallstone pancreatitis severity (GSP) is challenging in clinical practice. We aimed to investigate the efficacy of CT features and radiomics for the early prediction of acute GSP severity. METHODS We retrospectively recruited GSP patients who underwent CT imaging within 48 h of admission from tertiary referral centre. Radiomics and CT features were extracted from CT scans. The clinical and CT features were selected by the random forest algorithm to develop the ML GSP model for the identification of severity of GSP (mild or severe), and its predictive efficacy was compared with radiomics model. The predictive performance was assessed by the area under operating characteristic curve. Calibration curve and decision curve analysis were performed to demonstrate the classification performance and clinical efficacy. Furthermore, we built a web-based open access GSP severity calculator. The study was registered with ClinicalTrials.gov (NCT05498961). RESULTS A total of 301 patients were enrolled. They were randomly assigned into the training (n = 210) and validation (n = 91) cohorts at a ratio of 7:3. The random forest algorithm identified the level of calcium ions, WBC count, urea level, combined cholecystitis, gallbladder wall thickening, gallstones, and hydrothorax as the seven predictive factors for severity of GSP. In the validation cohort, the areas under the curve for the radiomics model and ML GSP model were 0.841 (0.757-0.926) and 0.914 (0.851-0.978), respectively. The calibration plot shows that the ML GSP model has good consistency between the prediction probability and the observation probability. Decision curve analysis showed that the ML GSP model had high clinical utility. CONCLUSIONS We built the ML GSP model based on clinical and CT image features and distributed it as a free web-based calculator. Our results indicated that the ML GSP model is useful for predicting the severity of GSP.
Collapse
Affiliation(s)
- Yuhu Ma
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Ping Yue
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jinduo Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jinqiu Yuan
- Clinical Research Center, Big Data Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhaoqing Liu
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zixian Chen
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Hengwei Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Chao Zhang
- Department of Orthopedics, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yong Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Chunlu Dong
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yanyan Lin
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yatao Liu
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Shuyan Li
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenbo Meng
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
143
|
Song R, Chen W, Zhang J, Zhang J, Du Y, Ren J, Shi L, Cui Y, Yang X. Multiparametric MRI-based Radiomics Analysis for Prediction of Lymph Node Metastasis and Survival Outcome in Gastric Cancer: A Dual-center Study. Acad Radiol 2024; 31:4900-4911. [PMID: 38849259 DOI: 10.1016/j.acra.2024.05.032] [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: 03/24/2024] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/09/2024]
Abstract
RATIONALE AND OBJECTIVES Gastric cancer (GC) is highly heterogeneous, and accurate preoperative assessment of lymph node status remains challenging. We aimed to develop a multiparametric MRI-based model for predicting lymph node metastasis (LNM) in GC and to explore its prognostic implications. MATERIALS AND METHODS In this dual-center retrospective study, 479 GC patients undergoing preoperative multiparametric MRI before radical gastrectomy were enrolled. 1595 imaging features were extracted from T2-weighted imaging, apparent diffusion coefficient maps, and contrast-enhanced T1-weighted imaging (cT1WI), respectively. Feature selection steps, including the Boruta and Simulated Annealing algorithms, were conducted to identify key features. Different radiomics models (RMs) based on the single- and multiple-sequence were constructed. The performance of various RMs in predicting LNM was assessed in terms of discrimination, calibration, and clinical usefulness. Additionally, Kaplan-Meier survival curves were employed to estimate differences in disease-free survival (DFS) and overall survival (OS). RESULTS The multi-sequence radiomics model (MRM) achieved area under the curves (AUCs) of 0.774 [95 % confidence interval (CI), 0.703-0.845], 0.721 (95 % CI, 0.593-0.850), and 0.720 (95 % CI, 0.639-0.801) in the training and two validation cohorts, respectively, outperforming the single-sequence RMs. Notably, the RM derived from cT1WI demonstrated superior performance compared to the other two single-sequence models. Furthermore, the proposed MRM exhibited a significant association with DFS and OS in GC patients (both P < 0.05). CONCLUSION The multiparametric MRI-based radiomics model, derived from primary lesions, demonstrated moderate performance in predicting LNM and survival outcomes in patients with GC, which could provide valuable insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Ruirui Song
- Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China; Department of Radiology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
| | - Wujie Chen
- Department of Radiology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Junjie Zhang
- Department of Radiology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
| | - Jianxin Zhang
- Department of Radiology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
| | - Yan Du
- Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
| | | | - Lei Shi
- Department of Radiology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yanfen Cui
- Department of Radiology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
| | - Xiaotang Yang
- Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China; Department of Radiology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China.
| |
Collapse
|
144
|
Shahidi R, Hassannejad E, Baradaran M, Klontzas ME, ShahirEftekhar M, Shojaeshafiei F, HajiEsmailPoor Z, Chong W, Broomand N, Alizadeh M, Mozafari N, Sadeghsalehi H, Teimoori S, Farhadi A, Nouri H, Shobeiri P, Sotoudeh H. Diagnostic performance of radiomics in prediction of Ki-67 index status in non-small cell lung cancer: A systematic review and meta-analysis. J Med Imaging Radiat Sci 2024; 55:101746. [PMID: 39276704 DOI: 10.1016/j.jmir.2024.101746] [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: 06/17/2024] [Revised: 08/03/2024] [Accepted: 08/07/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND Lung cancer's high prevalence and invasiveness make it a major global health concern. The Ki-67 index, which indicates cellular proliferation, is crucial for assessing lung cancer aggressiveness. Radiomics, which extracts quantifiable features from medical images using algorithms, may provide insights into tumor behavior. This systematic review and meta-analysis evaluate the effectiveness of radiomics in predicting Ki-67 status in Non-Small Cell Lung Cancer (NSCLC) using CT scans. METHODS AND MATERIALS A comprehensive search was conducted in PubMed/MEDLINE, Embase, Scopus, and Web of Science databases from inception until April 19, 2024. Original studies discussing the performance of CT-based radiomics for predicting Ki-67 status in NSCLC cohorts were included. The quality assessment involved quality assessment of diagnostic accuracy studies (QUADAS-2), radiomics quality score (RQS) and METhodological RadiomICs Score (METRICS). Quantitative meta-analysis, using R, assessed pooled diagnostic odds ratio, sensitivity, and specificity in NSCLC cohorts. RESULTS We identified 10 studies that met the inclusion criteria, involving 2279 participants, with 9 of these studies included in quantitative meta-analysis. The pooled sensitivity and specificity of radiomics-based models for predicting Ki-67 status in NSCLC were 0.783 (95 % CI: 0.732 - 0.827) and 0.796 (95 % CI: 0.707 - 0.864) in training cohorts, and 0.803 (95 % CI: 0.744 - 0.851) and 0.696 (95 % CI: 0.613 - 0.768) in validation cohorts. It was identified in subgroup analysis that utilizing ITK-SNAP as a segmentation software contributed to a significantly higher pooled sensitivity. CONCLUSION This meta-analysis indicates promising diagnostic accuracy of radiomics in predicting Ki-67 in NSCLC.
Collapse
Affiliation(s)
- Ramin Shahidi
- School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Ehsan Hassannejad
- Department of Radiology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran.
| | - Mansoureh Baradaran
- Department of Radiology, Imam Ali Hospital, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Michail E Klontzas
- Department of Medical Imaging, University Hospital of Heraklion, Heraklion 71110, Crete, Greece; Department of Radiology, School of Medicine, University of Crete, Heraklion, 71003, Crete, Greece.
| | - Mohammad ShahirEftekhar
- Department of Radiology, Imam Ali Hospital, North Khorasan University of Medical Sciences, Bojnurd, Iran; Department of Surgery, School of Medicine, Qom University of Medical Sciences, Qom, Iran.
| | | | | | - Weelic Chong
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States of America.
| | - Nima Broomand
- Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | | | - Navid Mozafari
- School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Hamidreza Sadeghsalehi
- Department of Artificial Intelligence in Medical Sciences, Faculty of Advanced Technologies in Medicine, Iran University Of Medical Sciences, Tehran, Iran.
| | - Soraya Teimoori
- Young Researchers and Elites Club, Faculty of Medicine, Islamic Azad University, Yazd Branch, Yazd, Iran.
| | - Akram Farhadi
- Persian Gulf Tropical Medicine Research Center, Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Hamed Nouri
- School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Parnian Shobeiri
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, 10065, NY, United States.
| | - Houman Sotoudeh
- Neuroradiology Section, Department of Radiology and Neurology, The University of Alabama at Birmingham, Alabama, United States.
| |
Collapse
|
145
|
Luo L, Wang X, Xie H, Liang H, Gao J, Li Y, Xia Y, Zhao M, Shi F, Shen C, Duan X. Role of [ 18F]-PSMA-1007 PET radiomics for seminal vesicle invasion prediction in primary prostate cancer. Comput Biol Med 2024; 183:109249. [PMID: 39388841 DOI: 10.1016/j.compbiomed.2024.109249] [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: 04/26/2024] [Revised: 09/23/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
PURPOSE The purpose of this study is to investigate the diagnostic utility of [18F]-PSMA-1007 PET radiomics combined with machine learning methods to predict seminal vesicle invasion (SVI) after radical prostatectomy (RP) in prostate cancer (PCa) patients. METHODS This is a post hoc retrospective analysis for a prospective clinical trial that included a consecutive sample of PCa patients (n = 140) who had [18F]-PSMA-1007 PET/CT prior to RP. The intraprostatic lesion's volume of interest (VOI) was semi-automatically sketched using a threshold of 40 % maximum standardized uptake value (SUVmax), namely 40%SUVmax-VOI, and seminal vesicle glands were manually contoured, namely SV-VOI. Models were built using a variety of machine learning methods such as logistic regression, random forest, and support vector machine. The area under the receiver operating characteristic curve (AUC) was calculated for different models, and the prediction performances of radiomics models were compared against the radiologists' assessment. Kaplan-Meier analysis was utilized to assess the effectiveness of selected radiomics features to determine the progression-free survival (PFS) probability. RESULTS The training set had 112 patients and the test set had 28 patients. The highest AUC for the PET radiomics model of 40%SUVmax-VOI and the PET radiomics model of SV-VOI were 0.85 and 0.96 in the test set, respectively. The PET radiomics model of SV-VOI had a significantly higher AUC compared to the radiologists' assessment (P < 0.05). The Kaplan-Meier analysis showed that PET radiomics features were associated with PFS in patients with PCa. CONCLUSION Radiomics models developed by preoperative [18F]-PSMA-1007 PET were proven useful in predicting SVI, and PSMA PET radiomics features were correlated with PFS, suggesting that the PSMA PET radiomics might be an accurate tool for PCa characterization.
Collapse
Affiliation(s)
- Liang Luo
- PET/CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyi Wang
- PET/CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongjun Xie
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hua Liang
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jungang Gao
- PET/CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yang Li
- PET/CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuwei Xia
- Shanghai United Imaging Intelligence, Shanghai, China
| | - Mengmeng Zhao
- Shanghai United Imaging Intelligence, Shanghai, China
| | - Feng Shi
- Shanghai United Imaging Intelligence, Shanghai, China
| | - Cong Shen
- PET/CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyi Duan
- PET/CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
146
|
Sadeghi MH, Sina S, Alavi M, Giammarile F, Yeong CH. PET/CT-based 3D multi-class semantic segmentation of ovarian cancer and the stability of the extracted radiomics features. Phys Eng Sci Med 2024; 47:1739-1749. [PMID: 39312120 DOI: 10.1007/s13246-024-01485-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 08/28/2024] [Indexed: 12/25/2024]
Abstract
Accurate segmentation of ovarian cancer (OC) lesions in PET/CT images is essential for effective disease management, yet manual segmentation for radiomics analysis is labor-intensive and time-consuming. This study introduces the application of a 3D U-Net deep learning model, leveraging advanced 3D networks, for multi-class semantic segmentation of OC in PET/CT images and assesses the stability of the extracted radiomics features. Utilizing a dataset of 3120 PET/CT images from 39 OC patients, the dataset was divided into training (70%), validation (15%), and test (15%) subsets to optimize and evaluate the model's performance. The 3D U-Net model, especially with a VGG16 backbone, achieved notable segmentation accuracy with a Dice score of 0.74, Precision of 0.76, and Recall of 0.78. Additionally, the study demonstrated high stability in radiomics features, with over 85% of PET and 84% of CT image features showing high intraclass correlation coefficients (ICCs > 0.8). These results underscore the potential of automated 3D U-Net-based segmentation to significantly enhance OC diagnosis and treatment planning. The reliability of the extracted radiomics features from automated segmentation supports its application in clinical decision-making and personalized medicine. This research marks a significant advancement in oncology diagnostics, providing a robust and efficient method for segmenting OC lesions in PET/CT images. By addressing the challenges of manual segmentation and demonstrating the effectiveness of 3D networks, this study contributes to the growing body of evidence supporting the application of artificial intelligence in improving diagnostic accuracy and patient outcomes in oncology.
Collapse
Affiliation(s)
- Mohammad Hossein Sadeghi
- Nuclear Engineering Department, School of Mechanical Engineering, Shiraz University, Shiraz, Iran
| | - Sedigheh Sina
- Nuclear Engineering Department, School of Mechanical Engineering, Shiraz University, Shiraz, Iran.
- Radiation Research Center, School of Mechanical Engineering, Shiraz University, Shiraz, Iran.
| | - Mehrosadat Alavi
- Ionizing and Non-Ionizing Radiation protection research center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Francesco Giammarile
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, International Atomic Energy Agency, Vienna, Austria
| | - Chai Hong Yeong
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
- Digital Health and Medical Advancement Impact Lab, Taylor's University, Subang Jaya, Malaysia
| |
Collapse
|
147
|
Sun J, Wang Z, Zhu H, Yang Q, Sun Y. Advanced Gastric Cancer: CT Radiomics Prediction of Lymph Modes Metastasis After Neoadjuvant Chemotherapy. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:2910-2919. [PMID: 38886288 PMCID: PMC11612076 DOI: 10.1007/s10278-024-01148-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024]
Abstract
This study aims to create and assess machine learning models for predicting lymph node metastases following neoadjuvant treatment in advanced gastric cancer (AGC) using baseline and restaging computed tomography (CT). We evaluated CT images and pathological data from 158 patients with resected stomach cancer from two institutions in this retrospective analysis. Patients were eligible for inclusion if they had histologically proven gastric cancer. They had received neoadjuvant chemotherapy, with at least 15 lymph nodes removed. All patients received baseline and preoperative abdominal CT and had complete clinicopathological reports. They were divided into two cohorts: (a) the primary cohort (n = 125) for model creation and (b) the testing cohort (n = 33) for evaluating models' capacity to predict the existence of lymph node metastases. The diagnostic ability of the radiomics-model for lymph node metastasis was compared to traditional CT morphological diagnosis by radiologist. The radiomics model based on the baseline and preoperative CT images produced encouraging results in the training group (AUC 0.846) and testing cohort (AUC 0.843). In the training cohort, the sensitivity and specificity were 81.3% and 77.8%, respectively, whereas in the testing cohort, they were 84% and 75%. The diagnostic sensitivity and specificity of the radiologist were 70% and 42.2% (using baseline CT) and 46.3% and 62.2% (using preoperative CT). In particular, the specificity of radiomics model was higher than that of conventional CT in diagnosing N0 cases (no lymph node metastasis). The CT-based radiomics model could assess lymph node metastasis more accurately than traditional CT imaging in AGC patients following neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Jia Sun
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 GongtiSouth Road, Chaoyang District, Beijing, Beijing, 100020, China
| | - Zhilong Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Haitao Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Qi Yang
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 GongtiSouth Road, Chaoyang District, Beijing, Beijing, 100020, China.
| | - Yingshi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
148
|
Hu Y, Liu S, Ren W, Dalbeth N, Zhou R, Chen Y, Pan Y, He Y, Liu Z, Jia Z, Ge Y, Du Y, Han L. Dual-energy computed tomography-based radiomics for differentiating patients with and without gout flares. Clin Rheumatol 2024; 43:3869-3877. [PMID: 39367919 DOI: 10.1007/s10067-024-07166-1] [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: 05/01/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND There is a current lack of data pertaining to the potential link between gout flares and dual-energy computed tomography radiomic features. This study aimed to construct and validate a comprehensive dual-energy computed tomography-based radiomics model for differentiating patients with and without gout flares. METHODS The analysis included 200 patients, of whom 150 were confirmed to have experienced at least one flare in the past 12 months; the remaining 50 patients did not experience flares. The radiomic features of the tophi at the bilateral first metatarsophalangeal joints were extracted and analyzed. Optimal radiomic features were selected using the least absolute shrinkage and selection operator method, and logistic regression analysis was used to screen clinical characteristics and establish a clinical model. The optimal radiomic features were then combined with the identified independent clinical variables to develop a comprehensive model. The performances of the radiomic, clinical, and comprehensive models were evaluated using receiver operating characteristic curve analysis, calibration curves, and decision curve analysis. RESULTS Four radiomic features distinguished patients with at least one flare from those without flares and were used to establish the radiomic model. Disease duration and hypertension were independent factors that differentiated flare occurrences. The radiomic, clinical, and comprehensive models showed favorable discrimination, with areas under the receiver operating characteristic curves of 0.76 (95% CI, 0.69-0.83), 0.72(95% CI, 0.63-0.80), and 0.79(95% CI, 0.73-0.86), respectively. The calibration curves (P > 0.05) showed that the differentiated values of the comprehensive model agreed well with the actual values. Decision curve analysis demonstrated that the comprehensive model achieved higher net clinical benefits than the use of either the radiomic or clinical model alone. CONCLUSION The results of this study suggest that a radiomics model can distinguish patients with and without gout flares. Our proposed clinical radiomics nomogram can increase the efficacy of differentiating flare occurrence, which may facilitate the clinical decision-making process.
Collapse
Affiliation(s)
- Yabin Hu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Peking University People's Hospital, Qingdao, China
- Women and Children's Hospital, Qingdao University, Qingdao, China
| | - Shunli Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Ren
- Gout Laboratory, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Nicola Dalbeth
- Department of Medicine, Room 502-201D, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Rui Zhou
- Gout Laboratory, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Yizhe Chen
- Institute of Robotics and Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Intelligent Robotics, Nankai University, Tianjin, China
- Institute of Intelligence Technology and Robotic Systems, Shenzhen Research Institute of Nankai University, Tianjin, China
| | - Yuehai Pan
- Department of Hand and Foot Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuwei He
- Gout Laboratory, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Zhen Liu
- Gout Laboratory, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Zhaotong Jia
- Gout Laboratory, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | | | - Yue Du
- Institute of Robotics and Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Intelligent Robotics, Nankai University, Tianjin, China
- Institute of Intelligence Technology and Robotic Systems, Shenzhen Research Institute of Nankai University, Tianjin, China
| | - Lin Han
- Gout Laboratory, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China.
| |
Collapse
|
149
|
He Q, Huo R, Sun Y, Zheng Z, Xu H, Zhao S, Ni Y, Yu Q, Jiao Y, Zhang W, Zhao J, Cao Y. Cerebral vascular malformations: pathogenesis and therapy. MedComm (Beijing) 2024; 5:e70027. [PMID: 39654683 PMCID: PMC11625509 DOI: 10.1002/mco2.70027] [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: 08/04/2024] [Revised: 10/30/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Cerebral vascular malformations (CVMs), particularly cerebral cavernous malformations and cerebral arteriovenous malformations, pose significant neurological challenges due to their complex etiologies and clinical implications. Traditionally viewed as congenital conditions with structural abnormalities, CVMs have been treated primarily through resection, embolization, and stereotactic radiosurgery. While these approaches offer some efficacy, they often pose risks to neurological integrity due to their invasive nature. Advances in next-generation sequencing, particularly high-depth whole-exome sequencing and bioinformatics, have facilitated the identification of gene variants from neurosurgically resected CVMs samples. These advancements have deepened our understanding of CVM pathogenesis. Somatic mutations in key mechanistic pathways have been identified as causative factors, leading to a paradigm shift in CVM treatment. Additionally, recent progress in noninvasive and minimally invasive techniques, including gene imaging genomics, liquid biopsy, or endovascular biopsies (endovascular sampling of blood vessel lumens), has enabled the identification of gene variants associated with CVMs. These methods, in conjunction with clinical data, offer potential for early detection, dynamic monitoring, and targeted therapies that could be used as monotherapy or adjuncts to surgery. This review highlights advancements in CVM pathogenesis and precision therapies, outlining the future potential of precision medicine in CVM management.
Collapse
Affiliation(s)
- Qiheng He
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Ran Huo
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yingfan Sun
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Zhiyao Zheng
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Research Unit of Accurate DiagnosisTreatment, and Translational Medicine of Brain Tumors Chinese Academy of Medical Sciences and Peking Union Medical College Beijing ChinaBeijingChina
- Department of Neurosurgery Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical College Beijing ChinaBeijingChina
| | - Hongyuan Xu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Shaozhi Zhao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yang Ni
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Qifeng Yu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yuming Jiao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Wenqian Zhang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Jizong Zhao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yong Cao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
- Collaborative Innovation CenterBeijing Institute of Brain DisordersBeijingChina
| |
Collapse
|
150
|
Shen K, Su W, Liang C, Shi D, Sun J, Yu R. Differentiating small (< 2 cm) pancreatic ductal adenocarcinoma from neuroendocrine tumors with multiparametric MRI-based radiomic features. Eur Radiol 2024; 34:7553-7563. [PMID: 38869639 DOI: 10.1007/s00330-024-10837-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: 03/19/2024] [Revised: 04/08/2024] [Accepted: 05/02/2024] [Indexed: 06/14/2024]
Abstract
OBJECTIVES To assess MR-based radiomic analysis in preoperatively discriminating small (< 2 cm) pancreatic ductal adenocarcinomas (PDACs) from neuroendocrine tumors (PNETs). METHODS A total of 197 patients (146 in the training cohort, 51 in the validation cohort) from two centers were retrospectively collected. A total of 7338 radiomics features were extracted from T2-weighted, diffusion-weighted, T1-weighted, arterial phase, portal venous phase and delayed phase imaging. The optimal features were selected by the Mann-Whitney U test, Spearman's rank correlation test and least absolute shrinkage and selection operator method and used to construct the radiomic score (Rad-score). Conventional radiological and clinical features were also assessed. Multivariable logistic regression was used to construct a radiological model, a radiomic model and a fusion model. RESULTS Nine optimal features were identified and used to build the Rad-score. The radiomic model based on the Rad-score achieved satisfactory results with AUCs of 0.905 and 0.930, sensitivities of 0.780 and 0.800, specificities of 0.906 and 0.952 and accuracies of 0.836 and 0.863 for the training and validation cohorts, respectively. The fusion model, incorporating CA19-9, tumor margins, pancreatic duct dilatation and the Rad-score, exhibited the best performance with AUCs of 0.977 and 0.941, sensitivities of 0.914 and 0.852, specificities of 0.954 and 0.950, and accuracies of 0.932 and 0.894 for the training and validation cohorts, respectively. CONCLUSIONS The MR-based Rad-score is a novel image biomarker for discriminating small PDACs from PNETs. A fusion model combining radiomic, radiological and clinical features performed very well in differentially diagnosing these two tumors. CLINICAL RELEVANCE STATEMENT A fusion model combining MR-based radiomic, radiological, and clinical features could help differentiate between small pancreatic ductal adenocarcinomas and pancreatic neuroendocrine tumors. KEY POINTS Preoperatively differentiating small pancreatic ductal adenocarcinomas (PDACs) and pancreatic neuroendocrine tumors (PNETs) is challenging. Multiparametric MRI-based Rad-score can be used for discriminating small PDACs from PNETs. A fusion model incorporating radiomic, radiological, and clinical features differentiated small PDACs from PNETs well.
Collapse
Affiliation(s)
- Keren Shen
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Weijie Su
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Chunmiao Liang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Dan Shi
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Risheng Yu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|