51
|
Zhao B, Cao B, Xia T, Zhu L, Yu Y, Lu C, Tang T, Wang Y, Ju S. Multiparametric MRI for Assessment of the Biological Invasiveness and Prognosis of Pancreatic Ductal Adenocarcinoma in the Era of Artificial Intelligence. J Magn Reson Imaging 2025. [PMID: 39781607 DOI: 10.1002/jmri.29708] [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: 10/07/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/12/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the deadliest malignant tumor, with a grim 5-year overall survival rate of about 12%. As its incidence and mortality rates rise, it is likely to become the second-leading cause of cancer-related death. The radiological assessment determined the stage and management of PDAC. However, it is a highly heterogeneous disease with the complexity of the tumor microenvironment, and it is challenging to adequately reflect the biological aggressiveness and prognosis accurately through morphological evaluation alone. With the dramatic development of artificial intelligence (AI), multiparametric magnetic resonance imaging (mpMRI) using specific contrast media and special techniques can provide morphological and functional information with high image quality and become a powerful tool in quantifying intratumor characteristics. Besides, AI has been widespread in the field of medical imaging analysis. Radiomics is the high-throughput mining of quantitative image features from medical imaging that enables data to be extracted and applied for better decision support. Deep learning is a subset of artificial neural network algorithms that can automatically learn feature representations from data. AI-enabled imaging biomarkers of mpMRI have enormous promise to bridge the gap between medical imaging and personalized medicine and demonstrate huge advantages in predicting biological characteristics and the prognosis of PDAC. However, current AI-based models of PDAC operate mainly in the realm of a single modality with a relatively small sample size, and the technical reproducibility and biological interpretation present a barrage of new potential challenges. In the future, the integration of multi-omics data, such as radiomics and genomics, alongside the establishment of standardized analytical frameworks will provide opportunities to increase the robustness and interpretability of AI-enabled image biomarkers and bring these biomarkers closer to clinical practice. EVIDENCE LEVEL: 3 TECHNICAL EFFICACY: Stage 4.
Collapse
Affiliation(s)
- Ben Zhao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Buyue Cao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Tianyi Xia
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Liwen Zhu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Yaoyao Yu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Chunqiang Lu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Tianyu Tang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Yuancheng Wang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| |
Collapse
|
52
|
Fusco R, Granata V, Setola SV, Trovato P, Galdiero R, Mattace Raso M, Maio F, Porto A, Pariante P, Cerciello V, Sorgente E, Pecori B, Castaldo M, Izzo F, Petrillo A. The application of radiomics in cancer imaging with a focus on lung cancer, renal cell carcinoma, gastrointestinal cancer, and head and neck cancer: A systematic review. Phys Med 2025; 130:104891. [PMID: 39787678 DOI: 10.1016/j.ejmp.2025.104891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
PURPOSE To study the application of radiomics in cancer imaging with a focus on lung cancer, renal cell carcinoma, gastrointestinal cancer, and head and neck cancer. METHODS Different electronic databases were considered. Articles published in the last five years were analyzed (January 2019 and December 2023). Papers were selected by two investigators with over 15 years of experience in Radiomics analysis in cancer imaging. The methodological quality of each radiomics study was performed using the Radiomic Quality Score (RQS) by two different readers in consensus and then by a third operator to solve disagreements between the two readers. RESULTS 19 articles are included in the review. Among the analyzed studies, only one study achieved an RQS of 18 reporting multivariable analyzes with also non-radiomics features and using the validation phase considering two datasets from two distinct institutes and open science and data domain. CONCLUSION This informative review has brought attention to the increasingly consolidated potential of Radiomics, although there are still several aspects to be evaluated before the transition to routine clinical practice. There are several challenges to address, including the need for standardization at all stages of the workflow and the potential for cross-site validation using heterogeneous real-world datasets. It will be necessary to establish and promote an imaging data acquisition protocol, conduct multicenter prospective quality control studies, add scanner differences and vendor-dependent characteristics; to collect images of individuals at additional time points, to report calibration statistics.
Collapse
Affiliation(s)
- Roberta Fusco
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Vincenza Granata
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy.
| | - Sergio Venanzio Setola
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Piero Trovato
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Galdiero
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Mauro Mattace Raso
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Francesca Maio
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Annamaria Porto
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Paolo Pariante
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Vincenzo Cerciello
- Division of Health Physics, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Eugenio Sorgente
- Division of Radiation protection and innovative technology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Biagio Pecori
- Division of Radiation protection and innovative technology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Mimma Castaldo
- Unit of "Progettazione e Manutenzione Edile ed impianti", Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale, IRCCS di Napoli, 80131 Naples, Italy
| |
Collapse
|
53
|
Sghedoni R, Origgi D, Cucurachi N, Minischetti GC, Alio D, Savini G, Botta F, Marzi S, Aiello M, Rancati T, Cusumano D, Politi LS, Didonna V, Massafra R, Petrillo A, Esposito A, Imparato S, Anemoni L, Bortolotto C, Preda L, Boldrini L. Stability of radiomic features in magnetic resonance imaging of the female pelvis: A multicentre phantom study. Phys Med 2025; 130:104895. [PMID: 39793255 DOI: 10.1016/j.ejmp.2025.104895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Affiliation(s)
- Roberto Sghedoni
- Medical Physics Unit, Azienda USL - IRCCS di Reggio Emilia, Viale Risorgimento 80, Reggio Emilia, Italy.
| | - Daniela Origgi
- Medical Physics Unit, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, Milano, Italy
| | - Noemi Cucurachi
- Medical Physics Unit, Azienda USL - IRCCS di Reggio Emilia, Viale Risorgimento 80, Reggio Emilia, Italy
| | - Giuseppe Castiglioni Minischetti
- Medical Physics Unit, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, Milano, Italy; School of Medical Physics, University of Milan, Via Celoria 16, 20133 Milan, Italy
| | - Davide Alio
- Medical Physics Unit, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, Milano, Italy; School of Medical Physics, University of Milan, Via Celoria 16, 20133 Milan, Italy
| | - Giovanni Savini
- Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy; Neuroradiology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Francesca Botta
- Medical Physics Unit, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, Milano, Italy
| | - Simona Marzi
- Medical Physics Laboratory, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Roma, Italy
| | - Marco Aiello
- IRCCS SYNLAB SDN, Via Francesco Crispi, 8, 80121 Napoli, Italy
| | - Tiziana Rancati
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milano, Italy
| | - Davide Cusumano
- UO Fisica Medica e Radioprotezione, Mater Olbia Hospital, SS 125 Orientale Sarda, 07026 Olbia, Italy
| | - Letterio Salvatore Politi
- Department of Biomedical Sciences, Humanitas University, Via R. Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy; Neuroradiology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Vittorio Didonna
- I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, Bari 70124, Italy
| | - Raffaella Massafra
- I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, Bari 70124, Italy
| | - Antonella Petrillo
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Via M. Semmola, 52, 80131 Napoli, Italy
| | - Antonio Esposito
- Experimetal Imaging Center, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milano, Italy; Vita-Salute San Raffaele University, School of Medicine, Via Olgettina, 58, 20132 Milano, Italy
| | - Sara Imparato
- Unità di Diagnostica per Immagini, CNAO, Via Erminio Borloni, 1, 27100 Pavia, Italy
| | - Luca Anemoni
- Unità di Diagnostica per Immagini, CNAO, Via Erminio Borloni, 1, 27100 Pavia, Italy
| | - Chandra Bortolotto
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy; Radiology Institute, Fondazione IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy
| | - Lorenzo Preda
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy; Radiology Institute, Fondazione IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy
| | - Luca Boldrini
- Dipartimento di Diagnostica per Immagini e Radioterapia Oncologica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Largo Agostino Gemelli 8, 00168 Roma, Italy
| |
Collapse
|
54
|
Shen Y, Chen L, Liu J, Chen H, Wang C, Ding H, Zhang Q. PADS-Net: GAN-based radiomics using multi-task network of denoising and segmentation for ultrasonic diagnosis of Parkinson disease. Comput Med Imaging Graph 2025; 120:102490. [PMID: 39808869 DOI: 10.1016/j.compmedimag.2024.102490] [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: 09/29/2024] [Revised: 12/05/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Parkinson disease (PD) is a prevalent neurodegenerative disorder, and its accurate diagnosis is crucial for timely intervention. We propose the PArkinson disease Denoising and Segmentation Network (PADS-Net), to simultaneously denoise and segment transcranial ultrasound images of midbrain for accurate PD diagnosis. The PADS-Net is built upon generative adversarial networks and incorporates a multi-task deep learning framework aimed at optimizing the tasks of denoising and segmentation for ultrasound images. A composite loss function including the mean absolute error, the mean squared error and the Dice loss, is adopted in the PADS-Net to effectively capture image details. The PADS-Net also integrates radiomics techniques for PD diagnosis by exploiting high-throughput features from ultrasound images. A four-branch ensemble diagnostic model is designed by utilizing two "wings" of the butterfly-shaped midbrain regions on both ipsilateral and contralateral images to enhance the accuracy of PD diagnosis. Experimental results demonstrate that the PADS-Net not only reduced speckle noise, achieving the edge-to-noise ratio of 16.90, but also attained a Dice coefficient of 0.91 for midbrain segmentation. The PADS-Net finally achieved an area under the receiver operating characteristic curve as high as 0.87 for diagnosis of PD. Our PADS-Net excels in transcranial ultrasound image denoising and segmentation and offers a potential clinical solution to accurate PD assessment.
Collapse
Affiliation(s)
- Yiwen Shen
- The SMART (Smart Medicine and AI-based Radiology Technology) Lab, Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China; Key Laboratory of Specialty Fiber Optics and Optical Access Networks, School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Li Chen
- Department of Ultrasound, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jieyi Liu
- The SMART (Smart Medicine and AI-based Radiology Technology) Lab, Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China; Key Laboratory of Specialty Fiber Optics and Optical Access Networks, School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Haobo Chen
- The SMART (Smart Medicine and AI-based Radiology Technology) Lab, Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China; Key Laboratory of Specialty Fiber Optics and Optical Access Networks, School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Changyan Wang
- The SMART (Smart Medicine and AI-based Radiology Technology) Lab, Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China; Key Laboratory of Specialty Fiber Optics and Optical Access Networks, School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Hong Ding
- Department of Ultrasound, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| | - Qi Zhang
- The SMART (Smart Medicine and AI-based Radiology Technology) Lab, Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China; Key Laboratory of Specialty Fiber Optics and Optical Access Networks, School of Communication and Information Engineering, Shanghai University, Shanghai, China.
| |
Collapse
|
55
|
Liu X, Lin F, Li D, Lei N. The accuracy of radiomics in diagnosing tumor deposits and perineural invasion in rectal cancer: a systematic review and meta-analysis. Front Oncol 2025; 14:1425665. [PMID: 39845326 PMCID: PMC11750663 DOI: 10.3389/fonc.2024.1425665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Background Radiomics has emerged as a promising approach for diagnosing, treating, and evaluating the prognosis of various diseases in recent years. Some investigators have utilized radiomics to create preoperative diagnostic models for tumor deposits (TDs) and perineural invasion (PNI) in rectal cancer (RC). However, there is currently a lack of comprehensive, evidence-based support for the diagnostic performance of these models. Thus, the accuracy of radiomic models was assessed in diagnosing preoperative RC TDs and PNI in this study. Methods PubMed, EMBASE, Web of Science, and Cochrane Library were searched for relevant articles from their establishment up to December 11, 2023. The radiomics quality score (RQS) was used to evaluate the risk of bias in the methodological quality and research level of the included studies. Results This meta-analysis included 15 eligible studies, most of which employed logistic regression models (LRMs). For diagnosing TDs, the c-index, sensitivity, and specificity of models based on radiomic features (RFs) alone were 0.85 (95% CI: 0.79 - 0.90), 0.85 (95% CI: 0.75 - 0.91), and 0.82 (95% CI: 0.70 - 0.89); in the validation set, the c-index, sensitivity, and specificity of models based on both RFs and interpretable CFs were 0.87 (95% CI: 0.83 - 0.91), 0.91 (95% CI: 0.72 - 0.99), and 0.65 (95% CI: 0.53 - 0.76), respectively. For diagnosing PNI, the c-index, sensitivity, and specificity of models based on RFs alone were 0.80 (95% CI: 0.74 - 0.86), 0.64 (95% CI: 0.44 - 0.80), and 0.79 (95% CI: 0.68 - 0.87) in the validation set; in the validation set, the c-index, sensitivity, and specificity of models based on both RFs and interpretable CFs were 0.83 (95% CI: 0.77 - 0.89), 0.60 (95% CI: 0.48 - 0.71), and 0.90 (95% CI: 0.84 - 0.94), respectively. Conclusions Diagnostic models based on both RFs and CFs have proven effective in preoperatively diagnosing TDs and PNI in RC. This non-invasive method shows promise as a new approach. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=498660, identifier CRD42024498660.
Collapse
Affiliation(s)
| | | | | | - Nan Lei
- Radiology Department, The People’s Hospital of Lezhi,
Ziyang, Sichuan, China
| |
Collapse
|
56
|
Prinzi F, Militello C, Zarcaro C, Bartolotta TV, Gaglio S, Vitabile S. Rad4XCNN: A new agnostic method for post-hoc global explanation of CNN-derived features by means of Radiomics. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 260:108576. [PMID: 39798282 DOI: 10.1016/j.cmpb.2024.108576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/11/2024] [Accepted: 12/25/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND AND OBJECTIVE In recent years, machine learning-based clinical decision support systems (CDSS) have played a key role in the analysis of several medical conditions. Despite their promising capabilities, the lack of transparency in AI models poses significant challenges, particularly in medical contexts where reliability is a mandatory aspect. However, it appears that explainability is inversely proportional to accuracy. For this reason, achieving transparency without compromising predictive accuracy remains a key challenge. METHODS This paper presents a novel method, namely Rad4XCNN, to enhance the predictive power of CNN-derived features with the inherent interpretability of radiomic features. Rad4XCNN diverges from conventional methods based on saliency maps, by associating intelligible meaning to CNN-derived features by means of Radiomics, offering new perspectives on explanation methods beyond visualization maps. RESULTS Using a breast cancer classification task as a case study, we evaluated Rad4XCNN on ultrasound imaging datasets, including an online dataset and two in-house datasets for internal and external validation. Some key results are: (i) CNN-derived features guarantee more robust accuracy when compared against ViT-derived and radiomic features; (ii) conventional visualization map methods for explanation present several pitfalls; (iii) Rad4XCNN does not sacrifice model accuracy for their explainability; (iv) Rad4XCNN provides a global explanation enabling the physician to extract global insights and findings. CONCLUSIONS Our method can mitigate some concerns related to the explainability-accuracy trade-off. This study highlighted the importance of proposing new methods for model explanation without affecting their accuracy.
Collapse
Affiliation(s)
- Francesco Prinzi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, 90127, Italy.
| | - Carmelo Militello
- Institute for High-Performance Computing and Networking (ICAR-CNR), Italian National Research Council, Palermo, 90146, Italy.
| | - Calogero Zarcaro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, 90127, Italy.
| | - Tommaso Vincenzo Bartolotta
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, 90127, Italy.
| | - Salvatore Gaglio
- Institute for High-Performance Computing and Networking (ICAR-CNR), Italian National Research Council, Palermo, 90146, Italy; Department of Engineering, University of Palermo, Palermo, 90128, Italy.
| | - Salvatore Vitabile
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, 90127, Italy.
| |
Collapse
|
57
|
Abbas S, Shafik R, Soomro N, Heer R, Adhikari K. AI predicting recurrence in non-muscle-invasive bladder cancer: systematic review with study strengths and weaknesses. Front Oncol 2025; 14:1509362. [PMID: 39839785 PMCID: PMC11746116 DOI: 10.3389/fonc.2024.1509362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
Background Non-muscle-invasive Bladder Cancer (NMIBC) is notorious for its high recurrence rate of 70-80%, imposing a significant human burden and making it one of the costliest cancers to manage. Current prediction tools for NMIBC recurrence rely on scoring systems that often overestimate risk and lack accuracy. Machine learning (ML) and artificial intelligence (AI) are transforming oncological urology by leveraging molecular and clinical data to enhance predictive precision. Methods This comprehensive review critically examines ML-based frameworks for predicting NMIBC recurrence. A systematic literature search was conducted, focusing on the statistical robustness and algorithmic efficacy of studies. These were categorised by data modalities (e.g., radiomics, clinical, histopathological, genomic) and types of ML models, such as neural networks, deep learning, and random forests. Each study was analysed for strengths, weaknesses, performance metrics, and limitations, with emphasis on generalisability, interpretability, and cost-effectiveness. Results ML algorithms demonstrate significant potential, with neural networks achieving accuracies of 65-97.5%, particularly with multi-modal datasets, and support vector machines averaging around 75%. Models combining multiple data types consistently outperformed single-modality approaches. However, challenges include limited generalisability due to small datasets and the "black-box" nature of advanced models. Efforts to enhance explainability, such as SHapley Additive ExPlanations (SHAP), show promise but require refinement for clinical use. Conclusion This review illuminates the nuances, complexities and contexts that influence the real-world advancement and adoption of these AI-driven techniques in precision oncology. It equips researchers with a deeper understanding of the intricacies of the ML algorithms employed. Actionable insights are provided for refining algorithms, optimising multimodal data utilisation, and bridging the gap between predictive accuracy and clinical utility. This rigorous analysis serves as a roadmap to advance real-world AI applications in oncological care, highlighting the collaborative efforts and robust datasets necessary to translate these advancements into tangible benefits for patient management.
Collapse
Affiliation(s)
- Saram Abbas
- School of Engineering, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Rishad Shafik
- School of Engineering, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Naeem Soomro
- Department of Urology, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Rakesh Heer
- Division of Surgery, Imperial College London, London, United Kingdom
- Centre for Cancer, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kabita Adhikari
- School of Engineering, Newcastle University, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
58
|
Li J, Jiang N, Zhang J, Sun W, Wang Z, Sun L, Wang X. Computed tomography-based absolute delta radiomics nomogram for predicting perineural invasion in hypopharyngeal squamous cell carcinoma. Eur J Radiol 2025; 183:111912. [PMID: 39809043 DOI: 10.1016/j.ejrad.2024.111912] [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: 09/29/2024] [Revised: 12/04/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
OBJECTIVE To assess the efficacy of computed tomography (CT)-based radiomics nomogram in predicting perineural invasion (PNI) in patients with hypopharyngeal squamous cell carcinoma (HPSCC). MATERIALS AND METHODS Overall, 146 patients were retrospectively recruited and divided into training and test cohorts at a 7:3 ratio. Radiomics features were extracted and delta and absolute delta radiomics features were calculated. Feature selection was performed using maximum relevance minimum redundancy and least absolute shrinkage and selection operator methods. Preliminary models were built using logistic regression, and the optimal one was selected as the radiomics signature. A nomogram was constructed by combining independent clinical factors and the radiomics signature. Its performance was evaluated using the area under the curve (AUC) values of receiver operating characteristic curves, decision curve analysis (DCA), and calibration curves. RESULTS The radiomics signature comprised 14 absolute delta radiomics features. The nomogram, incorporating tumor thickness and radiomics signature, outperformed the other models (AUC = 0.79 and 0.78, training and test cohorts, respectively). The Delong test demonstrated that the nomogram's predictive performance was significantly higher than that of the clinical model (p < 0.05) in both cohorts. Calibration curves indicated good calibration, and the Hosmer-Lemeshow test confirmed a good fit (p = 0.969 and 0.429, training and test cohorts, respectively). DCA highlighted the nomogram's considerable clinical usefulness. CONCLUSION The CT-based absolute delta radiomics nomogram can noninvasively and preoperatively predict PNI status in patients with HPSCC, providing a valuable tool for clinical decision making and individualized treatment plans.
Collapse
Affiliation(s)
- Jinyan Li
- Department of Radiology, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Nan Jiang
- Department of Pathology, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Juntao Zhang
- GE Healthcare PDX GMS Medical Affairs, Jinan, China
| | - Wenyue Sun
- Department of Radiology, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Zhan Wang
- Department of Radiology, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Lixin Sun
- Department of Radiology, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, China.
| |
Collapse
|
59
|
Hao L, Chen Y, Su X, Ma B. Application of Ultrasound Radiomics in Differentiating Benign from Malignant Breast Nodules in Women with Post-Silicone Breast Augmentation. Curr Oncol 2025; 32:29. [PMID: 39851945 PMCID: PMC11764215 DOI: 10.3390/curroncol32010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
PURPOSE To evaluate the diagnostic value of ultrasound radiomics in distinguishing between benign and malignant breast nodules in women who have undergone silicone breast augmentation. METHODS A retrospective study was conducted of 99 breast nodules detected by ultrasound in 93 women who had undergone silicone breast augmentation. The ultrasound data were collected between 1 January 2006 and 1 September 2023. The nodules were allocated into a training set (n = 69) and a validation set (n = 30). Regions of interest (ROIs) were manually delineated using 3D Slicer software, and radiomic features were extracted and selected using Python programming. Eight machine learning algorithms were applied to build predictive models, and their performance was assessed using sensitivity, specificity, area under the ROC curve (AUC), accuracy, Brier score, and log loss. Model performance was further evaluated using ROC curves and calibration curves, while clinical utility was assessed via decision curve analysis (DCA). RESULTS The random forest model exhibited superior performance in differentiating benign from malignant nodules in the validation set, achieving sensitivity of 0.765, specificity of 0.838, and an AUC of 0.787 (95% CI: 0.561-0.960). The model's accuracy, Brier score, and log loss were 0.796, 0.197, and 0.599, respectively. DCA suggested potential clinical utility of the model. CONCLUSION Ultrasound radiomics demonstrates promising diagnostic accuracy in differentiating benign from malignant breast nodules in women with silicone breast prostheses. This approach has the potential to serve as an additional diagnostic tool for patients following silicone breast augmentation.
Collapse
Affiliation(s)
| | | | | | - Buyun Ma
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China; (L.H.); (Y.C.); (X.S.)
| |
Collapse
|
60
|
Yu N, Ge X, Zuo L, Cao Y, Wang P, Liu W, Deng L, Zhang T, Wang W, Wang J, Lv J, Xiao Z, Feng Q, Zhou Z, Bi N, Zhang W, Wang X. Multi-Centered Pre-Treatment CT-Based Radiomics Features to Predict Locoregional Recurrence of Locally Advanced Esophageal Cancer After Definitive Chemoradiotherapy. Cancers (Basel) 2025; 17:126. [PMID: 39796752 PMCID: PMC11720276 DOI: 10.3390/cancers17010126] [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: 10/20/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 01/13/2025] Open
Abstract
Purpose: We constructed a prediction model to predict a 2-year locoregional recurrence based on the clinical features and radiomic features extracted from the machine learning method using computed tomography (CT) before definite chemoradiotherapy (dCRT) in locally advanced esophageal cancer. Patients and methods: A total of 264 patients (156 in Beijing, 87 in Tianjin, and 21 in Jiangsu) were included in this study. All those locally advanced esophageal cancer patients received definite radiotherapy and were randomly divided into five subgroups with a similar number and divided into training groups and validation groups by five cross-validations. The esophageal tumor and extratumoral esophagus were segmented to extract radiomic features from the gross tumor volume (GTV) drawn by radiation therapists before radiotherapy, and six clinical features associated with prognosis were added. T stage, N stage, M stage, total TNM stage, GTV, and GTVnd volume were included to construct a prediction model to predict the 2-year locoregional recurrence of patients after definitive radiotherapy. Results: A total of 264 patients were enrolled from August 2012 to April 2018, with a median age of 62 years and 81% were males. The 2-year locoregional recurrence rate was 52.6%, and the 2-year overall survival rate was 45.6%. About 66% of patients received concurrent chemotherapy. In total, we extracted 786 radiomic features from CT images and the Principal Component Analysis (PCA) method was used to screen out the maximum 30 features. Finally, the Support Vector Machine (SVM) method was used to construct the integrated prediction model combining radiomics and clinical features. In the five training groups for predicting locoregional recurrence, the mean value of C-index was 0.9841 (95%CI, 0.9809-0.9873), and in the five validation groups, the mean value was 0.744 (95%CI, 0.7437-0.7443). Conclusions: The integrated radiomics model could predict the 2-year locoregional recurrence after dCRT. The model showed promising results and could help guide treatment decisions by identifying high-risk patients and enabling strategies to prevent early recurrence.
Collapse
Affiliation(s)
- Nuo Yu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Xiaolin Ge
- Department of Radiation Oncology, Jiangsu Province Hospital/The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Lijing Zuo
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Ying Cao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Peipei Wang
- Department of Radiation Oncology, Jiangsu Province Hospital/The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wenyang Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Lei Deng
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Tao Zhang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Wenqing Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Jianyang Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Jima Lv
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Zefen Xiao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Qinfu Feng
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Zongmei Zhou
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| | - Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institution & Hospital, Tianjin 300060, China
| | - Xin Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (N.Y.)
| |
Collapse
|
61
|
Peng YT, Pang JS, Lin P, Chen JM, Wen R, Liu CW, Wen ZY, Wu YQ, Peng JB, Zhang L, Yang H, Wen DY, He Y. Preoperative prediction of lymph node metastasis in intrahepatic cholangiocarcinoma: an integrative approach combining ultrasound-based radiomics and inflammation-related markers. BMC Med Imaging 2025; 25:4. [PMID: 39748308 PMCID: PMC11697736 DOI: 10.1186/s12880-024-01542-8] [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: 10/05/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVES To develop ultrasound-based radiomics models and a clinical model associated with inflammatory markers for predicting intrahepatic cholangiocarcinoma (ICC) lymph node (LN) metastasis. Both are integrated for enhanced preoperative prediction. METHODS This study retrospectively enrolled 156 surgically diagnosed ICC patients. A region of interest (ROI) was manually identified on the ultrasound image of the tumor to extract radiomics features. In the training cohort, we performed a Wilcoxon test to screen for differentially expressed features, and then we used 12 machine learning algorithms to develop 107 models within the cross-validation framework and determine the optimal radiomics model through receiver operating characteristic (ROC) curve analysis. Multivariable logistic regression analysis was used to identify independent risk factors to construct a clinical model. The combined model was established by combining ultrasound-based radiomics and clinical parameters. The Delong test and decision curve analysis (DCA) were used to compare the diagnostic efficacy and clinical utility of different models. RESULTS A total of 1239 radiomics features were extracted from the ROIs of tumors. Among the 107 prediction models, the model (Stepglm + LASSO) utilizing 10 radiomics features ultimately yielded the highest average area under the receiver operating characteristic curve (AUC) of 0.872, with an AUC of 0.916 in the training cohort and 0.827 in the validation cohort. The combined model, which incorporates the optimal radiomics score, clinical N stage, and platelet-to-lymphocyte ratio (PLR), achieved an AUC of 0.882 in the validation cohort, significantly outperforming the clinical model with an AUC of 0.687 (P = 0.009). According to the DCA analysis, the combined model also showed better clinical benefits. CONCLUSIONS The combined model incorporating ultrasound-based radiomics features and the PLR marker offers an effective, noninvasive intelligence-assisted tool for preoperative LN metastasis prediction in ICC patients. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Yu-Ting Peng
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jin-Shu Pang
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Peng Lin
- Department of Medical Ultrasound, Fujian Medical University Union Hospital, No.29 Xinquan road, Fuzhou, Fujian Province, China
| | - Jia-Min Chen
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Rong Wen
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Chang-Wen Liu
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Yuan Wen
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yu-Quan Wu
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jin-Bo Peng
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Lu Zhang
- Department of Medical Pathology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Hong Yang
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Dong-Yue Wen
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China.
| | - Yun He
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong, Road, Nanning, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
62
|
Corti A, Lo Iacono F, Ronchetti F, Mushtaq S, Pontone G, Colombo GI, Corino VDA. Enhancing cardiovascular risk stratification: Radiomics of coronary plaque and perivascular adipose tissue - Current insights and future perspectives. Trends Cardiovasc Med 2025; 35:47-59. [PMID: 38960074 DOI: 10.1016/j.tcm.2024.06.003] [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: 05/10/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/05/2024]
Abstract
Radiomics, the quantitative extraction and mining of features from radiological images, has recently emerged as a promising source of non-invasive image-based cardiovascular biomarkers, potentially revolutionizing diagnostics and risk assessment. This review explores its application within coronary plaques and pericoronary adipose tissue, particularly focusing on plaque characterization and cardiac events prediction. By shedding light on the current state-of-the-art, achievements, and prospective avenues, this review contributes to a deeper understanding of the evolving landscape of radiomics in the context of coronary arteries. Finally, open challenges and existing gaps are emphasized to underscore the need for future efforts aimed at ensuring the robustness and reliability of radiomics studies, facilitating their clinical translation.
Collapse
Affiliation(s)
- Anna Corti
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan 20133, Italy.
| | - Francesca Lo Iacono
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan 20133, Italy
| | - Francesca Ronchetti
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Saima Mushtaq
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Gianluca Pontone
- Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino IRCCS, Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Gualtiero I Colombo
- Unit of Immunology and Functional Genomics, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Valentina D A Corino
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan 20133, Italy; Department of Perioperative Cardiology and Cardiovascular Imaging, Centro Cardiologico Monzino IRCCS, Milan, Italy
| |
Collapse
|
63
|
Russo L, Bottazzi S, Kocak B, Zormpas-Petridis K, Gui B, Stanzione A, Imbriaco M, Sala E, Cuocolo R, Ponsiglione A. Evaluating the quality of radiomics-based studies for endometrial cancer using RQS and METRICS tools. Eur Radiol 2025; 35:202-214. [PMID: 39014086 PMCID: PMC11632020 DOI: 10.1007/s00330-024-10947-6] [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: 04/08/2024] [Revised: 05/15/2024] [Accepted: 06/19/2024] [Indexed: 07/18/2024]
Abstract
OBJECTIVE To assess the methodological quality of radiomics-based models in endometrial cancer using the radiomics quality score (RQS) and METhodological radiomICs score (METRICS). METHODS We systematically reviewed studies published by October 30th, 2023. Inclusion criteria were original radiomics studies on endometrial cancer using CT, MRI, PET, or ultrasound. Articles underwent a quality assessment by novice and expert radiologists using RQS and METRICS. The inter-rater reliability for RQS and METRICS among radiologists with varying expertise was determined. Subgroup analyses were performed to assess whether scores varied according to study topic, imaging technique, publication year, and journal quartile. RESULTS Sixty-eight studies were analysed, with a median RQS of 11 (IQR, 9-14) and METRICS score of 67.6% (IQR, 58.8-76.0); two different articles reached maximum RQS of 19 and METRICS of 90.7%, respectively. Most studies utilised MRI (82.3%) and machine learning methods (88.2%). Characterisation and recurrence risk stratification were the most explored outcomes, featured in 35.3% and 19.1% of articles, respectively. High inter-rater reliability was observed for both RQS (ICC: 0.897; 95% CI: 0.821, 0.946) and METRICS (ICC: 0.959; 95% CI: 0.928, 0.979). Methodological limitations such as lack of external validation suggest areas for improvement. At subgroup analyses, no statistically significant difference was noted. CONCLUSIONS Whilst using RQS, the quality of endometrial cancer radiomics research was apparently unsatisfactory, METRICS depicts a good overall quality. Our study highlights the need for strict compliance with quality metrics. Adhering to these quality measures can increase the consistency of radiomics towards clinical application in the pre-operative management of endometrial cancer. CLINICAL RELEVANCE STATEMENT Both the RQS and METRICS can function as instrumental tools for identifying different methodological deficiencies in endometrial cancer radiomics research. However, METRICS also reflected a focus on the practical applicability and clarity of documentation. KEY POINTS The topic of radiomics currently lacks standardisation, limiting clinical implementation. METRICS scores were generally higher than the RQS, reflecting differences in the development process and methodological content. A positive trend in METRICS score may suggest growing attention to methodological aspects in radiomics research.
Collapse
Affiliation(s)
- Luca Russo
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Silvia Bottazzi
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Burak Kocak
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Konstantinos Zormpas-Petridis
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Benedetta Gui
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Massimo Imbriaco
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Evis Sala
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Renato Cuocolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy.
| | - Andrea Ponsiglione
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
64
|
Zhu ZN, Feng QX, Li Q, Xu WY, Liu XS. Machine learning-based CT radiomics approach for predicting occult peritoneal metastasis in advanced gastric cancer preoperatively. Clin Radiol 2025; 80:106727. [PMID: 39571365 DOI: 10.1016/j.crad.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/29/2024] [Accepted: 10/14/2024] [Indexed: 01/18/2025]
Abstract
AIM To develop a machine learning-based CT radiomics model to preoperatively diagnose occult peritoneal metastasis (OPM) in advanced gastric cancer (AGC) patients. MATERIALS AND METHODS A total of 177 AGC patients were retrospectively analyzed. Four regions of interest (ROIs) along the largest area of tumor (core ROI) and corresponding tumor mesenteric fat space (peri ROI) were manually delineated on the arterial (A-core and A-peri) and venous phase (V-core and V-peri) of CT images. A total of 1316 radiomics features were extracted from each ROI. Then, ten machine learning classification algorithms were used to develop the model. An integrated radiomics nomogram was established to predict OPM individually. RESULTS For the radiomics of tumor mesenteric fat space, the AUCs of A-peri in training and test sets were 0.881 and 0.800, respectively. And the AUCs of V-peri were 0.838 and 0.815, respectively. In terms of primary tumor' s radiomics signature, the AUCs of A-core in training and test sets were 0.862 and 0.691, respectively. The AUCs of V-core were 0.831 and 0.620. Integrated radiomics model showed the highest AUC value when it compared to each single radiomics score in the training (0.943 vs 0.831-0.881) and test set (0.835 vs 0.620-0.815). Radiomics nomogram demonstrated good diagnostic accuracy with a C-index of 0.948. CONCLUSION Both the radiomics of tumor mesenteric fat space and primary tumor were associated with OPM. A CT radiomics nomogram had a relatively good predictive performance for detecting OPM in patients with AGC.
Collapse
Affiliation(s)
- Z-N Zhu
- Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, China.
| | - Q-X Feng
- Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, China.
| | - Q Li
- Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, China.
| | - W-Y Xu
- Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, China.
| | - X-S Liu
- Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, China.
| |
Collapse
|
65
|
Gallifant J, Afshar M, Ameen S, Aphinyanaphongs Y, Chen S, Cacciamani G, Demner-Fushman D, Dligach D, Daneshjou R, Fernandes C, Hansen LH, Landman A, Lehmann L, McCoy LG, Miller T, Moreno A, Munch N, Restrepo D, Savova G, Umeton R, Gichoya JW, Collins GS, Moons KGM, Celi LA, Bitterman DS. The TRIPOD-LLM reporting guideline for studies using large language models. Nat Med 2025; 31:60-69. [PMID: 39779929 DOI: 10.1038/s41591-024-03425-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025]
Abstract
Large language models (LLMs) are rapidly being adopted in healthcare, necessitating standardized reporting guidelines. We present transparent reporting of a multivariable model for individual prognosis or diagnosis (TRIPOD)-LLM, an extension of the TRIPOD + artificial intelligence statement, addressing the unique challenges of LLMs in biomedical applications. TRIPOD-LLM provides a comprehensive checklist of 19 main items and 50 subitems, covering key aspects from title to discussion. The guidelines introduce a modular format accommodating various LLM research designs and tasks, with 14 main items and 32 subitems applicable across all categories. Developed through an expedited Delphi process and expert consensus, TRIPOD-LLM emphasizes transparency, human oversight and task-specific performance reporting. We also introduce an interactive website ( https://tripod-llm.vercel.app/ ) facilitating easy guideline completion and PDF generation for submission. As a living document, TRIPOD-LLM will evolve with the field, aiming to enhance the quality, reproducibility and clinical applicability of LLM research in healthcare through comprehensive reporting.
Collapse
Affiliation(s)
- Jack Gallifant
- Laboratory for Computational Physiology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Critical Care, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA
| | - Majid Afshar
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Saleem Ameen
- Laboratory for Computational Physiology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Yindalon Aphinyanaphongs
- Department of Population Health, NYU Grossman School of Medicine and Langone Health, New York, NY, USA
| | - Shan Chen
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, MA, USA
| | - Giovanni Cacciamani
- USC Institute of Urology and Catherine and Joseph Aresty Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Artificial Intelligence Center, USC Institute of Urology, University of Southern California, Los Angeles, CA, USA
| | | | - Dmitriy Dligach
- Department of Computer Science, Loyola University, Chicago, IL, USA
| | - Roxana Daneshjou
- Department of Dermatology, Stanford School of Medicine, Redwood City, CA, USA
- Department of Biomedical Data Science, Stanford School of Medicine, Redwood City, CA, USA
| | - Chrystinne Fernandes
- Laboratory for Computational Physiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lasse Hyldig Hansen
- Laboratory for Computational Physiology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Cognitive Science, Aarhus University, Jens Chr. Skou 2, Aarhus, Denmark
| | | | | | - Liam G McCoy
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Timothy Miller
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amy Moreno
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nikolaj Munch
- Laboratory for Computational Physiology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Cognitive Science, Aarhus University, Jens Chr. Skou 2, Aarhus, Denmark
| | - David Restrepo
- Laboratory for Computational Physiology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Departamento de Telematica, Universidad del Cauca, Popayan, Colombia
| | - Guergana Savova
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Judy Wawira Gichoya
- Department of Radiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gary S Collins
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- UK EQUATOR Centre, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Karel G M Moons
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
- Health Innovation Netherlands (HINL), Utrecht, the Netherlands
| | - Leo A Celi
- Laboratory for Computational Physiology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Danielle S Bitterman
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA.
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
66
|
Yang J, Dong X, Jin S, Wang S, Wang Y, Zhang L, Wei Y, Wu Y, Wang L, Zhu L, Feng Y, Gan M, Hu H, Ji W. Radiomics Model of Dynamic Contrast-Enhanced MRI for Evaluating Vessels Encapsulating Tumor Clusters and Microvascular Invasion in Hepatocellular Carcinoma. Acad Radiol 2025; 32:146-156. [PMID: 39025700 DOI: 10.1016/j.acra.2024.07.007] [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: 04/16/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024]
Abstract
RATIONALE AND OBJECTIVES To develop and validate a clinical-radiomics model of dynamic contrast-enhanced MRI (DCE-MRI) for the preoperative discrimination of Vessels encapsulating tumor clusters (VETC)- microvascular invasion (MVI) and prognosis of hepatocellular carcinoma (HCC). MATERIALS AND METHODS 219 HCC patients from Institution 1 were split into internal training and validation groups, with 101 patients from Institution 2 assigned to external validation. Histologically confirmed VETC-MVI pattern categorizing HCC into VM-HCC+ (VETC+/MVI+, VETC-/MVI+, VETC+/MVI-) and VM-HCC- (VETC-/MVI-). The regions of intratumor and peritumor were segmented manually in the arterial, portal-venous and delayed phase (AP, PP, and DP, respectively) of DCE-MRI. Six radiomics models (intratumor and peritumor in AP, PP, and DP of DCE-MRI) and one clinical model were developed for assessing VM-HCC. Establishing intra-tumoral and peri-tumoral models through combining intratumor and peritumor features. The best-performing radiomics model and the clinical model were then integrated to create a Combined model. RESULTS In institution 1, pathological VM-HCC+ were confirmed in 88 patients (training set: 61, validation set: 27). In internal testing, the Combined model had an AUC of 0.85 (95% CI: 0.76-0.93), which reached an AUC of 0.75 (95% CI: 0.66-0.85) in external validation. The model's predictions were associated with early recurrence and progression-free survival in HCC patients. CONCLUSIONS The clinical-radiomics model offers a non-invasive approach to discern VM-HCC and predict HCC patients' prognosis preoperatively, which could offer clinicians valuable insights during the decision-making phase.
Collapse
Affiliation(s)
- Jiawen Yang
- Department of Radiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China; Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China.
| | - Xue Dong
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Shengze Jin
- Department of Radiology, Taizhou Hospital of Zhejiang Province, Shaoxing University, Taizhou 318000 Zhejiang, China.
| | - Sheng Wang
- Department of Radiology, Taizhou First People's Hospital, Wenzhou Medical College, Taizhou 318020 Zhejiang, China.
| | - Yanna Wang
- Department of Radiology, Taizhou Central Hospital,Wenzhou Medical University, Taizhou 318000 Zhejiang,China.
| | - Limin Zhang
- Department of Radiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Yuguo Wei
- Precision Health Institution, GE Healthcare, 310000 Xihu District, Hangzhou, China.
| | - Yitian Wu
- Department of Radiology, Taizhou Hospital of Zhejiang Province, Shaoxing University, Taizhou 318000 Zhejiang, China.
| | - Lingxia Wang
- Department of Radiology, Taizhou Hospital, Zhejiang University, Taizhou 318000 Zhejiang, China.
| | - Lingwei Zhu
- Department of Radiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China.
| | - Yuyi Feng
- Department of Radiology, Taizhou Hospital of Zhejiang Province, Shaoxing University, Taizhou 318000 Zhejiang, China.
| | - Meifu Gan
- Department of Pathology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China.
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, People's Republic of China.
| | - Wenbin Ji
- Department of Radiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China; Key Laboratory of evidence-based Radiology of Taizhou, Linhai 317000, Zhejiang, China.
| |
Collapse
|
67
|
Lopez-Ramirez F, Soleimani S, Azadi JR, Sheth S, Kawamoto S, Javed AA, Tixier F, Hruban RH, Fishman EK, Chu LC. Radiomics machine learning algorithm facilitates detection of small pancreatic neuroendocrine tumors on CT. Diagn Interv Imaging 2025; 106:28-40. [PMID: 39278763 DOI: 10.1016/j.diii.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024]
Abstract
PURPOSE The purpose of this study was to develop a radiomics-based algorithm to identify small pancreatic neuroendocrine tumors (PanNETs) on CT and evaluate its robustness across manual and automated segmentations, exploring the feasibility of automated screening. MATERIALS AND METHODS Patients with pathologically confirmed T1 stage PanNETs and healthy controls undergoing dual-phase CT imaging were retrospectively identified. Manual segmentation of pancreas and tumors was performed, then automated pancreatic segmentations were generated using a pretrained neural network. A total of 1223 radiomics features were independently extracted from both segmentation volumes, in the arterial and venous phases separately. Ten final features were selected to train classifiers to identify PanNETs and controls. The cohort was divided into training and testing sets, and performance of classifiers was assessed using area under the receiver operator characteristic curve (AUC), specificity and sensitivity, and compared against two radiologists blinded to the diagnoses. RESULTS A total of 135 patients with 142 PanNETs, and 135 healthy controls were included. There were 168 women and 102 men, with a mean age of 55.4 ± 11.6 (standard deviation) years (range: 20-85 years). Median PanNET size was 1.3 cm (Q1, 1.0; Q3, 1.5; range: 0.5-1.9). The arterial phase LightGBM model achieved the best performance in the test set, with 90 % sensitivity (95 % confidence interval [CI]: 80-98), 76 % specificity (95 % CI: 62-88) and an AUC of 0.87 (95 % CI: 0.79-0.94). Using features from the automated segmentations, this model achieved an AUC of 0.86 (95 % CI: 0.79-0.93). In comparison, the two radiologists achieved a mean 50 % sensitivity and 100 % specificity using arterial phase CT images. CONCLUSION Radiomics features identify small PanNETs, with stable performance when extracted using automated segmentations. These models demonstrate high sensitivity, complementing the high specificity of radiologists, and could serve as opportunistic screeners.
Collapse
Affiliation(s)
- Felipe Lopez-Ramirez
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sahar Soleimani
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Javad R Azadi
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sheila Sheth
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Satomi Kawamoto
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ammar A Javed
- Department of Surgery, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Florent Tixier
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ralph H Hruban
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elliot K Fishman
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Linda C Chu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
68
|
Chen B, Ngremmadji MA, Morel O. Editorial for "A Hybrid Model for Fetal Growth Restriction Assessment by Automatic Placental Radiomics on T2-Weighted MRI and Multifeature Fusion". J Magn Reson Imaging 2025; 61:505-506. [PMID: 38708929 DOI: 10.1002/jmri.29418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 05/07/2024] Open
Affiliation(s)
- Bailiang Chen
- CIC-IT 1433, CHRU Nancy, Vandœuvre-lès-Nancy, France
- INSERM U1254, IADI, Université de Lorraine, Nancy, France
| | | | - Olivier Morel
- INSERM U1254, IADI, Université de Lorraine, Nancy, France
- Obstetrics and Fetal Medicine Unit, CHRU of Nancy, Nancy, France
| |
Collapse
|
69
|
Shakya MR, Zheng C, Fu F, Sun S, Lu J. Development and validation of the nomogram model derived non-contrast CT score to predict hematoma expansion in patients with spontaneous intracerebral hemorrhage. Clin Radiol 2025; 80:106694. [PMID: 39520934 DOI: 10.1016/j.crad.2024.08.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 11/16/2024]
Abstract
AIMS Develop and validate new non-contrast computed tomography (NCCT) score to predict hematoma expansion (HE) in spontaneous intracerebral hemorrhage (SICH) patients based on hematoma's shape irregularity and density heterogeneity. MATERIALS AND METHODS Retrospective study was conducted among 136 patients for development and 90 patients for validation at two separate hospitals. SICH patients with NCCT scanned within 6 hours of symptoms and follow-up NCCT scanned within 24 hours were enrolled. Black hole sign and blend sign were integrated as combined heterogeneity; likewise, satellite sign and island sign were integrated as combined irregularity. Binary logistic regression analysis screened the covariates associated with HE. Nomogram was generated using the predicted value of binary logistic regression model to derive NCCT score to predict HE. RESULTS A total of 65 patients had HE in developmental cohort, where history of hypertension [odds ratio (OR) 2.56; 95% CI 1.169-5.607; P=0.019], initial NCCT time ≤ 3 hours (OR 2.50; 95% CI 1.169-5.327; P=0.018), combined heterogeneity (OR 2.50; 95% CI 1.160-5.365; P=0.019), and combined irregularity (OR 2.63; 95% CI 1.164-5.942; P=0.020) were independently associated with HE. A score was derived and a single point was allocated to each independently associated variable. HE was observed in 35 patients in validation cohort, which showed a proportional increase in the probability of HE with an increase in score accumulated. CONCLUSION New four-point NCCT score to predict HE was developed and validated, which may be regarded as fair predictive score where advance facilities are rarely available.
Collapse
Affiliation(s)
- M R Shakya
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing, China
| | - C Zheng
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing, China
| | - F Fu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing, China; Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiaotong University, No.197 Ruijinerlu, Huangpu District, Shanghai, China
| | - S Sun
- Neuroradiology Department, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, No.119 Nansihuanxilu, Fengtai District, Beijing, China
| | - J Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing, China.
| |
Collapse
|
70
|
Jiang T, Wang H, Li J, Wang T, Zhan X, Wang J, Wang N, Nie P, Cui S, Zhao X, Hao D. Development and validation of a CT-based deep learning radiomics signature to predict lymph node metastasis in oropharyngeal squamous cell carcinoma: a multicentre study. Dentomaxillofac Radiol 2025; 54:77-87. [PMID: 39271161 DOI: 10.1093/dmfr/twae051] [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: 07/29/2024] [Revised: 08/30/2024] [Accepted: 09/07/2024] [Indexed: 09/15/2024] Open
Abstract
OBJECTIVES Lymph node metastasis (LNM) is a pivotal determinant that influences the treatment strategies and prognosis for oropharyngeal squamous cell carcinoma (OPSCC) patients. This study aims to establish and verify a deep learning (DL) radiomics model for the prediction of LNM in OPSCCs using contrast-enhanced computed tomography (CECT). METHODS A retrospective analysis included 279 OPSCC patients from 3 institutions. CECT images were used for handcrafted (HCR) and DL feature extraction. Dimensionality reduction for HCR features used recursive feature elimination (RFE) and least absolute shrinkage and selection operator (LASSO) algorithms, whereas DL feature dimensionality reduction used variance-threshold and RFE algorithms. Radiomics signatures were constructed using six machine learning classifiers. A combined model was then constructed using the screened DL, HCR, and clinical features. The area under the receiver operating characteristic curve (AUC) served to quantify the model's performance, and calibration curves were utilized to assess its calibration. RESULTS The combined model exhibited robust performance, achieving AUC values of 0.909 (95% CI, 0.861-0.957) in the training cohort, 0.884 (95% CI, 0.800-0.968) in the internal validation cohort, and 0.865 (95% CI, 0.791-0.939) in the external validation cohort. It outperformed both the clinical model and best-performing radiomics model. Moreover, calibration was deemed satisfactory. CONCLUSIONS The combined model based on CECT demonstrates the potential to predict LNM in OPSCCs preoperatively, offering a valuable tool for more precise and tailored treatment strategies. ADVANCES IN KNOWLEDGE This study presents a novel combined model integrating clinical factors with DL radiomics, significantly enhancing preoperative LNM prediction in OPSCC.
Collapse
Affiliation(s)
- Tianzi Jiang
- Department of Radiology, The Affiliated Hospital of Qingdao University, School of Medicine, Qingdao University, Qingdao, Shandong 266003, China
| | - Hexiang Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, School of Medicine, Qingdao University, Qingdao, Shandong 266003, China
| | - Jie Li
- Department of Radiology, The Affiliated Hospital of Qingdao University, School of Medicine, Qingdao University, Qingdao, Shandong 266003, China
| | - Tongyu Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, School of Medicine, Qingdao University, Qingdao, Shandong 266003, China
| | - Xiaohong Zhan
- Department of Pathology, The Affiliated Hospital of Qingdao University, School of Medicine, Qingdao University, Qingdao, Shandong 266003, China
| | - Jingqun Wang
- Department of Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Ning Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, School of Medicine, Shandong First Medical University, Jinan, Shandong 250000, China
| | - Pei Nie
- Department of Radiology, The Affiliated Hospital of Qingdao University, School of Medicine, Qingdao University, Qingdao, Shandong 266003, China
| | - Shiyu Cui
- Department of Radiology, The Affiliated Hospital of Qingdao University, School of Medicine, Qingdao University, Qingdao, Shandong 266003, China
| | - Xindi Zhao
- Department of Radiology, The Affiliated Hospital of Qingdao University, School of Medicine, Qingdao University, Qingdao, Shandong 266003, China
| | - Dapeng Hao
- Department of Radiology, The Affiliated Hospital of Qingdao University, School of Medicine, Qingdao University, Qingdao, Shandong 266003, China
| |
Collapse
|
71
|
Figiel S, Bates A, Braun DA, Eapen R, Eckstein M, Manley BJ, Milowsky MI, Mitchell TJ, Bryant RJ, Sfakianos JP, Lamb AD. Clinical Implications of Basic Research: Exploring the Transformative Potential of Spatial 'Omics in Uro-oncology. Eur Urol 2025; 87:8-14. [PMID: 39227262 DOI: 10.1016/j.eururo.2024.08.025] [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: 05/16/2024] [Revised: 07/17/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024]
Abstract
New spatial molecular technologies are poised to transform our understanding and treatment of urological cancers. By mapping the spatial molecular architecture of tumours, these platforms uncover the complex heterogeneity within and around individual malignancies, offering novel insights into disease development, progression, diagnosis, and treatment. They enable tracking of clonal phylogenetics in situ and immune-cell interactions in the tumour microenvironment. A whole transcriptome/genome/proteome-level spatial analysis is hypothesis generating, particularly in the areas of risk stratification and precision medicine. Current challenges include reagent costs, harmonisation of protocols, and computational demands. Nonetheless, the evolving landscape of the technology and evolving machine learning applications have the potential to overcome these barriers, pushing towards a future of personalised cancer therapy, leveraging detailed spatial cellular and molecular data. PATIENT SUMMARY: Tumours are complex and contain many different components. Although we have been able to observe some of these differences visually under the microscope, until recently, we have not been able to observe the genetic changes that underpin cancer development. Scientists are now able to explore molecular/genetic differences using approaches such as "spatial transcriptomics" and "spatial proteomics", which allow them to see genetic and cellular variation across a region of normal and cancerous tissue without destroying the tissue architecture. Currently, these technologies are limited by high associated costs, and a need for powerful and complex computational analysis workflows. Future advancements and results through these new technologies may assist patients and their doctors as they make decisions about treating their cancer.
Collapse
Affiliation(s)
- Sandy Figiel
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Anthony Bates
- Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - David A Braun
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Renu Eapen
- Department of Genitourinary Oncology & Division of Cancer Surgery, Peter MacCallum Cancer Centre, The University of Melbourne, Victoria, Australia
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg & Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Brandon J Manley
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Matthew I Milowsky
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Tom J Mitchell
- Early Detection Centre, University of Cambridge, Cambridge, UK
| | - Richard J Bryant
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - John P Sfakianos
- Department of Urology, Ichan School of Medicine at the Mount Sinai Hospital, New York, NY, USA
| | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
72
|
Viganò L, Zanuso V, Fiz F, Cerri L, Laino ME, Ammirabile A, Ragaini EM, Viganò S, Terracciano LM, Francone M, Ieva F, Di Tommaso L, Rimassa L. CT-based radiogenomics of intrahepatic cholangiocarcinoma. Dig Liver Dis 2025; 57:118-124. [PMID: 39003163 DOI: 10.1016/j.dld.2024.06.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is an aggressive disease with increasing incidence and its genetic alterations could be the target of systemic therapies. AIMS To elucidate if radiomics extracted from computed tomography (CT) may non-invasively predict ICC genetic alterations. METHODS All consecutive patients with a diagnosis of a mass-forming ICC (01/2016-06/2022) were considered. Inclusion criteria were availability of a high-quality contrast-enhanced CT and molecular profiling by NGS or FISH for FGFR2 fusion/rearrangement. The CT scan at diagnosis was considered. Genetic analyses were performed on surgical specimens (resectable patients) or biopsies (unresectable ones). The radiomic features were extracted using the LifeX software. Multivariate predictive models of the commonest genetic alterations were built. RESULTS In the 90 enrolled patients (58 NGS/32 FISH, median age 65 years), the most common genetic alterations were FGFR2 (20/90), IDH1 (10/58), and KRAS (9/58). At internal validation, the combined clinical-radiomic models achieved the best performance for the prediction of FGFR2 (AUC = 0.892) and IDH1 status (AUC = 0.819), outperforming the pure clinical and radiomic models. The radiomic model for predicting KRAS mutations achieved an AUC = 0.767 (vs. 0.660 of the clinical model) without further improvements with the addition of clinical features. CONCLUSIONS CT-based radiomics provides a reliable non-invasive prediction of ICC genetic status with a major impact on therapeutic strategies.
Collapse
Affiliation(s)
- Luca Viganò
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Hepatobiliary Unit, Department of Minimally Invasive General & Oncologic Surgery, Humanitas Gavazzeni University Hospital, Bergamo, Italy.
| | - Valentina Zanuso
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesco Fiz
- Nuclear Medicine Unit, Department of Diagnostic Imaging, Ente Ospedaliero "Ospedali Galliera", Genoa, Italy; Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital, Tübingen, Germany
| | - Luca Cerri
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | | | - Angela Ammirabile
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Department of Radiology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Elisa Maria Ragaini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Samuele Viganò
- MOX laboratory, Department of Mathematics, Politecnico di Milano, Milan, Italy
| | - Luigi Maria Terracciano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Pathology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Marco Francone
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Department of Radiology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesca Ieva
- MOX laboratory, Department of Mathematics, Politecnico di Milano, Milan, Italy; CHDS - Center for Health Data Science, Human Technopole, Milan, Italy
| | - Luca Di Tommaso
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Pathology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
73
|
Zhang H, Zhang H, Jiang M, Li J, Li J, Zhou H, Song X, Fan X. Radiomics in ophthalmology: a systematic review. Eur Radiol 2025; 35:542-557. [PMID: 39033472 DOI: 10.1007/s00330-024-10911-4] [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: 11/10/2023] [Revised: 04/03/2024] [Accepted: 05/12/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Radiomics holds great potential in medical image analysis for various ophthalmic diseases. In recent times, there have been numerous endeavors in this area of research. This systematic review aims to provide a comprehensive assessment of the strengths and limitations of radiomics in ophthalmology. METHOD Conforming to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines, we conducted a systematic review with a pre-registered protocol (PROSPERO: CRD42023446317). We explored the PubMed, Embase, and Cochrane databases for original studies on this topic and made a comprehensive descriptive integration. Furthermore, the included studies underwent quality assessment by the radiomics quality score (RQS). RESULTS A total of 41 articles from an initial search of 227 studies were finally selected for further analysis. These articles included research across five disease categories and covered seven imaging modalities. The radiomics models demonstrated robust performance, with area under the curve (AUC) values mostly falling within 0.7-1.0. The moderate RQS (mean score: 11.17/36) indicated that most studies were retrospectively, single-center analyses without external validation. CONCLUSIONS Radiomics holds promising utility in the field of ophthalmology, assisting diagnosis, early-stage screening, and prognostication of treatment response. Artificial intelligence algorithms significantly contribute to the construction of radiomics models in ophthalmology. This study highlights the strengths and challenges of radiomics in ophthalmology and suggests potential avenues for future improvement. CLINICAL RELEVANCE STATEMENT Radiomics represents a valuable approach for generating innovative imaging markers, enhancing efficiency in clinical diagnosis and treatment, and aiding decision-making in clinical contexts of many ophthalmic diseases, thereby improving overall patient prognosis. KEY POINTS Radiomics has attracted extensive attention in the field of ophthalmology. Articles included five disease categories over seven imaging modalities, consistently yielding AUCs mostly above 0.7. Current research has few prospective and multi-center studies, underlining the necessity for future high-quality studies.
Collapse
Affiliation(s)
- Haiyang Zhang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Huijie Zhang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Mengda Jiang
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaxin Li
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jipeng Li
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Huifang Zhou
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Xuefei Song
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| |
Collapse
|
74
|
Meng A, Zhuang Y, Huang Q, Tang L, Yang J, Gong P. Development and validation of a cross-modality tensor fusion model using multi-modality MRI radiomics features and clinical radiological characteristics for the prediction of microvascular invasion in hepatocellular carcinoma. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109364. [PMID: 39536525 DOI: 10.1016/j.ejso.2024.109364] [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: 08/13/2024] [Revised: 10/29/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES To develop and validate a cross-modality tensor fusion (CMTF) model using multi-modality MRI radiomics features and clinical radiological characteristics for the prediction of microvascular invasion (MVI) in hepatocellular carcinoma (HCC). MATERIALS AND METHODS This study included 174 HCC patients (47 MVI-positive and 127 MVI-negative) confirmed by postoperative pathology. The synthetic minority over-sampling technique was used to augment MVI-positive samples. The amplified dataset of 254 samples (127 MVI-positive and 127 MVI-negative) was randomly divided into training and test cohorts in a 7:3 ratio. Radiomics features were respectively extracted from arterial phase, delayed phase, diffusion-weighted imaging, and fat-suppressed T2-weighted imaging. The least absolute shrinkage and selection operator was used for feature selection. Univariate and multivariate logistic regression analyses were employed to identify clinical and radiological independent predictors. The selected multi-modality MRI radiomics features, clinical and radiological characteristics were used to construct the CMTF model, single modality (SM) model, early fusion (EF) model. RESULTS The CMTF model demonstrated superior performance in predicting MVI compared to the SM and EF models. When integrating four MRI modalities, the CMTF model achieved a high area under the curve (AUC) with 95 % confidence interval (95 % CI) of 0.894 (0.820-0.968). Additionally, incorporating clinical and radiological characteristics further enhanced the predictive performance of CMTF model, the AUC (95 % CI) value increased to 0.945 (0.892-0.998). CONCLUSION The CMTF model showed promising performance in preoperative MVI prediction, providing a more effective non-invasive detection tool for HCC patients.
Collapse
Affiliation(s)
- Ao Meng
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yinping Zhuang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Qian Huang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Li Tang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jing Yang
- Department of Interventional Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Ping Gong
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
75
|
Liu Z, Yao Y, Zhao M, Zhao Q, Xue J, Huang Y, Qin S. Radiomics Models Derived From Arterial-Phase-Enhanced CT Reliably Predict Both PD-L1 Expression and Immunotherapy Prognosis in Non-small Cell Lung Cancer: A Retrospective, Multicenter Cohort Study. Acad Radiol 2025; 32:493-505. [PMID: 39084935 DOI: 10.1016/j.acra.2024.07.028] [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: 06/03/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 08/02/2024]
Abstract
RATIONALE AND OBJECTIVES Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of non-small cell lung cancer (NSCLC) and programmed cell death-ligand 1 (PD-L1) is a companion biomarker. This study aims to use baseline arterial-phase enhanced CT (APECT) to construct efficient radiomic models for predicting PD-L1 expression and immunotherapy prognosis in NSCLC. MATERIALS AND METHODS We extracted radiomics features from the baseline APECT images of 204 patients enrolled in a published multicenter clinical trial that commenced on August 23, 2018, and concluded on November 15, 2019 (ClinicalTrials.gov: NCT03607539). Of these patients, 146 patients from selected centers were assigned to the training cohort. The least absolute shrinkage and selection operator (LASSO) method was used to reduce dimensionality of radiomics features and calculate tumor scores. Models were created using naive bayes, decision trees, XGBoost, and random forest algorithms according to tumor scores. These models were then validated in an independent validation cohort comprising 58 patients from the remaining centers. RESULTS The random forest algorithm outperformed the other methods. In the three-classification scenario, the random forest model achieving the area under the curve (AUC) values of 0.98 and 0.94 in the training and validation cohorts, respectively. In the two-classification scenario, the random forest model achieved AUCs of 0.99 (95%CI: 0.97-1.0, P < 0.0001) and 0.93 (95%CI: 0.83-0.98, P < 0.0001) in the training and validation cohorts, respectively. Furthermore, patients classified as PD-L1 high-expression by this model can predict treatment response (AUC=0.859, 95%CI: 0.7-0.96, P < 0.001) and improved survival (HR=0.2, 95%CI: 0.08-0.53, P = 0.001) only in validation sintilimab arm. CONCLUSION Radiomics models based on APECT represent a potential non-invasive approach to robustly predict PD-L1 expression and ICI treatment outcomes in patients with NSCLC, which could significantly improve precision cancer immunotherapy.
Collapse
Affiliation(s)
- Zhenhua Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215000, China; Department of Radiotherapy, Yancheng First Hospital Affiliated Hospital of Nanjing University Medical School, The First people's Hospital of Yancheng, 66 Renmin Road, Yancheng 224005, China; National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| | - Yimin Yao
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215000, China
| | - Miaomiao Zhao
- Department of Ultrasound, Yancheng First Hospital Affiliated Hospital of Nanjing University Medical School, The First people's Hospital of Yancheng, 66 Renmin Road, Yancheng 224005, China
| | - Qi Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215000, China
| | - Jiao Xue
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215000, China
| | - Yuhui Huang
- National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| | - Songbing Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215000, China.
| |
Collapse
|
76
|
Corallo C, Al-Adhami AS, Jamieson N, Valle J, Radhakrishna G, Moir J, Albazaz R. An update on pancreatic cancer imaging, staging, and use of the PACT-UK radiology template pre- and post-neoadjuvant treatment. Br J Radiol 2025; 98:13-26. [PMID: 39460945 DOI: 10.1093/bjr/tqae217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/01/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma continues to have a poor prognosis, although recent advances in neoadjuvant treatments (NATs) have provided some hope. Imaging assessment of suspected tumours can be challenging and requires a specific approach, with pancreas protocol CT being the primary imaging modality for staging with other modalities used as problem-solving tools to facilitate appropriate management. Imaging assessment post NAT can be particularly difficult due to a current lack of robust radiological criteria to predict response and differentiate treatment induced fibrosis/inflammation from residual tumour. This review aims to provide an update of pancreatic ductal adenocarcinoma with particular focus on three points: tumour staging pre- and post-NAT including vascular assessment, structured reporting with introduction of the PAncreatic Cancer reporting Template-UK (PACT-UK) radiology template, and the potential future role of artificial intelligence in the diagnosis and staging of pancreatic cancer.
Collapse
Affiliation(s)
- Carmelo Corallo
- Department of Radiology, St James's University Hospital, Leeds LS9 7TF, United Kingdom
| | - Abdullah S Al-Adhami
- Department of Radiology, Glasgow Royal Infirmary, Glasgow G31 2ER, United Kingdom
| | - Nigel Jamieson
- HPB Unit, Glasgow Royal Infirmary, Glasgow G31 2ER, United Kingdom
| | - Juan Valle
- Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, United Kingdom
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4 BX, United Kingdom
| | | | - John Moir
- HPB Unit, Freeman Hospital, Newcastle Upon Tyne NE7 7DN, United Kingdom
| | - Raneem Albazaz
- Department of Radiology, St James's University Hospital, Leeds LS9 7TF, United Kingdom
- University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
77
|
Wang X, Ye W, Gu Y, Gao Y, Wang H, Zhou Y, Pan D, Ge X, Liu W, Cai W. Predicting Secondary Vertebral Compression Fracture After Vertebral Augmentation via CT-Based Machine Learning Radiomics-Clinical Model. Acad Radiol 2025; 32:298-310. [PMID: 38991868 DOI: 10.1016/j.acra.2024.06.041] [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: 05/16/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024]
Abstract
RATIONALE AND OBJECTIVES Secondary vertebral compression fractures (SVCF) are very common in patients after vertebral augmentation (VA). The aim of this study was to establish a radiomic-based model to predict SVCF and specify appropriate treatment strategies. MATERIALS AND METHODS Patients diagnosed with osteoporotic vertebral compression fracture (OVCF) and undergoing VA surgery at our center between 2017 and 2021 were subject to a retrospective analysis. Radiological features of the T6-L5 vertebrae were derived from CT images. Clustering analysis, t-test, and LASSO (least absolute shrinkage and selection operator) regression were used to identify the optimization characteristics. A radiological signature model was constructed through the best combination of 13 machine learning algorithms. Radiomics signature was integrated with clinical characteristics into a nomogram for clinical applications. The model reliability was assessed by receiver operating characteristic (ROC) curve, calibration curve, clinical decision analysis (DCA), log-rank test, and confusion matrix. RESULTS A total of 470 eligible patients (81 with SVCF and 389 without) were identified in the clinical cohort. Eight radiomics features were identified and incorporated into machine learning, and "XGBoost" model showed the best performance. Final logistic nomogram included radiomics signature (P < 0.001), bone cement volume (P = 0.034), and T-scores of L1-L4 (P = 0.001), and showed satisfactory prediction capability in training set (0.986, 95%CI 0.969-1.000) and verification set (0.884, 95%CI 0.823-0.946). CONCLUSION Our radiomics-clinical model based on machine learning showed potential to prospectively predict SVCF after VA and provide precise treatment strategies.
Collapse
Affiliation(s)
- Xiaokun Wang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China (X.W., W.Y., Y.G., Y.G., H.W., Y.Z., D.P., X.G., W.C.)
| | - Wu Ye
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China (X.W., W.Y., Y.G., Y.G., H.W., Y.Z., D.P., X.G., W.C.)
| | - Yao Gu
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China (X.W., W.Y., Y.G., Y.G., H.W., Y.Z., D.P., X.G., W.C.)
| | - Yu Gao
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China (X.W., W.Y., Y.G., Y.G., H.W., Y.Z., D.P., X.G., W.C.)
| | - Haofan Wang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China (X.W., W.Y., Y.G., Y.G., H.W., Y.Z., D.P., X.G., W.C.)
| | - Yitong Zhou
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China (X.W., W.Y., Y.G., Y.G., H.W., Y.Z., D.P., X.G., W.C.)
| | - Dishui Pan
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China (X.W., W.Y., Y.G., Y.G., H.W., Y.Z., D.P., X.G., W.C.)
| | - Xuhui Ge
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China (X.W., W.Y., Y.G., Y.G., H.W., Y.Z., D.P., X.G., W.C.)
| | - Wei Liu
- Department of Orthopedics, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China (W.L.)
| | - Weihua Cai
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China (X.W., W.Y., Y.G., Y.G., H.W., Y.Z., D.P., X.G., W.C.).
| |
Collapse
|
78
|
Xu G, Feng F, Chen W, Xiao Y, Fu Y, Zhou S, Duan S, Li M. Development and External Validation of a CT-Based Radiomics Nomogram to Predict Perineural Invasion and Survival in Gastric Cancer: A Multi-institutional Study. Acad Radiol 2025; 32:120-131. [PMID: 39127522 DOI: 10.1016/j.acra.2024.07.051] [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: 04/06/2024] [Revised: 07/20/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
RATIONALE AND OBJECTIVES To develop and validate a radiomics nomogram utilizing CT data for predicting perineural invasion (PNI) and survival in gastric cancer (GC) patients. MATERIALS AND METHODS A retrospective analysis of 408 GC patients from two institutions: 288 patients from Institution I were divided 7:3 into a training set (n = 203) and a testing set (n = 85); 120 patients from Institution II served as an external validation set. Radiomics features were extracted and screened from CT images. Independent radiomics, clinical, and combined models were constructed to predict PNI. Model discrimination, calibration, clinical utility, and prognostic significance were evaluated using area under the curve (AUC), calibration curves, decision curves analysis, and Kaplan-Meier curves, respectively. RESULTS 15 radiomics features and three clinical factors were included in the final analysis. The AUCs of the radiomics model in the training, testing, and external validation sets were 0.843 (95% CI: 0.788-0.897), 0.831 (95% CI: 0.741-0.920), and 0.802 (95% CI: 0.722-0.882), respectively. A nomogram was developed by integrating significant clinical factors with radiomics features. The AUCs of the nomogram in the training, testing, and external validation sets were 0.872 (95% CI: 0.823-0.921), 0.862 (95% CI: 0.780-0.944), and 0.837 (95% CI: 0.767-0.908), respectively. Survival analysis revealed that the nomogram could effectively stratify patients for recurrence-free survival (Hazard Ratio: 4.329; 95% CI: 3.159-5.934; P < 0.001). CONCLUSION The radiomics-derived nomogram presented a promising tool for predicting PNI in GC and held significant prognostic implications. IMPORTANT FINDINGS The nomogram functioned as a non-invasive biomarker for determining the PNI status. The predictive performance of the nomogram surpassed that of the clinical model (P < 0.05). Furthermore, patients in the high-risk group stratified by the nomogram had a significantly shorter RFS (P < 0.05).
Collapse
Affiliation(s)
- Guodong Xu
- Department of Radiology, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng 224006, Jiangsu Province, China
| | - Feng Feng
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Wang Chen
- Department of Radiology, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng 224006, Jiangsu Province, China
| | - Yong Xiao
- Department of Radiology, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng 224006, Jiangsu Province, China
| | - Yigang Fu
- Department of Radiology, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng 224006, Jiangsu Province, China
| | - Siyu Zhou
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | | | - Manman Li
- Department of Radiology, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng 224006, Jiangsu Province, China.
| |
Collapse
|
79
|
Ye J, Chen Y, Pan J, Qiu Y, Luo Z, Xiong Y, He Y, Chen Y, Xie F, Huang W. US-based Radiomics Analysis of Different Machine Learning Models for Differentiating Benign and Malignant BI-RADS 4A Breast Lesions. Acad Radiol 2025; 32:67-78. [PMID: 39191562 DOI: 10.1016/j.acra.2024.08.024] [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: 06/07/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024]
Abstract
RATIONALE AND OBJECTIVES To investigate and authenticate the effectiveness of various radiomics models in distinguishing between benign and malignant BI-RADS 4A lesions. METHODS A total of 936 patients with pathologically confirmed 4A lesions were included in the study (training cohort: n = 655; test cohort: n = 281). Radiomic features were derived from greyscale US images. Following dimensionality reduction and feature selection, radiomics models were developed using logistic regression (LR), support vector machine (SVM), random forest (RF), eXtreme gradient boosting (XGBoost) and multilayer perceptron (MLP) algorithms. Univariate and multivariable logistic regression analyses were employed to investigate clinical-radiological characteristics and determine variables for creating a clinical model. Five combined models integrating radiomic and clinical parameters were constructed by using each algorithm, and comparison with radiologists' performance was performed. SHapley Additive exPlanations (SHAP) approach was used to elucidate the radiomic model by ranking the significance of features based on their contribution to the evaluation. RESULTS A total of 1561 radiomic features were extracted. Thirty-six features were deemed significant by dimensionality reduction and selection. The radiomic models showed good performance with AUCs of 0.829-0.945 in training cohort; and 0.805-0.857 in test cohort. The combined model developed by using LR showed the best performance (AUC, training cohort: 0.909; test cohort: 0.905), which is superior to radiologists' performance. Decision curve analysis (DCA) of this combined model indicated better clinical efficacy than clinical and radiomic models. CONCLUSIONS The combined model integrating radiomic and clinical features demonstrated excellent performance in differentiating between benign and malignant 4A lesions. It may offer a non-invasive and efficient approach to aid in clinical decision-making.
Collapse
Affiliation(s)
- Jieyi Ye
- Division of Interventional Ultrasound, Department of Medical Ultrasonics, Foshan First People's Hospital, 81 Lingnan North Road, Foshan 528000, Guangdong, China (J.Y., Y.C., Y.Q., Z.L., Y.X., Y.H., W.H.)
| | - Yinting Chen
- Division of Interventional Ultrasound, Department of Medical Ultrasonics, Foshan First People's Hospital, 81 Lingnan North Road, Foshan 528000, Guangdong, China (J.Y., Y.C., Y.Q., Z.L., Y.X., Y.H., W.H.)
| | - Jiawei Pan
- Department of Information Science, Foshan First People's Hospital, 81 Lingnan North Road, Foshan 528000, Guangdong, China (J.P.)
| | - Yide Qiu
- Division of Interventional Ultrasound, Department of Medical Ultrasonics, Foshan First People's Hospital, 81 Lingnan North Road, Foshan 528000, Guangdong, China (J.Y., Y.C., Y.Q., Z.L., Y.X., Y.H., W.H.)
| | - Zhuoru Luo
- Division of Interventional Ultrasound, Department of Medical Ultrasonics, Foshan First People's Hospital, 81 Lingnan North Road, Foshan 528000, Guangdong, China (J.Y., Y.C., Y.Q., Z.L., Y.X., Y.H., W.H.)
| | - Yue Xiong
- Division of Interventional Ultrasound, Department of Medical Ultrasonics, Foshan First People's Hospital, 81 Lingnan North Road, Foshan 528000, Guangdong, China (J.Y., Y.C., Y.Q., Z.L., Y.X., Y.H., W.H.)
| | - Yanping He
- Division of Interventional Ultrasound, Department of Medical Ultrasonics, Foshan First People's Hospital, 81 Lingnan North Road, Foshan 528000, Guangdong, China (J.Y., Y.C., Y.Q., Z.L., Y.X., Y.H., W.H.)
| | - Yingyu Chen
- Department of Radiology and Medical Ultrasonics, Leping Hospital Affiliated to Foshan First People's Hospital, 10 Lenan Road, Foshan 528100, Guangdong, China (Y.C., F.X.)
| | - Fuqing Xie
- Department of Radiology and Medical Ultrasonics, Leping Hospital Affiliated to Foshan First People's Hospital, 10 Lenan Road, Foshan 528100, Guangdong, China (Y.C., F.X.)
| | - Weijun Huang
- Division of Interventional Ultrasound, Department of Medical Ultrasonics, Foshan First People's Hospital, 81 Lingnan North Road, Foshan 528000, Guangdong, China (J.Y., Y.C., Y.Q., Z.L., Y.X., Y.H., W.H.).
| |
Collapse
|
80
|
Wang J, Serafini A, Kuker R, Ayubcha C, Cohen G, Nadel H, McKinney A, Alavi A, Yu JQ. The State-of-the-Art PET Tracers in Glioblastoma and High-grade Gliomas and Implications for Theranostics. PET Clin 2025; 20:147-164. [PMID: 39482219 DOI: 10.1016/j.cpet.2024.09.009] [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: 11/03/2024]
Abstract
MR imaging is currently the main imaging modality used for the diagnosis and post therapeutic assessment of glioblastomas. Recently, several innovative PET radioactive tracers have been investigated for the evaluation of glioblastomas (GBM). These radiotracers target several biochemical and pathophysiological processes seen in tumors. These include glucose metabolism, DNA synthesis and cell proliferation, amino acid transport, cell membrane biosynthesis, specific membrane antigens such as prostatic specific membrane antigens, fibroblast activation protein inhibitor, translocator protein and hypoxia sensing agents, and antibodies targeting specific cell receptor antigen. This review aims to discuss the clinical value of these PET radiopharmaceuticals in the evaluation and treatment of GBMs.
Collapse
Affiliation(s)
- Jiaqiong Wang
- Division of Nuclear Medicine, Department of Radiology, Temple University Health System, Fox Chase Cancer Center, Philadelphia, PA 19140, USA.
| | - Aldo Serafini
- Division of Nuclear Medicine, Department of Radiology, University of Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA
| | - Russ Kuker
- Division of Nuclear Medicine, Department of Radiology, University of Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA
| | - Cyrus Ayubcha
- Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gary Cohen
- Department of Radiology, Temple University Health System, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Helen Nadel
- Department of Radiology, Lucile Packard Children's Hospital at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander McKinney
- Department of Radiology, University of Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA
| | - Abass Alavi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jian Q Yu
- Division of Nuclear Medicine, Department of Radiology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
81
|
Hotton J, Beddok A, Moubtakir A, Papathanassiou D, Morland D. [ 18F]FDG PET/CT Radiomics in Cervical Cancer: A Systematic Review. Diagnostics (Basel) 2024; 15:65. [PMID: 39795593 PMCID: PMC11720459 DOI: 10.3390/diagnostics15010065] [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: 10/28/2024] [Revised: 12/06/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: Cervical cancer is a significant global health concern, with high incidence and mortality rates, especially in less-developed regions. [18F]FDG PET/CT is now indicated at various stages of management, but its analysis is essentially based on SUVmax, a measure of [18F]FDG uptake. Radiomics, by extracting a multitude of parameters, promises to improve the diagnostic and prognostic performance of the examination. However, studies remain heterogeneous, both in terms of patient numbers and methods, so a synthesis is needed. Methods: This systematic review was conducted following PRISMA-P guidelines and registered in PROSPERO (CRD42024584123). Eligible studies on PET/CT radiomics in cervical cancer were identified through PubMed and Scopus and assessed for quality using the Radiomics Quality Score (RQS v2.0), with data extraction focusing on study design, population characteristics, radiomic methods, and model performances. Results: The review identified 22 studies on radiomics in cervical cancer, 19 of which focused specifically on locally advanced cervical cancer (LACC) and assessed various clinical outcomes, such as survival, relapse, treatment response, and lymph node involvement prediction. They reported significant associations between prognostic indicators and radiomic features, indicating the potential of radiomics to improve the predictive accuracy for patient outcomes in LACC; however, the overall quality of the studies was relatively moderate, with a median RQS of 12/36. Conclusions: While radiomic analysis in cervical cancer presents promising opportunities for survival prediction and personalized care, further well-designed studies are essential to provide stronger evidence for its clinical utility.
Collapse
Affiliation(s)
- Judicael Hotton
- Department of Surgical Oncology, Institut Godinot, 51100 Reims, France
- CReSTIC, UR 3804, Université de Reims Champagne-Ardenne, 51687 Reims, France; (A.B.); (D.P.); (D.M.)
| | - Arnaud Beddok
- CReSTIC, UR 3804, Université de Reims Champagne-Ardenne, 51687 Reims, France; (A.B.); (D.P.); (D.M.)
- Department of Radiation Therapy, Institut Godinot, 51100 Reims, France
| | | | - Dimitri Papathanassiou
- CReSTIC, UR 3804, Université de Reims Champagne-Ardenne, 51687 Reims, France; (A.B.); (D.P.); (D.M.)
- Department of Nuclear Medicine, Institut Godinot, 51100 Reims, France;
| | - David Morland
- CReSTIC, UR 3804, Université de Reims Champagne-Ardenne, 51687 Reims, France; (A.B.); (D.P.); (D.M.)
- Department of Nuclear Medicine, Institut Godinot, 51100 Reims, France;
| |
Collapse
|
82
|
Cavallo AU, Stanzione A, Ponsiglione A, Trotta R, Fanni SC, Ghezzo S, Vernuccio F, Klontzas ME, Triantafyllou M, Ugga L, Kalarakis G, Cannella R, Cuocolo R. Prostate cancer MRI methodological radiomics score: a EuSoMII radiomics auditing group initiative. Eur Radiol 2024:10.1007/s00330-024-11299-x. [PMID: 39739041 DOI: 10.1007/s00330-024-11299-x] [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: 04/13/2024] [Revised: 09/05/2024] [Accepted: 10/10/2024] [Indexed: 01/02/2025]
Abstract
OBJECTIVES To evaluate the quality of radiomics research in prostate MRI for the evaluation of prostate cancer (PCa) through the assessment of METhodological RadiomICs (METRICS) score, a new scoring tool recently introduced with the goal of fostering further improvement in radiomics and machine learning methodology. MATERIALS AND METHODS A literature search was conducted from July 1st, 2019, to November 30th, 2023, to identify original investigations assessing MRI-based radiomics in the setting of PCa. Seven readers with varying expertise underwent a quality assessment using METRICS. Subgroup analyses were performed to assess whether the quality score varied according to papers' categories (diagnosis, staging, prognosis, technical) and quality ratings among these latter. RESULTS From a total of 1106 records, 185 manuscripts were available. Overall, the average METRICS total score was 52% ± 16%. ANOVA and chi-square tests revealed no statistically significant differences between subgroups. Items with the lowest positive scores were adherence to guidelines/checklists (4.9%), handling of confounding factors (14.1%), external testing (15.1%), and the availability of data (15.7%), code (4.3%), and models (1.6%). Conversely, most studies clearly defined patient selection criteria (86.5%), employed a high-quality reference standard (89.2%), and utilized a well-described (85.9%) and clinically applicable (87%) imaging protocol as a radiomics data source. CONCLUSION The quality of MRI-based radiomics research for PCa in recent studies demonstrated good homogeneity and overall moderate quality. KEY POINTS Question To evaluate the quality of MRI-based radiomics research for PCa, assessed through the METRICS score. Findings The average METRICS total score was 52%, reflecting moderate quality in MRI-based radiomics research for PCa, with no statistically significant differences between subgroups. Clinical relevance Enhancing the quality of radiomics research can improve diagnostic accuracy for PCa, leading to better patient outcomes and more informed clinical decision-making.
Collapse
Affiliation(s)
| | - Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Andrea Ponsiglione
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.
| | - Romina Trotta
- Department of Radiology, Fatima Hospital, Seville, Spain
| | | | | | - Federica Vernuccio
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Michail E Klontzas
- Artificial Intelligence and Translational Imaging (ATI) Lab, Department of Radiology, School of Medicine, University of Crete, Heraklion, Greece
- Department of Medical Imaging, University Hospital of Heraklion, Heraklion, Greece
- Division of Radiology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Matthaios Triantafyllou
- Artificial Intelligence and Translational Imaging (ATI) Lab, Department of Radiology, School of Medicine, University of Crete, Heraklion, Greece
- Department of Medical Imaging, University Hospital of Heraklion, Heraklion, Greece
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Georgios Kalarakis
- Division of Radiology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Roberto Cannella
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Renato Cuocolo
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy
| |
Collapse
|
83
|
Chen Q, Wang L, Deng Z, Wang R, Wang L, Jian C, Zhu YM. Cooperative multi-task learning and interpretable image biomarkers for glioma grading and molecular subtyping. Med Image Anal 2024; 101:103435. [PMID: 39778265 DOI: 10.1016/j.media.2024.103435] [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: 04/08/2024] [Revised: 11/12/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025]
Abstract
Deep learning methods have been widely used for various glioma predictions. However, they are usually task-specific, segmentation-dependent and lack of interpretable biomarkers. How to accurately predict the glioma histological grade and molecular subtypes at the same time and provide reliable imaging biomarkers is still challenging. To achieve this, we propose a novel cooperative multi-task learning network (CMTLNet) which consists of a task-common feature extraction (CFE) module, a task-specific unique feature extraction (UFE) module and a unique-common feature collaborative classification (UCFC) module. In CFE, a segmentation-free tumor feature perception (SFTFP) module is first designed to extract the tumor-aware features in a classification manner rather than a segmentation manner. Following that, based on the multi-scale tumor-aware features extracted by SFTFP module, CFE uses convolutional layers to further refine these features, from which the task-common features are learned. In UFE, based on orthogonal projection and conditional classification strategies, the task-specific unique features are extracted. In UCFC, the unique and common features are fused with an attention mechanism to make them adaptive to different glioma prediction tasks. Finally, deep features-guided interpretable radiomic biomarkers for each glioma prediction task are explored by combining SHAP values and correlation analysis. Through the comparisons with recent reported methods on a large multi-center dataset comprising over 1800 cases, we demonstrated the superiority of the proposed CMTLNet, with the mean Matthews correlation coefficient in validation and test sets improved by (4.1%, 10.7%), (3.6%, 23.4%), and (2.7%, 22.7%) respectively for glioma grading, 1p/19q and IDH status prediction tasks. In addition, we found that some radiomic features are highly related to uninterpretable deep features and that their variation trends are consistent in multi-center datasets, which can be taken as reliable imaging biomarkers for glioma diagnosis. The proposed CMTLNet provides an interpretable tool for glioma multi-task prediction, which is beneficial for glioma precise diagnosis and personalized treatment.
Collapse
Affiliation(s)
- Qijian Chen
- Key Laboratory of Advanced Medical Imaging and Intelligent Computing of Guizhou Province, Engineering Research Center of Text Computing, Ministry of Education, State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China
| | - Lihui Wang
- Key Laboratory of Advanced Medical Imaging and Intelligent Computing of Guizhou Province, Engineering Research Center of Text Computing, Ministry of Education, State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China.
| | - Zeyu Deng
- Key Laboratory of Advanced Medical Imaging and Intelligent Computing of Guizhou Province, Engineering Research Center of Text Computing, Ministry of Education, State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China
| | - Rongpin Wang
- Radiology department, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Li Wang
- Key Laboratory of Advanced Medical Imaging and Intelligent Computing of Guizhou Province, Engineering Research Center of Text Computing, Ministry of Education, State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China
| | - Caiqing Jian
- Key Laboratory of Advanced Medical Imaging and Intelligent Computing of Guizhou Province, Engineering Research Center of Text Computing, Ministry of Education, State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China
| | - Yue-Min Zhu
- University Lyon, INSA Lyon, CNRS, Inserm, CREATIS UMR5220, U1206, Lyon 69621, France
| |
Collapse
|
84
|
Brancato V, Garbino N, Aiello M, Salvatore M, Cavaliere C. Exploratory Analysis of Radiomics and Pathomics in Uterine Corpus Endometrial Carcinoma. Sci Rep 2024; 14:30727. [PMID: 39730425 DOI: 10.1038/s41598-024-78987-y] [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: 08/14/2024] [Accepted: 11/05/2024] [Indexed: 12/29/2024] Open
Abstract
Uterine corpus endometrial carcinoma (EC) is one of the most common malignancies in the female reproductive system, characterized by tumor heterogeneity at both radiological and pathological scales. Both radiomics and pathomics have the potential to assess this heterogeneity and support EC diagnosis. This study examines the correlation between radiomics features from Apparent Diffusion Coefficient (ADC) maps and post-contrast T1 (T1C) images with pathomic features from pathology images in 32 patients from the CPTAC-UCEC database. 91 radiomics features were extracted from ADC maps and T1C images, and 566 pathomic features from cell detections and cell density maps at four different resolutions. Spearman's correlation and Bayes Factor analysis were used to evaluate radio-pathomic correlations. Significant cross-scale correlations were found, with strengths ranging from 0.57 to 0.89 in absolute value (9.47 × 104 < BF < 4.77 × 1014) for the ADC task, and from 0.64 and 0.70 (1.80 × 104 < BF < 5.69 × 105) for the T1C task. Most significant and high cross-scale associations were observed between ADC textural features and features from cell density maps. Correlations involving morphometric features and ADC and T1C first-order features were also observed, reflecting variations in tumor aggressiveness and tissue composition. These findings suggest that correlating radiomic features from ADC and T1C features with histopathological features can enhance understanding of EC intratumoral heterogeneity.
Collapse
Affiliation(s)
| | - Nunzia Garbino
- IRCCS SYNLAB SDN, Via E. Gianturco 113, 80143, Naples, Italy
| | - Marco Aiello
- IRCCS SYNLAB SDN, Via E. Gianturco 113, 80143, Naples, Italy
| | - Marco Salvatore
- IRCCS SYNLAB SDN, Via E. Gianturco 113, 80143, Naples, Italy
| | - Carlo Cavaliere
- IRCCS SYNLAB SDN, Via E. Gianturco 113, 80143, Naples, Italy
| |
Collapse
|
85
|
Cao X, Xiong M, Liu Z, Yang J, Kan YB, Zhang LQ, Liu YH, Xie MG, Hu XF. Update report on the quality of gliomas radiomics: An integration of bibliometric and radiomics quality score. World J Radiol 2024; 16:794-805. [PMID: 39801663 PMCID: PMC11718527 DOI: 10.4329/wjr.v16.i12.794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/04/2024] [Accepted: 11/25/2024] [Indexed: 12/27/2024] Open
Abstract
BACKGROUND Despite the increasing number of publications on glioma radiomics, challenges persist in clinical translation. AIM To assess the development and reporting quality of radiomics in brain gliomas since 2019. METHODS A bibliometric analysis was conducted to reveal trends in brain glioma radiomics research. The Radiomics Quality Score (RQS), a metric for evaluating the quality of radiomics studies, was applied to assess the quality of adult-type diffuse glioma studies published since 2019. The total RQS score and the basic adherence rate for each item were calculated. Subgroup analysis by journal type and research objective was performed, correlating the total RQS score with journal impact factors. RESULTS The radiomics research in glioma was initiated in 2011 and has witnessed a surge since 2019. Among the 260 original studies, the median RQS score was 11, correlating with a basic compliance rate of 46.8%. Subgroup analysis revealed significant differences in domain 1 and its subitems (multiple segmentations) across journal types (P = 0.039 and P = 0.03, respectively). The Spearman correlation coefficients indicated that the total RQS score had a negative correlation with the Journal Citation Report category (-0.69) and a positive correlation with the five-year impact factors (0.318) of journals. CONCLUSION Glioma radiomics research quality has improved since 2019 but necessitates further advancement with higher publication standards.
Collapse
Affiliation(s)
- Xu Cao
- Department of Radiology, The People's Hospital of Shifang, Deyang 618400, Sichuan Province, China
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, Chongqing, China
| | - Ming Xiong
- Department of Digital Medicine, School of Biomedical Engineering and Medical Imaging, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhi Liu
- Department of Radiology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China
| | - Jing Yang
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu-Bo Kan
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Li-Qiang Zhang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yan-Hui Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Ming-Guo Xie
- Department of Radiology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 500643, Sichuan Province, China
| | - Xiao-Fei Hu
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, Chongqing, China
- Glioma Medicine Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, Chongqing, China
| |
Collapse
|
86
|
Wang B, Han X, Zhang Z, Song H, Song Y, Liu R, Li Z, Liu S. Longitudinal CT Radiomics to Predict Progression-free Survival in Patients with Locally Advanced Gastric Cancer After Neoadjuvant Chemotherapy. Acad Radiol 2024:S1076-6332(24)00943-7. [PMID: 39732617 DOI: 10.1016/j.acra.2024.11.068] [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: 09/20/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 12/30/2024]
Abstract
RATIONALE AND OBJECTIVES To develop and validate a radiomics signature, utilizing baseline and restaging CT, for preoperatively predicting progression-free survival (PFS) after neoadjuvant chemotherapy (NAC) in locally advanced gastric cancer (LAGC). METHODS A total of 316 patients with LAGC who received NAC followed by gastrectomy were retrospectively included in this single-center study; these patients were split into two cohorts, one for training (n = 243) and the other for validation (n = 73), based on the different districts of our hospital. A total of 1316 radiomics features were extracted from the volume of interest of the gastric-cancer lesion on venous phase CT images. Four radiomics signatures were built for predicting PFS based on baseline CT (Pre-Rad), restaging CT (Post-Rad), delta radiomics (Delta-Rad) and multi-time radiomics (PrePost-Rad), respectively. Then the PrePost-Rad was combined with clinical factors to establish a nomogram (Rad-clinical model). Kaplan-Meier survival curves with log-rank tests were used to assess the prognostic usefulness of the Rad-clinical model. RESULTS All baseline characteristics were not statistically different between the two cohorts. The PrePost-Rad achieved improved predictive value by a C-index of 0.724 (95% CI: 0.639-0.809) in the validation cohort [Pre-Rad: 0.715 (0.632-0.798); Post-Rad: 0.632 (0.538-0.725), Delta-Rad: 0.549 (0.447-0.651)]. In terms of clinical benefit, calibration capability, and prediction efficacy, the Rad-clinical model performed well for PFS prediction, with a C-index of 0.754 (95% CI: 0.707-0.800) and 0.719 (95% CI: 0.639-0.800) in the training and validation cohorts, respectively, superior to the clinical model (cN stage and CA199) but comparable to the PrePost-Rad. Moreover, the Rad-clinical model could accurately classify gastric-cancer patients after NAC into three PFS risk groups in both training and validation cohorts. The risk stratification also performed well in most subgroups (good responders, poor responders, ypTNM Ⅱ, and ypTNM Ⅲ/Ⅳ). CONCLUSIONS The Rad-clinical model integrating longitudinal radiomics score and clinical factors performed well in preoperatively predicting PFS of LAGC patients after NAC and surgery.
Collapse
Affiliation(s)
- Bo Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (B.W., X.H., Z.Z., Z.L., S.L.)
| | - Xiaomeng Han
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (B.W., X.H., Z.Z., Z.L., S.L.)
| | - Zaixian Zhang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (B.W., X.H., Z.Z., Z.L., S.L.)
| | - Hongzheng Song
- Department of Radiology, Qingdao Municipal Hospital, Shandong Province, Qingdao, Shandong Province, China (H.S.)
| | - Yaolin Song
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (Y.S.)
| | - Ruiqing Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (R.L.)
| | - Zhiming Li
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (B.W., X.H., Z.Z., Z.L., S.L.)
| | - Shunli Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (B.W., X.H., Z.Z., Z.L., S.L.).
| |
Collapse
|
87
|
Zhang Y, Huang Y, Xu M, Zhuang J, Zhou Z, Zheng S, Zhu B, Guan G, Chen H, Liu X. Pathomics-based machine learning models for predicting pathological complete response and prognosis in locally advanced rectal cancer patients post-neoadjuvant chemoradiotherapy: insights from two independent institutional studies. BMC Cancer 2024; 24:1580. [PMID: 39725903 DOI: 10.1186/s12885-024-13328-w] [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: 08/13/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Accurate prediction of pathological complete response (pCR) and disease-free survival (DFS) in locally advanced rectal cancer (LARC) patients undergoing neoadjuvant chemoradiotherapy (NCRT) is essential for formulating effective treatment plans. This study aimed to construct and validate the machine learning (ML) models to predict pCR and DFS using pathomics. METHOD A retrospective analysis was conducted on 294 patients who received NCRT from two independent institutions. Pathomics from pre-NCRT H&E stains were extracted, and five ML models were developed and validated across two centers using ROC, Kaplan-Meier, time-dependent ROC, and nomogram analyses. RESULT Among the five ML models, the Xgboost (XGB) model demonstrated superior performance in predicting pCR, achieving an AUC of 1.000 (p < 0.001) on the internal data-set and an AUC of 0.950 (p = 0.001) on the external data-set.The XGB model effectively differentiated between high-risk and low-risk prognosis patients across all five centers: internal dataset (DFS, p = 0.002; OS, p = 0.004) and external dataset (DFS, p = 0.074; OS, p = 0.224).Furthermore, the COX regression demonstrated that the tumor length (HR = 1.230, 95%CI: 1.050-1.440, p = 0.010), post-NCRT CEA (HR = 1.716, 95%CI: 1.031- 2.858, p = 0.038), and XGB model score (HR = 0.128, 95%CI: 0.026-0.636, p = 0.012) were independent predictors of DFS after NCRT in the internal data-set.Using COX regression, the nomogram model and time-dependent AUC analysis demonstrated strong predictive discrimination for DFS in LARC patients across two independent institutions. CONCLUSION The ML model based on pathomics demonstrated effective prediction of pCR and prognosis in LARC patients. Further validation in larger cohorts is warranted to confirm the findings of this study.
Collapse
Affiliation(s)
- Yiyi Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ying Huang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Meifang Xu
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiazheng Zhuang
- Department of Gastrointestinal Surgery, The Quanzhou First Hospital Affiliated of Fujian Medical University, Quanzhou, China
| | - Zhibo Zhou
- Fujian Medical University, Fuzhou, China
| | | | - Bingwang Zhu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guoxian Guan
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, Fujian, 350001, China.
| | - Hong Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, Fujian, 350001, China.
| | - Xing Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, Fujian, 350001, China.
| |
Collapse
|
88
|
Ahmadzadeh AM, Lomer NB, Ashoobi MA, Bathla G, Sotoudeh H. MRI-derived radiomics and end-to-end deep learning models for predicting glioma ATRX status: a systematic review and meta-analysis of diagnostic test accuracy studies. Clin Imaging 2024; 119:110386. [PMID: 39742798 DOI: 10.1016/j.clinimag.2024.110386] [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: 09/17/2024] [Revised: 11/06/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025]
Abstract
We aimed to systematically review and meta-analyze the predictive value of magnetic resonance imaging (MRI)-derived radiomics/end-to-end deep learning (DL) models in predicting glioma alpha thalassemia/mental retardation syndrome X-linked (ATRX) status. We conducted a comprehensive search across four major databases-Web of Science, PubMed, Scopus, and Embase. All the studies that assessed the performance of radiomics and/or end-to-end DL models for predicting glioma ATRX status were included. Quality assessment was performed using the Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) criteria and the METhodological RadiomICs Score (METRICS). Pooled estimates for performance metrics were calculated. I-squared was used to assess heterogeneity, while subgroup and sensitivity analyses were performed to find its potential sources. Publication bias was assessed using Deeks' funnel plots. Seventeen and eleven studies were included in the systematic review and meta-analysis, respectively. Most of the studies had a low risk of bias and low concern for applicability according to the QUADAS-2. Also, most of them had good quality according to the METRICS. Meta-analysis showed a pooled sensitivity of 0.80 (95%CI: 0.71-0.96), a specificity of 0.82 (95%CI: 0.67-0.93), a positive diagnostic likelihood ratio (DLR) of 6.77 (95%CI: 4.67-9.82), a negative DLR of 0.15 (95%CI: 0.06-0.38), a diagnostic odds ratio of 30.36 (95%CI: 15.87-58.05), and an area under the curve (AUC) of 0.92 (95%CI: 0.89-0.94). Subgroup analysis revealed significant intergroup differences based on several factors. Radiomics models can accurately predict ATRX status in gliomas, enhancing non-invasive tumor characterization and guiding treatment strategies.
Collapse
Affiliation(s)
- Amir Mahmoud Ahmadzadeh
- Department of Radiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nima Broomand Lomer
- Medical Image Processing Group, Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Girish Bathla
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Houman Sotoudeh
- Department of Radiology, Neuroradiology Section, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
89
|
Lopes Costa GL, Tasca Petroski G, Machado LG, Eulalio Santos B, de Oliveira Ramos F, Feuerschuette Neto LM, De Luca Canto G. Accuracy of machine learning models for pre-diagnosis and diagnosis of pancreatic ductal adenocarcinoma in contrast-CT images: a systematic review and meta-analysis. Abdom Radiol (NY) 2024:10.1007/s00261-024-04771-1. [PMID: 39720966 DOI: 10.1007/s00261-024-04771-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 12/26/2024]
Abstract
PURPOSE To evaluate the diagnostic ability and methodological quality of ML models in detecting Pancreatic Ductal Adenocarcinoma (PDAC) in Contrast CT images. METHOD Included studies assessed adults diagnosed with PDAC, confirmed by histopathology. Metrics of tests were interpreted by ML algorithms. Studies provided data on sensitivity and specificity. Studies that did not meet the inclusion criteria, segmentation-focused studies, multiple classifiers or non-diagnostic studies were excluded. PubMed, Cochrane Central Register of Controlled Trials, and Embase were searched without restrictions. Risk of bias was assessed using QUADAS-2, methodological quality was evaluated using Radiomics Quality Score (RQS) and a Checklist for AI in Medical Imaging (CLAIM). Bivariate random-effects models were used for meta-analysis of sensitivity and specificity, I2 values and subgroup analysis used to assess heterogeneity. RESULTS Nine studies were included and 12,788 participants were evaluated, of which 3,997 were included in the meta-analysis. AI models based on CT scans showed an accuracy of 88.7% (IC 95%, 87.7%-89.7%), sensitivity of 87.9% (95% CI, 82.9%-91.6%), and specificity of 92.2% (95% CI, 86.8%-95.5%). The average score of six radiomics studies was 17.83 RQS points. Nine ML methods had an average CLAIM score of 30.55 points. CONCLUSIONS Our study is the first to quantitatively interpret various independent research, offering insights for clinical application. Despite favorable sensitivity and specificity results, the studies were of low quality, limiting definitive conclusions. Further research is necessary to validate these models before widespread adoption.
Collapse
Affiliation(s)
- Geraldo Lucas Lopes Costa
- Federal University of Santa Catarina, Florianópolis, Brazil.
- Brazilian Center for Evidence-Based Research, Federal University of Santa Catarina, Florianópolis, Brazil.
| | - Guido Tasca Petroski
- Federal University of Santa Catarina, Florianópolis, Brazil
- Brazilian Center for Evidence-Based Research, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Luis Guilherme Machado
- Federal University of Santa Catarina, Florianópolis, Brazil
- Brazilian Center for Evidence-Based Research, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | | | | | - Graziela De Luca Canto
- Brazilian Center for Evidence-Based Research, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
90
|
Zhu J, Zhu X, Lv S, Guo D, Li H, Zhao Z. Incremental Value of Pericoronary Adipose Tissue Radiomics Models in Identifying Vulnerable Plaques. J Comput Assist Tomogr 2024:00004728-990000000-00402. [PMID: 39724572 DOI: 10.1097/rct.0000000000001704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Inflammatory characteristics in pericoronary adipose tissue (PCAT) may enhance the diagnostic capability of radiomics techniques for identifying vulnerable plaques. This study aimed to evaluate the incremental value of PCAT radiomics scores in identifying vulnerable plaques defined by intravascular ultrasound imaging (IVUS). METHODS In this retrospective study, a PCAT radiomics model was established and validated using IVUS as the reference standard. The dataset consisted of patients with coronary artery disease who underwent both coronary computed tomography angiography and IVUS examinations at a tertiary hospital between March 2023 and January 2024. The dataset was randomly assigned to the training and validation sets in a 7:3 ratio. The diagnostic performance of various models was evaluated on both sets using the area under the curve (AUC). RESULTS From 88 lesions in 79 patients, we selected 9 radiomics features (5 texture features, 1 shape feature, 1 gray matrix feature, and 2 first-order features) from the training cohort (n = 61) to build the PCAT model. The PCAT radiomics model demonstrated moderate to high AUCs (0.847 and 0.819) in both the training and test cohorts. Furthermore, the AUC of the PCAT radiomics model was significantly higher than that of the fat attenuation index model (0.847 vs 0.659, P < 0.05). The combined model had a higher AUC than the clinical model (0.925 vs 0.714, P < 0.01). CONCLUSIONS The PCAT radiomics signature of coronary CT angiography enabled the detection of vulnerable plaques defined by IVUS.
Collapse
Affiliation(s)
- Jinke Zhu
- From the School of Medicine, Shaoxing University, Shaoxing, Zhejiang, Shaoxing, Zhejiang, China
| | - Xiucong Zhu
- From the School of Medicine, Shaoxing University, Shaoxing, Zhejiang, Shaoxing, Zhejiang, China
| | - Sangying Lv
- Department of radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| | - Danling Guo
- Department of radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| | - Huaifeng Li
- Department of radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| | - Zhenhua Zhao
- Department of radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| |
Collapse
|
91
|
Zeng Q, Jia F, Tang S, He H, Fu Y, Wang X, Zhang J, Tan Z, Tang H, Wang J, Yi X, Chen BT. Ensemble learning-based radiomics model for discriminating brain metastasis from glioblastoma. Eur J Radiol 2024; 183:111900. [PMID: 39733718 DOI: 10.1016/j.ejrad.2024.111900] [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: 04/26/2024] [Revised: 10/24/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024]
Abstract
OBJECTIVE Differentiating between brain metastasis (BM) and glioblastoma (GBM) preoperatively is challenging due to their similar imaging features on conventional brain MRI. This study aimed to enhance diagnostic accuracy through a machine learning model based on MRI radiomics data. METHODS This retrospective study included 235 patients with confirmed solitary BM and 273 patients with GBM. Patients were randomly assigned to the training (n = 356) or the validation (n = 152) cohort. Conventional brain MRI sequences including T1-weighted imaging (T1WI), contrast-enhanced_T1WI, and T2-weighted imaging (T2WI) were acquired. Brain tumors were delineated on all three sequences and segmented. Features were selected from demographic, clinical, and radiomic data. An integrated ensemble machine learning model, i.e., the elastic regression-SVM-SVM model (ERSS) and a multivariable logistic regression (LR) model combining demographic, clinical, and radiomic data were built for predictive modeling. Model efficiency was evaluated using discrimination, calibration, and decision curve analyses. Additionally, external validation was performed using an independent cohort consisting of 47 patients with GBM and 43 patients with isolated BM to assess the ERSS model generalizability. RESULTS The ERSS model demonstrated more optimal classification performance (AUC: 0.9548, 95% CI: 0.9337-0.9734 in training cohort; AUC: 0.9716, 95% CI: 0.9485-0.9895 in validation cohort) as compared to the LR model according to the receiver operating characteristic (ROC) curve and decision curve for the internal cohort. The external validation cohort had less optimal but still robust performance (AUC: 0.7174, 95% CI: 0.6172-0.8024). The ERSS model with integration of multiple classifiers, including elastic net, random forest and support vector machine, produced robust predictive performance and outperformed the LR method. CONCLUSION The results suggested that the integrated machine learning model, i.e., the ERSS model, had the potential for efficient and accurate preoperative differentiation of BM from GBM, which may improve clinical decision-making and outcomes of patients with brain tumors.
Collapse
Affiliation(s)
- Qi Zeng
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Fangxu Jia
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Shengming Tang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Haoling He
- Department of Radiology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Yan Fu
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha 410008, Hunan, PR China
| | - Xueying Wang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Jinfan Zhang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zeming Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Haiyun Tang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.
| | - Jing Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.
| | - Xiaoping Yi
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Department of Radiology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, PR China.
| | - Bihong T Chen
- Department of Diagnostic Radiology, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
92
|
Valizadeh P, Jannatdoust P, Ghadimi DJ, Bagherieh S, Hassankhani A, Amoukhteh M, Adli P, Gholamrezanezhad A. Predicting lymph node metastasis in thyroid cancer: systematic review and meta-analysis on the CT/MRI-based radiomics and deep learning models. Clin Imaging 2024; 119:110392. [PMID: 39742800 DOI: 10.1016/j.clinimag.2024.110392] [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: 08/26/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Thyroid cancer, a common endocrine malignancy, has seen increasing incidence, making lymph node metastasis (LNM) a critical factor for recurrence and survival. Radiomics and deep learning (DL) advancements offer the potential for improved LNM prediction using CT and MRI, though challenges in diagnostic accuracy remain. METHODS A systematic review and meta-analysis were conducted per established guidelines, with searches across PubMed, Scopus, Web of Science, and Embase up to February 15, 2024. Studies developing CT/MRI-based radiomics and/or DL models for preoperative LNM assessment in thyroid cancer patients were included. Data were extracted and analyzed using R software. RESULTS Sixteen studies were analyzed. In internal validation sets, sensitivity was 81.1 % (95 % CI: 75.6 %-85.6 %) and specificity 76.4 % (95 % CI: 68.4 %-82.9 %). Training sets showed a sensitivity of 84.4 % (95 % CI: 81.5 %-87 %) and a specificity of 84.7 % (95 % CI: 74.4 %-91.4 %). The pooled AUC was 86 % for internal validation and 87 % for training. Handcrafted radiomics had a sensitivity of 79.4 % and specificity of 69.2 %, while DL models showed 80.8 % sensitivity and 78.7 % specificity. Subgroup analysis revealed that models for papillary thyroid cancer (PTC) had a pooled specificity of 76.3 %, while those including other or unspecified cancers showed 68.3 % specificity. Despite heterogeneity, significant differences (p = 0.037) were noted between models with and without clinical data. CONCLUSION Radiomics and DL models show promising potential for detecting LNM in thyroid cancer, particularly in PTC. However, study heterogeneity underscores the need for further research to optimize these imaging tools.
Collapse
Affiliation(s)
- Parya Valizadeh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Payam Jannatdoust
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Delaram J Ghadimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Bagherieh
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Hassankhani
- Department of Radiology, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA; Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Melika Amoukhteh
- Department of Radiology, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA; Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Paniz Adli
- College of Letters and Science, University of California, Berkeley, CA, USA
| | - Ali Gholamrezanezhad
- Department of Radiology, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA.
| |
Collapse
|
93
|
Wen B, Li C, Cai Q, Shen D, Bu X, Zhou F. Multimodal MRI radiomics-based stacking ensemble learning model with automatic segmentation for prognostic prediction of HIFU ablation of uterine fibroids: a multicenter study. Front Physiol 2024; 15:1507986. [PMID: 39759109 PMCID: PMC11695313 DOI: 10.3389/fphys.2024.1507986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Objectives To evaluate the effectiveness of an MRI radiomics stacking ensemble learning model, which combines T2-weighted imaging (T2WI) and contrast-enhanced T1-weighted imaging (CE-T1WI) with deep learning-based automatic segmentation, for preoperative prediction of the prognosis of high-intensity focused ultrasound (HIFU) ablation of uterine fibroids. Methods This retrospective study collected data from 360 patients with uterine fibroids who underwent HIFU treatment. The dataset was sourced from Center A (training set: N = 240; internal test set: N = 60) and Center B (external test set: N = 60). Patients were categorized into favorable and unfavorable prognosis groups based on the post-treatment non-perfused volume ratio. Automated segmentation of uterine fibroids was performed using a V-net deep learning models. Radiomics features were extracted from T2WI and CE-T1WI, followed by data preprocessing including normalization and scaling. Feature selection was performed using t-test, Pearson correlation, and LASSO to identify the most predictive features for preoperative prognosis Support Vector Machine (SVM), Random Forest (RF), Light Gradient Boosting Machine (LightGBM), and Multilayer Perceptron (MLP) were employed as base learners to construct base predictive models. These models were integrated into a stacking ensemble model, with Logistic Regression serving as the meta-learner to combine the outputs of the base models. The performance of the models was assessed using the area under the receiver operating characteristic curve (AUC). Results Among the base models developed using T2WI and CE-T1WI features, the MLP model exhibited superior performance, achieving an AUC of 0.858 (95% CI: 0.756-0.959) in the internal test set and 0.828 (95% CI: 0.726-0.930) in the external test set. It was followed by the SVM, LightGBM, and RF, which obtained AUC values of 0.841 (95% CI: 0.737-0.946), 0.823 (95% CI: 0.711-0.934), and 0.750 (95% CI: 0.619-0.881), respectively. The stacking ensemble learning model, which integrated these five algorithms, demonstrated a notable enhancement in performance, with an AUC of 0.897 (95% CI: 0.818-0.977) in the internal test set and 0.854 (95% CI: 0.759-0.948) in the external test set. Conclusion The DL based automatic segmentation MRI radiomics stacking ensemble learning model demonstrated high accuracy in predicting the prognosis of HIFU ablation of uterine fibroids.
Collapse
Affiliation(s)
- Bing Wen
- Department of Radiology, Yiyang Central Hospital, Yiyang, China
| | - Chengwei Li
- Department of Radiology, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Qiuyi Cai
- Department of Radiology, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Dan Shen
- Department of Radiology, Yiyang Central Hospital, Yiyang, China
| | - Xinyi Bu
- Department of Radiology, Yiyang Central Hospital, Yiyang, China
| | - Fuqiang Zhou
- Department of Radiology, Yiyang Central Hospital, Yiyang, China
| |
Collapse
|
94
|
Wang Y, Zhang J, Li Q, Sun L, Zheng Y, Gao C, Dong C. An MRI-based radiomics nomogram for preoperative prediction of Ki-67 index in nasopharyngeal carcinoma: a two-center study. Front Oncol 2024; 14:1423304. [PMID: 39759139 PMCID: PMC11695239 DOI: 10.3389/fonc.2024.1423304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/27/2024] [Indexed: 01/07/2025] Open
Abstract
Background The expression level of Ki-67 in nasopharyngeal carcinoma (NPC) affects the prognosis and treatment options of patients. Our study developed and validated an MRI-based radiomics nomogram for preoperative evaluation of Ki-67 expression levels in nasopharyngeal carcinoma (NPC). Methods In all, 133 patients with pathologically-confirmed (post-operatively) NPC who underwent MRI examination in one of two medical centers. Data from one medical center (n=105; Ki-67: ≥50% [n=57], <50% [n=48]) formed the training set, while data from another medical center (n=28; Ki-67: ≥50% [n=15], <50% [n=13]) formed the test set. Clinical data and routine MRI results were reviewed to determine significant predictive factors. The minimum absolute shrinkage and selection operator method was used to select key radiomics features to form a radiomics signatures from resonance imaging (MRI), and a radiomics score (Rad-score) was calculated. Subsequently, a radiomics nomogram was established using a logistic regression (LR) algorithm. The predictive performance of the nomogram was evaluated using operating characteristics curve (ROC), decision curve analysis (DCA), and the area under the curve (AUC). Results Five radiomics features were selected to build the radiomics signature. The radiomics nomogram incorporating the clinical factors and radiomics signature showed favorable predictive value for expression level of Ki-67, with AUC 0.841 (95% confidence intervals: 0.654 -0.951) for the test set. Decision curve analysis showed that the nomogram outperformed a clinical model in terms of clinical usefulness. Conclusions The radiomics nomogram based on MRI effectively predicted the pre-surgical expression level of Ki-67.
Collapse
Affiliation(s)
- Yao Wang
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Zhang
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qiyuan Li
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Sun
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingmei Zheng
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chuanping Gao
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cheng Dong
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
95
|
Netti S, D'Ecclesiis O, Corso F, Botta F, Origgi D, Pesapane F, Agazzi GM, Rotili A, Gaeta A, Scalco E, Rizzo G, Jereczek-Fossa BA, Cassano E, Curigliano G, Gandini S, Raimondi S. Methodological issues in radiomics: impact on accuracy of MRI for predicting response to neoadjuvant chemotherapy in breast cancer. Eur Radiol 2024:10.1007/s00330-024-11260-y. [PMID: 39702630 DOI: 10.1007/s00330-024-11260-y] [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: 05/08/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
AIM To investigate whether methodological aspects may influence the performance of MRI-radiomic models to predict response to neoadjuvant treatment (NAT) in breast cancer (BC) patients. MATERIALS AND METHODS We conducted a systematic review until March 2023. A random-effects meta-analysis was performed to combine the area under the receiver operating characteristic curve (AUC) values. Publication bias was assessed using Egger's test and heterogeneity was estimated by I2. A meta-regression was conducted to investigate the impact of various factors, including scanner, features' number/transformation/type, pixel/voxel scaling, etc. RESULTS: Forty-two studies were included. The summary AUC was 0.77 (95% CI: 0.74-0.81). Substantial heterogeneity was observed (I2 = 81%) with no publication bias (p = 0.35). Radiomic model accuracy was influenced by the scanner vendor, with lower AUCs in studies using mixed scanner vendors (AUC; 95% CI: 0.70; 0.61-0.78) compared to studies including images obtained from the same scanner (AUC (95% CI): 0.83 (0.77-0.88), 0.74 (0.67-0.82), 0.83 (0.78-0.89) for three different vendors; vendors 1, 2, and 3, respectively; p-value = 0.03 for comparison with vendor 1). Feature type also seemed to have an impact on the AUC, with higher prediction accuracy observed for studies using 3D than 2D/2.5D images (AUC; 95% CI: 0.81; 0.78-0.85 and 0.73; 0.65-0.81, respectively, p-value = 0.03). Non-significant between-study heterogeneity was observed in the studies including 3D images (I2 = 33%) and Vendor 1 scanners (I2 = 40%). CONCLUSION MRI-radiomics has emerged as a potential method for predicting the response to NAT in BC patients, showing promising outcomes. Nevertheless, it is important to acknowledge the diversity among the methodological choices applied. Further investigations should prioritize achieving standardized protocols, and enhancing methodological rigor in MRI-radiomics. KEY POINTS Question Do methodological aspects influence the performance of MRI-radiomic models in predicting response to NAT in BC patients? Findings Radiomic model accuracy was influenced by the scanner vendor and feature type. Clinical relevance Methodological discrepancies affect the performance of MRI-radiomic models. Developing standardized protocols and enhancing methodological rigor in these studies should be prioritized.
Collapse
Affiliation(s)
- Sofia Netti
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | - Oriana D'Ecclesiis
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Corso
- Department of Mathematics (DMAT), Politecnico di Milano, Milan, Italy
- Centre for Health Data Science (CHDS), Human Techonopole, Milan, Italy
| | - Francesca Botta
- Medical Physics Unit, IEO, European Institute of Oncology IRCCS*, Milan, Italy
| | - Daniela Origgi
- Medical Physics Unit, IEO, European Institute of Oncology IRCCS*, Milan, Italy.
| | - Filippo Pesapane
- Breast Imaging Division, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Anna Rotili
- Breast Imaging Division, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Aurora Gaeta
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Elisa Scalco
- Institute of Biomedical Technologies, Segrate, Italy
| | - Giovanna Rizzo
- Institute of Intelligent Industrial Systems and Technologies for Advanced Manufacturing, Milan, Italy
| | | | - Enrico Cassano
- Breast Imaging Division, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Gandini
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Sara Raimondi
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| |
Collapse
|
96
|
Zhang H, Xu L, Yang L, Su Z, Kang H, Xie X, He X, Zhang H, Zhang Q, Cao X, He X, Zhang T, Zhao F. Deep learning-based intratumoral and peritumoral features for differentiating ocular adnexal lymphoma and idiopathic orbital inflammation. Eur Radiol 2024:10.1007/s00330-024-11275-5. [PMID: 39702637 DOI: 10.1007/s00330-024-11275-5] [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: 05/26/2024] [Revised: 10/16/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
OBJECTIVES To evaluate the value of deep-learning-based intratumoral and peritumoral features for differentiating ocular adnexal lymphoma (OAL) and idiopathic orbital inflammation (IOI). METHODS Nighty-seven patients with histopathologically confirmed OAL (n = 43) and IOI (n = 54) were randomly divided into training (n = 79) and test (n = 18) groups. DL-based intratumoral and peritumoral features were extracted to characterize the differences in heterogeneity and tissue invasion between different lesions, respectively. Subsequently, an attention-based fusion model was employed to fuse the features extracted from intra- and peritumoral regions and multiple MR sequences. A comprehensive comparison was conducted among different methods for extracting intratumoral, peritumoral, and fused features. Area under the curve (AUC) was used to evaluate the performance under a 10-fold cross-validation and independent test. Chi-square and student's t-test were used to compare discrete and continuous variables, respectively. RESULTS Fused intra-peritumoral features achieved AUC values of 0.870-0.930 and 0.849-0.924 on individual MR sequences in the validation and test sets, respectively. This was significantly higher than those using intratumoral features (p < 0.05), but not significantly different than those using peritumoral features (p > 0.05). By combining multiple MR sequences, AUC values of the intra-peritumoral features were boosted to 0.943 and 0.940, higher than those obtained from each sequence alone. Moreover, intra-peritumoral features yielded higher AUC values compared to entire orbital cone features extracted by either the intra- or the peritumoral DL model, although no significant difference was found from the latter (p > 0.05). CONCLUSION DL-based intratumoral, peritumoral, and especially fused intra-peritumoral features may help differentiate between OAL and IOI. KEY POINTS Question What is the diagnostic value of the peritumoral region and its combination with the intratumoral region for radiomic analysis of orbital lymphoproliferative disorders? Findings Fused intra- and peritumoral features achieved significantly higher performance than intratumoral features, but had no significant difference to the peritumoral features. Clinical relevance Peritumoral contextual features, which characterize the invasion patterns of orbital lesions within the surrounding areas of the entire orbital cone, might serve as an independent imaging marker for differentiating between OAL and IOI.
Collapse
Affiliation(s)
- Huachen Zhang
- Xi'an Key Lab of Radiomics and Intelligent Perception, School of Information Science and Technology, Northwest University, Xi'an, China
| | - Li Xu
- Department of Computer Science and Technology, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Lijuan Yang
- Department of Radiology, Xi'an Fourth Hospital, Xi'an, China
| | - Zhiming Su
- Xi'an Key Lab of Radiomics and Intelligent Perception, School of Information Science and Technology, Northwest University, Xi'an, China
| | - Haobei Kang
- Xi'an Key Lab of Radiomics and Intelligent Perception, School of Information Science and Technology, Northwest University, Xi'an, China
| | - Xiaoyang Xie
- Xi'an Key Lab of Radiomics and Intelligent Perception, School of Information Science and Technology, Northwest University, Xi'an, China
| | - Xuelei He
- Xi'an Key Lab of Radiomics and Intelligent Perception, School of Information Science and Technology, Northwest University, Xi'an, China
| | - Hui Zhang
- Department of Radiology, Xi'an Fourth Hospital, Xi'an, China
| | - Qiufang Zhang
- Department of Radiology, Xi'an Fourth Hospital, Xi'an, China
| | - Xin Cao
- Xi'an Key Lab of Radiomics and Intelligent Perception, School of Information Science and Technology, Northwest University, Xi'an, China
| | - Xiaowei He
- Xi'an Key Lab of Radiomics and Intelligent Perception, School of Information Science and Technology, Northwest University, Xi'an, China
| | - Tao Zhang
- Xi'an Key Lab of Radiomics and Intelligent Perception, School of Information Science and Technology, Northwest University, Xi'an, China.
| | - Fengjun Zhao
- Xi'an Key Lab of Radiomics and Intelligent Perception, School of Information Science and Technology, Northwest University, Xi'an, China.
| |
Collapse
|
97
|
Hao P, Xin R, Li Y, Na X, Lv X. Developmental trends and knowledge frameworks in the application of radiomics in prostate cancer: a bibliometric analysis from 2000 to 2024. Discov Oncol 2024; 15:781. [PMID: 39692833 DOI: 10.1007/s12672-024-01678-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND This research utilized the bibliometrics method to analyze the published literature related to prostate cancer (PCa) imaging. Furthermore, current knowledge and research hotspots of radiomics in PCa diagnosis and treatment were comprehensively reviewed, as well as progress and emerging trends in field were explored. METHODS In this investigation, the relevant literature on radiomics, and PCa was retrieved from Web of Science Core Collection (WoSCC) databases from 2000 and 2024. Furthermore, a comprehensive bibliometric analysis was carried out using advanced tools like CiteSpace6.2, VOS viewer, and the 'bibliometrix' package of R software to visualize the annual distribution of publications across various aspects such as authors, countries, journals, institutions, and keywords. RESULTS This analysis included 593 from 58 countries including China and the United States. Chinese Academy of Sciences and Frontiers in Oncology were the institutions and journals that publish the most relevant articles, -while Radiology journal had the greatest number of co-cited publications. Furthermore, 3,621 authors published on this topic, of which Madabhushi Anant and Stoyanova Radka had the highest contributions. Moreover, Lambin, P. had the most co-citations. In addition, the diagnostic characteristics of radiomics in PCa imaging and treatment strategies are the current research focal points. The establishment of multi-functional imaging techniques and independent factor models warrants future investigation. CONCLUSIONS In summary, this analysis revealed that the research on PCa imaging is developing vigorously, focusing on the diagnostic methods and intervention measures of imaging in PCa diagnosis and treatment. In the future, there is an urgent need for improved collaboration and communication among countries and institutions.
Collapse
Affiliation(s)
- Pan Hao
- Medical Imaging Center, LuHe Hospital, Capital Medical University, Beijing, China
| | - Ruiqiang Xin
- Medical Imaging Center, LuHe Hospital, Capital Medical University, Beijing, China.
| | - Yancui Li
- Medical Imaging Center, LuHe Hospital, Capital Medical University, Beijing, China
| | - Xu Na
- Medical Imaging Center, LuHe Hospital, Capital Medical University, Beijing, China
| | - Xiaoyong Lv
- Medical Imaging Center, LuHe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
98
|
Zhao X, Wang Y, Xue M, Ding Y, Zhang H, Wang K, Ren J, Li X, Xu M, Lv J, Wang Z, Sun D. Preoperative assessment of tertiary lymphoid structures in stage I lung adenocarcinoma using CT radiomics: a multicenter retrospective cohort study. Cancer Imaging 2024; 24:167. [PMID: 39696659 DOI: 10.1186/s40644-024-00813-5] [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: 08/24/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE To develop a multimodal predictive model, Radiomics Integrated TLSs System (RAITS), based on preoperative CT radiomic features for the identification of TLSs in stage I lung adenocarcinoma patients and to evaluate its potential in prognosis stratification and guiding personalized treatment. METHODS The most recent preoperative chest CT thin-slice scans and postoperative hematoxylin and eosin-stained pathology sections of patients diagnosed with stage I LUAD were retrospectively collected. Tumor segmentation was achieved using an automatic virtual adversarial training segmentation algorithm based on a three-dimensional U-shape convolutional neural network (3D U-Net). Radiomic features were extracted from the tumor and peritumoral areas, with extensions of 2 mm, 4 mm, 6 mm, and 8 mm, respectively, and deep learning image features were extracted through a convolutional neural network. Subsequently, the RAITS was constructed. The performance of RAITS was then evaluated in both the train and validation cohorts. RESULTS RAITS demonstrated superior AUC, sensitivity, and specificity in both the training and external validation cohorts, outperforming traditional unimodal models. In the validation cohort, RAITS achieved an AUC of 0.78 (95% CI, 0.69-0.88) and showed higher net benefits across most threshold ranges. RAITS exhibited strong discriminative ability in risk stratification, with p < 0.01 in the training cohort and p = 0.02 in the validation cohort, consistent with the actual predictive performance of TLSs, where TLS-positive patients had significantly higher recurrence-free survival (RFS) compared to TLS-negative patients (p = 0.04 in the training cohort, p = 0.02 in the validation cohort). CONCLUSION As a multimodal predictive model based on preoperative CT radiomic features, RAITS demonstrated excellent performance in identifying TLSs in stage I LUAD and holds potential value in clinical decision-making.
Collapse
Affiliation(s)
| | - Yuhang Wang
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China
| | - Mengli Xue
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
- Department of Pathology, Tianjin Chest Hospital, Tianjin, China
| | - Yun Ding
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Han Zhang
- Chest hospital, Tianjin University, Tianjin, China
| | - Kai Wang
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China
| | - Jie Ren
- Department of Thoracic Surgery, Tianjin Jinnan Hospital, Tianjin, China
| | - Xin Li
- Chest hospital, Tianjin University, Tianjin, China
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China
| | - Meilin Xu
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
- Department of Pathology, Tianjin Chest Hospital, Tianjin, China
| | - Jun Lv
- Department of Imaging, Tianjin Chest Hospital, Tianjin, China
| | - Zixiao Wang
- Department of Thoracic Surgery, Qinhuangdao First Hospital, Hebei Province, China
| | - Daqiang Sun
- Chest hospital, Tianjin University, Tianjin, China.
- Department of Thoracic Surgery, Tianjin Chest Hospital, No. 261, Taierzhuang South Road, Jinnan District, Tianjin, 300222, China.
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
99
|
Chen J, Liu S, Lin Y, Hu W, Shi H, Liao N, Zhou M, Gao W, Chen Y, Shi P. The quality and accuracy of radiomics model in diagnosing osteoporosis: a systematic review and meta-analysis. Acad Radiol 2024:S1076-6332(24)00940-1. [PMID: 39701845 DOI: 10.1016/j.acra.2024.11.065] [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: 10/09/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024]
Abstract
RATIONALE AND OBJECTIVES The purpose of this study is to conduct a meta-analysis to evaluate the diagnostic performance of current radiomics models for diagnosing osteoporosis, as well as to assess the methodology and reporting quality of these radiomics studies. METHODS According to PRISMA guidelines, four databases including MEDLINE, Web of Science, Embase and the Cochrane Library were searched systematically to select relevant studies published before July 18, 2024. The articles that used radiomics models for diagnosing osteoporosis were considered eligible. The Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2) tool and radiomics quality score (RQS) were used to assess the quality of included studies. The pooled diagnostic odds ratio (DOR), sensitivity, specificity, area under the summary receiver operator characteristic curve (AUC) were calculated to estimated diagnostic efficiency of pooled model. RESULTS A total of 25 studies were included, of which 24 provided usable data that were utilized for the meta-analysis, including 1553 patients with osteoporosis and 2200 patients without osteoporosis. The mean RQS score of included studies was 11.48 ± 4.92, with an adherence rate of 31.89%. The pooled DOR, sensitivity and specificity for model to diagnose osteoporosis were 81.72 (95% CI: 51.08 - 130.73), 0.90 (95% CI: 0.87-0.93) and 0.90 (95% CI: 0.87-0.93), respectively. The AUC was 0.96, indicating a high diagnostic capability. Subgroup analysis revealed that the use of different imaging modalities to construct radiomics models might be one source of heterogeneity. Radiomics models built using CT images and deep learning algorithms demonstrated higher diagnostic accuracy for osteoporosis. CONCLUSION Radiomics models for the diagnosis of osteoporosis have high diagnostic efficacy. In the future, radiomics models for diagnosing osteoporosis will be an efficient instrument to assist clinical doctors in screening osteoporosis patients. However, relevant guidelines should be followed strictly to improve the quality of radiomics studies.
Collapse
Affiliation(s)
- Jianan Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China (J.C., S.L., Y.L., W.H., H.S., N.L., W.G., Y.C.)
| | - Song Liu
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China (J.C., S.L., Y.L., W.H., H.S., N.L., W.G., Y.C.)
| | - Youxi Lin
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China (J.C., S.L., Y.L., W.H., H.S., N.L., W.G., Y.C.)
| | - Wenjun Hu
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China (J.C., S.L., Y.L., W.H., H.S., N.L., W.G., Y.C.)
| | - Huihong Shi
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China (J.C., S.L., Y.L., W.H., H.S., N.L., W.G., Y.C.)
| | - Nianchun Liao
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China (J.C., S.L., Y.L., W.H., H.S., N.L., W.G., Y.C.)
| | - Miaomiao Zhou
- Department of Endocrinology, People's Hospital of Dingbian, Dingbian, Shanxi, PR China (M.Z.)
| | - Wenjie Gao
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China (J.C., S.L., Y.L., W.H., H.S., N.L., W.G., Y.C.)
| | - Yanbo Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China (J.C., S.L., Y.L., W.H., H.S., N.L., W.G., Y.C.)
| | - Peijie Shi
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China (P.S.).
| |
Collapse
|
100
|
Li X, Ding R, Liu Z, Teixeira WMS, Ye J, Tian L, Li H, Guo S, Yao K, Ma Z, Liu Z. A predictive system comprising serum microRNAs and radiomics for residual retroperitoneal masses in metastatic nonseminomatous germ cell tumors. Cell Rep Med 2024; 5:101843. [PMID: 39672156 PMCID: PMC11722113 DOI: 10.1016/j.xcrm.2024.101843] [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: 05/28/2024] [Revised: 09/18/2024] [Accepted: 11/06/2024] [Indexed: 12/15/2024]
Abstract
Predicting the histopathology of residual retroperitoneal masses (RMMs) before post-chemotherapy retroperitoneal lymph node dissection in metastatic nonseminomatous germ cell tumors (NSGCTs) can guide individualized treatment and minimize complications. Previous single approach-based models perform poorly in validation. Herein, we introduce a machine learning model that evolves from a single-dimensional tumor diameter to incorporate high-dimensional radiomic features, with its effectiveness assessed using the macro-average area under the receiver operating characteristic curves (AUCs). In addition, we utilize more precise and specific microRNAs (miRNAs), not common clinical indicators, to construct an integrated radiomics-miRNA predictive system, achieving an AUC of 0.91 (0.80-0.99) in the prospective test set. We further develop a web-based dynamic nomogram for swift and precise calculation of the histopathological probabilities of RMMs based on radiomic scores and serum miRNA levels. The radiomics-miRNA integrated system offers a promising tool to select personalized treatments for patients with metastatic NSGCT.
Collapse
Affiliation(s)
- Xiangdong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Renjie Ding
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Zhenhua Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Wilhem M S Teixeira
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Jingwei Ye
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Li Tian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Haojiang Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Shengjie Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Kai Yao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Zikun Ma
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Zhuowei Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| |
Collapse
|