1
|
Chang CP, Huang YC, Tsai YH, Lin LC, Yang JT, Wu KH, Wu PH, Peng SJ. Radiomics-based MRI model to predict hypoperfusion in lacunar infarction. Magn Reson Imaging 2025; 120:110366. [PMID: 40122187 DOI: 10.1016/j.mri.2025.110366] [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: 07/16/2024] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Approximately 20-30 % of patients with acute ischemic stroke due to lacunar infarction experience early neurological deterioration (END) within the first three days after onset, leading to disability or more severe sequelae. Hemodynamic perfusion deficits may play a crucial role in END, causing growth in the infarcted area and functional impairments, and even poor long-term prognosis. Therefore, it is vitally important to predict which patients may be at risk of perfusion deficits to initiate treatment and close monitoring early, preparing for potential reperfusion. Our goal is to utilize radiomic features from magnetic resonance imaging (MRI) and machine learning techniques to develop a predictive model for hypoperfusion. METHOD During January 2011 to December 2020, a retrospective collection of 92 patients with lacunar stroke was conducted, who underwent MRI within 48 h, had clinical laboratory values, follow-up prognosis records, and advanced perfusion image to confirm the presence of hypoperfusion. Using the initial MRI of these patients, radiomics features were extracted and selected from Diffusion Weighted Imaging (DWI), Apparent Diffusion Coefficient (ADC), and Fluid Attenuated Inversion Recovery (FLAIR) sequences. The data was divided into an 80 % training set and a 20 % testing set, and a hypoperfusion prediction model was developed using machine learning. RESULT Tthe model trained on DWI + FLAIR sequence showed superior performance with an accuracy of 84.1 %, AUC 0.92, recall 79.5 %, specificity 87.8 %, precision 83.8 %, and F1 score 81.2. Statistically significant clinical factors between patients with and without hypoperfusion included the NIHSS scores and the size of the lacunar infarction. Combining these two features with the top seven weighted radiomics features from DWI + FLAIR sequence, a total of nine features were used to develop a new prediction model through machine learning. This model in test set achieved an accuracy of 88.9 %, AUC 0.91, recall 87.5 %, specificity 90.0 %, precision 87.5 %, and F1 score 87.5. CONCLUSION Utilizing radiomics techniques on DWI and FLAIR sequences from MRI of patients with lacunar stroke, it is possible to predict the presence of hypoperfusion, necessitating close monitoring to prevent the deterioration of clinical symptoms. Incorporating stroke volume and NIHSS scores into the prediction model enhances its performance. Future studies of a larger scale are required to validate these findings.
Collapse
Affiliation(s)
- Chia-Peng Chang
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan; Department of Nursing, Chang Gung University of Science and Technology, Chiayi Campus, Chiayi, Taiwan; In-service Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University
| | - Yen-Chu Huang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurology, Chang Gung Memorial Hospital, Chiayi, Taiwan.
| | - Yuan-Hsiung Tsai
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Leng-Chieh Lin
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.
| | - Jen-Tsung Yang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Chiayi, Taiwan.
| | - Kai-Hsiang Wu
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan; Department of Nursing, Chang Gung University of Science and Technology, Chiayi Campus, Chiayi, Taiwan
| | - Po-Han Wu
- Department of Emergency Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Syu-Jyun Peng
- In-service Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University; Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
2
|
Zhou C, Zhang YF, Yang ZJ, Huang YQ, Da MX. Computed tomography-based deep learning radiomics model for preoperative prediction of tumor immune microenvironment in colorectal cancer. World J Gastrointest Oncol 2025; 17:106103. [DOI: 10.4251/wjgo.v17.i5.106103] [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: 02/15/2025] [Revised: 03/08/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading cause of cancer-related death globally, with the tumor immune microenvironment (TIME) influencing prognosis and immunotherapy response. Current TIME evaluation relies on invasive biopsies, limiting its clinical application. This study hypothesized that computed tomography (CT)-based deep learning (DL) radiomics models can non-invasively predict key TIME biomarkers: Tumor-stroma ratio (TSR), tumor-infiltrating lymphocytes (TILs), and immune score (IS).
AIM To develop a non-invasive DL approach using preoperative CT radiomics to evaluate TIME components in CRC patients.
METHODS In this retrospective study, preoperative CT images of 315 pathologically confirmed CRC patients (220 in training cohort and 95 in validation cohort) were analyzed. Manually delineated regions of interest were used to extract DL features. Predictive models (DenseNet-121/169) for TSR, TILs, IS, and TIME classification were constructed. Performance was evaluated via receiver operating characteristic curves, calibration curves, and decision curve analysis (DCA).
RESULTS The DL-DenseNet-169 model achieved area under the curve (AUC) values of 0.892 [95% confidence interval (CI): 0.828-0.957] for TSR and 0.772 (95%CI: 0.674-0.870) for TIME score. The DenseNet-121 model yielded AUC values of 0.851 (95%CI: 0.768-0.933) for TILs and 0.852 (95%CI: 0.775-0.928) for IS. Calibration curves demonstrated strong prediction-observation agreement, and DCA confirmed clinical utility across threshold probabilities (P < 0.05 for all models).
CONCLUSION CT-based DL radiomics provides a reliable non-invasive method for preoperative TIME evaluation, enabling personalized immunotherapy strategies in CRC management.
Collapse
Affiliation(s)
- Chuan Zhou
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Yun-Feng Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Zhi-Jun Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Yu-Qian Huang
- Center of Medical Cosmetology, Chengdu Second People’s Hospital, Chengdu 610017, Sichuan Province, China
| | - Ming-Xu Da
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
3
|
Stawarska A, Bamburowicz-Klimkowska M, Bystrzejewski M, Kasprzak A, Grudzinski IP. Carbon-Encapsulated Iron Nanoparticles Seeking Integrins in Murine Glioma. Int J Nanomedicine 2025; 20:5475-5488. [PMID: 40321805 PMCID: PMC12048782 DOI: 10.2147/ijn.s511286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose Targeting integrin receptors for MRI represents a novel method in diagnosing glioblastoma. In the present study carbon-encapsulated iron nanoparticles to explore murine glioma tracking based upon specific direct targeting with monoclonal antibodies against the beta-3 subunit (CD61) of the integrin αVβ3 receptor are described. Methods The carbon arc discharge method was used to synthesize nanoparticles and amidation-type reaction were applied to attach monoclonal antibody (anti-CD61) with acidic group functionalized nanoparticles to lead two types of bioconjugates (Fe@C-CONH-anti-CD61 and Fe@C-(CH2)2-CONH-anti-CD61). The as-synthesized bioconjugates were tested on murine glioma cells (GL261) using MTT, LDH and calcein AM/propidium iodide assays. Relaxometry measurements were performed with a 1.5 T (63 MHz) MRI scanner using both GL261 cells and C57BL/6 mice bearing GL261 tumors. Results The results showed that Fe@C-CONH-anti-CD61 and Fe@C-(CH2)2-CONH-anti-CD61 nanoparticles have higher binding affinity towards GL261 cells compared to pristine nanoparticles without antibodies. Studies evidenced that the antibody-decorated nanoparticles did not produce any severe cytotoxic effects on murine glioma cells. Preclinical MRI studies demonstrated that the Fe@C-(CH2)2-CONH-anti-CD61 nanoparticle-based construct specifically targeted murine glioma in animals. Conclusion The carbon-encapsulated iron nanoparticles functionalized with monoclonal antibodies recognizing the beta-3 subunit of the integrin αVβ3 receptor can be considered as a potential contrast agent for MRI-based tracking glioblastoma.
Collapse
Affiliation(s)
- Agnieszka Stawarska
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | | | - Michał Bystrzejewski
- Department of Physical Chemistry, Faculty of Chemistry, Warsaw University, Warsaw, Poland
| | - Artur Kasprzak
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Ireneusz P Grudzinski
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
4
|
Xu J, Gao S, Zhu Q, Dai F, Sun C, Lee W, Ye Y, Deng G, Huang Z, Li X, Li J, Cheong S, Huang Q, Di J. Machine learning-based multiparametric CT radiomics for predicting microvascular invasion before nephrectomy in clear cell renal cell carcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04956-2. [PMID: 40249552 DOI: 10.1007/s00261-025-04956-2] [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: 09/10/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
PURPOSE This study aimed to investigate the value of integrating computed tomography (CT)-based tumor radiomics features with clinical parameters for preoperative prediction of microvascular invasion (MVI) in clear cell renal cell carcinoma (ccRCC). METHODS We retrospectively analyzed data from a single-center cohort of ccRCC patients. Radiomics features were extracted from preoperative multiphasic CT scans (unenhanced, corticomedullary, and nephrographic phases). Following dimensionality reduction and feature selection, eight machine learning algorithms were evaluated to identify the optimal radiomics model. Independent clinical predictors were determined through univariate and multivariate analyses. A nomogram integrating the radiomics signature (rad-score) with significant clinical parameters was subsequently developed. Model performance was assessed using the area under the curve (AUC), decision curve analysis (DCA), and calibration curve analysis (CAC). RESULTS Of 143 initially enrolled patients, 110 met inclusion criteria after screening, with 5502 radiomics features extracted. The support vector classifier (SVM) model demonstrated the highest discriminative ability, achieving mean AUCs of 0.976 (training cohort) and 0.892 (test cohort), significantly outperforming the clinical model (training AUC = 0.935, test AUC = 0.933). The nomogram showed superior diagnostic performance, with AUCs of 0.958 (test). DCA and CAC confirmed its clinical utility and robustness. CONCLUSIONS Multiparametric CT radiomics models enable non-invasive prediction of MVI status in ccRCC, with the SVM-based algorithm showing optimal performance. The integrated nomogram provides excellent and consistent diagnostic accuracy, offering a valuable preoperative tool for clinical decision-making.
Collapse
Affiliation(s)
- Jinbin Xu
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuntian Gao
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qin Zhu
- First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fuyang Dai
- Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ciming Sun
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Weijen Lee
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuedian Ye
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Gengguo Deng
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhansen Huang
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoming Li
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiang Li
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Samun Cheong
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qunxiong Huang
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Jinming Di
- Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
5
|
Wang Y, Han Q, Wen B, Yang B, Zhang C, Song Y, Zhang L, Xian J. Development and validation of a prediction model for malignant sinonasal tumors based on MR radiomics and machine learning. Eur Radiol 2025; 35:2074-2083. [PMID: 39210161 DOI: 10.1007/s00330-024-11033-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/23/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVES This study aimed to utilize MR radiomics-based machine learning classifiers on a large-sample, multicenter dataset to develop an optimal model for predicting malignant sinonasal tumors and tumor-like lesions. METHODS This study included 1711 adult patients (875 benign and 836 malignant) with sinonasal tumors or tumor-like lesions from three institutions. Patients from institution 1 (n = 1367) constituted both the training and validation cohorts, while those from institution 2 and 3 (n = 158/186) made up the test cohorts. Manual segmentation of the region of interest of the tumor was performed on T1WI, T2WI, and contrast-enhanced T1WI (CE-T1WI). Data normalization, dimensional reductions, feature selection, and classifications were performed using ten machine-learning classifiers. Four fusion models, namely T1WI + T2WI, T1WI + CE-T1WI, T2WI + CE-T1WI, and T1WI + T2WI + CE-T1WI, were constructed using the top ten features with the highest contribution in feature selection in the optimal models of T1WI, T2WI, and CE-T1WI. The Delong test compared areas under the curve (AUC) between models. RESULTS The AUCs of training/validation/test1/test2 datasets for T1WI, T2WI, and CE-T1WI were 0.900/0.842/0.872/0.839, 0.876/0.789/0.842/0.863, and 0.899/0.824/0.831/0.707, respectively. The fusion model from T1WI + T2WI + CE-T1WI had the highest AUC. The AUCs of training/validation/test1/test2 datasets were 0.947/0.849/0.871/0.887. The T1WI + T2WI + CE-T1WI model demonstrated a significantly higher AUC than the T2WI + CE-T1WI model in both cohorts (p < 0.05) and outperformed the T2WI model in test 1 (p = 0.008) and the T1WI model in test 2 (p = 0.006). CONCLUSIONS This fusion model based on radiomics from T1WI + T2WI + CE-T1WI images and machine learning can improve the power in predicting malignant sinonasal tumors with high accuracy, resilience, and robustness. CLINICAL RELEVANCE STATEMENT Our study proposes a radiomics-based machine learning fusion model from T1- and T2-weighted images and contrast-enhanced T1-weighted images, which can non-invasively identify the nature of sinonasal tumors and improve the performance in predicting malignant sinonasal tumors. KEY POINTS Differentiating benign and malignant sinonasal tumors is difficult due to similar clinical presentations. A radiomics model from T1 + T2 + contrast-enhanced T1 images can identify the nature of sinonasal tumors. This model can help distinguish benign and malignant sinonasal tumors.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qinghe Han
- Department of Radiology, The Second Hospital of Jilin University, Changchun, China
| | - Baohong Wen
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bingbing Yang
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Chen Zhang
- MR Research Collaboration Team, Siemens Healthcare, Beijing, China
| | - Yang Song
- MR Research Collaboration Team, Siemens Healthcare, Beijing, China
| | - Luo Zhang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otorhinolaryngology, Beijing, China.
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China.
- Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Junfang Xian
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Deng Z, Jin D, Huang P, Wang C, Deng Y, Xu R, Fan B. Predictive models of epidermal growth factor receptor mutation in lung adenocarcinoma using PET/CT-based radiomics features. Med Phys 2025. [PMID: 40165657 DOI: 10.1002/mp.17780] [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: 04/19/2024] [Revised: 02/10/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma (LAC) comprises a substantial subset of non-small cell lung cancer (NSCLC) diagnoses, where epidermal growth factor receptor (EGFR) mutations play a pivotal role as indicators for therapeutic intervention with targeted agents. The emerging field of radiomics, which involves the extraction of numerous quantitative attributes from medical imaging, when coupled with positron emission tomography/ computed tomography (PET/CT) technology, has demonstrated promise in the prognostication of EGFR mutation status. The objective of this investigation is to construct and validate predictive models for EGFR mutations in LAC by leveraging PET/CT-derived radiomics features, thereby refining diagnostic precision and facilitating tailored treatment strategies. PURPOSE The aim of this study was to develop a non-invasive radiomics model based on PET/CT with excellent performance for predicting the EGFR mutation status in LAC. Thus, it can provide the basis for the individualized treatment decision of patients. METHODS Positron emission tomography (PET), computed tomography (CT), clinical and pathological data of 112 patients with LAC admitted to our hospital from January 2019 to June 2023 were retrospectively analyzed. This research cohort encompassed 54 LAC patients with EGFR wild type and 58 LAC patients with EGFR mutated type. The participants were randomly assigned to the training group (n = 78) and the validation group (n = 34) in a 7:3 ratio. A sum of 3562 radiomics attributes were derived from PET/CT scans. The minimal absolute shrinkage and selection operator method was employed to identify 13 notable features. Based on these characteristics, support vector machine (SVM), gradient boosting decision tree (GBDT), random forest (RF) and extreme gradient boosting (XGBOOST) were constructed. The forecasting effectiveness of the model was assessed using the area under the receiver operating characteristic (ROC) Curve, the DeLong test, and decision curve analysis (DCA). RESULTS SVM performance in PET/CT radiomics model was higher than that of other machine learning models (training group areas under the curve [AUC] of 0.916 and validation group AUC of 0.945, respectively). The integration of radiomics and clinical data did not yield a superior predictive performance compared to the radiomics model alone in terms of estimating EGFR mutation status (AUC: 0.916 vs. 0.921, 0.945 vs. 0.955, p> 0.05, in both the training and validation groups). CONCLUSIONS The SVM model has emerged as a commendable non-invasive technique, showing high precision and dependability in forecasting EGFR mutation statuses in individuals with LAC. The radiomics model derived from PET/CT scans holds promise as a prognostic indicator of EGFR mutations in LAC, offering a valuable tool that could refine personalized therapeutic strategies and ultimately enhance the prognosis for LAC patients.
Collapse
Affiliation(s)
- Zhikang Deng
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, China
| | - Di Jin
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Pei Huang
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Changchun Wang
- Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | | | - Rong Xu
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Bing Fan
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
7
|
Wang Y, Hu Z, Wang H. The clinical implications and interpretability of computational medical imaging (radiomics) in brain tumors. Insights Imaging 2025; 16:77. [PMID: 40159380 PMCID: PMC11955438 DOI: 10.1186/s13244-025-01950-6] [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: 10/17/2024] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
Radiomics has widespread applications in the field of brain tumor research. However, radiomic analyses often function as a 'black box' due to their use of complex algorithms, which hinders the translation of brain tumor radiomics into clinical applications. In this review, we will elaborate extensively on the application of radiomics in brain tumors. Additionally, we will address the interpretability of handcrafted-feature radiomics and deep learning-based radiomics by integrating biological domain knowledge of brain tumors with interpretability methods. Furthermore, we will discuss the current challenges and prospects concerning the interpretability of brain tumor radiomics. Enhancing the interpretability of radiomics may make it more understandable for physicians, ultimately facilitating its translation into clinical practice. CRITICAL RELEVANCE STATEMENT: The interpretability of brain tumor radiomics empowers neuro-oncologists to make well-informed decisions from radiomic models. KEY POINTS: Radiomics makes a significant impact on the management of brain tumors in several key clinical areas. Transparent models, habitat analysis, and feature attribute explanations can enhance the interpretability of traditional handcrafted-feature radiomics in brain tumors. Various interpretability methods have been applied to explain deep learning-based models; however, there is a lack of biological mechanisms underlying these models.
Collapse
Affiliation(s)
- Yixin Wang
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
| | - Zongtao Hu
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China
| | - Hongzhi Wang
- Department of Brain Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, P. R. China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P. R. China.
| |
Collapse
|
8
|
Wang L, Zhang P, Feng Y, Lv W, Min X, Liu Z, Li J, Feng Z. Identification of testicular cancer with T2-weighted MRI-based radiomics and automatic machine learning. BMC Cancer 2025; 25:563. [PMID: 40155850 PMCID: PMC11951623 DOI: 10.1186/s12885-025-13844-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 02/28/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Distinguishing between benign and malignant testicular lesions on clinical magnetic resonance imaging (MRI) is crucial for guiding treatment planning. However, conventional MRI-based radiomics to identify testicular cancer requires expert machine learning knowledge. This study aims to investigate the potential of utilizing automatic machine learning (AutoML) based on MRI to diagnose testicular lesions without the need for expert algorithm optimization. METHODS Retrospective preoperative MRI scans from 115 patients diagnosed with testicular disease through pathology were obtained. A total of 1781 radiomics features were extracted from each lesion on the T2-weighted images. Intraclass and interclass correlation coefficients were used to evaluate the intra-observer and interobserver agreements for each radiomics feature. We developed an AutoML method based on the tree-based pipeline optimization tool (TPOT) algorithm to construct a discriminant model. The best pipeline was determined through 100 repeated operations using a 5-fold cross-validation algorithm in TPOT. The model was evaluated for accuracy, sensitivity, and specificity using the area under the curve (AUC) value of the receiver operating characteristic (ROC) curve. Shapley Additive exPlanations were used to illustrate the optimization results. RESULTS Utilizing the TPOT method, 100 diagnostic models were developed to identify testicular lesions. The best model was determined based on the highest AUC in the training cohort. The prediction model yielded AUC values of 0.989 (95% confidence interval [CI]: 0.985-0.993) and 0.909 (95% CI: 0.893-0.923) in the training and testing cohorts, respectively. CONCLUSIONS AutoML, based on the TPOT algorithm, holds potential as a noninvasive method for effectively discriminating between benign and malignant testicular lesions.
Collapse
Affiliation(s)
- Liang Wang
- Computer Center, Tongji Hospital, Tongji Medical College, Uazhong University of Science and Technology, Wuhan, China
| | - PeiPei Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanhui Feng
- Computer Center, Tongji Hospital, Tongji Medical College, Uazhong University of Science and Technology, Wuhan, China
- School of Medicine and Health Management, Huazhong University of Science and Technology, Wuhan, China
| | - Wenzhi Lv
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Britton Chance Center, MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
| | - Xiangde Min
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyong Liu
- School of Medicine and Health Management, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Li
- Computer Center, Tongji Hospital, Tongji Medical College, Uazhong University of Science and Technology, Wuhan, China.
| | - Zhaoyan Feng
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
9
|
Shan Y, Xue Y, Zhu J, Vande Vyvere T, Pisică D, Maas A, Zhang S, Gao G. Development and validation of intracranial hypertension prediction models based on radiomic features in patients with traumatic brain injury: an exploratory study based on CENTER-TBI data. Crit Care 2025; 29:100. [PMID: 40050978 PMCID: PMC11887306 DOI: 10.1186/s13054-025-05328-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Head computed tomography (CT) is a routinely performed examination to assess the intracranial condition of patients with traumatic brain injury (TBI), and radiological findings can help to indicate the presence of intracranial hypertension. At present, the prediction of intracranial hypertension is mainly based on manual discrimination of imaging characteristics. The aim of our study was to establish a model to predict intracranial hypertension via fully automatic CT image segmentation, rigorous radiomic feature extraction and reliable model development and validation. METHODS Patients admitted to the intensive care unit (ICU) who underwent intracranial pressure (ICP) monitoring were included in our study. For the development cohort, we extracted data from the CENTER-TBI database and randomly divided the data into a training group and a test group. For the validation cohort, we extracted data from patients admitted to the Shanghai General Hospital. Patients whose initial recorded ICP value was greater than or equal to 20 mmHg were defined as having intracranial hypertension. Radiological features, including imaging characteristics and three categories of radiomic features, were extracted from the head CT. Feature selection was performed for all radiological findings. A morphological model was built on the basis of selected imaging characteristics. First-order, second-order and third-order models were built on the basis of selected radiomic features. A comprehensive model was built on the basis of all selected radiological findings. The performances of these five models were assessed by four classifiers, including logistic regression (LR), random forest (RF), multilayer perceptron (MLP), and extreme gradient boosting (XGB), from which the best classifier was selected. After the process of model training and external validation, we ultimately used the optimal classifier to generate a prediction model with greater predictive power and stability. RESULTS Five models were built, including a morphological model, first-order model, second-order model, third-order model and comprehensive model. The optimal classifier was the logistic regression (LR) classifier, with which the morphological, first-order, second-order, third-order and comprehensive models had AUCs of 0.75, 0.77, 0.76, 0.86, and 0.83 and F1 scores of 0.54, 0.73, 0.63, 0.72, and 0.75, respectively, in the external validation group. CONCLUSIONS We successfully established a model for predicting intracranial hypertension on the basis of radiomic features. This model may serve as an approach for intracranial hypertension prediction in TBI patients.
Collapse
Affiliation(s)
- Yingchi Shan
- Department of Neurosurgery, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yajun Xue
- Department of Neurosurgery, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Zhu
- Department of Neurosurgery, Zhongda Hospital, Southeast University, Nanjing, China
| | - Thijs Vande Vyvere
- Department of Radiology, Antwerp University Hospital, Antwerp, Belgium
- Department of Molecular Imaging and Radiology (MIRA), Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
| | - Dana Pisică
- Department of Neurosurgery, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Public Health, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andrew Maas
- Department of Neurosurgery, Antwerp University Hospital, Edegem, Belgium
- University of Antwerp, Edegem, Belgium
| | - Shuo Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Guoyi Gao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
10
|
Wu Y, Xu D, Zha Z, Gu L, Chen J, Fang J, Dou Z, Zhang P, Zhang C, Wang J. Integrating radiomics into predictive models for low nuclear grade DCIS using machine learning. Sci Rep 2025; 15:7505. [PMID: 40033061 PMCID: PMC11876686 DOI: 10.1038/s41598-025-92080-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
Predicting low nuclear grade DCIS before surgery can improve treatment choices and patient care, thereby reducing unnecessary treatment. Due to the high heterogeneity of DCIS and the limitations of biopsies in fully characterizing tumors, current diagnostic methods relying on invasive biopsies face challenges. Here, we developed an ensemble machine learning model to assist in the preoperative diagnosis of low nuclear grade DCIS. We integrated preoperative clinical data, ultrasound images, mammography images, and Radiomic scores from 241 DCIS cases. The ensemble model, based on Elastic Net, Generalized Linear Models with Boosting (glmboost), and Ranger, improved the ability to predict low nuclear grade DCIS preoperatively, achieving an AUC of 0.92 on the validation set, outperforming the model using clinical data alone. The comprehensive model also demonstrated notable enhancements in integrated discrimination improvement and net reclassification improvement (p < 0.001). Furthermore, the Radiomic ensemble model effectively stratified DCIS patients by risk based on disease-free survival. Our findings emphasize the importance of integrating Radiomic into DCIS prediction models, offering fresh perspectives for personalized treatment and clinical management of DCIS.
Collapse
Affiliation(s)
- Yimin Wu
- Department of Ultrasound, WuHu Hospital, East China Normal University (The Second People's Hospital, WuHu), No.6 Duchun Road, Jinghu District, Wuhu, 241000, Anhui, China
| | - Daojing Xu
- Department of Ultrasound, WuHu Hospital, East China Normal University (The Second People's Hospital, WuHu), No.6 Duchun Road, Jinghu District, Wuhu, 241000, Anhui, China
| | - Zongyu Zha
- Department of Ultrasound, WuHu Hospital, East China Normal University (The Second People's Hospital, WuHu), No.6 Duchun Road, Jinghu District, Wuhu, 241000, Anhui, China
| | - Li Gu
- Department of Ultrasound, WuHu Hospital, East China Normal University (The Second People's Hospital, WuHu), No.6 Duchun Road, Jinghu District, Wuhu, 241000, Anhui, China
| | - Jieqing Chen
- Department of Ultrasound, WuHu Hospital, East China Normal University (The Second People's Hospital, WuHu), No.6 Duchun Road, Jinghu District, Wuhu, 241000, Anhui, China
| | - Jiagui Fang
- Department of Ultrasound, WuHu Hospital, East China Normal University (The Second People's Hospital, WuHu), No.6 Duchun Road, Jinghu District, Wuhu, 241000, Anhui, China
| | - Ziyang Dou
- Department of Ultrasound, WuHu Hospital, East China Normal University (The Second People's Hospital, WuHu), No.6 Duchun Road, Jinghu District, Wuhu, 241000, Anhui, China
| | - Pingyang Zhang
- Department of Echocardiography, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210006, Jiangsu, China.
| | - Chaoxue Zhang
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China.
| | - Junli Wang
- Department of Ultrasound, WuHu Hospital, East China Normal University (The Second People's Hospital, WuHu), No.6 Duchun Road, Jinghu District, Wuhu, 241000, Anhui, China.
| |
Collapse
|
11
|
Yu M, Liu J, Zhou W, Gu X, Yu S. MRI radiomics based on machine learning in high-grade gliomas as a promising tool for prediction of CD44 expression and overall survival. Sci Rep 2025; 15:7433. [PMID: 40032983 PMCID: PMC11876340 DOI: 10.1038/s41598-025-90128-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
We aimed to predict CD44 expression and assess its prognostic significance in patients with high-grade gliomas (HGG) using non-invasive radiomics models based on machine learning. Enhanced magnetic resonance imaging, along with the corresponding gene expression and clinicopathological data, was downloaded from online database. Kaplan-Meier survival curves, univariate and multivariate COX analyses, and time-dependent receiver operating characteristic were used to assess the prognostic value of CD44. Following the screening of radiomic features using repeat least absolute shrinkage and selection operator, two radiomics models were constructed utilizing logistic regression and support vector machine for validation purposes. The results indicated that CD44 protein levels were higher in HGG compared to normal brain tissues, and CD44 expression emerged as an independent biomarker of diminished overall survival (OS) in patients with HGG. Moreover, two predictive models based on seven radiomic features were built to predict CD44 expression levels in HGG, achieving areas under the curves (AUC) of 0.809 and 0.806, respectively. Calibration and decision curve analysis validated the fitness of the models. Notably, patients with high radiomic scores presented worse OS (p < 0.001). In summary, our results indicated that the radiomics models effectively differentiate CD44 expression level and OS in patients with HGG.
Collapse
Affiliation(s)
- Mingjun Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
- Department of Medical Affairs, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Jinliang Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Wen Zhou
- Department of Pain Management, Dalian Municipal Central Hospital, Dalian, 116033, People's Republic of China
| | - Xiao Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
| | - Shijia Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
12
|
Wang Z, Wang L, Wang Y. Radiomics in glioma: emerging trends and challenges. Ann Clin Transl Neurol 2025; 12:460-477. [PMID: 39901654 PMCID: PMC11920724 DOI: 10.1002/acn3.52306] [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: 10/17/2024] [Revised: 12/11/2024] [Accepted: 12/31/2024] [Indexed: 02/05/2025] Open
Abstract
Radiomics is a promising neuroimaging technique for extracting and analyzing quantitative glioma features. This review discusses the application, emerging trends, and challenges associated with using radiomics in glioma. Integrating deep learning algorithms enhances various radiomics components, including image normalization, region of interest segmentation, feature extraction, feature selection, and model construction and can potentially improve model accuracy and performance. Moreover, investigating specific tumor habitats of glioblastomas aids in a better understanding of glioblastoma aggressiveness and the development of effective treatment strategies. Additionally, advanced imaging techniques, such as diffusion-weighted imaging, perfusion-weighted imaging, magnetic resonance spectroscopy, magnetic resonance fingerprinting, functional MRI, and positron emission tomography, can provide supplementary information for tumor characterization and classification. Furthermore, radiomics analysis helps understand the glioma immune microenvironment by predicting immune-related biomarkers and characterizing immune responses within tumors. Integrating multi-omics data, such as genomics, transcriptomics, proteomics, and pathomics, with radiomics, aids the understanding of the biological significance of the underlying radiomics features and improves the prediction of genetic mutations, prognosis, and treatment response in patients with glioma. Addressing challenges, such as model reproducibility, model generalizability, model interpretability, and multi-omics data integration, is crucial for the clinical translation of radiomics in glioma.
Collapse
Affiliation(s)
- Zihan Wang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Lei Wang
- Department of NeurosurgeryGuiqian International General HospitalGuiyangGuizhouChina
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
13
|
Zhong Z, Yu HF, Tong Y, Li J. Development and Validation of a Non-Invasive Prediction Model for Glioma-Associated Epilepsy: A Comparative Analysis of Nomogram and Decision Tree. Int J Gen Med 2025; 18:1111-1125. [PMID: 40026809 PMCID: PMC11872099 DOI: 10.2147/ijgm.s512814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/15/2025] [Indexed: 03/05/2025] Open
Abstract
Objective Glioma-associated epilepsy (GAE) is a common neurological symptom in glioma patients, which can worsen the condition and increase the risk of death on the basis of primary injury. Given this, accurate prediction of GAE is crucial, and this study aims to develop and validate a GAE warning recognition prediction model. Methods We retrospectively collected MRI scan imaging data and urine samples from 566 glioma patients at the Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science from August 2016 to December 2023. Least Absolute Shrinkage and Selection Operator (LASSO) regression and multivariate logistic regression analysis are used to determine independent risk factors for GAE. The nomogram and decision tree GAE visualization prediction model were constructed based on independent risk factors. The discrimination, calibration, and clinical usefulness of GAE prediction models were evaluated through receiver operating characteristic (ROC) curve, calibration curve, and decision curve analysis (DCA), respectively. Results In the training and validation datasets, the incidence of GAE was 34.50% and 33.00%, respectively. Nomogram and decision tree were composed of five independent radiomic predictors and three differential protein molecules derived from urine. The discrimination rate of area under the curve (AUC) was 0.897 (95% CI: 0.840-0.954), slightly decreased in the validation data set, reaching 0.874 (95% CI: 8.817-0.931). The calibration curve showed a high degree of consistency between the predicted GAE probability and the actual probability. In addition, DCA analysis showed that in machine learning prediction models, decision trees have higher overall net returns within the threshold probability range. Conclusion We have introduced a machine learning prediction model for GAE detection in glioma patients based on multiomics data. This model can improve the prognosis of GAE by providing early warnings and actionable feedback and prevent or reduce pathological damage and neurobiochemical changes by implementing early interventions.
Collapse
Affiliation(s)
- Zian Zhong
- Department of Neurology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| | - Hong-Fei Yu
- Department of Neurology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| | - Yanfei Tong
- Department of Neurology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| | - Jie Li
- Department of Neurology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| |
Collapse
|
14
|
Deng X, Li P, Tian K, Zhang F, Yan Y, Fan Y, Wu Z, Zhang J, Du J, Chen W, Wang L. Radiogenomic method combining DNA methylation profiles and magnetic resonance imaging radiomics predicts patient prognosis in skull base chordoma. Clin Epigenetics 2025; 17:23. [PMID: 39962547 PMCID: PMC11831810 DOI: 10.1186/s13148-025-01836-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Chordoma is a rare malignant bone tumor exhibiting poor survival and prognosis. Hence, it is crucial to develop a convenient and effective prognostic classification method for the rehabilitation and management of patients with chordoma. In this study, we combined DNA methylation profiles and magnetic resonance imaging (MRI) images to generate a radiogenomic signature to assess its effectiveness for prognosis classification in patients with skull base chordoma. RESULTS DNA methylation profiles from chordoma tissue samples of 40 patients were factorized into eight DNA methylation signatures. Among them, Signature 4 was identified as the prognosis-specific signature. Based on the Signature 4 loading values, the patients were categorized into low-signature (LLG) and high-signature (HLG) loading groups. HLG patients had higher progression-free survival times than LLG patients. Combined analysis with external single-cell RNA-seq data revealed higher tumor cell proportions and lower natural killer cell proportions in the HLG than in the LLG. Additionally, 2,553 radiomic features were extracted from T1, T2, and enhanced T1 MRI images of the patients, and a radiogenomic signature comprising 14 radiomic features was developed. In a validation cohort of 122 patients, the radiogenomic signature successfully distinguished between the two groups (P = 0.027). Furthermore, the existence of Signature 4 was confirmed in an additional dataset of 14 patients. CONCLUSION We developed a prognostic radiogenomic signature using a radiogenomic classification method, which leverages MRI images to extract features that reflect the DNA methylation signature associated with prognosis, enabling the stratification of patients based on their prognostic risk. This method offers the advantages of being noninvasive and convenient.
Collapse
Affiliation(s)
- Xiaoyu Deng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Engineering Medicine, Beihang University, Beijing, China
| | - Peiran Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kaibing Tian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fan Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Engineering Medicine, Beihang University, Beijing, China
| | - Yumeng Yan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Engineering Medicine, Beihang University, Beijing, China
| | - Yanghua Fan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Junting Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiang Du
- Beijing Neurosurgical Institute Capital Medical University, Beijing, China.
| | - Wei Chen
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Engineering Medicine, Beihang University, Beijing, China.
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
15
|
Shen Y, Wu S, Wu Y, Cui C, Li H, Yang S, Liu X, Chen X, Huang C, Wang X. Radiomics model building from multiparametric MRI to predict Ki-67 expression in patients with primary central nervous system lymphomas: a multicenter study. BMC Med Imaging 2025; 25:54. [PMID: 39962371 PMCID: PMC11834475 DOI: 10.1186/s12880-025-01585-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVES To examine the correlation of apparent diffusion coefficient (ADC), diffusion weighted imaging (DWI), and T1 contrast enhanced (T1-CE) with Ki-67 in primary central nervous system lymphomas (PCNSL). And to assess the diagnostic performance of MRI radiomics-based machine-learning algorithms in differentiating the high proliferation and low proliferation groups of PCNSL. METHODS 83 patients with PCNSL were included in this retrospective study. ADC, DWI and T1-CE sequences were collected and their correlation with Ki-67 was examined using Spearman's correlation analysis. The Kaplan-Meier method and log-rank test were used to compare the survival rates of the high proliferation and low proliferation groups. The radiomics features were extracted respectively, and the features were screened by machine learning algorithm and statistical method. Radiomics models of seven different sequence permutations were constructed. The area under the receiver operating characteristic curve (ROC AUC) was used to evaluate the predictive performance of all models. DeLong test was utilized to compare the differences of models. RESULTS Relative mean apparent diffusion coefficient (rADCmean) (ρ=-0.354, p = 0.019), relative mean diffusion weighted imaging (rDWImean) (b = 1000) (ρ = 0.273, p = 0.013) and relative mean T1 contrast enhancement (rT1-CEmean) (ρ = 0.385, p = 0.001) was significantly correlated with Ki-67. Interobserver agreements between the two radiologists were almost perfect for all parameters (rADCmean ICC = 0.978, 95%CI 0.966-0.986; rDWImean (b = 1000) ICC = 0.931, 95% CI 0.895-0.955; rT1-CEmean ICC = 0.969, 95% CI 0.953-0.980). The differences in PFS (p = 0.016) and OS (p = 0.014) between the low and high proliferation groups were statistically significant. The best prediction model in our study used a combination of ADC, DWI, and T1-CE achieving the highest AUC of 0.869, while the second ranked model used ADC and DWI, achieving an AUC of 0.828. CONCLUSION rDWImean, rADCmean and rT1-CEmean were correlated with Ki-67. The radiomics model based on MRI sequences combined is promising to distinguish low proliferation PCNSL from high proliferation PCNSL.
Collapse
Affiliation(s)
- Yelong Shen
- Department of Radiology, Shandong Provincial Hospital, No. 324, Jingwu Road, Jinan, 250021, Shandong, China
- Department of Radiology, Shandong University, No. 44, West Wenhua Road, Jinan, 250021, Shandong, China
| | - Siyu Wu
- Department of Radiology, Shandong Provincial Hospital, No. 324, Jingwu Road, Jinan, 250021, Shandong, China
- Department of Radiology, Shandong University, No. 44, West Wenhua Road, Jinan, 250021, Shandong, China
| | - Yanan Wu
- Department of Radiology, Shandong Provincial Hospital, No. 324, Jingwu Road, Jinan, 250021, Shandong, China
| | - Chao Cui
- Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, Shandong, China
| | - Haiou Li
- Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Shuang Yang
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University& Shandong Provincial Qianfoshan Hospital, Jinan, 250021, Shandong, China
| | - Xuejun Liu
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Xingzhi Chen
- Department of Research Collaboration, R&D center, Beijing Deepwise & League of PHD Technology Co., Ltd, 100080, Beijing, China
| | - Chencui Huang
- Department of Research Collaboration, R&D center, Beijing Deepwise & League of PHD Technology Co., Ltd, 100080, Beijing, China
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital, No. 324, Jingwu Road, Jinan, 250021, Shandong, China.
- Department of Radiology, Shandong University, No. 44, West Wenhua Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
16
|
Bijari S, Rezaeijo SM, Sayfollahi S, Rahimnezhad A, Heydarheydari S. Development and validation of a robust MRI-based nomogram incorporating radiomics and deep features for preoperative glioma grading: a multi-center study. Quant Imaging Med Surg 2025; 15:1125-1138. [PMID: 39995745 PMCID: PMC11847178 DOI: 10.21037/qims-24-1543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/28/2024] [Indexed: 02/26/2025]
Abstract
Background Gliomas, the most common primary brain tumors, are classified into low-grade glioma (LGG) and high-grade glioma (HGG) based on aggressiveness. Accurate preoperative differentiation is vital for effective treatment and prognosis, but traditional methods like biopsy have limitations, such as sampling errors and procedural risks. This study introduces a comprehensive model that combines radiomics features (RFs) and deep features (DFs) from magnetic resonance imaging (MRI) scans, integrating clinical factors with advanced imaging features to enhance diagnostic precision for preoperative glioma grading. Methods In this retrospective multi-center study [2017-2022], 582 patients underwent preoperative contrast-enhanced T1-weighted (CE-T1w) and T2-weighted fluid-attenuated inversion recovery (T2w FLAIR) MRI. The dataset, divided into 407 training and 175 testing cases, included 340 LGGs and 242 HGGs. RFs and DFs were extracted from CE-T1w images, and radiomic scores (rad-score) and deep scores (deep-score) were calculated. Additionally, a clinical model based on demographics and MRI findings (CE-T1w and T2w FLAIR imaging) was developed. A nomogram model integrating rad-score, deep-score, and clinical factors was constructed using multivariate logistic regression analysis. Decision curve analysis (DCA) was employed to evaluate the nomogram's clinical utility in distinguishing between HGGs and LGGs. Results The study included 582 patients (mean age: 52±14 years; 57.91% male). No significant differences in age or sex were found between the training and testing groups (P>0.05). For RFs, 73.02% of the 215 extracted features were selected based on inter-class correlation coefficients (ICCs), while for DFs, 38.27% of the 15,680 extracted features were selected. Optimal penalization coefficients lambda (λ) for RFs and DFs were determined using a five-fold cross-validation and minimal criteria process. The resulting receiver operating characteristic-area under the curve (ROC-AUC) values were 0.93 [95% confidence interval (CI): 0.91-0.94] for the training set and 0.91 (95% CI: 0.89-0.93) for the testing set. The Hosmer-Lemeshow test yielded P values of 0.619 and 0.547 for the training and testing sets, respectively, indicating satisfactory calibration. The nomogram demonstrated the highest net benefit (NB) up to a threshold of 0.7, followed by DFs and RFs. Conclusions This study underscores the efficacy of integrating RFs and DFs alongside clinical data to accurately predict the pathological grading of HGGs and LGGs, offering a comprehensive approach for clinical decision-making.
Collapse
Affiliation(s)
- Salar Bijari
- Department of Radiology, Faculty of Paramedical Sciences, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Seyed Masoud Rezaeijo
- Department of Medical Physics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sahar Sayfollahi
- Department of Neurosurgery, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Rahimnezhad
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sahel Heydarheydari
- Department of Medical Imaging and Radiation Sciences, Faculty of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
17
|
Li R, Bing X, Su X, Zhang C, Sun H, Dai Z, Ouyang A. The potential value of dual-energy CT radiomics in evaluating CD8 +, CD163 + and αSMA + cells in the tumor microenvironment of clear cell renal cell carcinoma. Clin Transl Oncol 2025; 27:716-726. [PMID: 39083142 DOI: 10.1007/s12094-024-03637-8] [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: 04/29/2024] [Accepted: 07/19/2024] [Indexed: 02/01/2025]
Abstract
PURPOSE This study aims to develop radiomics models and a nomogram based on machine learning techniques, preoperative dual-energy computed tomography (DECT) images, clinical and pathological characteristics, to explore the tumor microenvironment (TME) of clear cell renal cell carcinoma (ccRCC). METHODS We retrospectively recruited of 87 patients diagnosed with ccRCC through pathological confirmation from Center I (training set, n = 69; validation set, n = 18), and collected their DECT images and clinical information. Feature selection was conducted using variance threshold, SelectKBest, and the least absolute shrinkage and selection operator (LASSO). Radiomics models were then established using 14 classifiers to predict TME cells. Subsequently, we selected the most predictive radiomics features to calculate the radiomics score (Radscore). A combined model was constructed through multivariate logistic regression analysis combining the Radscore and relevant clinical characteristics, and presented in the form of a nomogram. Additionally, 17 patients were recruited from Center II as an external validation cohort for the nomogram. The performance of the models was assessed using methods such as the area under the receiver operating characteristic curve (AUC), calibration curve, and decision curve analysis (DCA). RESULTS The validation set AUC values for the radiomics models assessing CD8+, CD163+, and αSMA+ cells were 0.875, 0.889, and 0.864, respectively. Additionally, the external validation cohort AUC value for the nomogram reaches 0.849 and shows good calibration. CONCLUSION Radiomics models could allow for non-invasive assessment of TME cells from DECT images in ccRCC patients, promising to enhance our understanding and management of the tumor.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/diagnostic imaging
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/immunology
- Kidney Neoplasms/diagnostic imaging
- Kidney Neoplasms/pathology
- Kidney Neoplasms/immunology
- Male
- Female
- Tumor Microenvironment/immunology
- Middle Aged
- Retrospective Studies
- Tomography, X-Ray Computed/methods
- Nomograms
- Aged
- CD163 Antigen
- Antigens, CD/analysis
- Antigens, Differentiation, Myelomonocytic/analysis
- Antigens, Differentiation, Myelomonocytic/metabolism
- Receptors, Cell Surface/analysis
- Adult
- Machine Learning
- Radiomics
Collapse
Affiliation(s)
- Ruobing Li
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, 105 JieFang Road, Jinan, 250013, China
- Shandong First Medical University, Jinan, 250117, China
| | - Xue Bing
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, 105 JieFang Road, Jinan, 250013, China
| | - Xinyou Su
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Chunling Zhang
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, 105 JieFang Road, Jinan, 250013, China
| | - Haitao Sun
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, 105 JieFang Road, Jinan, 250013, China
| | - Zhengjun Dai
- Scientific Research Department, Huiying Medical Technology Co, Ltd, Beijing, 100192, China
| | - Aimei Ouyang
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, 105 JieFang Road, Jinan, 250013, China.
| |
Collapse
|
18
|
Qian X, Yang L, Shi Y. Investigation of the impact of magnetic resonance imaging-assisted surgery on immune cell cytokine levels and efficacy in patients with gliomas. Curr Probl Surg 2025; 63:101640. [PMID: 39922633 DOI: 10.1016/j.cpsurg.2024.101640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 02/10/2025]
Affiliation(s)
- Xueshan Qian
- Department of CT&MRI Room, Qianjiang Central Hospital, Qianjiang, Hubei Province, China
| | - Li Yang
- Department of Radiology, Mianyang Central Hospital, Mianyang, Sichuan Province, China
| | - Yonghui Shi
- Department of Radiology, the Affiliated Hospital of Tibet Minzu University, Xianyang, Shaanxi Province, China.
| |
Collapse
|
19
|
Yan T, Yan Z, Chen G, Xu S, Wu C, Zhou Q, Wang G, Li Y, Jia M, Zhuang X, Yang J, Liu L, Wang L, Wu Q, Wang B, Yan T. Survival outcome prediction of esophageal squamous cell carcinoma patients based on radiomics and mutation signature. Cancer Imaging 2025; 25:9. [PMID: 39891186 PMCID: PMC11783911 DOI: 10.1186/s40644-024-00821-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 12/29/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND The present study aimed to develop a nomogram model for predicting overall survival (OS) in esophageal squamous cell carcinoma (ESCC) patients. METHODS A total of 205 patients with ESCC were enrolled and randomly divided into a training cohort (n = 153) and a test cohort (n = 52) at a ratio of 7:3. Multivariate Cox regression was used to construct the radiomics model based on CT data. The mutation signature was constructed based on whole genome sequencing data and found to be significantly associated with the prognosis of patients with ESCC. A nomogram model combining the Rad-score and mutation signature was constructed. An integrated nomogram model combining the Rad-score, mutation signature, and clinical factors was constructed. RESULTS A total of 8 CT features were selected for multivariate Cox regression analysis to determine whether the Rad-score was significantly correlated with OS. The area under the curve (AUC) of the radiomics model was 0.834 (95% CI, 0.767-0.900) for the training cohort and 0.733 (95% CI, 0.574-0.892) for the test cohort. The Rad-score, S3, and S6 were used to construct an integrated RM nomogram. The predictive performance of the RM nomogram model was better than that of the radiomics model, with an AUC of 0. 830 (95% CI, 0.761-0.899) in the training cohort and 0.793 (95% CI, 0.653-0.934) in the test cohort. The Rad-score, TNM stage, lymph node metastasis status, S3, and S6 were used to construct an integrated RMC nomogram. The predictive performance of the RMC nomogram model was better than that of the radiomics model and RM nomogram model, with an AUC of 0. 862 (95% CI, 0.795-0.928) in the training cohort and 0. 837 (95% CI, 0.705-0.969) in the test cohort. CONCLUSION An integrated nomogram model combining the Rad-score, mutation signature, and clinical factors can better predict the prognosis of patients with ESCC.
Collapse
Affiliation(s)
- Ting Yan
- Second Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Zhenpeng Yan
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Guohui Chen
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Songrui Xu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Chenxuan Wu
- School of Life Science, Beijing Institute of Technology, Beijing, People's Republic of China
| | - Qichao Zhou
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Guolan Wang
- School of Computer Information Engineering, Shanxi Technology and Business University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Ying Li
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, Shanxi, 030024, People's Republic of China
| | - Mengjiu Jia
- School of Computer Information Engineering, Shanxi Technology and Business University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Xiaofei Zhuang
- Department of Thoracic Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, 030013, People's Republic of China
| | - Jie Yang
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Lili Liu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Lu Wang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Qinglu Wu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Bin Wang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, Shanxi, 030024, People's Republic of China.
| | - Tianyi Yan
- School of Life Science, Beijing Institute of Technology, Beijing, People's Republic of China.
| |
Collapse
|
20
|
Wei XX, Li CY, Yang HQ, Song P, Wu BL, Zhu FH, Hu J, Xu XY, Tian X. Cardiac computer tomography-derived radiomics in assessing myocardial characteristics at the connection between the left atrial appendage and the left atrium in atrial fibrillation patients. Front Cardiovasc Med 2025; 11:1442155. [PMID: 39872879 PMCID: PMC11769956 DOI: 10.3389/fcvm.2024.1442155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
Objectives To evaluate the feasibility of utilizing cardiac computer tomography (CT) images for extracting the radiomic features of the myocardium at the junction between the left atrial appendage (LAA) and the left atrium (LA) in patients with atrial fibrillation (AF) and to evaluate its asscociation with the risk of AF. Methods A retrospective analysis was conducted on 82 cases of AF and 56 cases in the control group who underwent cardiac CT at our hospital from May 2022 to May 2023, with recorded clinical information. The morphological parameters of the LAA were measured. A radiomics model, a clincal feature model and a model combining radiomics and clinical features were constructed. The radiomics model was built by extracting radiomic features of the myocardial tissue using Pyradiomics, and employing Least absolute shrinkage and selection operator (LASSO) method for feature selection, combining random forest with support vector machine (SVM) classifier. Results There were 82 cases in the AF group [44 males, 65.00 (59, 70)], and 56 cases in the control group (21 males, 61.09 ± 7.18). Age, BMI, hypertension, CHA2DS-VASC score, neutrophil to lymphocyte ratio (NLR), LAA volume, LA volume, the myocardial thickness at the junction of LAA and LA, the area, circumference, short diameter, and long diameter of the LAA opening, were significantly different between the AF group and the control group (P < 0.05). After conducting multivariate logistic regression analysis, it was found that BMI, the myocardial thickness at the junction of the LAA and the LA, LA volume, NLR and CHA2DS-VASC score were related to AF. 12 radiomics features of the myocardium at the junction of the LAA and the LA were extracted and identified. ROC curve analysis confirmed that the nomogram based on radiomics scores and clinical factors can effectively predict AF (AUC 0.869). Conclusion Radiomics enables the extraction of the myocardial characteristics at the junction of the LAA and the LA, which are related with AF, facilitating the assessment of its relationship with the risk of AF. The combination of radiomics with clinical characteristics enhances the evaluation capabilities significantly.
Collapse
Affiliation(s)
- Xiao-Xuan Wei
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Cai-Ying Li
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hai-Qing Yang
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Peng Song
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bai-Lin Wu
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fang-Hua Zhu
- Department of Statistical Investigation, Statistical Information Center of Hebei Health Commission, Shijiazhuang, China
| | - Jing Hu
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiao-Yu Xu
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xin Tian
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
21
|
Yang Q, Ke T, Wu J, Wang Y, Li J, He Y, Yang J, Xu N, Yang B. Preoperative prediction of pituitary neuroendocrine tumor invasion using multiparametric MRI radiomics. Front Oncol 2025; 14:1475950. [PMID: 39850814 PMCID: PMC11754205 DOI: 10.3389/fonc.2024.1475950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/03/2024] [Indexed: 01/25/2025] Open
Abstract
Objective The invasiveness of pituitary neuroendocrine tumor is an important basis for formulating individualized treatment plans and improving the prognosis of patients. Radiomics can predict invasiveness preoperatively. To investigate the value of multiparameter magnetic resonance imaging (mpMRI) radiomics in predicting pituitary neuroendocrine tumor invasion into the cavernous sinus (CS) before surgery. Patients and methods The clinical data of 133 patients with pituitary neuroendocrine tumor (62 invasive and 71 non-invasive) confirmed by surgery and pathology who underwent preoperative mpMRI examination were retrospectively analyzed. Data were divided into training set and testing set according to different field strength equipment. Radiomics features were extracted from the manually delineated regions of interest in T1WI, T2WI and CE-T1, and the best radiomics features were screened by LASSO algorithm. Single radiomics model (T1WI, T2WI, CE-T1) and combined radiomics model (T1WI+T2WI+CE-T1) were constructed respectively. In addition, clinical features were screened to establish clinical model. Finally, the prediction model was evaluated by ROC curve, calibration curve and decision curve analysis (DCA). Results A total of 10 radiomics features were selected from 306 primitive features. The combined radiomics model had the highest prediction efficiency. The area under curve (AUC) of the training set was 0.885 (95% CI, 0.819-0.952), and the accuracy, sensitivity, and specificity were 0.951,0.826, and 0.725. The AUC of the testing set was 0.864 (95% CI, 0.744-0.985), and the accuracy, sensitivity, and specificity were 0.829,0.952, and 0.700. DCA showed that the combined radiomics model had higher clinical net benefit. Conclusion The combined radiomics model based on mpMRI can effectively and accurately predict the invasiveness of pituitary neuroendocrine tumor to CS preoperatively, and provide decision-making basis for clinical individualized treatment.
Collapse
Affiliation(s)
- Qiuyuan Yang
- Department of Medical Imaging, The Second People’s Hospital of Dali Prefecture, Dali, China
| | - Tengfei Ke
- Department of Medical Imaging, Yunnan Cancer Hospital, Kunming, China
| | - Jialei Wu
- Medical Imaging Center, The First Hospital of Kunming, Kunming, China
| | - Yubo Wang
- Medical Imaging Center, The First Hospital of Kunming, Kunming, China
| | - Jiageng Li
- Medical Imaging Center, The First Hospital of Kunming, Kunming, China
| | - Yimin He
- Department of Medical Imaging, The Second People’s Hospital of Dali Prefecture, Dali, China
| | - Jianxian Yang
- Department of Medical Imaging, The Second People’s Hospital of Dali Prefecture, Dali, China
| | - Nan Xu
- Department of Radiology, Air Force Medical Center, Air Force Medical University, Beijing, China
| | - Bin Yang
- Medical Imaging Center, The First Hospital of Kunming, Kunming, China
| |
Collapse
|
22
|
Chen J, Wu Q, Berglund AE, Macaulay RJ, Etame AB. Comprehensive Analysis Identifies THEMIS2 as a Potential Prognostic and Immunological Biomarker in Glioblastoma. Cells 2025; 14:66. [PMID: 39851494 PMCID: PMC11764009 DOI: 10.3390/cells14020066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Glioblastoma (GBM) is a highly aggressive brain tumor characterized by its ability to evade the immune system, hindering the efficacy of current immunotherapies. Recent research has highlighted the important role of immunosuppressive macrophages in the tumor microenvironment (TME) in driving this immune evasion. In this study, we are the first to identify THEMIS2 as a key regulator of tumor-associated macrophage (TAM)-mediated immunosuppression in GBM. We found that a high THEMIS2 expression is associated with poor patient outcomes and increased infiltration of immune cells, particularly macrophages. Functional analyses revealed THEMIS2's critical involvement in immune-related pathways, including immune response activation, mononuclear cell differentiation, and the positive regulation of cytokine production. Additionally, single-cell RNA sequencing data demonstrated that macrophages with a high THEMIS2 expression were associated with increased phagocytosis, immune suppression, and enhanced tumor growth. These findings suggest that THEMIS2 could serve as both a prognostic marker and a therapeutic target for enhancing anti-tumor immunity in GBM.
Collapse
Affiliation(s)
- Jianan Chen
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (J.C.); (Q.W.)
| | - Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (J.C.); (Q.W.)
| | - Anders E. Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA;
| | - Robert J. Macaulay
- Departments of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA;
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (J.C.); (Q.W.)
| |
Collapse
|
23
|
Wang Z, Yuan Y, Cui T, Xu B, Zou Z, Xu Q, Yang J, Su H, Xiang C, Wang X, Yang J, Chang T, Chen S, Zeng Y, Deng L, Wang H, Zhang S, Yang Y, Hu X, Chen W, Yue Q, Liu Y. Survival and immune microenvironment prediction of glioma based on MRI imaging genomics method: a retrospective observational study. Neurosurg Rev 2025; 48:18. [PMID: 39751927 DOI: 10.1007/s10143-024-03164-8] [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: 10/09/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Glioma is characterized by high heterogeneity and poor prognosis. Attempts have been made to understand its diversity in both genetic expressions and radiomic characteristics, while few integrated the two omics in predicting survival of glioma. This study was intended to investigate the connection between glioma imaging and genome, and examine its predictive value in glioma mortality risk and tumor immune microenvironment (TIME). Clinical, transcriptomics and radiomics data were obtained from public datasets and patients in our center. Correlation analysis between gene expression and radiomic feature (RF) was performed, followed by survival analysis to select RF-related genes (RFRGs) and gene expression-related RFs (GRRFs). After that, RFRGs and GRRFs were used to construct mortality risk prediction model of all glioma and isocitrate dehydrogenase (IDH) wild type (WT) glioma. The association between RFRGs and TIME was explored. Six cohorts composed of 1,754 glioma patients were included. Thirty-five genes and eighty-two RFs demonstrated high correlation with each other. Gene score based on RFRGs was independent predictor of both glioma (P < 0.05) and IDH-WT glioma (P < 0.05). Same score based on GRRFs was also able to stratify risk of both glioma (P < 0.0001) and IDH-WT glioma (P < 0.0001), with nomograms constructed separately. The TIME of gliomas predicted with RFRGs' score found mismatched risk of death with immune response. RFRGs and GRRFs were able to predict glioma mortality risk and TIME. Further studies could validate our results and explore this genome-imaging interactions.
Collapse
Affiliation(s)
- Zhihao Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yunbo Yuan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Cui
- Chengdu Science and Technology Development Center of CAEP, Chengdu, China
| | - Biao Xu
- Chengdu Science and Technology Development Center of CAEP, Chengdu, China
| | - Zhubei Zou
- Chengdu Science and Technology Development Center of CAEP, Chengdu, China
| | - Qiuyi Xu
- Chengdu Science and Technology Development Center of CAEP, Chengdu, China
| | - Jie Yang
- Chengdu Science and Technology Development Center of CAEP, Chengdu, China
| | - Hang Su
- Chengdu Science and Technology Development Center of CAEP, Chengdu, China
| | - Chaodong Xiang
- School of Medicine, Chongqing University, Chongqing, China
- 7T Magnetic Resonance Translational Medicine Research Center, Department of Radiology, Southwest Hospital, Army Medical University, Third Military Medical University), Chongqing, China
| | - Xianqi Wang
- 7T Magnetic Resonance Translational Medicine Research Center, Department of Radiology, Southwest Hospital, Army Medical University, Third Military Medical University), Chongqing, China
| | - Jing Yang
- 7T Magnetic Resonance Translational Medicine Research Center, Department of Radiology, Southwest Hospital, Army Medical University, Third Military Medical University), Chongqing, China
| | - Tao Chang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Siliang Chen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yunhui Zeng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lanqin Deng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Haoyu Wang
- Department of Neurosurgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuxin Zhang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaofei Hu
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Glioma Medicine Research Center, The First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Wei Chen
- 7T Magnetic Resonance Translational Medicine Research Center, Department of Radiology, Southwest Hospital, Army Medical University, Third Military Medical University), Chongqing, China
| | - Qiang Yue
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.
| | - Yanhui Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
24
|
Feng Q, Xiang W, Fan Y, Li J, Jing X, Ji X, Han T, Xia S. Enhancement of the nontumor component in newly diagnosed glioblastoma as a more accurate predictor of local recurrence location: a multicenter study. Quant Imaging Med Surg 2025; 15:299-313. [PMID: 39839007 PMCID: PMC11744104 DOI: 10.21037/qims-24-1319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/26/2024] [Indexed: 01/23/2025]
Abstract
Background Although the spatial heterogeneity of glioblastoma (GBM) can be clearly mapped by the habitats generated by magnetic resonance imaging (MRI), the means to accurately predicting the spatial location of local recurrence (SLLR) remains a significant challenge. The aim of this study was to identify the different degrees enhancement of GBM, including the nontumor component and tumor component, and determine their relationship with SLLR. Methods A retrospective analysis was performed from three tertiary medical centers, totaling 728 patients with 109 radiation-induced temporal lobe necrosis (TLN) of nasopharyngeal carcinoma (NPC) and 619 with GBM. The spatial location of nontumor component enhancement (SLNTE) and the spatial location of tumor component enhancement (SLTE) for the preoperative images of patients with GBM were identified using TLN as the nontumor component reference by clustering analysis, and then their relationship with the SLLR was analyzed. Decision tree models of 10-fold cross-validation based on SLNTE and SLTE built to predict the SLLR. The area under the curve (AUC) was used to evaluate the predictive efficacy of these models. Results The SLNTE had a stronger spatial relationship with SLLR than did SLTE (χ2=4.77; P=0.029). In data set 3, both the SLNTE and SLTE were associated with the SLLR (rSLNTE=0.70, P<0.001; rSLTE=0.34, P=0.005). In data set 4, the SLLR was correlated with SLNTE but not with SLTE (rSLNTE=0.59, P=0.029; rSLTE=0.20, P=0.50). In data sets 3 and 4, the SLNTE-based decision tree models predicted the SLLR with 81% and 79% accuracy, respectively, and the AUC values were greater than 0.80 and 0.75, respectively. Meanwhile, the SLTE-based decision tree models predicted the SLLR with 72% and 50% accuracy, respectively, with AUC values of 0.70 and 0.60, respectively. Conclusions Radiation-induced TLN of NPC is a highly effective reference indicator for detecting nontumor components. The tumor periphery adjacent to the nontumor component enhancement of GBM may be associated with a higher risk of local recurrence, which may provide a more accurate imaging basis for performing supertotal resection.
Collapse
Affiliation(s)
- Quanzhi Feng
- Department of Radiology, The First Central Clinical School, Tianjin Medical University, Tianjin, China
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin University, Tianjin, China
| | - Wang Xiang
- Department of Radiology, The First Central Clinical School, Tianjin Medical University, Tianjin, China
- Department of Radiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yuhan Fan
- 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, China
| | - Jing Li
- Department of Radiology, Tianjin First Central Hospital, Tianjin, China
| | - Xiyue Jing
- Institute of Neurosurgery, Tianjin Huanhu Hospital, Tianjin University, Tianjin, China
| | - Xiaodong Ji
- Department of Radiology, Medical Imaging Institute of Tianjin, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Tong Han
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin University, Tianjin, China
| | - Shuang Xia
- Department of Radiology, Medical Imaging Institute of Tianjin, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
25
|
Adachi T, Nakamura M, Iwai T, Yoshimura M, Mizowaki T. Delta-Radiomics Approach Using Contrast-Enhanced and Noncontrast-Enhanced Computed Tomography Images for Predicting Distant Metastasis in Patients With Borderline Resectable Pancreatic Carcinoma. Adv Radiat Oncol 2025; 10:101669. [PMID: 39687476 PMCID: PMC11647472 DOI: 10.1016/j.adro.2024.101669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 10/21/2024] [Indexed: 12/18/2024] Open
Abstract
Purpose To predict distant metastasis (DM) in patients with borderline resectable pancreatic carcinoma using delta-radiomics features calculated from contrast-enhanced computed tomography (CECT) and non-CECT images. Methods and Materials Among 250 patients who underwent radiation therapy at our institution between February 2013 and December 2021, 67 patients were deemed eligible. A total of 11 clinical features and 3906 radiomics features were incorporated. Radiomics features were extracted from CECT and non-CECT images, and the differences between these features were calculated, resulting in delta-radiomics features. The patients were randomly divided into the training (70%) and test (30%) data sets for model development and validation. Predictive models were developed with clinical features (clinical model), radiomics features (radiomics model), and a combination of the abovementioned features (hybrid model) using Fine-Gray regression (FG) and random survival forest (RSF). Optimal hyperparameters were determined using stratified 5-fold cross-validation. Subsequently, the developed models were applied to the remaining test data sets, and the patients were divided into high- or low-risk groups based on their risk scores. Prognostic power was assessed using the concordance index, with 95% CIs obtained through 2000 bootstrapping iterations. Statistical significance between the above groups was assessed using Gray's test. Results At a median follow-up period of 23.8 months, 47 (70.1%) patients developed DM. The concordance indices of the FG-based clinical, radiomics, and hybrid models were 0.548, 0.603, and 0.623, respectively, in the test data set, whereas those of the RSF-based models were 0.598, 0.680, and 0.727, respectively. The RSF-based model, including delta-radiomics features, significantly divided the cumulative incidence curves into two risk groups (P < .05). The feature map of the gray-level size-zone matrix showed that the difference in feature values between CECT and non-CECT images correlated with the incidence of DM. Conclusions Delta-radiomics features obtained from CECT and non-CECT images using RSF successfully predict the incidence of DM in patients with borderline resectable pancreatic carcinoma.
Collapse
Affiliation(s)
- Takanori Adachi
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Mitsuhiro Nakamura
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
- Department of Advanced Medical Physics, Graduate School of Medicine, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Takahiro Iwai
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
26
|
Chong JK, Jain P, Prasad S, Dubey NK, Saxena S, Lo WC. Optimizing Glioblastoma, IDH-wildtype Treatment Outcomes : A Radiomics and Support Vector Machine-Based Approach to Overall Survival Estimation. J Korean Neurosurg Soc 2025; 68:7-18. [PMID: 39099033 PMCID: PMC11725457 DOI: 10.3340/jkns.2024.0100] [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: 05/18/2024] [Revised: 06/20/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024] Open
Abstract
OBJECTIVE Glioblastoma multiforme (GBM), particularly the isocitrate dehydrogenase (IDH)-wildtype type, represents a significant clinical challenge due to its aggressive nature and poor prognosis. Despite advancements in medical imaging and its modalities, survival rates have not improved significantly, demanding innovative treatment planning and outcome prediction approaches. METHODS This study utilizes a support vector machine (SVM) classifier using radiomics features to predict the overall survival (OS) of GBM, IDH-wildtype patients to short (<12 months) and long (≥12 months) survivors. A dataset comprising multi-parametric magnetic resonance imaging scans from 574 patients was analyzed. Radiomic features were extracted from T1, T2, fluid-attenuated inversion recovery, and T1 with gadolinium (T1GD) sequences. Low variance features were removed, and recursive feature elimination was used to select the most informative features. The SVM model was trained using a k-fold cross-validation approach. Furthermore, clinical parameters such as age, gender, and MGMT (O6-methylguanine-DNA methyltransferase) promoter methylation status were integrated to enhance prediction accuracy. RESULTS The model showed reasonable results in terms of cross-validated area under the curve of 0.84 (95% confidence interval, 0.80-0.90) with (p<0.001) effectively categorizing patients into short and long survivors. Log-rank test (chi-square statistics) analysis for the developed model was 0.00029 along with the 1.20 Cohen's d effect size. Most importantly, clinical data integration further refined the survival estimates, providing a more fitted prediction that considers individual patient characteristics by Kaplan-Meier curve with p-value <0.0001. CONCLUSION The proposed method significantly enhances the predictive accuracy of OS outcomes in GBM, IDH-wildtype patients. By integrating detailed imaging features with key clinical indicators, this model offers a robust tool for personalized treatment planning, potentially improving OS.
Collapse
Affiliation(s)
- Jiunn-Kai Chong
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Priyanka Jain
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, India
| | - Shivani Prasad
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, India
| | - Navneet Kumar Dubey
- Executive Programme in Healthcare Management, Indian Institute of Management, Lucknow, India
| | - Sanjay Saxena
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, India
| | - Wen-Cheng Lo
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
- Division of Neurosurgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
27
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408069. [PMID: 39535476 PMCID: PMC11727298 DOI: 10.1002/advs.202408069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Tianxiang Li
- Department of UltrasoundState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical. SciencesPeking Union Medical CollegeBeijing100730China
| | - Bingxin Gong
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and EngineeringSouthern University of Science and TechnologyShenzhen518055China
| | - Sichen Wang
- School of Life Science and TechnologyComputational Biology Research CenterHarbin Institute of TechnologyHarbin150001China
| | - Lian Yang
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Chuansheng Zheng
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| |
Collapse
|
28
|
Ohata M, Fukui R, Morimitsu Y, Kobayashi D, Yamauchi T, Akagi N, Honda M, Hayashi A, Hasegawa K, Kida K, Goto S, Hiraki T. Investigating the Effects of Reconstruction Conditions on Image Quality and Radiomic Analysis in Photon-counting Computed Tomography. J Med Phys 2025; 50:100-107. [PMID: 40256177 PMCID: PMC12005666 DOI: 10.4103/jmp.jmp_114_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/11/2024] [Accepted: 01/19/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction Photon-counting computed tomography (CT) is equipped with an adaptive iterative reconstruction method called quantum iterative reconstruction (QIR), which allows the intensity to be changed during image reconstruction. It is known that the reconstruction conditions of CT images affect the analysis results when performing radiomic analysis. The aim of this study is to investigate the effect of QIR intensity on image quality and radiomic analysis of renal cell carcinoma (RCC). Materials and Methods The QIR intensities were selected as off, 2 and 4. The image quality evaluation items considered were task-based transfer function (TTF), noise power spectrum (NPS), and low-contrast object specific contrast-to-noise ratio (CNRLO). The influence on radiomic analysis was assessed using the discrimination accuracy of clear cell RCC. Results For image quality evaluation, TTF and NPS values were lower and CNRLO values were higher with increasing QIR intensity; for radiomic analysis, sensitivity, specificity, and accuracy were higher with increasing QIR intensity. Principal component analysis and receiver operating characteristics analysis also showed higher values with increasing QIR intensity. Conclusion It was confirmed that the intensity of the QIR intensity affects both the image quality and the radiomic analysis.
Collapse
Affiliation(s)
- Miyu Ohata
- Department of Radiological Technology, Graduate School of Health Sciences, Okayama University, Okayama, Japan
| | - Ryohei Fukui
- Department of Radiological Technology, Faculty of Health Sciences, Okayama University, Okayama, Japan
| | - Yusuke Morimitsu
- Division of Radiological Technology, Okayama University Hospital, Okayama, Japan
| | - Daichi Kobayashi
- Division of Radiological Technology, Okayama University Hospital, Okayama, Japan
| | - Takatsugu Yamauchi
- Division of Radiological Technology, Okayama University Hospital, Okayama, Japan
| | - Noriaki Akagi
- Division of Radiological Technology, Okayama University Hospital, Okayama, Japan
| | - Mitsugi Honda
- Division of Radiological Technology, Okayama University Hospital, Okayama, Japan
| | - Aiko Hayashi
- Department of Radiology, Hiroshima University Hospital, Hiroshima, Japan
| | - Koshi Hasegawa
- Department of Radiological Technology, Graduate School of Health Sciences, Okayama University, Okayama, Japan
| | - Katsuhiro Kida
- Department of Radiological Technology, Faculty of Health Sciences, Okayama University, Okayama, Japan
| | - Sachiko Goto
- Department of Radiological Technology, Faculty of Health Sciences, Okayama University, Okayama, Japan
| | - Takao Hiraki
- Department of Radiology, Faculty of Medicine, Dentistry and Pharmaceutical, Okayama University, Okayama, Japan
| |
Collapse
|
29
|
Gu C, Chen X, Wu J, Zhang Y, Zhong L, Luo H, Luo W, Yang F. SOCS1: A potential diagnostic and prognostic marker for aggressive gliomas and a new target for immunotherapy. Medicine (Baltimore) 2024; 103:e40632. [PMID: 39654174 PMCID: PMC11630960 DOI: 10.1097/md.0000000000040632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Gliomas, the most common and deadly cancers of the central nervous system, present a unique immunological barrier that severely undermines the effectiveness of immunotherapies. Suppressor of cytokine signaling 1 (SOCS1), belonging to the SOCS protein family and playing a pivotal role in various cancer treatment strategies and is abundant in high-grade gliomas. This study conducted a comparative analysis of SOCS1 and glioma immune checkpoints. It underscores the feasibility of leveraging SOCS1 as a promising diagnostic and prognostic marker for aggressive gliomas, thus offering novel targets for glioma immunotherapy. Comprehensive gene expression analyses and clinical data validations were performed across multiple databases. The expression and biological functions of SOCS1 were examined through an array of techniques including pan-cancer analysis, functional enrichment, gene set variation analysis, and immune microenvironment examination. This was done alongside a comparison of the similarities between SOCS1 and various glioma immune checkpoints. Utilizing clinical information from patients, a bespoke predictive model was developed to further corroborate the prognostic capabilities of SOCS1. The investigation revealed considerable similarities between SOCS1 and several immune checkpoints such as CTLA4, demonstrating SOCS1's role as an independent prognostic factor positively influencing glioma patient outcomes. The inclusion of SOCS1 in the developed predictive model significantly enhanced its precision. Our findings highlight SOCS1's potential as an innovative target for glioma immunotherapy, providing a novel strategy to overcome the immunological barriers posed by gliomas. Furthermore, identifying SOCS1 as a viable diagnostic marker for aggressive gliomas improves the accuracy of prognostic predictions for affected patients.
Collapse
Affiliation(s)
- Chuanshen Gu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Xinyi Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiayan Wu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yiwen Zhang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Linyu Zhong
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Han Luo
- College of Acupuncture and Tuina, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Wenshu Luo
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Fuxia Yang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
30
|
Sun C, Jiang C, Wang X, Ma S, Zhang D, Jia W. MR-Based Radiomics Predicts CDK6 Expression and Prognostic Value in High-grade Glioma. Acad Radiol 2024; 31:5141-5153. [PMID: 38964985 DOI: 10.1016/j.acra.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024]
Abstract
RATIONALE AND OBJECTIVES This study aims to assess the prognostic value of Cyclin-dependent kinases 6 (CDK6) expression levels and establish a machine learning-based radiomics model for predicting the expression levels of CDK6 in high-grade gliomas (HGG). MATERIALS AND METHODS Clinical parameters and genomic data were extracted from 310 HGG patients in the Cancer Genome Atlas (TCGA) database and 27 patients in the Repository of Molecular Brain Neoplasia Data (REMBRANDT) database. Univariate and multivariate Cox regression, as well as Kaplan-Meier analysis, were performed for prognosis analysis. The correlation between immune cell Infiltration with CDK6 was assessed using spearman correlation analysis. Radiomic features were extracted from contrast-enhanced magnetic resonance imaging (CE-MRI) in the Cancer Imaging Archive (TCIA) database (n = 82) and REMBRANDT database (n = 27). Logistic regression (LR) and support vector machine (SVM) were employed to establish the radiomics model for predicting CDK6 expression. Receiver operating characteristic curve (ROC), calibration curve, and decision curve analysis (DCA) were utilized to assess the predictive performance of the radiomics model. Generate radiomic scores (RS) based on the LR model. An RS-based nomogram was constructed to predict the prognosis of HGG. RESULTS CDK6 was significantly overexpressed in HGG tissues and was related to lower overall survival. A significant elevation in infiltrating M0 macrophages was observed in the CDK6 high group (P < 0.001). The LR radiomics model for the prediction of CDK6 expression levels (AUC=0.810 in the training cohort, AUC = 0.784 after cross-validation, AUC=0.750 in the testing cohort) was established utilizing three radiomic features. The predictive efficiencies of the RS-based nomogram, as measured by AUC, were 0.769 for 1-year, 0.815 for 3-year, and 0.780 for 5-year, respectively. CONCLUSION The expression level of CDK6 can impact the prognosis of patients with HGG. The expression level of HGG can be noninvasively prognosticated utilizing a radiomics model.
Collapse
Affiliation(s)
- Chen Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Chenggang Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Xi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Dainan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China.
| |
Collapse
|
31
|
Kobayashi H, Nakata N, Izuka S, Hongo K, Nishikawa M. Using artificial intelligence and promoter-level transcriptome analysis to identify a biomarker as a possible prognostic predictor of cardiac complications in male patients with Fabry disease. Mol Genet Metab Rep 2024; 41:101152. [PMID: 39484074 PMCID: PMC11525769 DOI: 10.1016/j.ymgmr.2024.101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Fabry disease is the most frequently occurring form of lysosomal disease in Japan, and is characterized by a wide variety of conditions. Primarily, the three major types of concerns associated with Fabry disease observed during adulthood that must be prevented are central nervous system, renal, and cardiac complications. Cardiac complications, such as cardiomyopathy, cardiac muscle fibrosis, and severe arrhythmia, are the most common mortality causes in patients with Fabry disease. To predict cardiac complications of Fabry disease, we extracted RNA from the venous blood of patients for cap analysis of gene expression (CAGE), performed likelihood ratio tests for each RNA expression dataset obtained from individuals with and without cardiac complications, and analyzed the correlation between cardiac functional factors observed using magnetic resonance imaging data extracted using artificial intelligence algorithms and RNA expression. Our findings showed that CHN1 expression was significantly higher in male Fabry disease patients with cardiac complications and that it could be associated with many cardiac functional factors. CHN1 encodes a GTPase-activating protein, chimerin 1, which is specific to the GTP-binding protein Rac (involved in oxidative stress generation and the promotion of myocardial fibrosis). Thus, CHN1 is a potential predictive biomarker of cardiac complications in Fabry disease; however, further studies are required to confirm this observation.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Division of Gene Therapy, Research Center for Medical Sciences, The Jikei University of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
- Department of Pediatrics, The Jikei University of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Norio Nakata
- Division of Artificial Intelligence Medicine, Research Center for Medical Sciences, The Jikei University of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
- Department of Radiology, The Jikei University of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Sayoko Izuka
- Division of Gene Therapy, Research Center for Medical Sciences, The Jikei University of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Kenichi Hongo
- Division of Cardiology, Department of Internal Medicine, The Jikei University of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Masako Nishikawa
- Clinical Research Support Center, The Jikei University of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
32
|
Zhang Z, Li N, Qian Y, Cheng H. Establishment of an MRI-based radiomics model for distinguishing between intramedullary spinal cord tumor and tumefactive demyelinating lesion. BMC Med Imaging 2024; 24:317. [PMID: 39574000 PMCID: PMC11583559 DOI: 10.1186/s12880-024-01499-8] [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: 06/03/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
OBJECTIVE Differentiating intramedullary spinal cord tumor (IMSCT) from spinal cord tumefactive demyelinating lesion (scTDL) remains challenging with standard diagnostic approaches. This study aims to develop and evaluate the effectiveness of a magnetic resonance imaging (MRI)-based radiomics model for distinguishing scTDL from IMSCT before treatment initiation. METHODS A total of 75 patients were analyzed in this retrospective study, comprising 55 with IMSCT and 20 with scTDL. Radiomics features were extracted from T1- and T2-weighted imaging (T1&T2WI) scans upon admission. Ten classification algorithms were employed: logistic regression (LR); naive bayes (NaiveBayes); support vector machine (SVM); k nearest neighbors (KNN); random forest (RF); extra trees (ExtraTrees); eXtreme gradient boosting (XGBoost); light gradient boosting machine (LightGBM); gradient boosting (GradientBoosting); and multi-Layer perceptron (MLP). The performance of the optimal model was then compared to radiologists' assessments. RESULTS This study developed 30 predictive models using ten classifiers across two imaging sequences. The MLP model with two sequences (T1&T2WI) emerged as the most effective one, showing superior accuracy in MRI analysis with an area under the curve (AUC) of 0.991 in training and 0.962 in testing. Moreover, statistical analyses highlighted the radiomics model significantly outperformed radiologists' assessments (p < 0.05) in distinguishing between IMSCT and scTDL. CONCLUSION We present an MRI-based radiomics model with high diagnostic accuracy in differentiating IMSCT from scTDL. The model's performance was comparable to junior radiologists, highlighting its potential as an effective diagnostic aid in clinical practice.
Collapse
Affiliation(s)
- Zifeng Zhang
- School of Medicine, Southeast University, Nanjing, China
| | - Ning Li
- Department of Neurosurgery, Affiliated Zhongda Hospital, Southeast University, Nanjing, China
| | - Yuhang Qian
- School of Medicine, Southeast University, Nanjing, China
| | - Huilin Cheng
- School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
33
|
Zhang Y, Cheng X, Luo X, Sun R, Huang X, Liu L, Zhu M, Li X. Prediction of esophageal fistula in radiotherapy/chemoradiotherapy for patients with advanced esophageal cancer by a clinical-deep learning radiomics model : Prediction of esophageal fistula in radiotherapy/chemoradiotherapy patients. BMC Med Imaging 2024; 24:313. [PMID: 39558242 PMCID: PMC11571992 DOI: 10.1186/s12880-024-01473-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/21/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Esophageal fistula (EF), a rare and potentially fatal complication, can be better managed with predictive models for personalized treatment plans in esophageal cancers. We aim to develop a clinical-deep learning radiomics model for effectively predicting the occurrence of EF. METHODS The study involved esophageal cancer patients undergoing radiotherapy or chemoradiotherapy. Arterial phase enhanced CT images were used to extract handcrafted and deep learning radiomic features. Along with clinical information, a 3-step feature selection method (statistical tests, Least Absolute Shrinkage and Selection Operator, and Recursive Feature Elimination) was used to identify five feature sets in training cohort for constructing random forest EF prediction models. Model performance was compared and validated in both retrospective and prospective test cohorts. RESULTS One hundred seventy five patients (122 in training and 53 in test cohort)were retrospectively collected from April 2018 to June 2022. An additional 27 patients were enrolled as a prospective test cohort from June 2022 to December 2023. Post-selection in the training cohort, five feature sets were used for model construction: clinical, handcrafted radiomic, deep learning radiomic, clinical-handcrafted radiomic, and clinical-deep learning radiomic. The clinical-deep learning radiomic model excelled with AUC of 0.89 (95% Confidence Interval: 0.83-0.95) in the training cohort, 0.81 (0.65-0.94) in the test cohort, and 0.85 (0.71-0.97) in the prospective test cohort. Brier-score and calibration curve analyses validated its predictive ability. CONCLUSIONS The clinical-deep learning radiomic model can effectively predict EF in patients with advanced esophageal cancer undergoing radiotherapy or chemoradiotherapy.
Collapse
Affiliation(s)
- Yuxin Zhang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, P.R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, P.R. China
| | - Xu Cheng
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, P.R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, P.R. China
| | - Xianli Luo
- Department of Radiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ruixia Sun
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, P.R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, P.R. China
| | - Xiang Huang
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, P.R. China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, P.R. China.
| | - Lingling Liu
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, P.R. China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, P.R. China
| | - Min Zhu
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, P.R. China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, P.R. China.
| | - Xueling Li
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, P.R. China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, P.R. China.
| |
Collapse
|
34
|
Zhang M, Li X, Yang Y, Wang X, Li S, Yue Q, Wei Q, Hong J. The prognostic value and biological significance of MRI CE-T1-based radiomics models in CNS5-identified GBM: a multi-center study. Sci Rep 2024; 14:27551. [PMID: 39528608 PMCID: PMC11554799 DOI: 10.1038/s41598-024-78705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Following the publication of the 2021 WHO Classification of Central Nervous System Tumors (CNS5), Following the publication of the 2021 WHO Classification of Central Nervous System Tumors (CNS5), prognostic markers of glioblastoma (GBM) need to be further explored. Radiomics is a non-invasive and reproducible method for the prognostic assessment of multiple solid tumors. This study aimed to explore the prognostic value and biological significance of MRI T1-weighted enhancement (CE-T1) based radiomics in GBM (CNS5). A six-features radiomics prognostic model was created to calculate the radiomics score (RS). High RS (HR = 3.718, 95%CI: 2.222 - 6.220, P < 0.001) was an independent risk factor for overall survival (OS). The correlation between RS and OS was externally verified based on the First Affiliated Hospital of Fujian Medical University cohorts (n = 93; HR = 2.015, 95% CI: 1.079 - 3.762, P = 0.028) and the Second Affiliated Hospital of Zhejiang University School of Medicine cohorts (n = 126; HR = 1.779, 95% CI: 1.023 - 3.091, P = 0.041). Through biological significance exploration, RS was found to be significantly correlated with DNA repair (P = 0.009) and glycolysis (P = 0.001) pathway enrichment scores. RS was associated with γδT cell infiltration and the expression of LAG3. The MRI CE-T1 based radiomics models can predict GBM (CNS5) prognosis noninvasively. RS is relevant to DNA repair, and may guide the screening of radiosensitive populations.
Collapse
Affiliation(s)
- Mingwei Zhang
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaoxia Li
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yang Yang
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Xuezhen Wang
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Shan Li
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China.
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| | - Qiuyuan Yue
- Department of Radiology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350014, Fujian, China.
| | - Qichun Wei
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
| | - Jinsheng Hong
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China.
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China.
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
35
|
Xia Y, Yang M, Qian T, Zhou J, Bai M, Luo S, Lu C, Zhu Y, Wang L, Qiao Z. Prediction of feeding difficulties in neonates with hypoxic-ischemic encephalopathy using magnetic resonance imaging-derived radiomics features. Pediatr Radiol 2024; 54:2036-2045. [PMID: 39349660 DOI: 10.1007/s00247-024-06065-6] [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: 03/25/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND The mechanisms behind brain and spinal cord injuries in hypoxic-ischemic encephalopathy (HIE) and associated feeding difficulties are unclear, with previous magnetic resonance imaging (MRI) attempts yielding inconclusive results. OBJECTIVE We aim to evaluate an MRI radiomics model for predicting feeding difficulties in HIE infants. Additionally, we investigate changes in predictive capability after incorporating the duration of mechanical ventilation and the timing of MRI examination. MATERIALS AND METHODS Retrospective study with 151 HIE infants (January 2013 to December 2021), randomly divided into training and validation sets. Radiomics features extracted from basal ganglia-thalamus and brainstem in T1-weighted and T2-weighted MRI. Established single-modality, single-site, and multimodality/multisite models. Receiver operating characteristic analysis and area under the curve evaluated models. Decision curve analysis assessed changes in predictive capability. RESULTS The combined radiomics model of the basal ganglia-thalamus and brainstem regions on the T2-weighted imaging demonstrated superior performance (area under the curve: 0.958 and 0.875 for training and validation, respectively). Combining scores with duration of mechanical ventilation and MRI examination time in a calibration plot model improved and stabilized performance, showing high fitting and clinical utility. Decision curve analysis favored the combined calibration plot model. CONCLUSION The MRI-based radiomics model predicts feeding difficulties in HIE infants, with basal ganglia-thalamus and brainstem as relevant factors. The combined calibration plot model exhibits the highest clinical predictive efficacy.
Collapse
Affiliation(s)
- Yaqin Xia
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Mingshu Yang
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Tianyang Qian
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Jiayu Zhou
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Mei Bai
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Siqi Luo
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Chaogang Lu
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Yinghao Zhu
- Institute of Artificial Intelligence, Beihang University, Beijing, China
| | - Laishuan Wang
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Zhongwei Qiao
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China.
| |
Collapse
|
36
|
Yan Q, Hu Y, Liu Z, Liu X. Clinical characterization and immunomodulatory role of VSIG4 in glioma. Asian J Surg 2024; 47:4824-4825. [PMID: 38824014 DOI: 10.1016/j.asjsur.2024.05.147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/16/2024] [Indexed: 06/03/2024] Open
Affiliation(s)
- Qingzi Yan
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China.
| | - Yixiang Hu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China.
| | - Zheng Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China.
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China.
| |
Collapse
|
37
|
Wisnu Wardhana DP, Maliawan S, Mahadewa TGB, Rosyidi RM, Wiranata S. Radiomic Features as Artificial Intelligence Prognostic Models in Glioblastoma: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2024; 14:2354. [PMID: 39518322 PMCID: PMC11545697 DOI: 10.3390/diagnostics14212354] [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: 09/14/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Glioblastoma, the predominant primary tumor among all central nervous systems, accounts for around 80% of cases. Prognosis in neuro-oncology involves assessing the disease's progression in different individuals, considering the time between the initial pathological diagnosis and the time until the disease worsens. A noninvasive therapeutic approach called radiomic features (RFs), which involves the application of artificial intelligence in MRI, has been developed to address this issue. This study aims to systematically gather evidence and evaluate the prognosis significance of radiomics in glioblastoma using RFs. METHODS We conducted an extensive search across the PubMed, ScienceDirect, EMBASE, Web of Science, and Cochrane databases to identify relevant original studies examining the use of RFs to evaluate the prognosis of patients with glioblastoma. This thorough search was completed on 25 July 2024. Our search terms included glioblastoma, MRI, magnetic resonance imaging, radiomics, and survival or prognosis. We included only English-language studies involving human subjects, excluding case reports, case series, and review studies. The studies were classified into two quality categories: those rated 4-6 were considered moderate-, whereas those rated 7-9 were high-quality using the Newcastle-Ottawa Scale (NOS). Hazard ratios (HRs) and their 95% confidence intervals (CIs) for OS and PFS were combined using random effects models. RESULTS In total, 253 studies were found in the initial search across the five databases. After screening the articles, 40 were excluded due to not meeting the eligibility criteria, and we included only 14 studies. All twelve OS and eight PFS trials were considered, involving 1.639 and 747 patients, respectively. The random effects model was used to calculate the pooled HRs for OS and PFS. The HR for OS was 3.59 (95% confidence interval [CI], 1.80-7.17), while the HR for PFS was 4.20 (95% CI, 1.02-17.32). CONCLUSIONS An RF-AI-based approach offers prognostic significance for OS and PFS in patients with glioblastoma.
Collapse
Affiliation(s)
- Dewa Putu Wisnu Wardhana
- Neurosurgery Division, Department of Surgery, Faculty of Medicine, Universitas Udayana, Udayana University Hospital, Denpasar 80361, Indonesia
| | - Sri Maliawan
- Neurosurgery Division, Department of Surgery, Faculty of Medicine, Universitas Udayana, Prof. Dr. IGNG Ngoerah General Hospital, Denpasar 80113, Indonesia
| | - Tjokorda Gde Bagus Mahadewa
- Neurosurgery Division, Department of Surgery, Faculty of Medicine, Universitas Udayana, Prof. Dr. IGNG Ngoerah General Hospital, Denpasar 80113, Indonesia
| | - Rohadi Muhammad Rosyidi
- Department of Neurosurgery, Medical Faculty of Mataram University, West Nusa Tenggara General Hospital, Mataram 84371, Indonesia
| | - Sinta Wiranata
- Faculty of Medicine, Universitas Udayana, Denpasar 80232, Indonesia
| |
Collapse
|
38
|
Zhuo LY, Hao JW, Song ZJ, Meng H, Wang TD, Yang LL, Yang ZM, Ma JM, Shen D, Cui JJ, Chen WJ, Yang W, Zang LL, Wang JN, Yin XP. Predicting the severity of mycoplasma pneumoniae pneumonia in pediatric and adult patients: a multicenter study. Sci Rep 2024; 14:22978. [PMID: 39362944 PMCID: PMC11450145 DOI: 10.1038/s41598-024-74251-5] [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: 05/04/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
The purpose of this study is to develop a nomogram model for early prediction of the severe mycoplasma pneumoniae pneumonia (SMPP) in Pediatric and Adult Patients. A retrospective analysis was conducted on patients with MPP, classifying them into SMPP and non-severe MPP (NSMPP) groups. A total of 550 patients (NSMPP 374 and SMPP 176) were enrolled in the study and allocated to training, validation cohorts. 278 patients (NSMPP 224 and SMPP 54) were retrospectively collected from two institutions and allocated to testing cohort. The risk factors for SMPP were identified using univariate analysis. For radiomic feature selection, Spearman's correlation and the least absolute shrinkage and selection operator (LASSO) were utilized. Logistic regression was used to build different models, including clinical, imaging, radiomics, and integrated models (combining clinical, imaging, and radiomics features selected). The model's discrimination was evaluated using a receiver operating characteristic curve, its calibration with a calibration curve, and the results were visualized using the Hosmer-Lemeshow goodness-of-fit test. Thirteen clinical features and fourteen imaging features were selected for constructing the clinical and imaging models. Simultaneously, a set of twenty-five radiomics features were utilized to build the radiomics model. The integrated model demonstrated good calibration and discrimination in the training cohorts (AUC, 0.922; 95% CI: 0.900, 0.942), validation cohorts (AUC, 0.879; 95% CI: 0.806, 0.920), and testing cohorts (AUC, 0.877; 95% CI: 0.836, 0.916). The discriminatory and predictive efficacy of the clinical model in testing cohorts increased further after clinical and radiological features were incorporated (AUC, 0.849 vs. 0.922, P = 0.002). The model demonstrated exemplary predictive efficacy for SMPP by leveraging a comprehensive set of inputs, encompassing clinical data, quantitative and qualitative radiological features, along with radiomics features. The integration of these three aspects in the predictive model further enhanced the performance of the clinical model, indicating the potential for extensive clinical applications.
Collapse
Affiliation(s)
- Li-Yong Zhuo
- Department of Radiology and Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, The Affiliated Hospital of Hebei University, 212 Eastern Yuhua Road, Baoding, 071000, Hebei Province, China
| | - Jia-Wei Hao
- Department of Radiology and Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, The Affiliated Hospital of Hebei University, 212 Eastern Yuhua Road, Baoding, 071000, Hebei Province, China
| | - Zi-Jun Song
- Department of Critical Care Medicine, Baoding First Central Hospital, Lianchi District, No. 320, Changcheng North Street (Qianwei Road), Baoding, 071000, China
| | - Huan Meng
- Department of Radiology and Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, The Affiliated Hospital of Hebei University, 212 Eastern Yuhua Road, Baoding, 071000, Hebei Province, China
| | - Tian-Da Wang
- Department of Radiology and Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, The Affiliated Hospital of Hebei University, 212 Eastern Yuhua Road, Baoding, 071000, Hebei Province, China
| | - Lu-Lu Yang
- Department of Radiology and Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, The Affiliated Hospital of Hebei University, 212 Eastern Yuhua Road, Baoding, 071000, Hebei Province, China
| | - Zi-Mei Yang
- Department of Radiology and Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, The Affiliated Hospital of Hebei University, 212 Eastern Yuhua Road, Baoding, 071000, Hebei Province, China
| | - Jia-Mei Ma
- Department of Radiology and Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, The Affiliated Hospital of Hebei University, 212 Eastern Yuhua Road, Baoding, 071000, Hebei Province, China
| | - Dan Shen
- Department of Urology, the Affiliated Hospital of Hebei University, 212 Eastern Yuhua Road, Baoding City, 071000, Hebei Province, China
| | - Jing-Jing Cui
- Department of Research and Development, United Imaging Intelligence (Beijing) Co., Ltd.Yongteng North Road, Haidian District, Beijing, 100094, China
| | - Wen-Jing Chen
- Department of Research and Development, United Imaging Intelligence (Beijing) Co., Ltd.Yongteng North Road, Haidian District, Beijing, 100094, China
| | - Wei Yang
- Department of Pulmonary and Critical Care Medicine, Baoding First Central Hospital, Lianchi District, No. 320, Changcheng North Street (Qianwei Road), Baoding, 071000, China
| | - Li-Li Zang
- Department of Radiology, Baoding Children's Hospital, No. 103, East Baihua Road, Baoding, 071000, China
| | - Jia-Ning Wang
- Department of Radiology and Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, The Affiliated Hospital of Hebei University, 212 Eastern Yuhua Road, Baoding, 071000, Hebei Province, China.
| | - Xiao-Ping Yin
- Department of Radiology and Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, The Affiliated Hospital of Hebei University, 212 Eastern Yuhua Road, Baoding, 071000, Hebei Province, China.
| |
Collapse
|
39
|
Dai X, Zhao B, Zang J, Wang X, Liu Z, Sun T, Yu H, Sui X. Diagnostic Performance of Radiomics and Deep Learning to Identify Benign and Malignant Soft Tissue Tumors: A Systematic Review and Meta-analysis. Acad Radiol 2024; 31:3956-3967. [PMID: 38614826 DOI: 10.1016/j.acra.2024.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/24/2024] [Accepted: 03/24/2024] [Indexed: 04/15/2024]
Abstract
RATIONALE AND OBJECTIVES To systematically evaluate the application value of radiomics and deep learning (DL) in the differential diagnosis of benign and malignant soft tissue tumors (STTs). MATERIALS AND METHODS A systematic review was conducted on studies published up to December 11, 2023, that utilized radiomics and DL methods for the diagnosis of STTs. The methodological quality and risk of bias were evaluated using the Radiomics Quality Score (RQS) 2.0 system and Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) tool, respectively. A bivariate random-effects model was used to calculate the summarized sensitivity and specificity. To identify factors contributing to heterogeneity, meta-regression and subgroup analyses were performed to assess the following covariates: diagnostic modality, region/volume of interest, imaging examination, study design, and pathology type. The asymmetry of Deeks' funnel plot was used to assess publication bias. RESULTS A total of 21 studies involving 3866 patients were included, with 13 studies using independent test/validation sets included in the quantitative statistical analysis. The average RQS was 21.31, with substantial or near-perfect inter-rater agreement. The combined sensitivity and specificity were 0.84 (95% CI: 0.76-0.89) and 0.88 (95% CI: 0.69-0.96), respectively. Meta-regression and subgroup analyses showed that study design and the region/volume of interest were significant factors affecting study heterogeneity (P < 0.05). No publication bias was observed. CONCLUSION Radiomics and DL can accurately distinguish between benign and malignant STTs. Future research should concentrate on enhancing the rigor of study designs, conducting multicenter prospective validations, amplifying the interpretability of DL models, and integrating multimodal data to elevate the diagnostic accuracy and clinical utility of soft tissue tumor assessments.
Collapse
Affiliation(s)
- Xinpeng Dai
- Department of Ultrasound, Hebei Medical University Third Hospital, Hebei, China
| | - Bingxin Zhao
- Department of Ultrasound, Hebei Medical University Third Hospital, Hebei, China
| | - Jiangnan Zang
- Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinying Wang
- Department of Ultrasound, Hebei Medical University Third Hospital, Hebei, China
| | - Zongjie Liu
- Department of Ultrasound, Hebei Medical University Third Hospital, Hebei, China
| | - Tao Sun
- Department of Orthopaedic Oncology, Hebei Medical University Third Hospital, Hebei, China
| | - Hong Yu
- Department of CT/MR, Hebei Medical University Third Hospital, Hebei, China
| | - Xin Sui
- Department of Ultrasound, Hebei Medical University Third Hospital, No.139 Ziqiang road, Qiaoxi Area, Shijiazhuang, Hebei Province, China.
| |
Collapse
|
40
|
Mao Q, Qiao Z, Wang Q, Zhao W, Ju H. Construction and validation of a machine learning-based immune-related prognostic model for glioma. J Cancer Res Clin Oncol 2024; 150:439. [PMID: 39352539 PMCID: PMC11445300 DOI: 10.1007/s00432-024-05970-5] [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: 08/13/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Glioma stands as the most prevalent primary brain tumor found within the central nervous system, characterized by high invasiveness and treatment resistance. Although immunotherapy has shown potential in various tumors, it still faces challenges in gliomas. This study seeks to develop and validate a prognostic model for glioma based on immune-related genes, to provide new tools for precision medicine. METHODS Glioma samples were obtained from a database that includes the ImmPort database. Additionally, we incorporated ten machine learning algorithms to assess the model's performance using evaluation metrics like the Harrell concordance index (C-index). The model genes were further studied using GSCA, TISCH2, and HPA databases to understand their role in glioma pathology at the genomic, molecular, and single-cell levels, and validate the biological function of IKBKE in vitro experiments. RESULTS In this study, a total of 199 genes associated with prognosis were identified using univariate Cox analysis. Subsequently, a consensus prognostic model was developed through the application of machine learning algorithms. In which the Lasso + plsRcox algorithm demonstrated the best predictive performance. The model showed a good ability to distinguish two groups in both the training and test sets. Additionally, the model genes were closely related to immunity (oligodendrocytes and macrophages), and mutation burden. The results of in vitro experiments showed that the expression level of the IKBKE gene had a significant effect on the apoptosis and migration of GL261 glioma cells. Western blot analysis showed that down-regulation of IKBKE resulted in increased expression of pro-apoptotic protein Bax and decreased expression of anti-apoptotic protein Bcl-2, which was consistent with increased apoptosis rate. On the contrary, IKBKE overexpression caused a decrease in Bax expression an increase in Bcl-2 expression, and a decrease in apoptosis rate. Tunel results further confirmed that down-regulation of IKBKE promoted apoptosis, while overexpression of IKBKE reduced apoptosis. In addition, cells with down-regulated IKBKE had reduced migration in scratch experiments, while cells with overexpression of IKBKE had increased migration. CONCLUSION This study successfully constructed a glioma prognosis model based on immune-related genes. These findings provide new perspectives for glioma prognosis assessment and immunotherapy.
Collapse
Affiliation(s)
- Qi Mao
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Zhi Qiao
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Qiang Wang
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Wei Zhao
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Haitao Ju
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China.
| |
Collapse
|
41
|
Li X, Dong J, Li B, Aimei O, Sun Y, Wu X, Liu W, Li R, Li Z, Yang Y. Prediction of Neoadjuvant Chemoradiotherapy Sensitivity in Patients With Esophageal Squamous Cell Carcinoma Using CT-Based Radiomics Combined With Clinical Features. Dose Response 2024; 22:15593258241301525. [PMID: 39588071 PMCID: PMC11587189 DOI: 10.1177/15593258241301525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/21/2024] [Accepted: 10/03/2024] [Indexed: 11/27/2024] Open
Abstract
Background: For patients with resectable locally advanced esophageal squamous cell carcinoma (ESCC), the current standard treatment is neoadjuvant chemoradiotherapy (nCRT) plus radical surgery. Objective: This study aimed to establish a predictive model, based on computed tomography (CT) radiomics features and clinical parameters, to predict sensitivity to nCRT in patients with ESCC pre-treatment. The goal was to provide risk stratification and decision-making recommendations for clinical treatments and offer more valuable information for developing personalized therapies. Methods: This retrospective study involved 102 patients diagnosed with ESCC through biopsy who underwent nCRT. To select radiomics features, we used the least absolute shrinkage and selection operator (LASSO) algorithm. A combined model was constructed, integrating the selected clinically relevant parameters with the Rad-Score. To assess the performance of this combined model, we utilized calibration curves and receiver operating characteristic (ROC) curves. Results: Nine optimal radiomics features were selected using the LASSO algorithm. The support vector machine (SVM) classifier was identified as having the best predictive performance. The area under the curve (AUC) of the SVM training group was 0.937 (95% CI: 0.856-1.000), and of the validation group was 0.831 (95% CI: 0.679-0.983). Smoking and alcohol history, neutrophil to lymphocyte ratio, serum aspartate aminotransferase to alanine aminotransferase ratio, and carcinoembryonic antigen and fibrinogen levels were independent predictors of sensitivity to nCRT in patients with ESCC. The AUCs of the combined model for the training and validation groups were 0.870 (95% CI: 0.774-0.964) and 0.821 (95% CI: 0.669-0.972), respectively. The calibration curve showed that the nomogram's predictions were close to the actual clinical observations, indicating that the model exhibited good predictive performance. Conclusion: Our combined model based on Rad-Score and clinical characteristics showed high predictive performance for predicting sensitivity to nCRT in patients with ESCC. It may be useful for predicting treatment effects in clinical practice and demonstrates the significant potential of radiomics in predicting and optimizing treatment decisions.
Collapse
Affiliation(s)
- Xindi Li
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
- Department of Oncology, Shandong University, Shandong Provincial Third Hospital, Jinan, China
| | - Jigang Dong
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
- Qingdao Jiaozhou Central Hospital, Qingdao, China
| | - Baosheng Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ouyang Aimei
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yahong Sun
- Department of Oncology, Shandong University, Shandong Provincial Third Hospital, Jinan, China
| | - Xia Wu
- Department of Oncology, Shandong University, Shandong Provincial Third Hospital, Jinan, China
| | - Wenjuan Liu
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Jinan, China
| | - Ruobing Li
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhongyuan Li
- School of Medical Imaging, Shandong Second Medical University, Weifang, China
| | - Yu Yang
- Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
42
|
Shams A. Leveraging State-of-the-Art AI Algorithms in Personalized Oncology: From Transcriptomics to Treatment. Diagnostics (Basel) 2024; 14:2174. [PMID: 39410578 PMCID: PMC11476216 DOI: 10.3390/diagnostics14192174] [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: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Continuous breakthroughs in computational algorithms have positioned AI-based models as some of the most sophisticated technologies in the healthcare system. AI shows dynamic contributions in advancing various medical fields involving data interpretation and monitoring, imaging screening and diagnosis, and treatment response and survival prediction. Despite advances in clinical oncology, more effort must be employed to tailor therapeutic plans based on each patient's unique transcriptomic profile within the precision/personalized oncology frame. Furthermore, the standard analysis method is not compatible with the comprehensive deciphering of significant data streams, thus precluding the prediction of accurate treatment options. METHODOLOGY We proposed a novel approach that includes obtaining different tumour tissues and preparing RNA samples for comprehensive transcriptomic interpretation using specifically trained, programmed, and optimized AI-based models for extracting large data volumes, refining, and analyzing them. Next, the transcriptomic results will be scanned against an expansive drug library to predict the response of each target to the tested drugs. The obtained target-drug combination/s will be then validated using in vitro and in vivo experimental models. Finally, the best treatment combination option/s will be introduced to the patient. We also provided a comprehensive review discussing AI models' recent innovations and implementations to aid in molecular diagnosis and treatment planning. RESULTS The expected transcriptomic analysis generated by the AI-based algorithms will provide an inclusive genomic profile for each patient, containing statistical and bioinformatics analyses, identification of the dysregulated pathways, detection of the targeted genes, and recognition of molecular biomarkers. Subjecting these results to the prediction and pairing AI-based processes will result in statistical graphs presenting each target's likely response rate to various treatment options. Different in vitro and in vivo investigations will further validate the selection of the target drug/s pairs. CONCLUSIONS Leveraging AI models will provide more rigorous manipulation of large-scale datasets on specific cancer care paths. Such a strategy would shape treatment according to each patient's demand, thus fortifying the avenue of personalized/precision medicine. Undoubtedly, this will assist in improving the oncology domain and alleviate the burden of clinicians in the coming decade.
Collapse
Affiliation(s)
- Anwar Shams
- Department of Pharmacology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; or ; Tel.: +00966-548638099
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif 26432, Saudi Arabia
- High Altitude Research Center, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
43
|
Zheng B, Zhao Z, Zheng P, Liu Q, Li S, Jiang X, Huang X, Ye Y, Wang H. The current state of MRI-based radiomics in pituitary adenoma: promising but challenging. Front Endocrinol (Lausanne) 2024; 15:1426781. [PMID: 39371931 PMCID: PMC11449739 DOI: 10.3389/fendo.2024.1426781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/30/2024] [Indexed: 10/08/2024] Open
Abstract
In the clinical diagnosis and treatment of pituitary adenomas, MRI plays a crucial role. However, traditional manual interpretations are plagued by inter-observer variability and limitations in recognizing details. Radiomics, based on MRI, facilitates quantitative analysis by extracting high-throughput data from images. This approach elucidates correlations between imaging features and pituitary tumor characteristics, thereby establishing imaging biomarkers. Recent studies have demonstrated the extensive application of radiomics in differential diagnosis, subtype identification, consistency evaluation, invasiveness assessment, and treatment response in pituitary adenomas. This review succinctly presents the general workflow of radiomics, reviews pertinent literature with a summary table, and provides a comparative analysis with traditional methods. We further elucidate the connections between radiological features and biological findings in the field of pituitary adenoma. While promising, the clinical application of radiomics still has a considerable distance to traverse, considering the issues with reproducibility of imaging features and the significant heterogeneity in pituitary adenoma patients.
Collapse
Affiliation(s)
- Baoping Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pingping Zheng
- Department of Neurosurgery, People’s Hospital of Biyang County, Zhumadian, China
| | - Qiang Liu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Li
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Huang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Youfan Ye
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haijun Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Li M, Fu S, Du J, Han X, Duan C, Ren Y, Qiao Y, Tang Y. Preoperative MRI-based radiomic nomogram for distinguishing solitary fibrous tumor from angiomatous meningioma: a multicenter study. Front Oncol 2024; 14:1399270. [PMID: 39359426 PMCID: PMC11445187 DOI: 10.3389/fonc.2024.1399270] [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: 03/11/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
Purpose This study evaluates the efficacy of radiomics-based machine learning methodologies in differentiating solitary fibrous tumor (SFT) from angiomatous meningioma (AM). Materials and methods A retrospective analysis was conducted on 171 pathologically confirmed cases (94 SFT and 77 AM) spanning from January 2009 to September 2020 across four institutions. The study comprised a training set (n=137) and a validation set (n=34). All patients underwent contrast-enhanced T1-weighted (CE-T1WI) and T2-weighted(T2WI) MRI scans, from which 1166 radiomics features were extracted. Subsequently, seventeen features were selected through minimum redundancy maximum relevance (mRMR) and the least absolute shrinkage and selection operator (LASSO). Multivariate logistic regression analysis was employed to assess the independence of these features as predictors. A clinical model, established via both univariate and multivariate logistic regression based on MRI morphological features, was integrated with the optimal radiomics model to formulate a radiomics nomogram. The performance of the models was assessed utilizing the area under the receiver operating characteristic curve (AUC), accuracy (ACC), sensitivity (SEN), specificity (SPE), positive predictive value (PPV), and negative predictive value (NPV). Results The radiomics nomogram demonstrated exceptional discriminative performance in the validation set, achieving an AUC of 0.989. This outperformance was evident when compared to both the radiomics algorithm (AUC= 0.968) and the clinical model (AUC = 0.911) in the same validation sets. Notably, the radiomics nomogram exhibited impressive values for ACC, SEN, and SPE at 97.1%, 93.3%, and 100%, respectively, in the validation set. Conclusions The machine learning-based radiomic nomogram proves to be highly effective in distinguishing between SFT and AM.
Collapse
Affiliation(s)
- Mengjie Li
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shengli Fu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingjing Du
- Department of Radiology, Shizuishan First People's Hospital, Shizuishan, China
| | - Xiaoyu Han
- Department of Radiology, Qilu Hospital, Shandong University, Jinan, China
| | - Chongfeng Duan
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yande Ren
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaqian Qiao
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yueshan Tang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
45
|
Zhou Q, Zhang B, Xue C, Ren J, Zhang P, Ke X, Man J, Zhou J. Magnetic resonance imaging-based radiomics for predicting infiltration levels of CD68+ tumor-associated macrophages in glioblastomas. Strahlenther Onkol 2024:10.1007/s00066-024-02289-5. [PMID: 39269469 DOI: 10.1007/s00066-024-02289-5] [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: 12/21/2023] [Accepted: 07/29/2024] [Indexed: 09/15/2024]
Abstract
PURPOSE Tumor-associated macrophages (TAMs) are important biomarkers of tumor invasion and prognosis in patients with glioblastoma. We combined the imaging and radiomics features of preoperative MRI to predict CD68+ macrophage infiltration. METHODS Clinical, MRI image, and pathology data of 188 patients with glioblastoma were analyzed. Overall, 143 patients were included in the training (n = 101) and validation (n = 42) sets, whereas 45 patients were included in an independent test set. The optimal cut-off value (14.8%) was based on the minimum p-value formed by the Kaplan-Meier survival analysis and log-rank tests which divided patients into groups with high CD68+ TAMs (≥ 14.8%) and low CD68+ TAMs (< 14.8%). Regions of interest and radiomics features extraction were based on contrast-enhanced T1-weighted images (CE-T1WI) and T2WI. Multi-parameter stepwise regression was used to create the clinical, radiomics, and combined models, each evaluated using the receiver operating characteristic curve. Decision curve analysis was used to assess the clinical applicability of the nomogram. RESULTS A clinical model based on the minimum apparent diffusion coefficient (ADCmin) revealed an area under the curve (AUC) of 0.768, 0.764, and 0.624 for the training set, validation set, and test set, respectively. The 2D radiomics model, based on two features, revealed an AUC of 0.783, 0.724, and 0.789 for the training, validation, and test sets, respectively. The 3D radiomics model, based on three features, revealed AUCs of 0.823, 0.811, and 0.787 for the training, validation, and test sets, respectively. The combined model, with ADCmin and radiomics features, showed the best performance, with AUCs of 0.865, 0.822, and 0.776 for the training, validation, and test sets, respectively. The calibration curve of the combined model nomogram showed good agreement between the estimated and actual probabilities. CONCLUSION The combined model constructed using ADCmin, a quantitative imaging parameter, combined with five key radiomics features can be used to evaluate the extent of CD68+ macrophages before surgery.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, 730030, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, 730030, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, 730030, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | | | - Peng Zhang
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, 730030, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Jiangwei Man
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Department of Surgical, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, 730030, Lanzhou, Gansu, China.
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China.
| |
Collapse
|
46
|
Shu K, Wang K, Zhang R, Wang C, Cai Z, Liu K, Lin H, Zeng Y, Cao Z, Lai C, Yan Z, Lu Y. Pituitary MRI Radiomics Improves Diagnostic Performance of Growth Hormone Deficiency in Children Short Stature: A Multicenter Radiomics Study. Acad Radiol 2024; 31:3783-3792. [PMID: 38796401 DOI: 10.1016/j.acra.2024.05.009] [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: 04/09/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/28/2024]
Abstract
RATIONALE AND OBJECTIVES To develop an efficient machine-learning model using pituitary MRI radiomics and clinical data to differentiate growth hormone deficiency (GHD) from idiopathic short stature (ISS), making the diagnostic process more acceptable to patients and their families. MATERIALS AND METHODS A retrospective cohort of 297 GHD and 300 ISS children (4-12 years) were enrolled as training and validation cohorts (8:2 ratio). An external cohort from another institution (49 GHD and 51 ISS) was employed as the testing cohort. Radiomics features extracted from the anterior pituitary gland on sagittal T1-weighted image (1.5 T or 3.0 T) were used to develop a radiomics model after feature selection. Hematological biomarkers were selected to create a clinical model and combine with the optimal radiomics features to create a clinical-radiomics model. The area under the receive operating characteristic curve (AUC) and Delong test compared the diagnostic performance of the previously mentioned three models across different validation and testing cohorts. RESULTS 17 radiomics features were selected for the radiomics model, and total protein, total cholesterol, free triiodothyronine, and triglyceride were utilized for the clinical model. In the training and validation cohorts, the diagnostic performance of the clinical-radiomics model (AUC=0.820 and 0.801) was comparable to the radiomics model (AUC=0.812 and 0.779, both P >0.05), both outperforming the clinical model (AUC=0.575 and 0.593, P <0.001). In the testing cohort, the clinical-radiomics model exhibited the highest AUC of 0.762 than the clinical and radiomics model (AUC=0.604 and 0.741, respectively, P <0.05). In addition, the clinical and radiomics models demonstrated similar diagnostic performance in the testing cohort (P >0.05). CONCLUSION Integrating radiomics features from conventional pituitary MRI with clinical indicators offers a minimally invasive approach for identifying GHD and shows robustness in a multicenter setting.
Collapse
Affiliation(s)
- Kun Shu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Keren Wang
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ruifang Zhang
- Department of Radiology, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chenyan Wang
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zheng Cai
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Kun Liu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hu Lin
- Department of Endocrinology, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yan Zeng
- Department of Research Center, Shanghai United Imaging Intelligence Co., Ltd, China
| | - Zirui Cao
- Department of Research Center, Shanghai United Imaging Intelligence Co., Ltd, China
| | - Can Lai
- Department of Radiology, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Zhihan Yan
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang Province, China; Wenzhou Key Laboratory of Structural and Functional Imaging, Wenzhou, Zhejiang Province, China
| | - Yi Lu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang Province, China; Wenzhou Key Laboratory of Structural and Functional Imaging, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
47
|
Fu FX, Cai QL, Li G, Wu XJ, Hong L, Chen WS. The efficacy of using a multiparametric magnetic resonance imaging-based radiomics model to distinguish glioma recurrence from pseudoprogression. Magn Reson Imaging 2024; 111:168-178. [PMID: 38729227 DOI: 10.1016/j.mri.2024.05.003] [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: 01/17/2024] [Revised: 04/24/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
OBJECTIVE The early differential diagnosis of the postoperative recurrence or pseudoprogression (psPD) of a glioma is of great guiding significance for individualized clinical treatment. This study aimed to evaluate the ability of a multiparametric magnetic resonance imaging (MRI)-based radiomics model to distinguish between the postoperative recurrence and psPD of a glioma early on and in a noninvasive manner. METHODS A total of 52 patients with gliomas who attended the Hainan Provincial People's Hospital between 2000 and 2021 and met the inclusion criteria were selected for this study. 1137 and 1137 radiomic features were extracted from T1 enhanced and T2WI/FLAIR sequence images, respectively.After clearing some invalid information and LASSO screening, a total of 9 and 10 characteristic radiological features were extracted and randomly divided into the training set and the test set according to 7:3 ratio. Select-Kbest and minimum Absolute contraction and selection operator (LASSO) were used for feature selection. Support vector machine and logistic regression were used to form a multi-parameter model for training and prediction. The optimal sequence and classifier were selected according to the area under the curve (AUC) and accuracy. RESULTS Radiomic models 1, 2 and 3 based on T1WI, T2FLAIR and T1WI + T2T2FLAIR sequences have better performance in the identification of postoperative recurrence and false progression of T1 glioma. The performance of model 2 is more stable, and the performance of support vector machine classifier is more stable. The multiparameter model based on CE-T1 + T2WI/FLAIR sequence showed the best performance (AUC:0.96, sensitivity: 0.87, specificity: 0.94, accuracy: 0.89,95% CI:0.93-1). CONCLUSION The use of multiparametric MRI-based radiomics provides a noninvasive, stable, and accurate method for differentiating between the postoperative recurrence and psPD of a glioma, which allows for timely individualized clinical treatment.
Collapse
Affiliation(s)
- Fang-Xiong Fu
- Department of Radiology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Qin-Lei Cai
- Department of Radiology, Hainan General Hospital, Haikou 570311, China
| | - Guo Li
- Department of Radiology, Hainan General Hospital, Haikou 570311, China
| | - Xiao-Jing Wu
- Department of Radiology, Hainan General Hospital, Haikou 570311, China
| | - Lan Hong
- Department of Gynecology, Hainan General Hospital, Haikou 570311, China.
| | - Wang-Sheng Chen
- Department of Radiology, Hainan General Hospital, Haikou 570311, China.
| |
Collapse
|
48
|
Wu X, Zhang S, Zhang Z, He Z, Xu Z, Wang W, Jin Z, You J, Guo Y, Zhang L, Huang W, Wang F, Liu X, Yan D, Cheng J, Yan J, Zhang S, Zhang B. Biologically interpretable multi-task deep learning pipeline predicts molecular alterations, grade, and prognosis in glioma patients. NPJ Precis Oncol 2024; 8:181. [PMID: 39152182 PMCID: PMC11329669 DOI: 10.1038/s41698-024-00670-2] [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: 02/15/2024] [Accepted: 08/01/2024] [Indexed: 08/19/2024] Open
Abstract
Deep learning models have been developed for various predictions in glioma; yet, they were constrained by manual segmentation, task-specific design, or a lack of biological interpretation. Herein, we aimed to develop an end-to-end multi-task deep learning (MDL) pipeline that can simultaneously predict molecular alterations and histological grade (auxiliary tasks), as well as prognosis (primary task) in gliomas. Further, we aimed to provide the biological mechanisms underlying the model's predictions. We collected multiscale data including baseline MRI images from 2776 glioma patients across two private (FAHZU and HPPH, n = 1931) and three public datasets (TCGA, n = 213; UCSF, n = 410; and EGD, n = 222). We trained and internally validated the MDL model using our private datasets, and externally validated it using the three public datasets. We used the model-predicted deep prognosis score (DPS) to stratify patients into low-DPS and high-DPS subtypes. Additionally, a radio-multiomics analysis was conducted to elucidate the biological basis of the DPS. In the external validation cohorts, the MDL model achieved average areas under the curve of 0.892-0.903, 0.710-0.894, and 0.850-0.879 for predicting IDH mutation status, 1p/19q co-deletion status, and tumor grade, respectively. Moreover, the MDL model yielded a C-index of 0.723 in the TCGA and 0.671 in the UCSF for the prediction of overall survival. The DPS exhibits significant correlations with activated oncogenic pathways, immune infiltration patterns, specific protein expression, DNA methylation, tumor mutation burden, and tumor-stroma ratio. Accordingly, our work presents an accurate and biologically meaningful tool for predicting molecular subtypes, tumor grade, and survival outcomes in gliomas, which provides personalized clinical decision-making in a global and non-invasive manner.
Collapse
Affiliation(s)
- Xuewei Wu
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shuaitong Zhang
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zicong He
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zexin Xu
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhe Jin
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jingjing You
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yang Guo
- Department of Neurosurgery, The Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Lu Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Wenhui Huang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Fei Wang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Shuixing Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| | - Bin Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
49
|
Shi D, Li S, Liu F, Jiang X, Wu L, Chen L, Zheng Q, Bao H, Guo H, Xu J. Comprehensive characterization of tumor therapeutic response with simultaneous mapping cell size, density, and transcytolemmal water exchange. ARXIV 2024:arXiv:2408.01918v1. [PMID: 39130198 PMCID: PMC11312621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Early assessment of tumor therapeutic response is an important topic in precision medicine to optimize personalized treatment regimens and reduce unnecessary toxicity, cost, and delay. Although diffusion MRI (dMRI) has shown potential to address this need, its predictive accuracy is limited, likely due to its unspecific sensitivity to overall pathological changes. In this work, we propose a new quantitative dMRI-based method dubbed EXCHANGE (MRI of water Exchange, Confined and Hindered diffusion under Arbitrary Gradient waveform Encodings) for simultaneous mapping of cell size, cell density, and transcytolemmal water exchange. Such rich microstructural information comprehensively evaluates tumor pathologies at the cellular level. Validations using numerical simulations and in vitro cell experiments confirmed that the EXCHANGE method can accurately estimate mean cell size, density, and water exchange rate constants. The results from in vivo animal experiments show the potential of EXCHANGE for monitoring tumor treatment response. Finally, the EXCHANGE method was implemented in breast cancer patients with neoadjuvant chemotherapy, demonstrating its feasibility in assessing tumor therapeutic response in clinics. In summary, a new, quantitative dMRI-based EXCHANGE method was proposed to comprehensively characterize tumor microstructural properties at the cellular level, suggesting a unique means to monitor tumor treatment response in clinical practice.
Collapse
Affiliation(s)
- Diwei Shi
- Center for Nano and Micro Mechanics, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Sisi Li
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Fan Liu
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiaoyu Jiang
- Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lei Wu
- Qinghai University Affiliated Hospital, Qinghai, Xining 810000, China
| | - Li Chen
- Center for Nano and Micro Mechanics, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Quanshui Zheng
- Center for Nano and Micro Mechanics, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Haihua Bao
- Qinghai University Affiliated Hospital, Qinghai, Xining 810000, China
| | - Hua Guo
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Junzhong Xu
- Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
50
|
Yimit Y, Yasin P, Tuersun A, Wang J, Wang X, Huang C, Abudoubari S, Chen X, Ibrahim I, Nijiati P, Wang Y, Zou X, Nijiati M. Multiparametric MRI-Based Interpretable Radiomics Machine Learning Model Differentiates Medulloblastoma and Ependymoma in Children: A Two-Center Study. Acad Radiol 2024; 31:3384-3396. [PMID: 38508934 DOI: 10.1016/j.acra.2024.02.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 02/23/2024] [Accepted: 02/24/2024] [Indexed: 03/22/2024]
Abstract
RATIONALE AND OBJECTIVES Medulloblastoma (MB) and Ependymoma (EM) in children, share similarities in age group, tumor location, and clinical presentation. Distinguishing between them through clinical diagnosis is challenging. This study aims to explore the effectiveness of using radiomics and machine learning on multiparametric magnetic resonance imaging (MRI) to differentiate between MB and EM and validate its diagnostic ability with an external set. MATERIALS AND METHODS Axial T2 weighted image (T2WI) and contrast-enhanced T1weighted image (CE-T1WI) MRI sequences of 135 patients from two centers were collected as train/test sets. Volume of interest (VOI) was manually delineated by an experienced neuroradiologist, supervised by a senior. Feature selection analysis and the least absolute shrinkage and selection operator (LASSO) algorithm identified valuable features, and Shapley additive explanations (SHAP) evaluated their significance. Five machine-learning classifiers-extreme gradient boosting (XGBoost), Bernoulli naive Bayes (Bernoulli NB), Logistic Regression (LR), support vector machine (SVM), linear support vector machine (Linear SVC) classifiers were built based on T2WI (T2 model), CE-T1WI (T1 model), and T1 + T2WI (T1 + T2 model). A human expert diagnosis was developed and corrected by senior radiologists. External validation was performed at Sun Yat-Sen University Cancer Center. RESULTS 31 valuable features were extracted from T2WI and CE-T1WI. XGBoost demonstrated the highest performance with an area under the curve (AUC) of 0.92 on the test set and maintained an AUC of 0.80 during external validation. For the T1 model, XGBoost achieved the highest AUC of 0.85 on the test set and the highest accuracy of 0.71 on the external validation set. In the T2 model, XGBoost achieved the highest AUC of 0.86 on the test set and the highest accuracy of 0.82 on the external validation set. The human expert diagnosis had an AUC of 0.66 on the test set and 0.69 on the external validation set. The integrated T1 + T2 model achieved an AUC of 0.92 on the test set, 0.80 on the external validation set, achieved the best performance. Overall, XGBoost consistently outperformed in different classification models. CONCLUSION The combination of radiomics and machine learning on multiparametric MRI effectively distinguishes between MB and EM in childhood, surpassing human expert diagnosis in training and testing sets.
Collapse
Affiliation(s)
- Yasen Yimit
- Department of Radiology, The First People's Hospital of Kashi (Kashgar) Prefecture, Xinjiang, China, 844000; Xinjiang Key Laboratory of Artificial Intelligence assisted Imaging Diagnosis, Kashi (Kashgar), China, 844000
| | - Parhat Yasin
- Department of Spine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China, 830054
| | - Abudouresuli Tuersun
- Department of Radiology, The First People's Hospital of Kashi (Kashgar) Prefecture, Xinjiang, China, 844000; Xinjiang Key Laboratory of Artificial Intelligence assisted Imaging Diagnosis, Kashi (Kashgar), China, 844000
| | - Jingru Wang
- Department of Research Collaboration, R&D center, Beijing Deepwise & League of PHD Technology Co., Ltd, Beijing, PR China, 100080
| | - Xiaohong Wang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China, 510630
| | - Chencui Huang
- Department of Research Collaboration, R&D center, Beijing Deepwise & League of PHD Technology Co., Ltd, Beijing, PR China, 100080
| | - Saimaitikari Abudoubari
- Department of Radiology, The First People's Hospital of Kashi (Kashgar) Prefecture, Xinjiang, China, 844000; Xinjiang Key Laboratory of Artificial Intelligence assisted Imaging Diagnosis, Kashi (Kashgar), China, 844000
| | - Xingzhi Chen
- Department of Research Collaboration, R&D center, Beijing Deepwise & League of PHD Technology Co., Ltd, Beijing, PR China, 100080
| | - Irshat Ibrahim
- Department of General Surgery, The First People's Hospital of Kashi (Kashgar) Prefecture, Xinjiang, China, 844000
| | - Pahatijiang Nijiati
- Department of Radiology, The First People's Hospital of Kashi (Kashgar) Prefecture, Xinjiang, China, 844000; Xinjiang Key Laboratory of Artificial Intelligence assisted Imaging Diagnosis, Kashi (Kashgar), China, 844000
| | - Yunling Wang
- Department of Imaging Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China, 830054
| | - Xiaoguang Zou
- Xinjiang Key Laboratory of Artificial Intelligence assisted Imaging Diagnosis, Kashi (Kashgar), China, 844000; Clinical Medical Research Center, The First People's Hospital of Kashi (Kashgar) Prefecture, Xinjiang, China, 844000
| | - Mayidili Nijiati
- Department of Radiology, The First People's Hospital of Kashi (Kashgar) Prefecture, Xinjiang, China, 844000; Xinjiang Key Laboratory of Artificial Intelligence assisted Imaging Diagnosis, Kashi (Kashgar), China, 844000.
| |
Collapse
|