201
|
O'Sullivan NJ, Temperley HC, Horan MT, Curtain BMM, O'Neill M, Donohoe C, Ravi N, Corr A, Meaney JFM, Reynolds JV, Kelly ME. Computed tomography (CT) derived radiomics to predict post-operative disease recurrence in gastric cancer; a systematic review and meta-analysis. Curr Probl Diagn Radiol 2024; 53:717-722. [PMID: 39025746 DOI: 10.1067/j.cpradiol.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/10/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Radiomics offers the potential to predict oncological outcomes from pre-operative imaging in order to identify 'high risk' patients at increased risk of recurrence. The application of radiomics in predicting disease recurrence provides tailoring of therapeutic strategies. We aim to comprehensively assess the existing literature regarding the current role of radiomics as a predictor of disease recurrence in gastric cancer. METHODS A systematic search was conducted in Medline, EMBASE, and Web of Science databases. Inclusion criteria encompassed retrospective and prospective studies investigating the use of radiomics to predict post-operative recurrence in ovarian cancer. Study quality was assessed using the QUADAS-2 and Radiomics Quality Score tools. RESULTS Nine studies met the inclusion criteria, involving a total of 6,662 participants. Radiomic-based nomograms demonstrated consistent performance in predicting disease recurrence, as evidenced by satisfactory area under the receiver operating characteristic curve values (AUC range 0.72 - 1). The pooled AUCs calculated using the inverse-variance method for both the training and validation datasets were 0.819 and 0.789 respectively CONCLUSION: Our review provides good evidence supporting the role of radiomics as a predictor of post-operative disease recurrence in gastric cancer. Included studies noted good performance in predicting their primary outcome. Radiomics may enhance personalised medicine by tailoring treatment decision based on predicted prognosis.
Collapse
Affiliation(s)
- Niall J O'Sullivan
- Department of Radiology, St. James's Hospital, Dublin, Ireland; School of Medicine, Trinity College Dublin, Ireland; The National Centre for Advanced Medical Imaging (CAMI), St. James's Hospital, Dublin, Ireland.
| | - Hugo C Temperley
- Department of Radiology, St. James's Hospital, Dublin, Ireland; School of Medicine, Trinity College Dublin, Ireland
| | - Michelle T Horan
- Department of Radiology, St. James's Hospital, Dublin, Ireland; The National Centre for Advanced Medical Imaging (CAMI), St. James's Hospital, Dublin, Ireland
| | | | - Maeve O'Neill
- Department of Surgery, St. James's Hospital, Dublin, Ireland
| | - Claire Donohoe
- Department of Upper Gastrointestinal Surgery, St. James's Hospital, Dublin, Ireland
| | - Narayanasamy Ravi
- Department of Upper Gastrointestinal Surgery, St. James's Hospital, Dublin, Ireland
| | - Alison Corr
- Department of Radiology, St. James's Hospital, Dublin, Ireland
| | - James F M Meaney
- Department of Radiology, St. James's Hospital, Dublin, Ireland; School of Medicine, Trinity College Dublin, Ireland; The National Centre for Advanced Medical Imaging (CAMI), St. James's Hospital, Dublin, Ireland
| | - John V Reynolds
- School of Medicine, Trinity College Dublin, Ireland; Department of Upper Gastrointestinal Surgery, St. James's Hospital, Dublin, Ireland
| | - Michael E Kelly
- School of Medicine, Trinity College Dublin, Ireland; Department of Surgery, St. James's Hospital, Dublin, Ireland; Trinity St James Cancer Institute, Trinity College Dublin, Ireland
| |
Collapse
|
202
|
Wang Z, Li X, Zhang H, Duan T, Zhang C, Zhao T. Deep learning Radiomics Based on Two-Dimensional Ultrasound for Predicting the Efficacy of Neoadjuvant Chemotherapy in Breast Cancer. ULTRASONIC IMAGING 2024; 46:357-366. [PMID: 39257175 DOI: 10.1177/01617346241276168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
We investigate the predictive value of a comprehensive model based on preoperative ultrasound radiomics, deep learning, and clinical features for pathological complete response (pCR) after neoadjuvant chemotherapy (NAC) for the breast cancer. We enrolled 155 patients with pathologically confirmed breast cancer who underwent NAC. The patients were randomly divided into the training set and the validation set in the ratio of 7:3. The deep learning and radiomics features of pre-treatment ultrasound images were extracted, and the random forest recursive elimination algorithm and the least absolute shrinkage and selection operator were used for feature screening and DL-Score and Rad-Score construction. According to multifactorial logistic regression, independent clinical predictors, DL-Score, and Rad-Score were selected to construct the comprehensive prediction model DLRC. The performance of the model was evaluated in terms of its predictive effect, and clinical practicability. Compared to the clinical, radiomics (Rad-Score), and deep learning (DL-Score) models, the DLRC accurately predicted the pCR status, with an area under the curve (AUC) of 0.937 (95%CI: 0.895-0.970) in the training set and 0.914 (95%CI: 0.838-0.973) in the validation set. Moreover, decision curve analysis confirmed that the DLRC had the highest clinical value among all models. The comprehensive model DLRC based on ultrasound radiomics, deep learning, and clinical features can effectively and accurately predict the pCR status of breast cancer after NAC, which is conducive to assisting clinical personalized diagnosis and treatment plan.
Collapse
Affiliation(s)
- Zhan Wang
- Jintan Peoples Hospital, Jiangsu, Changzhou, China
| | - Xiaoqin Li
- Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Jiangsu, Changzhou, China
| | - Heng Zhang
- Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Jiangsu, Changzhou, China
| | - Tongtong Duan
- Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Jiangsu, Changzhou, China
| | - Chao Zhang
- Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Jiangsu, Changzhou, China
| | - Tong Zhao
- Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Jiangsu, Changzhou, China
| |
Collapse
|
203
|
Verstappen D, Pasquier D, Chen Y, Lambin P, Woodruff HC. Radiomic feature robustness in CT scans affected by fiducial marker induced streak artifacts for patients with hepatocellular carcinoma. Med Phys 2024; 51:8362-8370. [PMID: 39047166 DOI: 10.1002/mp.17329] [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: 02/10/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Stereotactic body radiation therapy for hepatocellular carcinoma necessitates the implantation of gold fiducial markers in the liver, resulting in artifacts on computed tomography (CT) scans, which affect radiomic feature values. PURPOSE This report aims to assess the effect of these artifacts on radiomic features and how removing CT slices affects radiomic features extracted from 3D regions of interest (ROI). METHODS First, the range variation in 38 tumor contours unaffected by artifacts was assessed after sequentially and randomly removing 25%, 50%, 75% of slices. Subsequently, the agreement of feature values before and after removing ROI slices containing artifacts from 186 patients' CT scans was assessed with Lin's concordance correlation coefficient (CCC) and a Wilcoxon signed-rank test. RESULTS In artifact-free tumor volumes, at least 71% of features remain robust with up to 50% of slices removed, while 56% remains robust with up to 75% of slices removed. When comparing contours before and after removing slices containing artifacts, around a third of features in the tumor contour and surrounding area remain robust (CCC > 0.9), compared to 44% in the healthy liver. Concerning the tumor, 13% (Gray Level Size Zone Matrix) to 61% (first order) of the features remain robust (CCC > 0.9). Over 90% of features differ significantly as assessed by Wilcoxon signed-rank test, however. CONCLUSIONS This study demonstrates that removing slices containing artifacts is a feasible solution for the CT fiducial problem in this patient population and provides insight into which features are affected.
Collapse
Affiliation(s)
- Damon Verstappen
- The D-Lab, Department of Precision Medicine, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - David Pasquier
- The D-Lab, Department of Precision Medicine, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Academic Department of Radiation Oncology, Centre Oscar Lambret, Lille, France
- CNRS, Centrale Lille, UMR 9189 - CRIStAL, Univ. Lille, Lille, France
| | - Yi Chen
- The D-Lab, Department of Precision Medicine, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Phillipe Lambin
- The D-Lab, Department of Precision Medicine, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Henry C Woodruff
- The D-Lab, Department of Precision Medicine, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
204
|
Li H, Zhao J, Jiang Z. Deep learning-based computer-aided detection of ultrasound in breast cancer diagnosis: A systematic review and meta-analysis. Clin Radiol 2024; 79:e1403-e1413. [PMID: 39217049 DOI: 10.1016/j.crad.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/05/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE The aim of this meta-analysis was to assess the diagnostic performance of deep learning (DL) and ultrasound in breast cancer diagnosis. Additionally, we categorized the included studies into two subgroups: B-mode ultrasound diagnostic subgroup and multimodal ultrasound diagnostic subgroup, and compared the performance differences of DL algorithms in breast cancer diagnosis using only B-mode ultrasound or multimodal ultrasound. METHODS We conducted a comprehensive search for relevant studies published from January 01, 2017 to July 31, 2023 in the MEDLINE and EMBASE databases. The quality of the included studies was evaluated using the QUADAS-2 tool and radiomics quality scores (RQS). Meta-analysis was performed using R software. Inter-study heterogeneity was assessed by I^2 values and Q-test P-values, with sources of heterogeneity analyzed through a random effects model based on test results. Summary receiver operating characteristics (SROC) curves were used for meta-analysis across multiple trials, while combined sensitivity, specificity, and AUC were calculated to quantify prediction accuracy. Subgroup analysis and sensitivity analyses were also conducted to identify potential sources of study heterogeneity. Publication bias was assessed using the funnel plot method. (PROSPERO identifier: CRD42024545758). RESULTS The 20 studies included a total of 14,955 cases, with 4197 cases used for model testing. Among these cases were 1582 breast cancer patients and 2615 benign or other breast lesions. The combined sensitivity, specificity, and AUC values across all studies were found to be 0.93, 0.90, and 0.732, respectively. In subgroup analysis, the multimodal subgroup demonstrated superior performance with combined sensitivity, specificity, and AUC values of 0.93, 0.88, and 0.787, respectively; whereas the combined sensitivity, specificity, and AUC value for the model B subgroup was at a level of 0.92, 0.91, and 0.642, respectively. CONCLUSIONS The integration of DL with ultrasound demonstrates high accuracy in the adjunctive diagnosis of breast cancer, while the fusion of DL and multimodal breast ultrasound exhibits superior diagnostic efficacy compared to B-mode ultrasound alone.
Collapse
Affiliation(s)
- H Li
- Department of Ultrasound, Changzheng Hospital, Naval Medical University (Second Medical University), No.415, Fengyang Rd, Shanghai, China.
| | - J Zhao
- Department of Ultrasound, Changzheng Hospital, Naval Medical University (Second Medical University), No.415, Fengyang Rd, Shanghai, China; Department of Ultrasound, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1279, Sanmen Rd, Shanghai, China.
| | - Z Jiang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No.516, Jungong Rd, Shanghai, China
| |
Collapse
|
205
|
Masson-Grehaigne C, Lafon M, Palussière J, Leroy L, Bonhomme B, Jambon E, Italiano A, Cousin S, Crombé A. Single- and multi-site radiomics may improve overall survival prediction for patients with metastatic lung adenocarcinoma. Diagn Interv Imaging 2024; 105:439-452. [PMID: 39191636 DOI: 10.1016/j.diii.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 07/09/2024] [Indexed: 08/29/2024]
Abstract
PURPOSE The purpose of this study was to assess whether single-site and multi-site radiomics could improve the prediction of overall survival (OS) of patients with metastatic lung adenocarcinoma compared to clinicopathological model. MATERIALS AND METHODS Adults with metastatic lung adenocarcinoma, pretreatment whole-body contrast-enhanced computed tomography examinations, and performance status (WHO-PS) ≤ 2 were included in this retrospective single-center study, and randomly assigned to training and testing cohorts. Radiomics features (RFs) were extracted from all measurable lesions with volume ≥ 1 cm3. Radiomics prognostic scores based on the largest tumor (RPSlargest) and the average RF values across all tumors per patient (RPSaverage) were developed in the training cohort using 5-fold cross-validated LASSO-penalized Cox regression. Intra-patient inter-tumor heterogeneity (IPITH) metrics were calculated to quantify the radiophenotypic dissimilarities among all tumors within each patient. A clinicopathological model was built in the training cohort using stepwise Cox regression and enriched with combinations of RPSaverage, RPSlargest and IPITH. Models were compared with the concordance index in the independent testing cohort. RESULTS A total of 300 patients (median age: 63.7 years; 40.7% women; median OS, 16.3 months) with 1359 lesions were included (200 and 100 patients in the training and testing cohorts, respectively). The clinicopathological model included WHO-PS = 2 (hazard ratio [HR] = 3.26; P < 0.0001), EGFR, ALK, ROS1 or RET mutations (HR = 0.57; P = 0.0347), IVB stage (HR = 1.65; P = 0.0211), and liver metastases (HR = 1.47; P = 0.0670). In the testing cohort, RPSaverage, RPSlargest and IPITH were associated with OS (HR = 85.50, P = 0.0038; HR = 18.83, P = 0.0082 and HR = 8.00, P = 0.0327, respectively). The highest concordance index was achieved with the combination of clinicopathological variables and RPSaverage, significantly better than that of the clinicopathological model (concordance index = 0.7150 vs. 0.695, respectively; P = 0.0049) CONCLUSION: Single-site and multi-site radiomics-based scores are associated with OS in patients with metastatic lung adenocarcinoma. RPSaverage improves the clinicopathological model.
Collapse
Affiliation(s)
- Cécile Masson-Grehaigne
- Department of Diagnostic and Interventional Oncologic Imaging, Institut Bergonié, 33076 Bordeaux, France; Department of Radiology, Pellegrin University Hospital, 33000 Bordeaux, France
| | - Mathilde Lafon
- Department of Medical Oncology, Institut Bergonié, 33076 Bordeaux, France
| | - Jean Palussière
- Department of Diagnostic and Interventional Oncologic Imaging, Institut Bergonié, 33076 Bordeaux, France
| | - Laura Leroy
- Department of Medical Oncology, Institut Bergonié, 33076 Bordeaux, France
| | | | - Eva Jambon
- Department of Radiology, Pellegrin University Hospital, 33000 Bordeaux, France
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, 33076 Bordeaux, France; SARCOTARGET Team, Bordeaux Institute of Oncology (BRIC) INSERM U1312, Bordeaux 33076, France
| | - Sophie Cousin
- Department of Medical Oncology, Institut Bergonié, 33076 Bordeaux, France
| | - Amandine Crombé
- Department of Diagnostic and Interventional Oncologic Imaging, Institut Bergonié, 33076 Bordeaux, France; Department of Radiology, Pellegrin University Hospital, 33000 Bordeaux, France; SARCOTARGET Team, Bordeaux Institute of Oncology (BRIC) INSERM U1312, Bordeaux 33076, France.
| |
Collapse
|
206
|
Mao Y, Jiang LP, Wang JL, Diao YH, Chen FQ, Zhang WP, Chen L, Liu ZX. Multi-feature Fusion Network on Gray Scale Ultrasonography: Effective Differentiation of Adenolymphoma and Pleomorphic Adenoma. Acad Radiol 2024; 31:4396-4407. [PMID: 38871552 DOI: 10.1016/j.acra.2024.05.023] [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: 04/02/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024]
Abstract
RATIONALE AND OBJECTIVES to develop a deep learning radiomics graph network (DLRN) that integrates deep learning features extracted from gray scale ultrasonography, radiomics features and clinical features, for distinguishing parotid pleomorphic adenoma (PA) from adenolymphoma (AL) MATERIALS AND METHODS: A total of 287 patients (162 in training cohort, 70 in internal validation cohort and 55 in external validation cohort) from two centers with histologically confirmed PA or AL were enrolled. Deep transfer learning features and radiomics features extracted from gray scale ultrasound images were input to machine learning classifiers including logistic regression (LR), support vector machines (SVM), KNN, RandomForest (RF), ExtraTrees, XGBoost, LightGBM, and MLP to construct deep transfer learning radiomics (DTL) models and Rad models respectively. Deep learning radiomics (DLR) models were constructed by integrating the two features and DLR signatures were generated. Clinical features were further combined with the signatures to develop a DLRN model. The performance of these models was evaluated using receiver operating characteristic (ROC) curve analysis, calibration, decision curve analysis (DCA), and the Hosmer-Lemeshow test. RESULTS In the internal validation cohort and external validation cohort, comparing to Clinic (AUC=0.767 and 0.777), Rad (AUC=0.841 and 0.748), DTL (AUC=0.740 and 0.825) and DLR (AUC=0.863 and 0.859), the DLRN model showed greatest discriminatory ability (AUC=0.908 and 0.908) showed optimal discriminatory ability. CONCLUSION The DLRN model built based on gray scale ultrasonography significantly improved the diagnostic performance for benign salivary gland tumors. It can provide clinicians with a non-invasive and accurate diagnostic approach, which holds important clinical significance and value. Ensemble of multiple models helped alleviate overfitting on the small dataset compared to using Resnet50 alone.
Collapse
Affiliation(s)
- Yi Mao
- Department of Ultrasonography, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Li-Ping Jiang
- Department of Ultrasonography, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Jing-Ling Wang
- Department of Ultrasonography, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Yu-Hong Diao
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Fang-Qun Chen
- Department of Ultrasonography, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Wei-Ping Zhang
- Department of Ultrasonography, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Li Chen
- Department of Ultrasonography, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Zhi-Xing Liu
- Department of Ultrasonography, The First Affiliated Hospital of Nanchang University, Nanchang, China; Department of Ultrasonography, GanJiang New District Peoples Hospital, Nanchang, China.
| |
Collapse
|
207
|
Huang Z, Lam S, Lin Z, Zhou L, Pei L, Song A, Wang T, Zhang Y, Qi R, Huang S. Predicting major adverse cardiac events using radiomics nomogram of pericoronary adipose tissue based on CCTA: A multi-center study. Med Phys 2024; 51:8348-8361. [PMID: 39042398 DOI: 10.1002/mp.17324] [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: 09/26/2023] [Revised: 06/19/2024] [Accepted: 07/06/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND The evolution of coronary atherosclerotic heart disease (CAD) is intricately linked to alterations in the pericoronary adipose tissue (PCAT). In recent epochs, characteristics of the PCAT have progressively ascended as focal points of research in CAD risk stratification and individualized clinical decision-making. Harnessing radiomic methodologies allows for the meticulous extraction of imaging features from these adipose deposits. Coupled with machine learning paradigms, we endeavor to establish predictive models for the onset of major adverse cardiovascular events (MACE). PURPOSE To appraise the predictive utility of radiomic features of PCAT derived from coronary computed tomography angiography (CCTA) in forecasting MACE. METHODS We retrospectively incorporated data from 314 suspected or confirmed CAD patients admitted to our institution from June 2019 to December 2022. An additional cohort of 242 patients from two external institutions was encompassed for external validation. The endpoint under consideration was the occurrence of MACE after a 1-year follow-up. MACE was delineated as cardiovascular mortality, newly diagnosed myocardial infarction, hospitalization (or re-hospitalization) for heart failure, and coronary target vessel revascularization occurring more than 30 days post-CCTA examination. All enrolled patients underwent CCTA scanning. Radiomic features were meticulously extracted from the optimal diastolic phase axial slices of CCTA images. Feature reduction was achieved through a composite feature selection algorithm, laying the groundwork for the radiomic signature model. Both univariate and multivariate analyses were employed to assess clinical variables. A multifaceted logistic regression analysis facilitated the crafting of a clinical-radiological-radiomic combined model (or nomogram). Receiver operating characteristic (ROC) curves, calibration, and decision curve analyses (DCA) were delineated, with the area under the ROC curve (AUCs) computed to gauge the predictive prowess of the clinical model, radiomic model, and the synthesized ensemble. RESULTS A total of 12 radiomic features closely associated with MACE were identified to establish the radiomic model. Multivariate logistic regression results demonstrated that smoking, age, hypertension, and dyslipidemia were significantly correlated with MACE. In the integrated nomogram, which amalgamated clinical, imaging, and radiomic parameters, the diagnostic performance was as follows: 0.970 AUC, 0.949 accuracy (ACC), 0.833 sensitivity (SEN), 0.981 specificity (SPE), 0.926 positive predictive value (PPV), and 0.955 negative predictive value (NPV). The calibration curve indicated a commendable concordance of the nomogram, and the decision curve analysis underscored its superior clinical utility. CONCLUSIONS The integration of radiomic signatures from PCAT based on CCTA, clinical indices, and imaging parameters into a nomogram stands as a promising instrument for prognosticating MACE events.
Collapse
Affiliation(s)
- Zhaoheng Huang
- Department of Radiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Saikit Lam
- Department of Biomedical Engineering, The Hong Kong Polytechnical University, Hong Kong, China
- Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China
| | - Zihe Lin
- Department of Computing, The Hong Kong Polytechnical University, Hong Kong, China
| | - Linjia Zhou
- Department of Medical Informatics, Nantong University, Nantong, China
| | - Liangchen Pei
- School of Automation, Southeast University, Nanjing, China
| | - Anyi Song
- Department of Radiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Tianle Wang
- Department of Radiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Yuanpeng Zhang
- Department of Medical Informatics, Nantong University, Nantong, China
| | - Rongxing Qi
- Department of Radiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Sheng Huang
- Department of Radiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
208
|
Sha X, Wang C, Qi S, Yuan X, Zhang H, Yang J. The efficacy of CBCT-based radiomics techniques in differentiating between conventional and unicystic ameloblastoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2024; 138:656-665. [PMID: 39227265 DOI: 10.1016/j.oooo.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 09/05/2024]
Abstract
OBJECTIVE The aim of this study was to develop a cone beam computed tomography (CBCT) radiomics-based model that differentiates between conventional and unicystic ameloblastoma (AB). METHODS In this retrospective study, CBCT images were collected from 100 patients who had ABs that were diagnosed histopathologically as conventional or unicystic AB after surgical treatment. The patients were randomly divided into training (70) and validation (30) cohorts. Radiomics features were extracted from the images, and the optimal features were incorporated into 5 models: Logistic Regression, Support Vector Machine, Linear Discriminant Analysis, Random Forest, and XGBoost for prediction of tumor type. Model performance was evaluated using the area under the curve (AUC) from receiver operating characteristic analysis, sensitivity, specificity, accuracy, calibration curves, and decision curve analysis (DCA). RESULTS The 20 optimal radiomics features were incorporated into the Logistic Regression (LR) model, which exhibited the best overall performance with AUC = 0.936 (95% confidence interval [CI] = 0.877-0.996) for the training cohort and AUC = 0.929 (95% CI = 0.832-1.000) for the validation cohort. The nomogram combined the clinical features and the radiomics signature and resulted in the best predictive performance. CONCLUSIONS The LR model demonstrated the ability of radiomics and the nomogram to distinguish between the 2 types of AB and may have the potential to replace biopsies under noninvasive conditions.
Collapse
Affiliation(s)
- Xiaoyan Sha
- Department of Oral and Maxillofacial Radiology, School of Stomatology, Capital Medical University, Beijing, China
| | - Chao Wang
- Department of Clinical Research, SinoUnion Healthcare Inc., Beijing, China
| | - Senrong Qi
- Department of Oral and Maxillofacial Radiology, School of Stomatology, Capital Medical University, Beijing, China
| | - Xiaohong Yuan
- Department of Oral and Maxillofacial Pathology, School of Stomatology, Capital Medical University, Beijing, China
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
209
|
Li S, Chen H, Chen J, Yang X, Zhong W, Zhou H, Meng X, Liao C, Zhang W. Predicting long-term outcomes in patients with classical trigeminal neuralgia following microvascular decompression with an MRI-based radiomics nomogram: a multicentre study. Eur Radiol 2024; 34:7349-7361. [PMID: 38717486 DOI: 10.1007/s00330-024-10775-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/20/2024] [Accepted: 03/09/2024] [Indexed: 10/29/2024]
Abstract
OBJECTIVES This study aimed to develop a clinical-radiomics nomogram to predict the long-term outcomes of patients with classical trigeminal neuralgia (CTN) following microvascular decompression (MVD). MATERIALS AND METHODS This retrospective study included 455 patients with CTN who underwent MVD from three independent institutions A total of 2030 radiomics features from the cistern segment of the trigeminal nerve were extracted computationally from the three-dimensional steady-state free precession and three-dimensional time-of-flight magnetic resonance angiography sequences. Using the least absolute shrinkage and selection operator regression, 16 features were chosen to develop radiomics signatures. A clinical-radiomics nomogram was subsequently developed in the development cohort of 279 patients via multivariate Cox regression. The predictive performance and clinical application of the nomogram were assessed in an external cohort consisting of 176 patients. RESULTS Sixteen highly outcome-related radiomics features extracted from multisequence images were used to construct the radiomics model, with concordance indices (C-index) of 0.804 and 0.796 in the development and test cohorts, respectively. Additionally, a clinical-radiomics nomogram was developed by incorporating both radiomics features and clinical characteristics (i.e., pain type and degree of neurovascular compression) and yielded higher C-indices of 0.865 and 0.834 in the development and test cohorts, respectively. K‒M survival analysis indicated that the nomogram successfully stratified patients with CTN into high-risk and low-risk groups for poor outcomes (hazard ratio: 37.18, p < 0.001). CONCLUSION Our study findings indicated that the clinical-radiomics nomogram exhibited promising performance in accurately predicting long-term pain outcomes following MVD. CLINICAL RELEVANCE STATEMENT This model had the potential to aid clinicians in making well-informed decisions regarding the treatment of patients with CTN. KEY POINTS Trigeminal neuralgia recurs in about one-third of patients after undergoing MVD. The clinical-radiomics nomogram stratified patients into high- and low-risk groups for poor surgical outcomes. Using this nomogram could better inform patients of recurrence risk and allow for discussion of alternative treatments.
Collapse
Affiliation(s)
- Shuo Li
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongjin Chen
- Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jiahao Chen
- The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xiaosheng Yang
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weijie Zhong
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han Zhou
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuchen Meng
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenlong Liao
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wenchuan Zhang
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
210
|
Hu K, Tan G, Liao X, Liu WV, Han W, Hu L, Jiang H, Yang L, Guo M, Deng Y, Meng Z, Liu X. Multi-parameter MRI radiomics model in predicting postoperative progressive cerebral edema and hemorrhage after resection of meningioma. Cancer Imaging 2024; 24:149. [PMID: 39487466 PMCID: PMC11529156 DOI: 10.1186/s40644-024-00796-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Postoperative progressive cerebral edema and hemorrhage (PPCEH) are major complications after meningioma resection, yet their preoperative predictive studies are limited. The aim is to develop and validate a multiparametric MRI machine learning model to predict PPCEH after meningioma resection. METHODS This retrospective study included 148 patients with meningioma. A stratified three-fold cross-validation was used to split the dataset into training and validation sets. Radiomics features from the tumor enhancement (TE) and peritumoral brain edema (PTBE) regions were extracted from T1WI, T2WI, and ADC maps. Support vector machine constructed different radiomics models, and logistic regression explored clinical risk factors. Prediction models, integrating clinical and radiomics features, were evaluated using the area under the curve (AUC), visualized in a nomogram. RESULTS The radiomics model based on TE and PTBE regions (training set mean AUC: 0.85 (95% CI: 0.78-0.93), validation set mean AUC: 0.77 (95%CI: 0.63-0.90)) outperformed the model with TE region solely (training set mean AUC: 0.83 (95% CI: 0.76-0.91), validation set mean AUC: 0.73 (95% CI: 0.58-0.87)). Furthermore, the combined model incorporating radiomics features, and clinical features of preoperative peritumoral edema and tumor boundary adhesion, had the best predictive performance, with AUC values of 0.87 (95% CI: 0.80-0.94) and 0.84 (95% CI: 0.72-0.95) for the training and validation set. CONCLUSIONS We developed a novel model based on clinical characteristics and multiparametric radiomics features derived from TE and PTBE regions, which can accurately and non-invasively predict PPCEH after meningioma resection. Additionally, our findings suggest the crucial role of PTBE radiomics features in understanding the potential mechanisms of PPCEH.
Collapse
Affiliation(s)
- Kangjian Hu
- Department of Radiology, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong Province, China
| | - Guirong Tan
- Department of Radiology, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong Province, China
- Advanced Neuroimaging Laboratory, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong Province, China
| | - Xueqing Liao
- Department of Radiology, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong Province, China
| | | | - Wenjing Han
- Yanjing Medical College, Capital Medical University, Beijing, China
| | - Lingjing Hu
- Yanjing Medical College, Capital Medical University, Beijing, China
| | - Haihui Jiang
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, China
| | - Lijuan Yang
- Department of Radiology, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong Province, China
| | - Ming Guo
- Department of Neurosurgery, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong Province, China
| | - Yaohong Deng
- Department of Research & Development, Yizhun Medical AI Co. Ltd, Beijing, China
| | - Zhihua Meng
- Department of Radiology, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong Province, China
| | - Xiang Liu
- Department of Radiology, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong Province, China.
- Advanced Neuroimaging Laboratory, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong Province, China.
- Advanced Neuroimaging Laboratory, Department of Radiology, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong Province, China.
| |
Collapse
|
211
|
Morelli L, Paganelli C, Marvaso G, Parrella G, Annunziata S, Vicini MG, Zaffaroni M, Pepa M, Summers PE, De Cobelli O, Petralia G, Jereczek-Fossa BA, Baroni G. Addressing intra- and inter-institution variability of a radiomic framework based on Apparent Diffusion Coefficient in prostate cancer. Med Phys 2024; 51:8096-8107. [PMID: 39172115 DOI: 10.1002/mp.17355] [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: 02/07/2024] [Revised: 06/27/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a highly heterogeneous disease, making tailored treatment approaches challenging. Magnetic resonance imaging (MRI), notably diffusion-weighted imaging (DWI) and the derived Apparent Diffusion Coefficient (ADC) maps, plays a crucial role in PCa characterization. In this context, radiomics is a very promising approach able to disclose insights from MRI data. However, the sensitivity of radiomic features to MRI settings, encompassing DWI protocols and multicenter variations, requires the development of robust and generalizable models. PURPOSE To develop a comprehensive radiomics framework for noninvasive PCa characterization using ADC maps, focusing on identifying reliable imaging biomarkers against intra- and inter-institution variations. MATERIALS AND METHODS Two patient cohorts, including an internal cohort (118 PCa patients) used for both training (75%) and hold-out testing (25%), and an external cohort (50 PCa patients) for independent testing, were employed in the study. DWI images were acquired with three different DWI protocols on two different MRI scanners: two DWI protocols acquired on a 1.5-T scanner for the internal cohort, and one DWI protocol acquired on a 3-T scanner for the external cohort. One hundred and seven radiomics features (i.e., shape, first order, texture) were extracted from ADC maps of the whole prostate gland. To address variations in DWI protocols and multicenter variability, a dedicated pipeline, including two-way ANOVA, sequential-feature-selection (SFS), and ComBat features harmonization was implemented. Mann-Whitney U-tests (α = 0.05) were performed to find statistically significant features dividing patients with different tumor characteristics in terms of Gleason score (GS) and T-stage. Support-Vector-Machine models were then developed to predict GS and T-stage, and the performance was assessed through the area under the curve (AUC) of receiver-operating-characteristic curves. RESULTS Downstream of ANOVA, two subsets of 38 and 41 features stable against DWI protocol were identified for GS and T-stage, respectively. Among these, SFS revealed the most predictive features, yielding an AUC of 0.75 (GS) and 0.70 (T-stage) in the hold-out test. Employing ComBat harmonization improved the external-test performance of the GS model, raising AUC from 0.72 to 0.78. CONCLUSION By incorporating stable features with a harmonization procedure and validating the model on an external dataset, model robustness, and generalizability were assessed, highlighting the potential of ADC and radiomics for PCa characterization.
Collapse
Affiliation(s)
- Letizia Morelli
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Chiara Paganelli
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Giulia Marvaso
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giovanni Parrella
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Simone Annunziata
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Maria Giulia Vicini
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
| | - Mattia Zaffaroni
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
| | - Matteo Pepa
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
| | | | - Ottavio De Cobelli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Urology, European Institute of Oncology (IEO), Milan, Italy
| | - Giuseppe Petralia
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Radiology, European Institute of Oncology (IEO), Milan, Italy
| | - Barbara Alicja Jereczek-Fossa
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Guido Baroni
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| |
Collapse
|
212
|
Li H, Zhuang Y, Yuan W, Gu Y, Dai X, Li M, Chen H, Zhou H. Radiomics in precision medicine for colorectal cancer: a bibliometric analysis (2013-2023). Front Oncol 2024; 14:1464104. [PMID: 39558950 PMCID: PMC11571149 DOI: 10.3389/fonc.2024.1464104] [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: 07/28/2024] [Accepted: 10/04/2024] [Indexed: 11/20/2024] Open
Abstract
Background The incidence and mortality of colorectal cancer (CRC) have been rising steadily. Early diagnosis and precise treatment are essential for improving patient survival outcomes. Over the past decade, the integration of artificial intelligence (AI) and medical imaging technologies has positioned radiomics as a critical area of research in the diagnosis, treatment, and prognosis of CRC. Methods We conducted a comprehensive review of CRC-related radiomics literature published between 1 January 2013 and 31 December 2023 using the Web of Science Core Collection database. Bibliometric tools such as Bibliometrix, VOSviewer, and CiteSpace were employed to perform an in-depth bibliometric analysis. Results Our search yielded 1,226 publications, revealing a consistent annual growth in CRC radiomics research, with a significant rise after 2019. China led in publication volume (406 papers), followed by the United States (263 papers), whereas the United States dominated in citation numbers. Notable institutions included General Electric, Harvard University, University of London, Maastricht University, and the Chinese Academy of Sciences. Prominent researchers in this field are Tian J from the Chinese Academy of Sciences, with the highest publication count, and Ganeshan B from the University of London, with the most citations. Journals leading in publication and citation counts are Frontiers in Oncology and Radiology. Keyword and citation analysis identified deep learning, texture analysis, rectal cancer, image analysis, and management as prevailing research themes. Additionally, recent trends indicate the growing importance of AI and multi-omics integration, with a focus on improving precision medicine applications in CRC. Emerging keywords such as deep learning and AI have shown rapid growth in citation bursts over the past 3 years, reflecting a shift toward more advanced technological applications. Conclusions Radiomics plays a crucial role in the clinical management of CRC, providing valuable insights for precision medicine. It significantly contributes to predicting molecular biomarkers, assessing tumor aggressiveness, and monitoring treatment efficacy. Future research should prioritize advancing AI algorithms, enhancing multi-omics data integration, and further expanding radiomics applications in CRC precision medicine.
Collapse
Affiliation(s)
- Hao Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yupei Zhuang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Weichen Yuan
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yutian Gu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinyan Dai
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Muhan Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Haibin Chen
- Science and Technology Department, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongguang Zhou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
213
|
Piccolo CL, Sarli M, Pileri M, Tommasiello M, Rofena A, Guarrasi V, Soda P, Beomonte Zobel B. Radiomics for Predicting Prognostic Factors in Breast Cancer: Insights from Contrast-Enhanced Mammography (CEM). J Clin Med 2024; 13:6486. [PMID: 39518625 PMCID: PMC11546631 DOI: 10.3390/jcm13216486] [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: 09/23/2024] [Revised: 10/16/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Objectives: To evaluate the correlation between radiomic features extracted from contrast-enhanced mammography (CEM) tumor lesions and peritumoral background with prognostic factors in breast cancer (BC). Methods: In this retrospective, single-center study, 134 women with histologically confirmed breast cancer underwent CEM examination. Radiomic features were extracted from manually segmented lesions and lesion contours were automatically delineated using PyRadiomics. The extracted features were categorized into seven classes: First-order Features, Shape Features (2D), Gray Level Co-occurrence Matrix (GLCM), Gray Level Run Length Matrix (GLRLM), Gray Level Size Zone Matrix (GLSZM), and Neighboring Gray Tone Difference Matrix (NGTDM). Histological examination assessed tumor type, grade, receptor structure (ER, PgR, HER2), Ki67 index, and lymph node involvement. Pearson correlation and multivariate regression were applied to evaluate associations between radiomic features and prognostic factors. Results: Significant correlations were found between First-order Features and prognostic factors such as ER, PgR, and Ki67 (p < 0.05). GLCM-based texture features showed strong associations with Ki67 and HER2 (p < 0.01). Radiomic features from peritumoral regions, especially shape and GLSZM metrics, were significantly correlated with Ki67 and lymph node involvement. Conclusions: Radiomic analysis of both tumor and peritumoral regions offers significant insights into BC prognosis. These findings support the integration of radiomics into personalized diagnostic and therapeutic strategies, potentially improving clinical decision making in BC management.
Collapse
Affiliation(s)
- Claudia Lucia Piccolo
- Operative Research Unit of Radiology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.L.P.); (M.S.); (M.T.); (B.B.Z.)
- Research Unit of Radiology, Department of Medicine and Surgery, Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Marina Sarli
- Operative Research Unit of Radiology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.L.P.); (M.S.); (M.T.); (B.B.Z.)
- Research Unit of Radiology, Department of Medicine and Surgery, Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Matteo Pileri
- Operative Research Unit of Radiology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.L.P.); (M.S.); (M.T.); (B.B.Z.)
- Research Unit of Radiology, Department of Medicine and Surgery, Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Manuela Tommasiello
- Operative Research Unit of Radiology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.L.P.); (M.S.); (M.T.); (B.B.Z.)
- Research Unit of Radiology, Department of Medicine and Surgery, Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Aurora Rofena
- Unit of Computer Systems & Bioinformatics, Department of Engineering, Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128 Rome, Italy; (A.R.); (V.G.); (P.S.)
| | - Valerio Guarrasi
- Unit of Computer Systems & Bioinformatics, Department of Engineering, Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128 Rome, Italy; (A.R.); (V.G.); (P.S.)
| | - Paolo Soda
- Unit of Computer Systems & Bioinformatics, Department of Engineering, Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128 Rome, Italy; (A.R.); (V.G.); (P.S.)
| | - Bruno Beomonte Zobel
- Operative Research Unit of Radiology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.L.P.); (M.S.); (M.T.); (B.B.Z.)
- Research Unit of Radiology, Department of Medicine and Surgery, Campus Bio-Medico University, Via Alvaro del Portillo 21, 00128 Rome, Italy
| |
Collapse
|
214
|
Jiang Y, Gao C, Shao Y, Lou X, Hua M, Lin J, Wu L, Gao C. The prognostic value of radiogenomics using CT in patients with lung cancer: a systematic review. Insights Imaging 2024; 15:259. [PMID: 39466334 PMCID: PMC11519241 DOI: 10.1186/s13244-024-01831-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/20/2024] [Indexed: 10/30/2024] Open
Abstract
This systematic review aimed to evaluate the effectiveness of combining radiomic and genomic models in predicting the long-term prognosis of patients with lung cancer and to contribute to the further exploration of radiomics. This study retrieved comprehensive literature from multiple databases, including radiomics and genomics, to study the prognosis of lung cancer. The model construction consisted of the radiomic and genomic methods. A comprehensive bias assessment was conducted, including risk assessment and model performance indicators. Ten studies between 2016 and 2023 were analyzed. Studies were mostly retrospective. Patient cohorts varied in size and characteristics, with the number of patients ranging from 79 to 315. The construction of the model involves various radiomic and genotic datasets, and most models show promising prediction performance with the area under the receiver operating characteristic curve (AUC) values ranging from 0.64 to 0.94 and the concordance index (C-index) values from 0.28 to 0.80. The combination model typically outperforms the single method model, indicating higher prediction accuracy and the highest AUC was 0.99. Combining radiomics and genomics in the prognostic model of lung cancer may improve the predictive performance. However, further research on standardized data and larger cohorts is needed to validate and integrate these findings into clinical practice. CRITICAL RELEVANCE STATEMENT: The combination of radiomics and genomics in the prognostic model of lung cancer improved prediction accuracy in most included studies. KEY POINTS: The combination of radiomics and genomics can improve model performance in most studies. The results of establishing prognosis models by different methods are discussed. The combination of radiomics and genomics may be helpful to provide better treatment for patients.
Collapse
Affiliation(s)
- Yixiao Jiang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuan Gao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yilin Shao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinjing Lou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meiqi Hua
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiangnan Lin
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Linyu Wu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Chen Gao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
215
|
Santinha J, Pinto Dos Santos D, Laqua F, Visser JJ, Groot Lipman KBW, Dietzel M, Klontzas ME, Cuocolo R, Gitto S, Akinci D'Antonoli T. ESR Essentials: radiomics-practice recommendations by the European Society of Medical Imaging Informatics. Eur Radiol 2024:10.1007/s00330-024-11093-9. [PMID: 39453470 DOI: 10.1007/s00330-024-11093-9] [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: 05/24/2024] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 10/26/2024]
Abstract
Radiomics is a method to extract detailed information from diagnostic images that cannot be perceived by the naked eye. Although radiomics research carries great potential to improve clinical decision-making, its inherent methodological complexities make it difficult to comprehend every step of the analysis, often causing reproducibility and generalizability issues that hinder clinical adoption. Critical steps in the radiomics analysis and model development pipeline-such as image, application of image filters, and selection of feature extraction parameters-can greatly affect the values of radiomic features. Moreover, common errors in data partitioning, model comparison, fine-tuning, assessment, and calibration can reduce reproducibility and impede clinical translation. Clinical adoption of radiomics also requires a deep understanding of model explainability and the development of intuitive interpretations of radiomic features. To address these challenges, it is essential for radiomics model developers and clinicians to be well-versed in current best practices. Proper knowledge and application of these practices is crucial for accurate radiomics feature extraction, robust model development, and thorough assessment, ultimately increasing reproducibility, generalizability, and the likelihood of successful clinical translation. In this article, we have provided researchers with our recommendations along with practical examples to facilitate good research practices in radiomics. KEY POINTS: Radiomics' inherent methodological complexity should be understood to ensure rigorous radiomic model development to improve clinical decision-making. Adherence to radiomics-specific checklists and quality assessment tools ensures methodological rigor. Use of standardized radiomics tools and best practices enhances clinical translation of radiomics models.
Collapse
Affiliation(s)
- João Santinha
- Digital Surgery LAB, Champalimaud Research, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal.
- Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001, Lisbon, Portugal.
| | - Daniel Pinto Dos Santos
- Department of Radiology, University Hospital of Cologne, Cologne, Germany
- Department of Radiology, University Hospital of Frankfurt, Frankfurt, Germany
| | - Fabian Laqua
- Department of Diagnostic and Interventional Radiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Jacob J Visser
- Department of Radiology & Nuclear Medicine, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Kevin B W Groot Lipman
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Matthias Dietzel
- Department of Radiology, University Hospital Erlangen, Maximiliansplatz 3, 91054, Erlangen, Germany
| | - Michail E Klontzas
- Department of Radiology, School of Medicine, University of Crete, Heraklion, Crete, Greece
- Department of Medical Imaging, University Hospital of Heraklion, Crete, Greece
- Division of Radiology, Department of Clinical Science Intervention and Technology (CLINTEC), Karolinska Institute, Solna, Sweden
| | - Renato Cuocolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Salvatore Gitto
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Tugba Akinci D'Antonoli
- Institute of Radiology and Nuclear Medicine, Cantonal Hospital Baselland, Liestal, Switzerland
| |
Collapse
|
216
|
Jin Z, Zou Q, Zhou T, Xue T. Preoperative prediction of early recurrence in patients with BRAF mutant colorectal cancer using a intergrated nomogram. Sci Rep 2024; 14:25320. [PMID: 39455810 PMCID: PMC11512039 DOI: 10.1038/s41598-024-77256-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024] Open
Abstract
Objective To explore the predictive value of radiomics nomogram combining with CT radiomics features and clinical features for postoperative early recurrence in patients with BRAF-mutant colorectal cancer. Methods A total of 220 patients with surgically and pathologically confirmed BRAF-mutant colorectal cancer from 2 institutions were retrospectively included. All patients from institution 1 were randomized at a 7:3 ratio into a training cohort (n = 108) and an internal validation cohort (n = 45), and patients from institution 2 were used as an external validation cohort (n = 67). The association between the radiomics features and early recurrence was assessed in the training cohort and verified in the validation cohort. Furthermore, the performance of the radiomics nomogram was evaluated by combining the rad-score and clinical risk factors. The predictive performance was evaluated by receiver operating characteristic curve analysis, calibration curve analysis, and decision curve analysis. Results The dierenees in the Lymphocyte/monocyte ratio (LMR) and Peripheral nerve infiltration (PNI) were statistically significant between the early recurrence in BRAF-mutant colorectal cancer groups and the early non-recurrence in BRAF-mutant colorectal cancer groups (P < 0.05); The two groups showed no significant differenee in clinical parameters including age, sex, and biochemistry serum markers (P > 0.05). Comparing with the pure radiomics or clinical data, combined models can be seen that the addition of LMR and PNI further improveed the predictive efficiency of the model. The rad-score based on LR, generated by 4 selected radiomics features, demonstrated a favorable ability to predict early recurrence in both the training (AUC 0.81), internal validation (AUC 0.73), external validation (AUC 0.63) cohorts. Subsequently, integrating two independent predictors into a nomogram exhibited more favorable discriminatory performance, with the AUC improved to 0.88 and 0.81 in both cohorts. Conclusions The proposed CT-based radiomics signature is associated with early recurrence among the patients with BRAF-mutant colorectal cancer. The present study also proposes a combined model can potentially be applied in the individual preoperative prediction of early recurrence in patients with BRAF-mutant colorectal cancer. Advances in knowledge CT-based radiomics showed satisfactory diagnostic significance for early recurrence in patients with BRAF-mutant colorectal cancer. Key baseline clinical characteristics were associated with early recurrence in patients with BRAF-mutant colorectal cancer. The combined model may be applied in the individual preoperative prediction of early recurrence in patients with BRAF-mutant colorectal cancer.
Collapse
Affiliation(s)
- Zhenbin Jin
- Department of Radiology, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, #60 Youth Middle Road, Chongchuan District, Nantong, 226000, Jiangsu, China
| | - Qin Zou
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Taohu Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Ting Xue
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
217
|
Lu H, Yuan Y, Liu M, Li Z, Ma X, Xia Y, Shi F, Lu Y, Lu J, Shen F. Predicting pathological complete response following neoadjuvant chemoradiotherapy (nCRT) in patients with locally advanced rectal cancer using merged model integrating MRI-based radiomics and deep learning data. BMC Med Imaging 2024; 24:289. [PMID: 39448917 PMCID: PMC11515279 DOI: 10.1186/s12880-024-01474-3] [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: 06/20/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND To construct and compare merged models integrating clinical factors, MRI-based radiomics features and deep learning (DL) models for predicting pathological complete response (pCR) to neoadjuvant chemoradiotherapy (nCRT) in patients with locally advanced rectal cancer (LARC). METHODS Totally 197 patients with LARC administered surgical resection after nCRT were assigned to cohort 1 (training and test sets); meanwhile, 52 cases were assigned to cohort 2 as a validation set. Radscore and DL models were established for predicting pCR applying pre- and post-nCRT MRI data, respectively. Different merged models integrating clinical factors, Radscore and DL model were constituted. Their predictive performances were validated and compared by receiver operating characteristic (ROC) and decision curve analyses (DCA). RESULTS Merged models were established integrating selected clinical factors, Radscore and DL model for pCR prediction. The areas under the ROC curves (AUCs) of the pre-nCRT merged model were 0.834 (95% CI: 0.737-0.931) and 0.742 (95% CI: 0.650-0.834) in test and validation sets, respectively. The AUCs of the post-nCRT merged model were 0.746 (95% CI: 0.636-0.856) and 0.737 (95% CI: 0.646-0.828) in test and validation sets, respectively. DCA showed that the pretreatment algorithm could yield enhanced clinically benefit than the post-nCRT approach. CONCLUSIONS The pre-nCRT merged model including clinical factors, Radscore and DL model constitutes an effective non-invasive tool for pCR prediction in LARC.
Collapse
Affiliation(s)
- Haidi Lu
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yuan Yuan
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Minglu Liu
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Zhihui Li
- Department of Radiology, RuiJin Hospital LuWan Branch, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaolu Ma
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yuwei Xia
- Shanghai United Imaging Intelligence, Co., Ltd, Shanghai, China
| | - Feng Shi
- Shanghai United Imaging Intelligence, Co., Ltd, Shanghai, China
| | - Yong Lu
- Department of Radiology, RuiJin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| | - Jianping Lu
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Fu Shen
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
218
|
Laukhtina E, Muin D, Shariat SF. Imaging for upper tract urothelial carcinoma: update of the evidence and a glimpse into the future. Curr Opin Urol 2024:00042307-990000000-00202. [PMID: 39444272 DOI: 10.1097/mou.0000000000001241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
PURPOSE OF REVIEW Upper tract urothelial carcinoma (UTUC) is a rare malignancy posing significant diagnostic and management challenges. This review provides an overview of the evidence supporting various imaging modalities and offers insights into future innovations in UTUC imaging. RECENT FINDINGS With the growing use of advancements in computed tomography (CT) technologies for both staging and follow-up of UTUC patients, continuous innovations aim to enhance performance and minimize the risk of excessive exposure to ionizing radiation and iodinated contrast medium. In patients unable to undergo CT, magnetic resonance imaging serves as an alternative imaging modality, though its sensitivity is lower than CT. Positron emission tomography, particularly with innovative radiotracers and theranostics, has the potential to significantly advance precision medicine in UTUC. Endoscopic imaging techniques including advanced modalities seem to be promising in improved visualization and diagnostic accuracy, however, evidence remains scarce. Radiomics and radiogenomics present emerging tools for noninvasive tumor characterization and prognosis. SUMMARY The landscape of imaging for UTUC is rapidly evolving, with significant advancements across various modalities promising improved diagnostic accuracy, patient outcomes, and safety.
Collapse
Affiliation(s)
- Ekaterina Laukhtina
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna
| | - Dina Muin
- Department of Biomedical Imaging and Image-guided Therapy, Devision of Nuclear Medicine, Medical University of Vienna
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna
- Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria
- Department of Urology, Weill Cornell Medical College, New York, New York
- Department of Urology, University of Texas Southwestern, Dallas, Texas, USA
- Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan
| |
Collapse
|
219
|
D’Anna A, Stella G, Gueli AM, Marino C, Pulvirenti A. Mitigating Interobserver Variability in Radiomics with ComBat: A Feasibility Study. J Imaging 2024; 10:270. [PMID: 39590734 PMCID: PMC11595722 DOI: 10.3390/jimaging10110270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
This study investigates Intraobserver Features Variability (IFV) in radiomics studies and assesses the effectiveness of the ComBat harmonization method in mitigating these effects. Methods: This study utilizes data from the NSCLC-Radiomics-Interobserver1 dataset, comprising CT scans of 22 Non-Small Cell Lung Cancer (NSCLC) patients, with multiple Gross Tumor Volume (GTV) delineations performed by five radiation oncologists. Segmentation was completed manually ("vis") or by autosegmentation with manual editing ("auto"). A total of 1229 radiomic features were extracted from each GTV, segmentation method, and oncologist. Features extracted included first order, shape, GLCM, GLRLM, GLSZM, and GLDM from original, wavelet-filtered, and LoG-filtered images. Results: Before implementing ComBat harmonization, 83% of features exhibited p-values below 0.05 in the "vis" approach; this percentage decreased to 34% post-harmonization. Similarly, for the "auto" approach, 75% of features demonstrated statistical significance prior to ComBat, but this figure declined to 33% after its application. Among a subset of three expert radiation oncologists, percentages changed from 77% to 25% for "vis" contouring and from 64% to 23% for "auto" contouring. This study demonstrates that ComBat harmonization could effectively reduce IFV, enhancing the feasibility of multicenter radiomics studies. It also highlights the significant impact of physician experience on radiomics analysis outcomes.
Collapse
Affiliation(s)
- Alessia D’Anna
- Department of Physics and Astronomy “E. Majorana”, University of Catania, Via Santa Sofia 64, 95123 Catania, Italy; (A.D.); (A.M.G.)
| | - Giuseppe Stella
- Department of Physics and Astronomy “E. Majorana”, University of Catania, Via Santa Sofia 64, 95123 Catania, Italy; (A.D.); (A.M.G.)
| | - Anna Maria Gueli
- Department of Physics and Astronomy “E. Majorana”, University of Catania, Via Santa Sofia 64, 95123 Catania, Italy; (A.D.); (A.M.G.)
| | - Carmelo Marino
- Department of Medical Phyisics, Humanitas Istituto Clinico Catanese (H-ICC), Contrada Cubba S.P. 54 n.11, 95045 Misterbianco, Italy;
| | - Alfredo Pulvirenti
- Department of Clinical and Experimental Medicine, University of Catania, Via Santa Sofia 64, 95123 Catania, Italy;
| |
Collapse
|
220
|
de Bloeme CM, Jansen RW, Cardoen L, Göricke S, van Elst S, Jessen JL, Ramasubramanian A, Skalet AH, Miller AK, Maeder P, Uner OE, Hubbard GB, Grossniklaus H, Boldt HC, Nichols KE, Brennan RC, Sen S, Koob M, Sirin S, Brisse HJ, Galluzzi P, Dommering CJ, Cysouw M, Boellaard R, Dorsman JC, Moll AC, de Jong MC, de Graaf P. Differentiating MYCN-amplified RB1 wild-type retinoblastoma from biallelic RB1 mutant retinoblastoma using MR-based radiomics: a retrospective multicenter case-control study. Sci Rep 2024; 14:25103. [PMID: 39443629 PMCID: PMC11499940 DOI: 10.1038/s41598-024-76933-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024] Open
Abstract
MYCN-amplified RB1 wild-type (MYCNampRB1+/+) retinoblastoma is a rare and aggressive subtype, often resistant to standard therapies. Identifying unique MRI features is crucial for diagnosing this subtype, as biopsy is not recommended. This study aimed to differentiate MYCNampRB1+/+ from the most prevalent RB1-/- retinoblastoma using pretreatment MRI and radiomics. Ninety-eight unilateral retinoblastoma patients (19 MYCN cases and 79 matched controls) were included. Tumors on T2-weighted MR images were manually delineated and validated by experienced radiologists. Radiomics analysis extracted 120 features per tumor. Several combinations of feature selection methods, oversampling techniques and machine learning (ML) classifiers were evaluated in a repeated fivefold cross-validation machine learning pipeline to yield the best-performing prediction model for MYCN. The best model used univariate feature selection, data oversampling (duplicating MYCN cases), and logistic regression classifier, achieving a mean AUC of 0.78 (SD 0.12). SHAP analysis highlighted lower sphericity, higher flatness, and greater gray-level heterogeneity as predictive for MYCNampRB1+/+ status, yielding an AUC of 0.81 (SD 0.11). This study shows the potential of MRI-based radiomics to distinguish MYCNampRB1+/+ and RB1-/- retinoblastoma subtypes.
Collapse
Affiliation(s)
- Christiaan M de Bloeme
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
- Department of Radiology and Nuclear Medicine, UMC location Vrije Universiteit Amsterdam, Amsterdam, 1007 MB, The Netherlands.
| | - Robin W Jansen
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, UMC location Vrije Universiteit Amsterdam, Amsterdam, 1007 MB, The Netherlands
| | - Liesbeth Cardoen
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands
- Imaging department Institut Curie, France and Paris-Siences-et-Lettres University, Paris, France
| | - Sophia Göricke
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Sabien van Elst
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, UMC location Vrije Universiteit Amsterdam, Amsterdam, 1007 MB, The Netherlands
| | | | | | - Alison H Skalet
- Casey Eye Institute Oregon Health & Science University , Portland, United States
- Knight Cancer Institute Oregon Health & Science University, Portland, United States
| | - Audra K Miller
- Casey Eye Institute Oregon Health & Science University , Portland, United States
| | - Philippe Maeder
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands
- Department of Radiology Centre Hospitalier Universitaire Vaudois (CHUV) , University of Lausanne , Lausanne, Switzerland
| | - Ogul E Uner
- Casey Eye Institute Oregon Health & Science University , Portland, United States
- Emory Eye Center Ocular Oncology Service, Atlanta, United States
| | - G Baker Hubbard
- Emory Eye Center Ocular Oncology Service, Atlanta, United States
| | | | - H Culver Boldt
- Department of Ophthalmology, University of Iowa Hospitals & Clinics, Iowa, United States
| | - Kim E Nichols
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, United States
| | - Rachel C Brennan
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, United States
- Department of Pediatric Hematology/Oncology, Logan Health, Kalispell (Montana), United States
| | - Saugata Sen
- Department of Radiology and Imaging Sciences, Tata Medical Center, Kolkata, India
| | - Mériam Koob
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands
- Department of Radiology Centre Hospitalier Universitaire Vaudois (CHUV) , University of Lausanne , Lausanne, Switzerland
| | - Selma Sirin
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands
- Department of Diagnostic Imaging, University Children's Hospital Zurich University of Zurich, Zurich, Switzerland
| | - Hervé J Brisse
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands
- Imaging department Institut Curie, France and Paris-Siences-et-Lettres University, Paris, France
| | - Paolo Galluzzi
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands
- Department of Neuroimaging unit, Siena University Hospital, Siena, Italy
| | - Charlotte J Dommering
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
| | - Matthijs Cysouw
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, UMC location Vrije Universiteit Amsterdam, Amsterdam, 1007 MB, The Netherlands
| | - Ronald Boellaard
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, UMC location Vrije Universiteit Amsterdam, Amsterdam, 1007 MB, The Netherlands
| | - Josephine C Dorsman
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Oncogenetics, UMC location Vrije Universiteit, Amsterdam, The Netherlands
| | - Annette C Moll
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Ophthalmology, UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marcus C de Jong
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, UMC location Vrije Universiteit Amsterdam, Amsterdam, 1007 MB, The Netherlands
| | - Pim de Graaf
- European Retinoblastoma Imaging Collaboration (ERIC), Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, UMC location Vrije Universiteit Amsterdam, Amsterdam, 1007 MB, The Netherlands
| |
Collapse
|
221
|
Chhabra R. Molecular and modular intricacies of precision oncology. Front Immunol 2024; 15:1476494. [PMID: 39507541 PMCID: PMC11537923 DOI: 10.3389/fimmu.2024.1476494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Precision medicine is revolutionizing the world in combating different disease modalities, including cancer. The concept of personalized treatments is not new, but modeling it into a reality has faced various limitations. The last decade has seen significant improvements in incorporating several novel tools, scientific innovations and governmental support in precision oncology. However, the socio-economic factors and risk-benefit analyses are important considerations. This mini review includes a summary of some commendable milestones, which are not just a series of successes, but also a cautious outlook to the challenges and practical implications of the advancing techno-medical era.
Collapse
Affiliation(s)
- Ravneet Chhabra
- Business Department, Biocytogen Boston Corporation, Waltham, MA, United States
| |
Collapse
|
222
|
Brunetti N, Campi C, Biddau G, Piana M, Picone I, Conti B, Cesano S, Starovatskyi O, Bozzano S, Rescinito G, Tosto S, Garlaschi A, Calabrese M, Stefano Tagliafico A. Radiomic and clinical model for predicting atypical ductal hyperplasia upgrades and potentially reduce unnecessary surgical treatments. Eur J Radiol 2024; 181:111799. [PMID: 39454425 DOI: 10.1016/j.ejrad.2024.111799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/09/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
OBJECTIVE To identify patients with atypical ductal hyperplasia (ADH) at low risk of upgrading to carcinoma. This study aims to assess the performance of radiomics combined with clinical factors to predict occult breast cancer among women diagnosed with ADH. METHODS This study retrospectively included microcalcification clusters of patients who underwent Mx and VABB with a diagnosis of ADH at a tertiary center from January 2015 to May 2023. Clinical and radiological data (age, cluster size, BI-RADS classification, mammographic density, breast cancer history, residual microcalcifications) were collected. Surgical outcomes were used to determine upgrade. Four logistic regression models were developed to predict the risk of upgrade. The performance was evaluated using the area under the receiver operating characteristic curve (AUC) and performance scores. RESULTS A total of 143 patients with 153 clusters were included. Twelve radiomic features and six clinical factors were selected for model development. The sample was split into 107 training and 46 test cases. Clinical features achieved an AUC of 0.72 (0.60-0.84), radiomic features an AUC of 0.73 (0.61-0.85). Radiomic features with "cluster size" and "age" improved the AUC to 0.79 (0.67-0.91). The best model, incorporating all data, achieved an AUC of 0.82 (0.71-0.92), a specificity of 0.89 (0.75, 0.97), and NPV of 0.92 (0.78-0.98). CONCLUSION This study demonstrates the potential of radiomic as a valuable tool for reducing unnecessary treatments for patient classified as "low risk of ADH upgrade". Combining radiomic information with clinical data improved the accuracy of risk prediction.
Collapse
Affiliation(s)
- Nicole Brunetti
- Department of Radiology, IRCCS - Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy; Department of Experimental Medicine (DIMES), University of Genova, Via L.B. Alberti 2, 16132 Genoa, Italy.
| | - Cristina Campi
- Department of Mathematics (DIMA), University of Genoa, Via Dodecaneso 35, 16146 Genoa, Italy; Life Science Computational Laboratory (LISCOMP), IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Giorgia Biddau
- Department of Mathematics (DIMA), University of Genoa, Via Dodecaneso 35, 16146 Genoa, Italy
| | - Michele Piana
- Department of Mathematics (DIMA), University of Genoa, Via Dodecaneso 35, 16146 Genoa, Italy; Life Science Computational Laboratory (LISCOMP), IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Ilaria Picone
- Radiology Section, Department of Health Sciences (DISSAL), University of Genova, Via L.B. Alberti 2, 16132 Genoa, Italy
| | - Benedetta Conti
- Radiology Section, Department of Health Sciences (DISSAL), University of Genova, Via L.B. Alberti 2, 16132 Genoa, Italy
| | - Sara Cesano
- Radiology Section, Department of Health Sciences (DISSAL), University of Genova, Via L.B. Alberti 2, 16132 Genoa, Italy
| | - Oleksandr Starovatskyi
- Scientific Director Office, IRCCS - Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Silvia Bozzano
- Division of Anatomical Pathology, Department of Integrated Surgical and Diagnostic Sciences (DISC), Viale Benedetto XV, 16132 Genoa, Italy
| | - Giuseppe Rescinito
- Department of Radiology, IRCCS - Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Simona Tosto
- Department of Radiology, IRCCS - Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Alessandro Garlaschi
- Department of Radiology, IRCCS - Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Massimo Calabrese
- Department of Radiology, IRCCS - Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Alberto Stefano Tagliafico
- Department of Radiology, IRCCS - Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy; Radiology Section, Department of Health Sciences (DISSAL), University of Genova, Via L.B. Alberti 2, 16132 Genoa, Italy
| |
Collapse
|
223
|
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
|
224
|
Barioni ED, Lopes SLPDC, Silvestre PR, Yasuda CL, Costa ALF. Texture Analysis in Volumetric Imaging for Dentomaxillofacial Radiology: Transforming Diagnostic Approaches and Future Directions. J Imaging 2024; 10:263. [PMID: 39590727 PMCID: PMC11595357 DOI: 10.3390/jimaging10110263] [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: 09/21/2024] [Revised: 10/19/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024] Open
Abstract
This narrative review explores texture analysis as a valuable technique in dentomaxillofacial diagnosis, providing an advanced method for quantification and characterization of different image modalities. The traditional imaging techniques rely primarily on visual assessment, which may overlook subtle variations in tissue structure. In contrast, texture analysis uses sophisticated algorithms to extract quantitative information from imaging data, thus offering deeper insights into the spatial distribution and relationships of pixel intensities. This process identifies unique "texture signatures", serving as markers for accurately characterizing tissue changes or pathological processes. The synergy between texture analysis and radiomics allows radiologists to transcend traditional size-based or semantic descriptors, offering a comprehensive understanding of imaging data. This method enhances diagnostic accuracy, particularly for the assessment of oral and maxillofacial pathologies. The integration of texture analysis with radiomics expands the potential for precise tissue characterization by moving beyond the limitations of human eye evaluations. This article reviews the current trends and methodologies in texture analysis within the field of dentomaxillofacial imaging, highlights its practical applications, and discusses future directions for research and dental clinical practice.
Collapse
Affiliation(s)
- Elaine Dinardi Barioni
- Postgraduate Program in Dentistry, Cruzeiro do Sul University (UNICSUL), São Paulo 1506-000, SP, Brazil;
| | - Sérgio Lúcio Pereira de Castro Lopes
- Department of Diagnosis and Surgery, São José dos Campos School of Dentistry, São Paulo State University (UNESP), São José dos Campos 2245-000, SP, Brazil; (S.L.P.d.C.L.); (P.R.S.)
| | - Pedro Ribeiro Silvestre
- Department of Diagnosis and Surgery, São José dos Campos School of Dentistry, São Paulo State University (UNESP), São José dos Campos 2245-000, SP, Brazil; (S.L.P.d.C.L.); (P.R.S.)
| | - Clarissa Lin Yasuda
- Laboratory of Neuroimaging, Department of Neurology, University of Campinas (UNICAMP), Campinas 13083-970, SP, Brazil;
| | - Andre Luiz Ferreira Costa
- Postgraduate Program in Dentistry, Cruzeiro do Sul University (UNICSUL), São Paulo 1506-000, SP, Brazil;
| |
Collapse
|
225
|
Doganay MT, Chakraborty P, Bommakanti SM, Jammalamadaka S, Battalapalli D, Madabhushi A, Draz MS. Artificial intelligence performance in testing microfluidics for point-of-care. LAB ON A CHIP 2024; 24:4998-5008. [PMID: 39360887 PMCID: PMC11448392 DOI: 10.1039/d4lc00671b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/16/2024] [Indexed: 10/06/2024]
Abstract
Artificial intelligence (AI) is revolutionizing medicine by automating tasks like image segmentation and pattern recognition. These AI approaches support seamless integration with existing platforms, enhancing diagnostics, treatment, and patient care. While recent advancements have demonstrated AI superiority in advancing microfluidics for point of care (POC) diagnostics, a gap remains in comparative evaluations of AI algorithms in testing microfluidics. We conducted a comparative evaluation of AI models specifically for the two-class classification problem of identifying the presence or absence of bubbles in microfluidic channels under various imaging conditions. Using a model microfluidic system with a single channel loaded with 3D transparent objects (bubbles), we challenged each of the tested machine learning (ML) (n = 6) and deep learning (DL) (n = 9) models across different background settings. Evaluation revealed that the random forest ML model achieved 95.52% sensitivity, 82.57% specificity, and 97% AUC, outperforming other ML algorithms. Among DL models suitable for mobile integration, DenseNet169 demonstrated superior performance, achieving 92.63% sensitivity, 92.22% specificity, and 92% AUC. Remarkably, DenseNet169 integration into a mobile POC system demonstrated exceptional accuracy (>0.84) in testing microfluidics at under challenging imaging settings. Our study confirms the transformative potential of AI in healthcare, emphasizing its capacity to revolutionize precision medicine through accurate and accessible diagnostics. The integration of AI into healthcare systems holds promise for enhancing patient outcomes and streamlining healthcare delivery.
Collapse
Affiliation(s)
- Mert Tunca Doganay
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Purbali Chakraborty
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Sri Moukthika Bommakanti
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Soujanya Jammalamadaka
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | | | - Anant Madabhushi
- Department of Biomedical Engineering, Emory University, Atlanta, GA, USA
- Atlanta Veterans Administration Medical Center, Atlanta, GA, USA
| | - Mohamed S Draz
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, 44106, USA
| |
Collapse
|
226
|
Yang C, Wu M, Zhang J, Qian H, Fu X, Yang J, Luo Y, Qin Z, Shi T. Radiomics based on MRI in predicting lymphovascular space invasion of cervical cancer: a meta-analysis. Front Oncol 2024; 14:1425078. [PMID: 39484029 PMCID: PMC11524797 DOI: 10.3389/fonc.2024.1425078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/26/2024] [Indexed: 11/03/2024] Open
Abstract
Objective The objective of this meta-analysis is to assess the efficacy of radiomics techniques utilizing magnetic resonance imaging (MRI) for predicting lymphovascular space invasion (LVSI) in patients with cervical cancer (CC). Methods A comprehensive literature search was conducted in databases including PubMed, Embase, Cochrane Library, Medline, Scopus, CNKI, and Wanfang, with studies published up to 08/04/2024, being considered for inclusion. The meta-analysis was performed using Stata 15 and Review Manager 5.4. The quality of the included studies was evaluated using the Quality Assessment of Diagnostic Accuracy Studies 2 and Radiomics Quality Score tools. The analysis encompassed the pooled sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), and diagnostic odds ratio (DOR). Summary ROC curves were constructed, and the AUC was calculated. Heterogeneity was investigated using meta-regression. Statistical significance was set at p ≤ 0.05. Results There were 13 studies involving a total of 2,245 patients that were included in the meta-analysis. The overall sensitivity and specificity of the MRI-based model in the Training set were 83% (95% CI: 77%-87%) and 72% (95% CI: 74%-88%), respectively. The AUC, DOR, PLR, and NLR of the MRI-based model in the Training set were 0.89 (95% CI: 0.86-0.91), 22 (95% CI: 12-40), 4.6 (95% CI: 3.1-7.0), and 0.21 (95% CI: 0.16-0.29), respectively. Subgroup analysis revealed that the AUC of the model combining radiomics with clinical factors [0.90 (95% CI: 0.87-0.93)] was superior to models based on T2-weighted imaging (T2WI) sequence [0.78 (95% CI: 0.74-0.81)], contrast-enhanced T1-weighted imaging (T1WI-CE) sequence [0.85 (95% CI: 0.82-0.88)], and multiple sequences [0.86 (95% CI: 0.82-0.89)] in the Training set. The pooled sensitivity and specificity of the model integrating radiomics with clinical factors [83% (95% CI: 73%-89%) and 86% (95% CI: 73%-93%)] surpassed those of models based on the T2WI sequence [79% (95% CI: 71%-85%) and 72% (95% CI: 67%-76%)], T1WI-CE sequence [78% (95% CI: 67%-86%) and 78% (95% CI: 68%-86%)], and multiple sequences [78% (95% CI: 67%-87%) and 79% (95% CI: 70%-87%)], respectively. Funnel plot analysis indicated an absence of publication bias (p > 0.05). Conclusion MRI-based radiomics demonstrates excellent diagnostic performance in predicting LVSI in CC patients. The diagnostic performance of models combing radiomics and clinical factors is superior to that of models utilizing radiomics alone. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/#myprospero, identifier CRD42024538007.
Collapse
Affiliation(s)
- Chongshuang Yang
- Department of Radiology, Tongren People’s Hospital, Tongren, China
| | - Min Wu
- Department of Radiology, Tongren People’s Hospital, Tongren, China
| | - Jiancheng Zhang
- Department of Radiology, Tongren People’s Hospital, Tongren, China
| | - Hongwei Qian
- Department of Radiology, Tongren People’s Hospital, Tongren, China
| | - Xiangyang Fu
- Department of Radiology, Tongren People’s Hospital, Tongren, China
- Department of Radiology, Wanshan District People’s Hospital, Tongren, China
| | - Jing Yang
- Department of Radiology, Tongren People’s Hospital, Tongren, China
| | - Yingbin Luo
- Department of Radiology, Tongren People’s Hospital, Tongren, China
| | - Zhihong Qin
- Department of Radiology, Tongren People’s Hospital, Tongren, China
| | - Tianliang Shi
- Department of Radiology, Tongren People’s Hospital, Tongren, China
| |
Collapse
|
227
|
Liu Y, Wang Z, Yang L, Zhang M, Li M, Zhang J, Tang L, Jiang Z, Li X, Deng J, Meng Q, Liu S, Wang K, Qi L. Identification of a rank-based radiomic signature with individualized prognostic value for lung adenocarcinoma in a multi-cohort study. Eur J Radiol 2024; 181:111782. [PMID: 39427495 DOI: 10.1016/j.ejrad.2024.111782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
OBJECTIVES Radiomics provides an opportunity to evaluate cancer prognosis noninvasively. However, the susceptibility of the radiomic quantitative features to multicenter effects, leads to the clinical dilemma of the radiomic signatures. This study aimed to develop a radiomic signature to circumvent multicenter effects, achieving the individualized prognostic assessment of lung adenocarcinoma (LUAD). METHODS Using computed tomography (CT) imaging of 234 stage I-IIIA LUAD patients derived from three public multicenter cohorts, we proposed a rank-based method that utilized the relative rank patterns of quantitative values between radiomic feature pairs within individual patients and established a feature pair signature for LUAD prognosis. We collected a new clinical cohort with 162 LUAD patients for independent validation. RESULTS A rank-based radiomic signature, consisting of 12 feature pairs, was developed, and it could determine the mortality risk for an individual according to the rank patterns of 12 feature pairs within the patient's CT imaging. The prognostic performance of the rank-based signature was effectively validated in the new clinical cohort (log-rank P = 0.0051, C-index = 0.73), whereas other signatures lost their prognostic ability across centers. The novel proposed radiomic nomogram significantly improved the prognostic performance of clinicopathological factors. The further radiogenomic analyses revealed the underlying biological characteristics (e.g., Stemness, Ferroptosis, 'ECM') reflected by the rank-based radiomic signature. CONCLUSIONS This multicenter study illustrates the accuracy and stability of the rank-based radiomic signature for LUAD prognosis, and demonstrates a unique advantage of clinical individualized application. The biological characteristics underlying the rank-based radiomic signature would accelerate its clinical application.
Collapse
Affiliation(s)
- Yixin Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China; Modern Education Technology Center, Harbin Medical University, Harbin, China
| | - Zhihui Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Liping Yang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, China
| | - Meng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Mengyue Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Juxuan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Lefan Tang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Zhiyun Jiang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Jiaxing Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Qingwei Meng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, China
| | - Shilong Liu
- Department of Thoracic Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin 150086, China.
| | - Kezheng Wang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Lishuang Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
228
|
Li W, Xiao J, Zhang C, Di X, Yao J, Li X, Huang J, Li Z. Pathomics models for CD40LG expression and prognosis prediction in glioblastoma. Sci Rep 2024; 14:24350. [PMID: 39420038 PMCID: PMC11487080 DOI: 10.1038/s41598-024-75018-8] [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: 04/14/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
Glioblastoma (GBM) is the most prevalent primary malignant tumor of the nervous system. In this study, we utilized pathomics analysis to explore the expression of CD40LG and its predictive value for the prognosis of GBM patients. We analyzed the expression differences of CD40LG in GBM tissue and normal brain tissue, along with performing survival prognosis analysis. Additionally, histopathological sections of GBM were used to screen for pathological features. Subsequently, SVM and LR pathomics models were constructed, and the models' performance was evaluated. The pathomics model was employed to predict CD40LG expression and patient prognosis. Furthermore, we investigated the potential molecular mechanisms through enrichment analysis, WGCNA analysis, immune correlation analysis, and immune checkpoint analysis. The expression level of CD40LG was significantly increased in GBM. Multivariate analysis demonstrated that high expression of CD40LG is a risk factor for overall survival (OS) in GBM patients. Five pathological features were identified, and SVM and LR pathomics models were constructed. Model evaluation showed promising predictive effects, with an AUC value of 0.779 for the SVM model, and the Hosmer-Lemeshow test confirmed the model's prediction probability consistency (P > 0.05). The LR model achieved an AUC value of 0.785, and the Hosmer-Lemeshow test indicated good agreement between the LR model's predicted probabilities and the true value (P > 0.05). Immune infiltration analysis revealed a significant correlation between the pathomics score (PS) and the degree of infiltration of activated DC cells, T cells CD4 naïve, Macrophages M2, Macrophages M1, and T cells CD4 memory resting. Our results demonstrate that the pathomics model exhibits predictability for CD40LG expression and GBM patient survival. These findings can be utilized to assist neurosurgeons in selecting optimal treatment strategies in clinical practice.
Collapse
Affiliation(s)
- Wenle Li
- Department of Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Jianqi Xiao
- Department of Neurosurgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Chunyu Zhang
- Department of Neurosurgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Xiaoqing Di
- Pathological Diagnosis and Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Jieqin Yao
- Department of Neurosurgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Xiaopeng Li
- Department of Neurosurgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Jincheng Huang
- Department of Neurosurgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Zhenzhe Li
- Department of Neurosurgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China.
| |
Collapse
|
229
|
Ende TVD, Kuijper SC, Widaatalla Y, Noortman WA, van Velden FHP, Woodruff HC, van der Pol Y, Moldovan N, Pegtel DM, Derks S, Bijlsma MF, Mouliere F, de Geus-Oei LF, Lambin P, van Laarhoven HWM. Integrating Clinical Variables, Radiomics, and Tumor-derived Cell-Free DNA for Enhanced Prediction of Resectable Esophageal Adenocarcinoma Outcomes. Int J Radiat Oncol Biol Phys 2024:S0360-3016(24)03468-0. [PMID: 39424077 DOI: 10.1016/j.ijrobp.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/13/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
PURPOSE The value of integrating clinical variables, radiomics, and tumor-derived cell-free DNA (cfDNA) for the prediction of survival and response to chemoradiation of patients with resectable esophageal adenocarcinoma is not yet known. Our aim was to investigate if radiomics and cfDNA metrics combined with clinical variables can improve personalized predictions. METHODS AND MATERIALS A cohort of 111 patients with resectable esophageal adenocarcinoma from 2 centers treated with neoadjuvant chemoradiation therapy was used for exploratory retrospective analyses. Models combining the clinical variables of the SOURCE survival model with radiomic features and cfDNA were built using elastic net regression and internally validated using 5-fold cross-validation. Model performance for overall survival (OS) and time to progression (TTP) were evaluated with the C-index and the area under the curve for pathologic complete response. RESULTS The best-performing baseline models for OS and TTP were based on the combination of SOURCE-cfDNA that reached a C-index of 0.55 and 0.59 compared with 0.44 to 0.45 with SOURCE alone. The addition of restaging positron emission tomography radiomics to SOURCE was the most promising addition for predicting OS (C-index: 0.65) and TTP (C-index: 0.60). Baseline risk stratification was achieved for OS and TTP by combining SOURCE with radiomics or cfDNA, log-rank P < .01. The best-performing combination model for the prediction of pathologic complete response reached an area under the curve of 0.61 compared with 0.47 with SOURCE variables alone. CONCLUSIONS The addition of radiomics and cfDNA can improve the performance of an established survival model. External validity needs to be further assessed in future studies together with the optimization of radiomic pipelines.
Collapse
Affiliation(s)
- Tom van den Ende
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Steven C Kuijper
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Yousif Widaatalla
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Wyanne A Noortman
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands; TechMed Centre, University of Twente, Enschede, The Netherlands
| | - Floris H P van Velden
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Henry C Woodruff
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ymke van der Pol
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Norbert Moldovan
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - D Michiel Pegtel
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sarah Derks
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Maarten F Bijlsma
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands; Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Florent Mouliere
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands; TechMed Centre, University of Twente, Enschede, The Netherlands; Department of Radiation Science & Technology, Delft University of Technology, Delft., The Netherlands
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| |
Collapse
|
230
|
Yan L, Chen Y, He J. Leveraging MRI radiomics signature for predicting the diagnosis of CXCL9 in breast cancer. Heliyon 2024; 10:e38640. [PMID: 39430466 PMCID: PMC11490775 DOI: 10.1016/j.heliyon.2024.e38640] [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: 02/24/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/22/2024] Open
Abstract
Objective A non-invasive predictive model was developed using radiomic features to forecast CXCL9 expression level in breast cancer patients. Methods CXCL9 expression data and MRI images of breast cancer patients were obtained from The Cancer Genome Atlas (TCGA) and The Cancer Imaging Archive (TCIA) databases, respectively. Local tissue samples from 20 breast cancer patients were collected to measure CXCL9 expression levels. Radiomic features were extracted from MRI images using 3DSlicer, and the minimum Redundancy Maximum Relevance and Recursive Feature Elimination (mRMR_RFE) method was employed to select the most pertinent radiomic features associated with CXCL9 expression levels. Support vector machine (SVM) and Logistic Regression (LR) models were utilized to construct the predictive model, and the area under the receiver operating characteristic curve (AUC) was calculated for performance evaluation. Results CXCL9 was found to be upregulated in breast cancer patients and linked to breast cancer prognosis. Nine radiomic features were ultimately selected using the mRMR_RFE method, and SVM and LR models were trained and validated. The SVM model achieved AUC values of 0.748 and 0.711 in the training and validation sets, respectively. The LR model obtained AUC values of 0.771 and 0.724 in the training and validation sets, respectively. Conclusion The utilization of MRI radiomic features for predicting CXCL9 expression level provides a novel non-invasive approach for breast cancer Prognostic research.
Collapse
Affiliation(s)
- Liping Yan
- Department of Breast Surgery, Maternal and Child Health Hospital of Jiangxi Province, Nanchang, China
- Department of Surgery, the First Affiliated Hospital of Guangxi Medical University, China
| | - Yuexia Chen
- Department of Pathology, The Third Hospital of Nanchang, Nanchang, China
| | - Jianxin He
- Department of Ultrasound Medicine, The First Affiliated Hospital of Nanchang University, China
| |
Collapse
|
231
|
Xu R, Wang K, Peng B, Zhou X, Wang C, Lu T, Shi J, Zhao J, Zhang L. Evaluating peritumoral and intratumoral radiomics signatures for predicting lymph node metastasis in surgically resectable non-small cell lung cancer. Front Oncol 2024; 14:1427743. [PMID: 39464711 PMCID: PMC11502299 DOI: 10.3389/fonc.2024.1427743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 09/18/2024] [Indexed: 10/29/2024] Open
Abstract
Background Whether lymph node metastasis in non-small cell lung cancer is critical to clinical decision-making. This study was to develop a non-invasive predictive model for preoperative assessing lymph node metastasis in patients with non-small cell lung cancer (NSCLC) using radiomic features from chest CT images. Materials & methods In this retrospective study, 247 patients with resectable non-small cell lung cancer (NSCLC) were enrolled. These individuals underwent preoperative chest CT scans that identified lung nodules, followed by lobectomies and either lymph node sampling or dissection. We extracted both intratumoral and peritumoral radiomic features from the CT images, which were used as covariates to predict the lymph node metastasis status. By using ROC curves, Delong tests, Calibration curve, and DCA curves, intra-tumoral-peri-tumoral model performance were compared with models using only intratumoral features or clinical information. Finally, we constructed a model that combined clinical information and radiomic features to increase clinical applicability. Results This study enrolled 247 patients (117 male and 130 females). In terms of predicting lymph node metastasis, the intra-tumoral-peri-tumoral model (0.953, 95%CI 0.9272-0.9792) has a higher AUC compared to the intratumoral radiomics model (0.898, 95%CI 0.8553-0.9402) and the clinical model (0.818, 95%CI 0.7653-0.8709). The DeLong test shows that the performance of the Intratumoral and Peritumoral radiomics models is superior to that of the Intratumoral or clinical feature model (p <0.001). In addition, to increase the clinical applicability of the model, we combined the intratumoral-peritumoral model and clinical information to construct a nomogram. Nomograms still have good predictive performance. Conclusion The radiomics-based model incorporating both peritumoral and intratumoral features from CT images can more accurately predict lymph node metastasis in NSCLC than traditional methods.
Collapse
Affiliation(s)
- Ran Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Second Clinical Medical College, Harbin Medical University, Harbin, China
| | - Kaiyu Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Second Clinical Medical College, Harbin Medical University, Harbin, China
| | - Bo Peng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Second Clinical Medical College, Harbin Medical University, Harbin, China
| | - Xiang Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Second Clinical Medical College, Harbin Medical University, Harbin, China
| | - Chenghao Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Second Clinical Medical College, Harbin Medical University, Harbin, China
| | - Tong Lu
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaxin Shi
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Second Clinical Medical College, Harbin Medical University, Harbin, China
| | - Jiaying Zhao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Second Clinical Medical College, Harbin Medical University, Harbin, China
| | - Linyou Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
232
|
Du Y, Zhang S, Jia X, Zhang X, Li X, Pan L, Li Z, Niu G, Liang T, Guo H. Radiomics Biomarkers to Predict Checkpoint Inhibitor Pneumonitis in Non-small Cell Lung Cancer. Acad Radiol 2024:S1076-6332(24)00703-7. [PMID: 39395887 DOI: 10.1016/j.acra.2024.09.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/14/2024]
Abstract
RATIONALE AND OBJECTIVES Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of non-small cell lung cancer (NSCLC). However, immune-related adverse events still occur, of which checkpoint inhibitor pneumonitis (CIP) is the most common. We aimed to construct and validate a contrast-enhanced computed tomography-based radiomic nomogram to predict the probability of CIP before ICIs treatment in NSCLC. MATERIALS AND METHODS We retrospectively analyzed 685 patients with NSCLC who were initially treated with ICIs. A total of 186 patients were included in our study, and an additional 52 patients from another hospital were considered for external validation. After radiomics feature extraction and selection, we applied a support vector machine classification model to distinguish CIP and used the probability as a radiomics signature. A radiomics-clinical logistic regression model was built using the filtered clinical parameters and a radiomic signature. Receiver operating characteristic, area under the curve (AUC), calibration curve, and decision curve analysis was used for inter-model comparison. RESULTS The combined radiomics-clinical model constructed using age, interstitial lung disease, emphysema at baseline, and radiomics signature showed an AUC of 0.935, 0.905, and 0.923 for the training, validation, and external validation cohorts, respectively. Compared with the clinical-only (AUC of 0.829, 0.826, and 0.809) and radiomics-only models (0.865, 0.847, and 0.841), the radiomics-clinical displayed better predictive power. CONCLUSION This combined radiomics-clinical model predicted the probability of CIP during ICIs treatment in patients with NSCLC with favorable accuracy and could therefore be used as an effective tool to guide clinical ICIs decisions.
Collapse
Affiliation(s)
- Yonghao Du
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (Y.D., S.Z., G.N., T.L.)
| | - Shuo Zhang
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (Y.D., S.Z., G.N., T.L.)
| | - Xiaohui Jia
- Phase I Clinical Trial Ward, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi 710004, PR China (X.J., H.G.)
| | - Xi Zhang
- Department of Thoracic Surgery, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (X.Z.)
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (X.L.)
| | - Libo Pan
- Department of Radiology, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (L.P.)
| | - Zhihao Li
- Department of Pharmaceuticals Diagnostic, GE Healthcare, Xi'an, Shaanxi 710076, PR China (Z.L.)
| | - Gang Niu
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (Y.D., S.Z., G.N., T.L.)
| | - Ting Liang
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China (Y.D., S.Z., G.N., T.L.)
| | - Hui Guo
- Phase I Clinical Trial Ward, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi 710004, PR China (X.J., H.G.); Department of Medical Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi 710004, PR China (H.G.); Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China (H.G.); Key Laboratory of Surgical Critical Care and Life Support, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, Shaanxi 710061, PR China (H.G.).
| |
Collapse
|
233
|
Ammari S, Quillent A, Elvira V, Bidault F, Garcia GCTE, Hartl DM, Balleyguier C, Lassau N, Chouzenoux É. Using Machine Learning on MRI Radiomics to Diagnose Parotid Tumours Before Comparing Performance with Radiologists: A Pilot Study. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024:10.1007/s10278-024-01255-y. [PMID: 39390287 DOI: 10.1007/s10278-024-01255-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/31/2024] [Accepted: 08/19/2024] [Indexed: 10/12/2024]
Abstract
The parotid glands are the largest of the major salivary glands. They can harbour both benign and malignant tumours. Preoperative work-up relies on MR images and fine needle aspiration biopsy, but these diagnostic tools have low sensitivity and specificity, often leading to surgery for diagnostic purposes. The aim of this paper is (1) to develop a machine learning algorithm based on MR images characteristics to automatically classify parotid gland tumours and (2) compare its results with the diagnoses of junior and senior radiologists in order to evaluate its utility in routine practice. While automatic algorithms applied to parotid tumours classification have been developed in the past, we believe that our study is one of the first to leverage four different MRI sequences and propose a comparison with clinicians. In this study, we leverage data coming from a cohort of 134 patients treated for benign or malignant parotid tumours. Using radiomics extracted from the MR images of the gland, we train a random forest and a logistic regression to predict the corresponding histopathological subtypes. On the test set, the best results are given by the random forest: we obtain a 0.720 accuracy, a 0.860 specificity, and a 0.720 sensitivity over all histopathological subtypes, with an average AUC of 0.838. When considering the discrimination between benign and malignant tumours, the algorithm results in a 0.760 accuracy and a 0.769 AUC, both on test set. Moreover, the clinical experiment shows that our model helps to improve diagnostic abilities of junior radiologists as their sensitivity and accuracy raised by 6 % when using our proposed method. This algorithm may be useful for training of physicians. Radiomics with a machine learning algorithm may help improve discrimination between benign and malignant parotid tumours, decreasing the need for diagnostic surgery. Further studies are warranted to validate our algorithm for routine use.
Collapse
Affiliation(s)
- Samy Ammari
- Biomaps, UMR1281 INSERM, CEA, CNRS, Université Paris-Saclay, 94805, Villejuif, France
- Department of Imaging, Gustave Roussy Cancer Campus, Université Paris Saclay, 94805, Villejuif, France
| | - Arnaud Quillent
- Centre de Vision Numérique, OPIS, CentraleSupélec, Inria, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Víctor Elvira
- School of Mathematics, University of Edinburgh, Edinburgh, EH9 3FD, UK
| | - François Bidault
- Biomaps, UMR1281 INSERM, CEA, CNRS, Université Paris-Saclay, 94805, Villejuif, France
- Department of Imaging, Gustave Roussy Cancer Campus, Université Paris Saclay, 94805, Villejuif, France
| | - Gabriel C T E Garcia
- Department of Imaging, Gustave Roussy Cancer Campus, Université Paris Saclay, 94805, Villejuif, France
| | - Dana M Hartl
- Department of Otolaryngology Head and Neck Surgery, Gustave Roussy Cancer Campus, Université Paris Saclay, 94805, Villejuif, France
| | - Corinne Balleyguier
- Biomaps, UMR1281 INSERM, CEA, CNRS, Université Paris-Saclay, 94805, Villejuif, France
- Department of Imaging, Gustave Roussy Cancer Campus, Université Paris Saclay, 94805, Villejuif, France
| | - Nathalie Lassau
- Biomaps, UMR1281 INSERM, CEA, CNRS, Université Paris-Saclay, 94805, Villejuif, France
- Department of Imaging, Gustave Roussy Cancer Campus, Université Paris Saclay, 94805, Villejuif, France
| | - Émilie Chouzenoux
- Centre de Vision Numérique, OPIS, CentraleSupélec, Inria, Université Paris-Saclay, 91190, Gif-sur-Yvette, France.
| |
Collapse
|
234
|
Crimì F, Turatto F, D'Alessandro C, Sussan G, Iacobone M, Torresan F, Tizianel I, Campi C, Quaia E, Caccese M, Ceccato F. Texture analysis can predict response to etoposide-doxorubicin-cisplatin in patients with adrenocortical carcinoma. J Endocrinol Invest 2024:10.1007/s40618-024-02476-2. [PMID: 39382628 DOI: 10.1007/s40618-024-02476-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND The adrenocortical carcinoma (ACC) is a rare and highly aggressive malignancy originating from the adrenal cortex. These patients usually undergo chemotherapy with etoposide, doxorubicin, cisplatin and mitotane (EDP-M) in case of locally advanced or metastatic ACC. Computed tomography (CT) radiomics showed to be useful in adrenal pathologies. The study aimed to analyze the association between response to EDP-M treatment and CT textural features at diagnosis in patients with locally advanced or metastatic ACCs. METHODS We enrolled 17 patients with advanced or metastatic ACC who underwent CT before and after EDP-M therapy. The response to treatment was evaluated according to RECIST 1.1, Choi, and volumetric criteria. Based on the aforementioned criteria, the patients were classified as responders and not responders. Textural features were extracted from the biggest lesion in contrast-enhanced CT images with LifeX software. ROC curves were drawn for the variables that were significantly different (p < 0.05) between the two groups. RESULTS Long-run high grey level emphasis (LRHGLE_GLRLM) and histogram kurtosis were significantly different between responder and not responder groups (p = 0.04) and the multivariate ROC curve combining the two features showed a very good AUC (0.900; 95%IC: 0.724-1.000) in discriminating responders from not responders. More heterogeneous tissue texture of initial staging CT in locally advanced or metastatic ACC could predict the positive response to EDP-M treatment. CONCLUSIONS Adrenal texture is able to predict the response to EDP-M therapy in patients with advanced ACC.
Collapse
Affiliation(s)
- Filippo Crimì
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Institute of Radiology, University-Hospital of Padova, Padova, Italy
| | - Francesca Turatto
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Institute of Radiology, University-Hospital of Padova, Padova, Italy
| | - Carlo D'Alessandro
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Institute of Radiology, University-Hospital of Padova, Padova, Italy
| | - Giovanni Sussan
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Institute of Radiology, University-Hospital of Padova, Padova, Italy
| | - Maurizio Iacobone
- Endocrine Surgery Unit, Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padova, 35128, Italy
| | - Francesca Torresan
- Endocrine Surgery Unit, Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padova, 35128, Italy
| | - Irene Tizianel
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Endocrinology Unit, Department of Medicine-DIMED, University of Padova, Via Ospedale Civile, Padova, 105 - 35128, Italy
| | - Cristina Campi
- Department of Mathematics, University of Genoa, Genoa, Italy
- Life Science Computational Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genoa, Italy
| | - Emilio Quaia
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Institute of Radiology, University-Hospital of Padova, Padova, Italy
| | - Mario Caccese
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Filippo Ceccato
- Department of Medicine-DIMED, University of Padova, Padova, Italy.
- Endocrinology Unit, Department of Medicine-DIMED, University of Padova, Via Ospedale Civile, Padova, 105 - 35128, Italy.
| |
Collapse
|
235
|
Park JH, Cho ES, Yoon J, Rhee HJ, Park J, Choi JY, Chung YE. MRI radiomics model differentiates small hepatic metastases and abscesses in periampullary cancer patients. Sci Rep 2024; 14:23541. [PMID: 39384874 PMCID: PMC11464643 DOI: 10.1038/s41598-024-74311-w] [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: 01/29/2024] [Accepted: 09/25/2024] [Indexed: 10/11/2024] Open
Abstract
This multi-center, retrospective study focused on periampullary cancer patients undergoing MRI for hepatic metastasis and abscess differentiation. T1-weighted, T2-weighted, and arterial phase images were utilized to create radiomics models. In the training-set, 112 lesions in 54 patients (median age [IQR, interquartile range], 73 [63-80]; 38 men) were analyzed, and 123 lesions in 55 patients (72 [66-78]; 34 men) comprised the validation set. The T1-weighted + T2-weighted radiomics model showed the highest AUC (0.82, 95% CI 0.75-0.89) in the validation set. Notably, < 30% T1-T2 size discrepancy in MRI findings predicted metastasis (Ps ≤ 0.037), albeit with AUCs of 0.64-0.68 for hepatic metastasis. The radiomics model enhanced radiologists' performance (AUCs, 0.85-0.87 vs. 0.80-0.84) and significantly increased diagnostic confidence (P < 0.001). Although the performance increase lacked statistical significance (P = 0.104-0.281), the radiomics model proved valuable in differentiating small hepatic lesions and enhancing diagnostic confidence. This study highlights the potential of MRI-based radiomics in improving accuracy and confidence in the diagnosis of periampullary cancer-related hepatic lesions.
Collapse
Affiliation(s)
- Jae Hyon Park
- Department of Radiology and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Radiology, Armed Forces Daejeon Hospital, Daejeon, Republic of Korea
| | - Eun-Suk Cho
- Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jongjin Yoon
- Department of Radiology and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyung-Jin Rhee
- Department of Radiology and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - June Park
- Department of Radiology and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin-Young Choi
- Department of Radiology and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Eun Chung
- Department of Radiology and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Department of Radiology, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
236
|
Zhang H, Dohopolski M, Stojadinovic S, Schmitt LG, Anand S, Kim H, Pompos A, Godley A, Jiang S, Dan T, Wardak Z, Timmerman R, Peng H. Multiomics-Based Outcome Prediction in Personalized Ultra-Fractionated Stereotactic Adaptive Radiotherapy (PULSAR). Cancers (Basel) 2024; 16:3425. [PMID: 39410044 PMCID: PMC11475788 DOI: 10.3390/cancers16193425] [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: 09/04/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Objectives: This retrospective study aims to develop a multiomics approach that integrates radiomics, dosiomics, and delta features to predict treatment responses in brain metastasis (BM) patients undergoing PULSAR. Methods: A retrospective study encompassing 39 BM patients with 69 lesions treated with PULSAR was undertaken. Radiomics, dosiomics, and delta features were extracted from both pre-treatment and intra-treatment MRI scans alongside dose distributions. Six individual models, alongside an ensemble feature selection (EFS) model, were evaluated. The classification task focused on distinguishing between two lesion groups based on whether they exhibited a volume reduction of more than 20% at follow-up. Performance metrics, including sensitivity, specificity, accuracy, precision, F1 score, and the area under the receiver operating characteristic (ROC) curve (AUC), were assessed. Results: The EFS model integrated the features from pre-treatment radiomics, pre-treatment dosiomics, intra-treatment radiomics, and delta radiomics. It outperformed six individual models, achieving an AUC of 0.979, accuracy of 0.917, and F1 score of 0.821. Among the top nine features of the EFS model, six features came from post-wavelet transformation and three from original images. Conclusions: The study demonstrated the feasibility of employing a data-driven multiomics approach to predict treatment outcomes in BM patients receiving PULSAR treatment. Integrating multiomics with intra-treatment decision support in PULSAR shows promise for optimizing patient management and reducing the risks of under- or over-treatment.
Collapse
Affiliation(s)
- Haozhao Zhang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Medical Artificial Intelligence and Automation Laboratory, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Michael Dohopolski
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Medical Artificial Intelligence and Automation Laboratory, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Strahinja Stojadinovic
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Luiza Giuliani Schmitt
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Soummitra Anand
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Heejung Kim
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Arnold Pompos
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Medical Artificial Intelligence and Automation Laboratory, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andrew Godley
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Medical Artificial Intelligence and Automation Laboratory, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Steve Jiang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Medical Artificial Intelligence and Automation Laboratory, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tu Dan
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zabi Wardak
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Robert Timmerman
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hao Peng
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Medical Artificial Intelligence and Automation Laboratory, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
237
|
Li J, Zhou M, Tong Y, Chen H, Su R, Tao Y, Zhang G, Sun Z. Tumor Growth Pattern and Intra- and Peritumoral Radiomics Combined for Prediction of Initial TACE Outcome in Patients with Primary Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:1927-1944. [PMID: 39398867 PMCID: PMC11471153 DOI: 10.2147/jhc.s480554] [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: 05/30/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
Purpose Non-invasive methods are urgently needed to assess the efficacy of transarterial chemoembolization (TACE) and to identify patients with hepatocellular carcinoma (HCC) who may benefit from this procedure. This study, therefore, aimed to investigate the predictive ability of tumor growth patterns and radiomics features from contrast-enhanced magnetic resonance imaging (CE-MRI) in predicting tumor response to TACE among patients with HCC. Patients and Methods A retrospective study was conducted on 133 patients with HCC who underwent TACE at three centers between January 2015 and April 2023. Enrolled patients were divided into training, testing, and validation cohorts. Rim arterial phase hyperenhancement (Rim APHE), tumor growth patterns, nonperipheral washout, markedly low apparent diffusion coefficient (ADC) value, intratumoral arteries, and clinical baseline features were documented for all patients. Radiomics features were extracted from the intratumoral and peritumoral regions across the three phases of CE-MRI. Seven prediction models were developed, and their performances were evaluated using receiver operating characteristic (ROC) and decision curve analysis (DCA). Results Tumor growth patterns and albumin-bilirubin (ALBI) score were significantly correlated with tumor response. Tumor growth patterns also showed a positive correlation with tumor burden (r = 0.634, P = 0.000). The Peritumor (AUC = 0.85, 0.71, and 0.77), Clinics_Peritumor (AUC = 0.86, 0.77, and 0.81), and Tumor_Peritumor (AUC = 0.87, 0.77, and 0.80) models significantly outperformed the Clinics and Tumor models (P < 0.05), while the Clinics_Tumor_Peritumor model (AUC = 0.88, 0.81, and 0.81) outperformed the Clinics (AUC = 0.67, 0.77, and 0.75), Tumor (AUC = 0.78, 0.72, and 0.68), and Clinics_Tumor (AUC = 0.82, 0.83, and 0.78) models (P < 0.05 or 0.053, respectively). The DCA curve demonstrated better predictive performance within a specific threshold probability range for Clinics_Tumor_Peritumor. Conclusion Combining tumor growth patterns, intra- and peri-tumoral radiomics features, and ALBI score could be a robust tool for non-invasive and personalized prediction of treatment response to TACE in patients with HCC.
Collapse
Affiliation(s)
- Jiaying Li
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, People's Republic of China
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Minhui Zhou
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Yahan Tong
- Department of Radiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, 310005, People's Republic of China
| | - Haibo Chen
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, People's Republic of China
| | - Ruisi Su
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Yinghui Tao
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Guodong Zhang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, People's Republic of China
| | - Zhichao Sun
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, People's Republic of China
| |
Collapse
|
238
|
Singh G, Singh A, Bae J, Manjila S, Spektor V, Prasanna P, Lignelli A. -New frontiers in domain-inspired radiomics and radiogenomics: increasing role of molecular diagnostics in CNS tumor classification and grading following WHO CNS-5 updates. Cancer Imaging 2024; 24:133. [PMID: 39375809 PMCID: PMC11460168 DOI: 10.1186/s40644-024-00769-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 08/31/2024] [Indexed: 10/09/2024] Open
Abstract
Gliomas and Glioblastomas represent a significant portion of central nervous system (CNS) tumors associated with high mortality rates and variable prognosis. In 2021, the World Health Organization (WHO) updated its Glioma classification criteria, most notably incorporating molecular markers including CDKN2A/B homozygous deletion, TERT promoter mutation, EGFR amplification, + 7/-10 chromosome copy number changes, and others into the grading and classification of adult and pediatric Gliomas. The inclusion of these markers and the corresponding introduction of new Glioma subtypes has allowed for more specific tailoring of clinical interventions and has inspired a new wave of Radiogenomic studies seeking to leverage medical imaging information to explore the diagnostic and prognostic implications of these new biomarkers. Radiomics, deep learning, and combined approaches have enabled the development of powerful computational tools for MRI analysis correlating imaging characteristics with various molecular biomarkers integrated into the updated WHO CNS-5 guidelines. Recent studies have leveraged these methods to accurately classify Gliomas in accordance with these updated molecular-based criteria based solely on non-invasive MRI, demonstrating the great promise of Radiogenomic tools. In this review, we explore the relative benefits and drawbacks of these computational frameworks and highlight the technical and clinical innovations presented by recent studies in the landscape of fast evolving molecular-based Glioma subtyping. Furthermore, the potential benefits and challenges of incorporating these tools into routine radiological workflows, aiming to enhance patient care and optimize clinical outcomes in the evolving field of CNS tumor management, have been highlighted.
Collapse
Affiliation(s)
- Gagandeep Singh
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA.
| | - Annie Singh
- Atal Bihari Vajpayee Institute of Medical Sciences, New Delhi, India
| | - Joseph Bae
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, USA
| | - Sunil Manjila
- Department of Neurological Surgery, Garden City Hospital, Garden City, MI, USA
| | - Vadim Spektor
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA
| | - Prateek Prasanna
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, USA
| | - Angela Lignelli
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
239
|
Xie J, He Y, Che S, Zhao W, Niu Y, Qin D, Li Z. Differential diagnosis of benign and lung adenocarcinoma presenting as larger solid nodules and masses based on multiscale CT radiomics. PLoS One 2024; 19:e0309033. [PMID: 39365772 PMCID: PMC11451992 DOI: 10.1371/journal.pone.0309033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/04/2024] [Indexed: 10/06/2024] Open
Abstract
PURPOSE To develop a better radiomic model for the differential diagnosis of benign and lung adenocarcinoma lesions presenting as larger solid nodules and masses based on multiscale computed tomography (CT) radiomics. MATERIALS AND METHODS This retrospective study enrolled 205 patients with solid nodules and masses from Center 1 between January 2010 and February 2022 and Center 2 between January 2019 and February 2022. After applying the inclusion and exclusion criteria, we retrospectively enrolled 165 patients from two centers and assigned them to the training dataset (n = 115) or the test dataset (n = 50). Radiomics features were extracted from volumes of interest on CT images. A gradient boosting decision tree (GBDT) was used for data dimensionality reduction to perform the final feature selection. Four models were developed using clinical data, conventional imaging features and radiomics features, namely, the clinical and image model (CIM), the plain CT radiomics model (PRM), the enhanced CT radiomics model (ERM) and the combined model (CM). Model performance was evaluated to determine the best model for identifying benign and lung adenocarcinoma presenting as larger solid nodules and masses. RESULTS In the training dataset, the areas under the curve (AUCs) for the CIM, PRM, ERM, and CM were 0.718, 0.806, 0.819, and 0.917, respectively. The differential diagnostic capability of the ERM was better than that of the PRM and the CIM. The CM was optimal. Intermediate and junior radiologists and respiratory physicians achieved improved obviously diagnostic results with the radiomics model. The senior radiologists showed slight improved diagnostic results after using the radiomics model. CONCLUSION Radiomics may have the potential to be used as a noninvasive tool for the differential diagnosis of benign and lung adenocarcinoma lesions presenting as larger solid nodules and masses.
Collapse
Affiliation(s)
- Jiayue Xie
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Yifan He
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Siyu Che
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Wenjing Zhao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Yuxin Niu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Dongxue Qin
- Department of Radiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Zhiyong Li
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
240
|
Lin A, Chen Y, Chen Y, Ye Z, Luo W, Chen Y, Zhang Y, Wang W. MRI radiomics combined with machine learning for diagnosing mild cognitive impairment: a focus on the cerebellar gray and white matter. Front Aging Neurosci 2024; 16:1460293. [PMID: 39430972 PMCID: PMC11489926 DOI: 10.3389/fnagi.2024.1460293] [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: 07/05/2024] [Accepted: 09/25/2024] [Indexed: 10/22/2024] Open
Abstract
Objective Mild Cognitive Impairment (MCI) is a recognized precursor to Alzheimer's Disease (AD), presenting a significant risk of progression. Early detection and intervention in MCI can potentially slow disease advancement, offering substantial clinical benefits. This study employed radiomics and machine learning methodologies to distinguish between MCI and Normal Cognition (NC) groups. Methods The study included 172 MCI patients and 183 healthy controls from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database, all of whom had 3D-T1 weighted MRI structural images. The cerebellar gray and white matter were segmented automatically using volBrain software, and radiomic features were extracted and screened through Pyradiomics. The screened features were then input into various machine learning models, including Random Forest (RF), Logistic Regression (LR), eXtreme Gradient Boosting (XGBoost), Support Vector Machines (SVM), K Nearest Neighbors (KNN), Extra Trees, Light Gradient Boosting Machine (LightGBM), and Multilayer Perceptron (MLP). Each model was optimized for penalty parameters through 5-fold cross-validation to construct radiomic models. The DeLong test was used to evaluate the performance of different models. Results The LightGBM model, which utilizes a combination of cerebellar gray and white matter features (comprising eight gray matter and eight white matter features), emerges as the most effective model for radiomics feature analysis. The model demonstrates an Area Under the Curve (AUC) of 0.863 for the training set and 0.776 for the test set. Conclusion Radiomic features based on the cerebellar gray and white matter, combined with machine learning, can objectively diagnose MCI, which provides significant clinical value for assisted diagnosis.
Collapse
Affiliation(s)
- Andong Lin
- Department of Neurology, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Yini Chen
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Yi Chen
- Department of Pharmacy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Zhinan Ye
- Department of Neurology, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Weili Luo
- Department of Neurology, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Ying Chen
- Department of Neurology, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Yaping Zhang
- Department of Neurology, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Wenjie Wang
- Department of Neurology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| |
Collapse
|
241
|
Zhang Y, Huang W, Jiao H, Kang L. PET radiomics in lung cancer: advances and translational challenges. EJNMMI Phys 2024; 11:81. [PMID: 39361110 PMCID: PMC11450131 DOI: 10.1186/s40658-024-00685-5] [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: 11/19/2023] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
Radiomics is an emerging field of medical imaging that aims at improving the accuracy of diagnosis, prognosis, treatment planning and monitoring non-invasively through the automated or semi-automated quantitative analysis of high-dimensional image features. Specifically in the field of nuclear medicine, radiomics utilizes imaging methods such as positron emission tomography (PET) and single photon emission computed tomography (SPECT) to evaluate biomarkers related to metabolism, blood flow, cellular activity and some biological pathways. Lung cancer ranks among the leading causes of cancer-related deaths globally, and radiomics analysis has shown great potential in guiding individualized therapy, assessing treatment response, and predicting clinical outcomes. In this review, we summarize the current state-of-the-art radiomics progress in lung cancer, highlighting the potential benefits and existing limitations of this approach. The radiomics workflow was introduced first including image acquisition, segmentation, feature extraction, and model building. Then the published literatures were described about radiomics-based prediction models for lung cancer diagnosis, differentiation, prognosis and efficacy evaluation. Finally, we discuss current challenges and provide insights into future directions and potential opportunities for integrating radiomics into routine clinical practice.
Collapse
Affiliation(s)
- Yongbai Zhang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Hao Jiao
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng Dist, Beijing, 100034, China.
| |
Collapse
|
242
|
Li Z, Lu J, Zhang B, Si J, Zhang H, Zhong Z, He S, Cai W, Li T. New Model and Public Online Prediction Platform for Risk Stratification of Vocal Cord Leukoplakia. Laryngoscope 2024; 134:4329-4337. [PMID: 38828682 DOI: 10.1002/lary.31555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/05/2024]
Abstract
OBJECTIVE To extract texture features from vocal cord leukoplakia (VCL) images and establish a VCL risk stratification prediction model using machine learning (ML) techniques. METHODS A total of 462 patients with pathologically confirmed VCL were retrospectively collected and divided into low-risk and high-risk groups. We use a 5-fold cross validation method to ensure the generalization ability of the model built using the included dataset and avoid overfitting. Totally 504 texture features were extracted from each laryngoscope image. After feature selection, 10 ML classifiers were utilized to construct the model. The SHapley Additive exPlanations (SHAP) was employed for feature analysis. To evaluate the model, accuracy, sensitivity, specificity, and the area under the receiver operating characteristic (ROC) curve (AUC) were utilized. In addition, the model was transformed into an online application for public use and further tested in an independent dataset with 52 cases of VCL. RESULTS A total of 12 features were finally selected, random forest (RF) achieved the best model performance, the mean accuracy, sensitivity, specificity, and AUC of the 5-fold cross validation were 92.2 ± 4.1%, 95.6 ± 4.0%, 85.8 ± 5.8%, and 90.7 ± 4.9%, respectively. The result is much higher than the clinicians (AUC between 63.1% and 75.2%). The SHAP algorithm ranks the importance of 12 texture features to the model. The test results of the additional independent datasets were 92.3%, 95.7%, 90.0%, and 93.3%, respectively. CONCLUSION The proposed VCL risk stratification prediction model, which has been developed into a public online prediction platform, may be applied in practical clinical work. LEVEL OF EVIDENCE 3 Laryngoscope, 134:4329-4337, 2024.
Collapse
Affiliation(s)
- Zufei Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jinghui Lu
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, U.S.A
| | - Baiwen Zhang
- Institute of Information and Artificial Intelligence Technology, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Joshua Si
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, U.S.A
| | - Hong Zhang
- Department of Pathology, Peking University First Hospital, Beijing, China
| | - Zhen Zhong
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University First Hospital, Beijing, China
| | - Shuai He
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wenli Cai
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, U.S.A
| | - Tiancheng Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
243
|
O'Sullivan NJ, Temperley HC, Horan MT, Kamran W, Corr A, O'Gorman C, Saadeh F, Meaney JM, Kelly ME. Role of radiomics as a predictor of disease recurrence in ovarian cancer: a systematic review. Abdom Radiol (NY) 2024; 49:3540-3547. [PMID: 38744703 PMCID: PMC11390851 DOI: 10.1007/s00261-024-04330-8] [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: 10/27/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 05/16/2024]
Abstract
Ovarian cancer is associated with high cancer-related mortality rate attributed to late-stage diagnosis, limited treatment options, and frequent disease recurrence. As a result, careful patient selection is important especially in setting of radical surgery. Radiomics is an emerging field in medical imaging, which may help provide vital prognostic evaluation and help patient selection for radical treatment strategies. This systematic review aims to assess the role of radiomics as a predictor of disease recurrence in ovarian cancer. A systematic search was conducted in Medline, EMBASE, and Web of Science databases. Studies meeting inclusion criteria investigating the use of radiomics to predict post-operative recurrence in ovarian cancer were included in our qualitative analysis. Study quality was assessed using the QUADAS-2 and Radiomics Quality Score tools. Six retrospective studies met the inclusion criteria, involving a total of 952 participants. Radiomic-based signatures demonstrated consistent performance in predicting disease recurrence, as evidenced by satisfactory area under the receiver operating characteristic curve values (AUC range 0.77-0.89). Radiomic-based signatures appear to good prognosticators of disease recurrence in ovarian cancer as estimated by AUC. The reviewed studies consistently reported the potential of radiomic features to enhance risk stratification and personalise treatment decisions in this complex cohort of patients. Further research is warranted to address limitations related to feature reliability, workflow heterogeneity, and the need for prospective validation studies.
Collapse
Affiliation(s)
- Niall J O'Sullivan
- Department of Radiology, St. James's Hospital, Dublin, Ireland.
- School of Medicine, Trinity College Dublin, Dublin, Ireland.
- The National Centre for Advanced Medical Imaging (CAMI), St. James's Hospital, Dublin, Ireland.
| | | | - Michelle T Horan
- Department of Radiology, St. James's Hospital, Dublin, Ireland
- The National Centre for Advanced Medical Imaging (CAMI), St. James's Hospital, Dublin, Ireland
| | - Waseem Kamran
- Department of Gynaecology, St. James's Hospital, Dublin, Ireland
| | - Alison Corr
- Department of Radiology, St. James's Hospital, Dublin, Ireland
| | | | - Feras Saadeh
- Department of Gynaecology, St. James's Hospital, Dublin, Ireland
| | - James M Meaney
- Department of Radiology, St. James's Hospital, Dublin, Ireland
- School of Medicine, Trinity College Dublin, Dublin, Ireland
- The National Centre for Advanced Medical Imaging (CAMI), St. James's Hospital, Dublin, Ireland
| | - Michael E Kelly
- Department of Radiology, St. James's Hospital, Dublin, Ireland
- Department of Surgery, St. James's Hospital, Dublin, Ireland
| |
Collapse
|
244
|
Lu J, Zhu K, Yang N, Chen Q, Liu L, Liu Y, Yang Y, Li J. Radiomics and Clinical Features for Distinguishing Kidney Stone-Associated Urinary Tract Infection: A Comprehensive Analysis of Machine Learning Classification. Open Forum Infect Dis 2024; 11:ofae581. [PMID: 39435322 PMCID: PMC11493090 DOI: 10.1093/ofid/ofae581] [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: 07/29/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024] Open
Abstract
Background This study investigated the abilities of radiomics and clinical feature models to distinguish kidney stone-associated urinary tract infections (KS-UTIs) using computed tomography. Methods A retrospective analysis was conducted on a single-center dataset comprising computed tomography (CT) scans and corresponding clinical information from 461 patients with kidney stones. Radiomics features were extracted from CT images and underwent dimensionality reduction and selection. Multiple machine learning (Three types of shallow learning and four types of deep learning) algorithms were employed to construct radiomics and clinical models in this study. Performance evaluation and optimal model selection were done using receiver operating characteristic (ROC) curve analysis and Delong test. Univariate and multivariate logistic regression analyzed clinical and radiomics features to identify significant variables and develop a clinical model. A combined model integrating radiomics and clinical features was established. Model performance was assessed by ROC curve analysis, clinical utility was evaluated through decision curve analysis, and the accuracy of the model was analyzed via calibration curve. Results Multilayer perceptron (MLP) showed higher classification accuracy than other classifiers (area under the curve (AUC) for radiomics model: train 0.96, test 0.94; AUC for clinical model: train 0.95, test 0.91. The combined radiomics-clinical model performed best (AUC for combined model: train 0.98, test 0.95). Decision curve and calibration curve analyses confirmed the model's clinical efficacy and calibration. Conclusions This study showed the effectiveness of combining radiomics and clinical features from CT scans to identify KS-UTIs. A combined model using MLP exhibited strong classification abilities.
Collapse
Affiliation(s)
- Jianjuan Lu
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Kun Zhu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ning Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiang Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lingrui Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanyan Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases, Anhui Medical University, Hefei, China
- Institute of Infectious Diseases, Anhui Medical University, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiabin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Infectious Diseases, Anhui Medical University, Hefei, China
- Institute of Infectious Diseases, Anhui Medical University, Hefei, China
- Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
- Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
245
|
Adachi T, Nakamura M, Matsuo Y, Karasawa K, Kokubo M, Sakamoto T, Hiraoka M, Mizowaki T. Prospective external validation of radiomics-based predictive model of distant metastasis after dynamic tumor tracking stereotactic body radiation therapy in patients with non-small-cell lung cancer: A multi-institutional analysis. J Appl Clin Med Phys 2024; 25:e14475. [PMID: 39178139 PMCID: PMC11466494 DOI: 10.1002/acm2.14475] [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: 02/22/2024] [Accepted: 07/03/2024] [Indexed: 08/25/2024] Open
Abstract
BACKGROUND AND PURPOSE This study aims to externally validate a predictive model for distant metastasis (DM) with computed tomography (CT)-based radiomics features in prospectively enrolled non-small-cell lung cancer patients undergoing dynamic tumor-tracking stereotactic body radiation therapy (DTT-SBRT). MATERIALS AND METHODS The study collected retrospective data from 567 patients across 11 institutions as the training dataset and prospectively enrolled 42 patients from four institutions as the external test dataset. Four clinical features were collected, and 944 CT-based radiomic features were extracted from gross tumor volumes. After standardization and feature selection, DM predictive models were developed using fine and gray regression (FG) and random survival forest (RSF), incorporating clinical and radiomic features, and their combinations within the training dataset. Then, the model was applied to the test dataset, dividing patients into high- and low-risk groups based on medians of risk scores. Model performance was assessed using the concordance index (C-index), and the statistical significance between groups was evaluated using Gray's test. RESULTS In the training dataset, 122 of 567 patients (21.5%) developed DM, compared to 9 of 42 patients (21.4%) in the test dataset. In the test dataset, the C-indices of the clinical, radiomics, and hybrid models with FG were 0.559, 0.544, and 0.560, respectively, whereas those with RSF were 0.576, 0.604, and 0.627, respectively. The hybrid model with RSF, which exhibited the best predictive performance of all models, identified 7 of 23 patients (30.4%) as high risk and 2 of 19 patients (10.5%) as low risk for DM incidence in the test dataset (p = 0.116). CONCLUSION Although predictive models for DM lack significance when applied to prospectively enrolled cases undergoing DTT-lung SBRT, the model with RSF exhibits a consistent capacity to effectively classify patients at a high risk of developing DM.
Collapse
Affiliation(s)
- Takanori Adachi
- Department of Radiation Oncology and Image‐Applied TherapyGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Mitsuhiro Nakamura
- Department of Radiation Oncology and Image‐Applied TherapyGraduate School of MedicineKyoto UniversityKyotoJapan
- Department of Advanced Medical PhysicsGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Yukinori Matsuo
- Department of Radiation Oncology and Image‐Applied TherapyGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Katsuyuki Karasawa
- Division of Radiation OncologyDepartmentof RadiologyTokyo Metropolitan Cancer and Infectious Diseases Center Komagome HospitalTokyoJapan
| | - Masaki Kokubo
- Department of Radiation OncologyKobe City Medical Center General HospitalHyogoJapan
| | - Takashi Sakamoto
- Department of Radiation OncologyKyoto Katsura HospitalKyotoJapan
| | - Masahiro Hiraoka
- Department of Radiation OncologyJapanese Red Cross Society Wakayama Medical CenterWakayamaJapan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image‐Applied TherapyGraduate School of MedicineKyoto UniversityKyotoJapan
| |
Collapse
|
246
|
Li T, Gan T, Wang J, Long Y, Zhang K, Liao M. "Application of CT radiomics in brain metastasis of lung cancer: A systematic review and meta-analysis". Clin Imaging 2024; 114:110275. [PMID: 39243496 DOI: 10.1016/j.clinimag.2024.110275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/16/2024] [Accepted: 08/25/2024] [Indexed: 09/09/2024]
Abstract
PURPOSE This study aimed to systematically assess the quality and performance of computed tomography (CT) radiomics studies in predicting brain metastasis (BM) among patients with lung cancer. METHODS The PubMed, Embase and Web of Science were searched for studies predicting BM in patients with lung cancer using CT-based radiomics features. Information regarding patients, imaging, and radiomics analysis was extracted from eligible studies. We assessed the quality of included studies using the Radiomics Quality Scoring (RQS) tool and the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2). A meta-analysis of studies regarding the prediction of BM in patients with lung cancer was performed. RESULTS Thirteen studies were identified, with sample sizes ranging from 75 to 602. The mean RQS of the studies was 12 (range 9-16), and the corresponding percentage of the score was 33.55 % (range 25.00-44.44 %). Four studies (30.8 %) were considered as low risk of bias, while the remaining nine studies (69.2 %) were considered to have unclear risks. The meta-analysis included twelve studies. The pooled sensitivity, specificity and Area Under the Curve (AUC) value with 95 % confidence intervals were 0.75 [0.69, 0.80], 0.76 [0.68, 0.82], and 0.81 [0.77-0.84], respectively. CONCLUSION CT radiomics-based models show promising results as a non-invasive method to predict BM in lung cancer patients. However, multicenter and prospective studies are warranted to enhance the stability and acceptance of radiomics.
Collapse
Affiliation(s)
- Ting Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China; The Second School of Clinical Medicine, Wuhan University, Wuhan, China.
| | - Tian Gan
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China; The Second School of Clinical Medicine, Wuhan University, Wuhan, China.
| | - Jingting Wang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China; The Second School of Clinical Medicine, Wuhan University, Wuhan, China.
| | - Yun Long
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China; The Second School of Clinical Medicine, Wuhan University, Wuhan, China.
| | - Kemeng Zhang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China; The Second School of Clinical Medicine, Wuhan University, Wuhan, China.
| | - Meiyan Liao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China; The Second School of Clinical Medicine, Wuhan University, Wuhan, China.
| |
Collapse
|
247
|
Becker J, Woźnicki P, Decker JA, Risch F, Wudy R, Kaufmann D, Canalini L, Wollny C, Scheurig-Muenkler C, Kroencke T, Bette S, Schwarz F. Radiomics signature for automatic hydronephrosis detection in unenhanced Low-Dose CT. Eur J Radiol 2024; 179:111677. [PMID: 39178684 DOI: 10.1016/j.ejrad.2024.111677] [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: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 08/26/2024]
Abstract
PURPOSE To investigate the diagnostic performance of an automatic pipeline for detection of hydronephrosis on kidney's parenchyma on unenhanced low-dose CT of the abdomen. METHODS This retrospective study included 95 patients with confirmed unilateral hydronephrosis in an unenhanced low-dose CT of the abdomen. Data were split into training (n = 67) and test (n = 28) cohorts. Both kidneys for each case were included in further analyses, whereas the kidney without hydronephrosis was used as control. Using the training cohort, we developed a pipeline consisting of a deep-learning model for automatic segmentation (a Convolutional Neural Network based on nnU-Net architecture) of the kidney's parenchyma and a radiomics classifier to detect hydronephrosis. The models were assessed using standard classification metrics, such as area under the ROC curve (AUC), sensitivity and specificity, as well as semantic segmentation metrics, including Dice coefficient and Jaccard index. RESULTS Using manual segmentation of the kidney's parenchyma, hydronephrosis can be detected with an AUC of 0.84, a sensitivity of 75% and a specificity of 82%, a PPV of 81% and a NPV of 77%. Automatic kidney segmentation achieved a mean Dice score of 0.87 and 0.91 for the right and left kidney, respectively. Additionally, automatic segmentation achieved an AUC of 0.83, a sensitivity of 86%, specificity of 64%, PPV of 71%, and NPV of 82%. CONCLUSION Our proposed radiomics signature using automatic kidney's parenchyma segmentation allows for accurate hydronephrosis detection on unenhanced low-dose CT scans of the abdomen independently of widened renal pelvis. This method could be used in clinical routine to highlight hydronephrosis to radiologists as well as clinicians, especially in patients with concurrent parapelvic cysts and might reduce time and costs associated with diagnosing hydronephrosis.
Collapse
Affiliation(s)
- Judith Becker
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - Piotr Woźnicki
- Diagnostic and Interventional Radiology, University Hospital Würzburg, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
| | - Josua A Decker
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - Franka Risch
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - Ramona Wudy
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - David Kaufmann
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - Luca Canalini
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - Claudia Wollny
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - Christian Scheurig-Muenkler
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - Thomas Kroencke
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany; Centre for Advanced Analytics and Predictive Sciences (CAAPS), University of Augsburg, Universitätsstr. 2, 86159 Augsburg, Germany.
| | - Stefanie Bette
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Stenglinstr. 2, 86156 Augsburg, Germany
| | - Florian Schwarz
- Centre for Diagnostic Imaging and Interventional Therapy, Donau-Isar-Klinikum, Perlasberger Straße 41, 94469 Deggendorf, Germany; Medical Faculty, Ludwig Maximilian University Munich, Bavariaring 19, 80336 Munich, Germany
| |
Collapse
|
248
|
Pan GS, Sun XM, Kong FF, Wang JZ, He XY, Lu XG, Hu CS, Dong SX, Ying HM. Delta magnetic resonance imaging radiomics features‑based nomogram predicts long‑term efficacy after induction chemotherapy in locoregionally advanced nasopharyngeal carcinoma. Oral Oncol 2024; 157:106987. [PMID: 39133972 DOI: 10.1016/j.oraloncology.2024.106987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 08/22/2024]
Abstract
PURPOSE To establish and validate a delta-radiomics-based model for predicting progression-free survival (PFS) in patients with locoregionally advanced nasopharyngeal carcinoma (LA-NPC) following induction chemotherapy (IC). METHODS AND MATERIALS A total of 250 LA-NPC patients (training cohort: n = 145; validation cohort: n = 105) were enrolled. Radiomic features were extracted from MRI scans taken before and after IC, and changes in these features were calculated. Following feature selection, a delta-radiomics signature was constructed using LASSO-Cox regression analysis. A prognostic nomogram incorporating independent clinical indicators and the delta-radiomics signature was developed and assessed for calibration and discrimination. Risk stratification by the nomogram was evaluated using Kaplan-Meier methods. RESULTS The delta-radiomics signature, consisting of 12 features, was independently associated with prognosis. The nomogram, integrating the delta-radiomics signature and clinical factors demonstrated excellent calibration and discrimination. The model achieved a Harrell's concordance index (C-index) of 0.848 in the training cohort and 0.820 in the validation cohort. Risk stratification identified two groups with significantly different PFS rates. The three-year PFS for high-risk patients who received concurrent chemoradiotherapy (CCRT) or radiotherapy plus adjuvant chemotherapy (RT+AC) after IC was significantly higher than for those who received RT alone, reaching statistical significance. In contrast, for low-risk patients, the three-year PFS after IC was slightly higher for those who received CCRT or RT+AC compared to those who received RT alone; however, this difference did not reach statistical significance. CONCLUSIONS Our delta MRI-based radiomics model could be useful for predicting PFS and may guide subsequent treatment decisions after IC in LA-NPC.
Collapse
Affiliation(s)
- Guang-Sen Pan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Xiao-Ming Sun
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Fang-Fang Kong
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Jia-Zhou Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Xia-Yun He
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Xue-Guan Lu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Chao-Su Hu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Si-Xue Dong
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China.
| | - Hong-Mei Ying
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China.
| |
Collapse
|
249
|
Maniaci A, Lavalle S, Gagliano C, Lentini M, Masiello E, Parisi F, Iannella G, Cilia ND, Salerno V, Cusumano G, La Via L. The Integration of Radiomics and Artificial Intelligence in Modern Medicine. Life (Basel) 2024; 14:1248. [PMID: 39459547 PMCID: PMC11508875 DOI: 10.3390/life14101248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
With profound effects on patient care, the role of artificial intelligence (AI) in radiomics has become a disruptive force in contemporary medicine. Radiomics, the quantitative feature extraction and analysis from medical images, offers useful imaging biomarkers that can reveal important information about the nature of diseases, how well patients respond to treatment and patient outcomes. The use of AI techniques in radiomics, such as machine learning and deep learning, has made it possible to create sophisticated computer-aided diagnostic systems, predictive models, and decision support tools. The many uses of AI in radiomics are examined in this review, encompassing its involvement of quantitative feature extraction from medical images, the machine learning, deep learning and computer-aided diagnostic (CAD) systems approaches in radiomics, and the effect of radiomics and AI on improving workflow automation and efficiency, optimize clinical trials and patient stratification. This review also covers the predictive modeling improvement by machine learning in radiomics, the multimodal integration and enhanced deep learning architectures, and the regulatory and clinical adoption considerations for radiomics-based CAD. Particular emphasis is given to the enormous potential for enhancing diagnosis precision, treatment personalization, and overall patient outcomes.
Collapse
Affiliation(s)
- Antonino Maniaci
- Faculty of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (S.L.); (C.G.)
| | - Salvatore Lavalle
- Faculty of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (S.L.); (C.G.)
| | - Caterina Gagliano
- Faculty of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (S.L.); (C.G.)
| | - Mario Lentini
- ASP Ragusa, Hospital Giovanni Paolo II, 97100 Ragusa, Italy;
| | - Edoardo Masiello
- Radiology Unit, Department Clinical and Experimental, Experimental Imaging Center, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Federica Parisi
- Department of Medical and Surgical Sciences and Advanced Technologies “GF Ingrassia”, ENT Section, University of Catania, Via S. Sofia, 78, 95125 Catania, Italy;
| | - Giannicola Iannella
- Department of ‘Organi di Senso’, University “Sapienza”, Viale dell’Università, 33, 00185 Rome, Italy;
| | - Nicole Dalia Cilia
- Department of Computer Engineering, University of Enna “Kore”, 94100 Enna, Italy;
- Institute for Computing and Information Sciences, Radboud University Nijmegen, 6544 Nijmegen, The Netherlands
| | - Valerio Salerno
- Department of Engineering and Architecture, Kore University of Enna, 94100 Enna, Italy;
| | - Giacomo Cusumano
- University Hospital Policlinico “G. Rodolico—San Marco”, 95123 Catania, Italy;
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy
| | - Luigi La Via
- University Hospital Policlinico “G. Rodolico—San Marco”, 95123 Catania, Italy;
| |
Collapse
|
250
|
Wang B, Liu J, Xie J, Zhang X, Wang Z, Cao Z, Wen D, Wan Hasan WZ, Harun Ramli HR, Dong X. Systematic review and meta-analysis of the prognostic value of 18F-Fluorodeoxyglucose ( 18F-FDG) positron emission tomography (PET) and/or computed tomography (CT)-based radiomics in head and neck cancer. Clin Radiol 2024; 79:757-772. [PMID: 38944542 DOI: 10.1016/j.crad.2024.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 07/01/2024]
Abstract
AIM Radiomics involves the extraction of quantitative data from medical images to facilitate the diagnosis, prognosis, and staging of tumors. This study provides a comprehensive overview of the efficacy of radiomics in prognostic applications for head and neck cancer (HNC) in recent years. It undertakes a systematic review of prognostic models specific to HNC and conducts a meta-analysis to evaluate their predictive performance. MATERIALS AND METHODS This study adhered rigorously to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for literature searches. The literature databases, including PubMed, Embase, Cochrane, and Scopus were systematically searched individually. The methodological quality of the incorporated studies underwent assessment utilizing the radiomics quality score (RQS) tool. A random-effects meta-analysis employing the Harrell concordance index (C-index) was conducted to evaluate the performance of all radiomics models. RESULTS Among the 388 studies retrieved, 24 studies encompassing a total of 6,978 cases were incorporated into the systematic review. Furthermore, eight studies, focusing on overall survival as an endpoint, were included in the meta-analysis. The meta-analysis revealed that the estimated random effect of the C-index for all studies utilizing radiomics alone was 0.77 (0.71-0.82), with a substantial degree of heterogeneity indicated by an I2 of 80.17%. CONCLUSIONS Based on this review, prognostic modeling utilizing radiomics has demonstrated enhanced efficacy for head and neck cancers; however, there remains room for improvement in this approach. In the future, advancements are warranted in the integration of clinical parameters and multimodal features, balancing multicenter data, as well as in feature screening and model construction within this field.
Collapse
Affiliation(s)
- B Wang
- Department of Electrical and Electronic Engineering, Faculty of Engineering, Universiti Putra Malaysia, Serdang, Malaysia; Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
| | - J Liu
- Department of Nursing, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia; Department of Nursing, Chengde Central Hospital, Chengde city, Hebei Province, China.
| | - J Xie
- Department of Automatic, Tsinghua University, Beijing, China.
| | - X Zhang
- Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
| | - Z Wang
- Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
| | - Z Cao
- Department of Radiology, The Affiliated Hospital of Chengde Medical University, Chengde City, Hebei Province, China.
| | - D Wen
- Institute of Artificial Intelligence, University of Science and Technology Beijing, Beijing, China.
| | - W Z Wan Hasan
- Department of Electrical and Electronic Engineering, Faculty of Engineering, Universiti Putra Malaysia, Serdang, Malaysia.
| | - H R Harun Ramli
- Department of Electrical and Electronic Engineering, Faculty of Engineering, Universiti Putra Malaysia, Serdang, Malaysia.
| | - X Dong
- Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China; Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde City, Hebei, China; Hebei International Research Center of Medical Engineering, Chengde Medical University, Hebei, China.
| |
Collapse
|