1
|
Zhu Q, Hu X, Ye Q, Wu C, Dong X, Li W, Lin F. Development and Validation of an Early Recurrence Prediction Model for High-Grade Glioma Integrating Temporalis Muscle and Tumor Features: Exploring the Prognostic Value of Temporalis Muscle. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025:10.1007/s10278-025-01491-w. [PMID: 40205255 DOI: 10.1007/s10278-025-01491-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/06/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
This study aimed to develop and validate a predictive model for early recurrence of high-grade glioma (HGG) within 180 days, assess the prognostic value of preoperative and postoperative temporalis muscle metrics (area and thickness), and explore their significance in postoperative follow-up. Seventy-one molecularly confirmed HGG patients were included, with data sourced from local data and TCIA (The Cancer Imaging Archive) RHUH-GBM (Río Hortega University Hospital Glioblastoma) dataset. Tumor segmentation was performed using deep learning, and radiomic features were extracted following comparison with manual segmentation. Feature selection was conducted using mutual information and recursive feature elimination. A comprehensive model integrating 3D tumor radiomics and temporalis muscle metrics was developed and compared with a tumor-only model to identify the optimal predictive framework. SHAP analysis was used to evaluate model interpretability and feature importance. The TM_Tumor_HistGradientBoosting model, incorporating 16 features including temporalis muscle metrics, outperformed the tumor-only model in accuracy (0.89), recall (0.87), and F1 score (0.88). SHAP analysis highlighted that preoperative temporalis muscle cross-sectional area was strongly associated with early recurrence risk, while postoperative temporalis muscle thickness significantly contributed to recurrence prediction. Combining temporalis muscle metrics with preoperative tumor MRI substantially improved the accuracy of early recurrence prediction in HGG. Temporalis muscle metrics serve as objective and sustainable prognostic indicators with significant clinical value in postoperative follow-up.
Collapse
Affiliation(s)
- Qianni Zhu
- Shenzhen University, Shenzhen, China
- Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Xiaocong Hu
- Shenzhen University, Shenzhen, China
- The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Qihui Ye
- Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, China
- Institute of Biopharmaceutics and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - ChunXiang Wu
- Shenzhen University, Shenzhen, China
- Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - XueWen Dong
- Shenzhen University, Shenzhen, China
- Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Weihua Li
- Shenzhen University, Shenzhen, China.
- Shenzhen Second People's Hospital, Shenzhen, 518035, China.
- Medical imaging department, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China.
| | - Fan Lin
- Shenzhen University, Shenzhen, China.
- Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| |
Collapse
|
2
|
Zhou Q, Ke X, Man J, Jiang J, Ren J, Xue C, Zhang B, Zhang P, Zhao J, Zhou J. Integrated MRI radiomics, tumor microenvironment, and clinical risk factors for improving survival prediction in patients with glioblastomas. Strahlenther Onkol 2025; 201:398-410. [PMID: 39249499 DOI: 10.1007/s00066-024-02283-x] [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: 12/27/2023] [Accepted: 07/14/2024] [Indexed: 09/10/2024]
Abstract
PURPOSE To construct a comprehensive model for predicting the prognosis of patients with glioblastoma (GB) using a radiomics method and integrating clinical risk factors, tumor microenvironment (TME), and imaging characteristics. MATERIALS AND METHODS In this retrospective study, we included 148 patients (85 males and 63 females; median age 53 years) with isocitrate dehydrogenase-wildtype GB between January 2016 and April 2022. Patients were randomly divided into the training (n = 104) and test (n = 44) sets. The best feature combination related to GB overall survival (OS) was selected using LASSO Cox regression analyses. Clinical, radiomics, clinical-radiomics, clinical-TME, and clinical-radiomics-TME models were established. The models' concordance index (C-index) was evaluated. The survival curve was drawn using the Kaplan-Meier method, and the prognostic stratification ability of the model was tested. RESULTS LASSO Cox analyses were used to screen the factors related to OS in patients with GB, including MGMT (hazard ratio [HR] = 0.642; 95% CI 0.414-0.997; P = 0.046), TERT (HR = 1.755; 95% CI 1.095-2.813; P = 0.019), peritumoral edema (HR = 1.013; 95% CI 0.999-1.027; P = 0.049), tumor purity (TP; HR = 0.982; 95% CI 0.964-1.000; P = 0.054), CD163 + tumor-associated macrophages (TAMs; HR = 1.049; 95% CI 1.021-1.078; P < 0.001), CD68 + TAMs (HR = 1.055; 95% CI 1.018-1.093; P = 0.004), and the six radiomics features. The clinical-radiomics-TME model had the best survival prediction ability, the C‑index was 0.768 (0.717-0.819). The AUC of 1‑, 2‑, and 3‑year OS prediction in the test set was 0.842, 0.844, and 0.795, respectively. CONCLUSION The clinical-radiomics-TME model is the most effective for predicting the survival of patients with GB. Radiomics features, TP, and TAMs play important roles in the prognostic model.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Jiangwei Man
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Department of Surgical, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Jian Jiang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Jialiang Ren
- Department of Pharmaceuticals Diagnostics, GE HealthCare, Beijing, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Peng Zhang
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Jun Zhao
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China.
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, 730030, Lanzhou, Gansu, China.
| |
Collapse
|
3
|
Mohammadzadeh I, Hajikarimloo B, Niroomand B, Eini P, Ghanbarnia R, Habibi MA, Albakr A, Borghei-Razavi H. Application of artificial intelligence in forecasting survival in high-grade glioma: systematic review and meta-analysis involving 79,638 participants. Neurosurg Rev 2025; 48:240. [PMID: 39954167 DOI: 10.1007/s10143-025-03419-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
High-grade glioma (HGG) is an aggressive brain tumor with poor survival rates. Predicting survival outcomes is critical for personalized treatment planning. In recent years, artificial intelligence (AI), particularly machine learning (ML) and deep learning (DL) models, has emerged as a promising approach for enhancing prognostic accuracy in HGG but this study especially focused on the potential of AI in the recurrence of HGG. A systematic review and meta-analysis were conducted to assess the performance of AI-based models in predicting survival outcomes for HGG patients. Relevant studies were retrieved from PubMed, Embase, Scopus, and Web of Science until 2 Dec 2024, using predefined keywords ("High-Grade Glioma", "Survival" and "Machine Learning") without date or language restrictions. Data extraction and quality assessment were performed in accordance with PRISMA and PROBAST guidelines. In this study were included. The pooled diagnostic metric, the area under the curve (AUC), was analyzed using random-effects models. A total of 39 studies with 29 various algorithms and 79,638 patients were included, with 15 studies contributing to the meta-analysis. The most commonly used algorithms were random forest (RF) and logistic regression (LR), which demonstrated robust predictive accuracy. The pooled AUCs for one-year, two-year, three-year and overall survival predictions were 0.816, 0.854, 0.871 and 0.789 respectively. Subgroup analysis revealed that RSF achieved the highest predictive accuracy with an AUC of 0.91 (95% CI: 0.84-0.98), while LR followed with an AUC of 0.89 (95% CI: 0.82-0.96). Models integrating clinical, radiomics, and genetic features consistently outperformed single-data-type models. MRI was the most frequently utilized imaging modality. AI-based models, particularly ML and DL algorithms, show significant potential for improving survival prediction in HGG patients. By integrating multimodal data, these models offer valuable tools for personalized treatment planning, although further validation in prospective, multicenter studies is needed to ensure clinical applicability.
Collapse
Affiliation(s)
- Ibrahim Mohammadzadeh
- Skull Base Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Neuroscience Lab, Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Bardia Hajikarimloo
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, USA
| | - Behnaz Niroomand
- Skull Base Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pooya Eini
- Toxicological Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramin Ghanbarnia
- Skull Base Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdulrahman Albakr
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA
- Department of Surgery, Division of Neurosurgery, King Saud University, Riyadh, Saudi Arabia
| | - Hamid Borghei-Razavi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA.
| |
Collapse
|
4
|
Awuah WA, Ben-Jaafar A, Roy S, Nkrumah-Boateng PA, Tan JK, Abdul-Rahman T, Atallah O. Predicting survival in malignant glioma using artificial intelligence. Eur J Med Res 2025; 30:61. [PMID: 39891313 PMCID: PMC11783879 DOI: 10.1186/s40001-025-02339-3] [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: 11/07/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025] Open
Abstract
Malignant gliomas, including glioblastoma, are amongst the most aggressive primary brain tumours, characterised by rapid progression and a poor prognosis. Survival analysis is an essential aspect of glioma management and research, as most studies use time-to-event outcomes to assess overall survival (OS) and progression-free survival (PFS) as key measures to evaluate patients. However, predicting survival using traditional methods such as the Kaplan-Meier estimator and the Cox Proportional Hazards (CPH) model has faced many challenges and inaccuracies. Recently, advances in artificial intelligence (AI), including machine learning (ML) and deep learning (DL), have enabled significant improvements in survival prediction for glioma patients by integrating multimodal data such as imaging, clinical parameters and molecular biomarkers. This study highlights the comparative effectiveness of imaging-based, non-imaging and combined AI models. Imaging models excel at identifying tumour-specific features through radiomics, achieving high predictive accuracy. Non-imaging approaches also excel in utilising clinical and genetic data to provide complementary insights, whilst combined methods integrate multiple data modalities and have the greatest potential for accurate survival prediction. Limitations include data heterogeneity, interpretability challenges and computational demands, particularly in resource-limited settings. Solutions such as federated learning, lightweight AI models and explainable AI frameworks are proposed to overcome these barriers. Ultimately, the integration of advanced AI techniques promises to transform glioma management by enabling personalised treatment strategies and improved prognostic accuracy.
Collapse
Affiliation(s)
| | - Adam Ben-Jaafar
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Subham Roy
- Hull York Medical School, University of York, York, UK
| | | | - Joecelyn Kirani Tan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | | | - Oday Atallah
- Department of Neurosurgery, Carl Von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
5
|
Chong JK, Jain P, Prasad S, Dubey NK, Saxena S, Lo WC. Optimizing Glioblastoma, IDH-wildtype Treatment Outcomes : A Radiomics and Support Vector Machine-Based Approach to Overall Survival Estimation. J Korean Neurosurg Soc 2025; 68:7-18. [PMID: 39099033 PMCID: PMC11725457 DOI: 10.3340/jkns.2024.0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/20/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024] Open
Abstract
OBJECTIVE Glioblastoma multiforme (GBM), particularly the isocitrate dehydrogenase (IDH)-wildtype type, represents a significant clinical challenge due to its aggressive nature and poor prognosis. Despite advancements in medical imaging and its modalities, survival rates have not improved significantly, demanding innovative treatment planning and outcome prediction approaches. METHODS This study utilizes a support vector machine (SVM) classifier using radiomics features to predict the overall survival (OS) of GBM, IDH-wildtype patients to short (<12 months) and long (≥12 months) survivors. A dataset comprising multi-parametric magnetic resonance imaging scans from 574 patients was analyzed. Radiomic features were extracted from T1, T2, fluid-attenuated inversion recovery, and T1 with gadolinium (T1GD) sequences. Low variance features were removed, and recursive feature elimination was used to select the most informative features. The SVM model was trained using a k-fold cross-validation approach. Furthermore, clinical parameters such as age, gender, and MGMT (O6-methylguanine-DNA methyltransferase) promoter methylation status were integrated to enhance prediction accuracy. RESULTS The model showed reasonable results in terms of cross-validated area under the curve of 0.84 (95% confidence interval, 0.80-0.90) with (p<0.001) effectively categorizing patients into short and long survivors. Log-rank test (chi-square statistics) analysis for the developed model was 0.00029 along with the 1.20 Cohen's d effect size. Most importantly, clinical data integration further refined the survival estimates, providing a more fitted prediction that considers individual patient characteristics by Kaplan-Meier curve with p-value <0.0001. CONCLUSION The proposed method significantly enhances the predictive accuracy of OS outcomes in GBM, IDH-wildtype patients. By integrating detailed imaging features with key clinical indicators, this model offers a robust tool for personalized treatment planning, potentially improving OS.
Collapse
Affiliation(s)
- Jiunn-Kai Chong
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Priyanka Jain
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, India
| | - Shivani Prasad
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, India
| | - Navneet Kumar Dubey
- Executive Programme in Healthcare Management, Indian Institute of Management, Lucknow, India
| | - Sanjay Saxena
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, India
| | - Wen-Cheng Lo
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
- Division of Neurosurgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
6
|
Lin J, Su CQ, Tang WT, Xia ZW, Lu SS, Hong XN. Radiomic features on multiparametric MRI for differentiating pseudoprogression from recurrence in high-grade gliomas. Acta Radiol 2024; 65:1390-1400. [PMID: 39380365 DOI: 10.1177/02841851241283781] [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] [Indexed: 10/10/2024]
Abstract
BACKGROUND Distinguishing between tumor recurrence and pseudoprogression (PsP) in high-grade glioma postoperatively is challenging. This study aims to enhance this differentiation using a combination of intratumoral and peritumoral radiomics. PURPOSE To assess the effectiveness of intratumoral and peritumoral radiomics in improving the differentiation between high-grade glioma recurrence and pseudoprogression after surgery. MATERIAL AND METHODS A total of 109 cases were randomly divided into training and validation sets, with 1316 features extracted from intratumoral and peritumoral volumes of interest (VOIs) on conventional magnetic resonance imaging (MRI) and apparent diffusion coefficient (ADC) maps. Feature selection was performed using the mRMR algorithm, resulting in intratumoral (100 features), peritumoral (100 features), and combined (200 features) subsets. Optimal features were then selected using PCC and RFE algorithms and modeled using LR, SVM, and LDA classifiers. Diagnostic performance was compared using area under the receiver operating characteristic curve (AUC), evaluated in the validation set. A nomogram was established using radscores from intratumoral, peritumoral, and combined models. RESULTS The combined model, utilizing 14 optimal features (8 peritumoral, 6 intratumoral) and LR as the best classifier, outperformed the single intratumoral and peritumoral models. In the training set, the AUC values for the combined model, intratumoral model, and peritumoral model were 0.938, 0.921, and 0.847, respectively; in the validation set, the AUC values were 0.841, 0.755, and 0.705. The nomogram model demonstrated AUCs of 0.960 (training set) and 0.850 (validation set). CONCLUSION The combination of intratumoral and peritumoral radiomics is effective in distinguishing high-grade glioma recurrence from pseudoprogression after surgery.
Collapse
Affiliation(s)
- Jie Lin
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Chun-Qiu Su
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Wen-Tian Tang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Zhi-Wei Xia
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Shan-Shan Lu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Xun-Ning Hong
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| |
Collapse
|
7
|
Jiang C, Sun C, Wang X, Ma S, Jia W, Zhang D. BTK Expression Level Prediction and the High-Grade Glioma Prognosis Using Radiomic Machine Learning Models. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1359-1374. [PMID: 38381384 PMCID: PMC11300408 DOI: 10.1007/s10278-024-01026-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/22/2024]
Abstract
We aimed to study whether the Bruton's tyrosine kinase (BTK) expression is correlated with the prognosis of patients with high-grade gliomas (HGGs) and predict its expression level prior to surgery, by constructing radiomic models. Clinical and gene expression data of 310 patients from The Cancer Genome Atlas (TCGA) were included for gene-based prognostic analysis. Among them, contrast-enhanced T1-weighted imaging (T1WI + C) from The Cancer Imaging Archive (TCIA) with genomic data was selected from 82 patients for radiomic models, including support vector machine (SVM) and logistic regression (LR) models. Furthermore, the nomogram incorporating radiomic signatures was constructed to evaluate its clinical efficacy. BTK was identified as an independent risk factor for HGGs through univariate and multivariate Cox regression analyses. Three radiomic features were selected to construct the SVM and LR models, and the validation set showed area under curve (AUCs) values of 0.711 (95% CI, 0.598-0.824) and 0.736 (95% CI, 0.627-0.844), respectively. The median survival times of the high Rad_score and low-Rad_score groups based on LR model were 15.53 and 23.03 months, respectively. In addition, the total risk score of each patient was used to construct a predictive nomogram, and the AUCs calculated from the corresponding time-dependent ROC curves were 0.533, 0.659, and 0.767 for 1, 3, and 5 years, respectively. BTK is an independent risk factor associated with poor prognosis in patients, and the radiomic model constructed in this study can effectively and non-invasively predict preoperative BTK expression levels and patient prognosis based on T1WI + C.
Collapse
Affiliation(s)
- Chenggang Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Chen Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Xi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Dainan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China.
| |
Collapse
|
8
|
Zhou Q, Wang Y, Zhang Q, Wei X, Yao Y, Xia L. Noninvasive prediction of CCL2 expression level in high-grade glioma patients. Cancer Med 2024; 13:e70016. [PMID: 39030882 PMCID: PMC11257997 DOI: 10.1002/cam4.70016] [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: 11/21/2023] [Revised: 06/21/2024] [Accepted: 07/05/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND Gliomas are recognized as the most frequent type of malignancies in the central nervous system, and efficacious prognostic indicators are essential to treat patients with gliomas and improve their clinical outcomes. The chemokine (C-C motif) ligand 2 (CCL2) is a promising predictor for glioma malignancy and progression. However, at present, the methods to evaluate CCL2 expression level are invasive and operator-dependent. OBJECTIVE It was expected to noninvasively predict CCL2 expression levels in malignant glioma tissues by magnetic resonance imaging (MRI)-based radiomics and assess the association between the developed radiomics model and prognostic indicators and related genes. METHODS MRI-based radiomics was used to predict CCL2 expression level using data obtained from The Cancer Imaging Archive (TCIA) and The Cancer Genome Atlas (TCGA) databases. A support vector machine (SVM)-based radiomics model and a logistic regression (LR)-based radiomics model were used to predict the radiomics score, and its correlation with CCL2 expression level was analyzed. RESULTS The results revealed that there was an association between CCL2 expression level and the overall survival of cases with gliomas, and bioinformatics correlation analysis showed that CCL2 expression level was highly correlated with disease-related pathways, such as mTOR signaling pathway, cGMP-PKG signaling pathway, and MAPK signaling pathway. Both SVM- and LR-based radiomics data robustly predicted CCL2 expression level, and radiomics scores could also be used to predict the overall survival of patients. Moreover, the high/low radiomics scores were highly correlated with the known glioma-related genes, including CD70, CD27, and PDCD1. CONCLUSION An MRI-based radiomics model was successfully developed, and its clinical benefits were confirmed, including the prediction of CCL2 expression level and patients' prognosis.
Collapse
Affiliation(s)
- Qingqing Zhou
- Department of NeurosurgeryThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Yamei Wang
- Department of NeurologyThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Qing Zhang
- Department of RadiologyThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - XiaoMing Wei
- Department of NeurosurgeryThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Yuan Yao
- Department of NeurosurgeryThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Liang Xia
- Department of NeurosurgeryThe Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of SciencesHangzhouPeople's Republic of China
| |
Collapse
|
9
|
Dhamdhere R, Bharadwaj S, Aggarwal A, Mutha P, Shi W, Marteau B, Madabhushi A, Wang MD. Interpretable Survival Risk Prediction for High-Grade Glioma Patients via Radiomic Features from Peritumoral Region. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-5. [PMID: 40040135 DOI: 10.1109/embc53108.2024.10782039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Peritumoral edema regions carry prognostic value in patients with high-grade glioma (HGG), the most invasive type of brain cancer. Recent findings have established the association of texture and shape features extracted from these regions with survival outcomes. However, no study has converged on a single feature that significantly correlates with survival outcomes. In this study, we develop an automated and interpretable brain tumor patient survival risk prediction model using radiomic features from the peritumoral region of HGG. First, the peritumoral edema regions are segmented from MRI scans imaged using multiple modalities (T1, T2, FLAIR, T1-contrast enhanced) and compiled into the BraTS-2020 dataset. Texture and shape features extracted from the segmented regions were analyzed to stratify patients based on a risk score. The proposed framework demonstrates the significance of a texture and shape feature to predict survival outcomes for a subset of 76 HGG patients with survival information. Moreover, we conduct univariate and multivariable analysis to further demonstrate the clinical utility of the extracted texture and shape features. The study provides evidence for the importance of texture and shape features extracted from peritumoral edema regions in predicting survival outcomes in HGG patients. It may facilitate personalized treatment and improve the prognostic accuracy of HGG patients in real-world clinical setting.
Collapse
|
10
|
Zhang H, Ouyang Y, Zhang H, Zhang Y, Su R, Zhou B, Yang W, Lei Y, Huang B. Sub-region based radiomics analysis for prediction of isocitrate dehydrogenase and telomerase reverse transcriptase promoter mutations in diffuse gliomas. Clin Radiol 2024; 79:e682-e691. [PMID: 38402087 DOI: 10.1016/j.crad.2024.01.030] [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: 08/25/2023] [Revised: 01/16/2024] [Accepted: 01/21/2024] [Indexed: 02/26/2024]
Abstract
AIM To enhance the prediction of mutation status of isocitrate dehydrogenase (IDH) and telomerase reverse transcriptase (TERT) promoter, which are crucial for glioma prognostication and therapeutic decision-making, via sub-regional radiomics analysis based on multiparametric magnetic resonance imaging (MRI). MATERIALS AND METHODS A retrospective study was conducted on 401 participants with adult-type diffuse gliomas. Employing the K-means algorithm, tumours were clustered into two to four subregions. Sub-regional radiomics features were extracted and selected using the Mann-Whitney U-test, Pearson correlation analysis, and least absolute shrinkage and selection operator, forming the basis for predictive models. The performance of model combinations of different sub-regional features and classifiers (including logistic regression, support vector machines, K-nearest neighbour, light gradient boosting machine, and multilayer perceptron) was evaluated using an external test set. RESULTS The models demonstrated high predictive performance, with area under the receiver operating characteristic curve (AUC) values ranging from 0.918 to 0.994 in the training set for IDH mutation prediction and from 0.758 to 0.939 for TERT promoter mutation prediction. In the external test sets, the two-cluster radiomics features and the logistic regression model yielded the highest prediction for IDH mutation, resulting in an AUC of 0.905. Additionally, the most effective predictive performance with an AUC of 0.803 was achieved using the four-cluster radiomics features and the support vector machine model, specifically for TERT promoter mutation prediction. CONCLUSION The present study underscores the potential of sub-regional radiomics analysis in predicting IDH and TERT promoter mutations in glioma patients. These models have the capacity to refine preoperative glioma diagnosis and contribute to personalised therapeutic interventions for patients.
Collapse
Affiliation(s)
- H Zhang
- Department of Radiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 517108, China; Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Y Ouyang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - H Zhang
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Y Zhang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - R Su
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - B Zhou
- Department of Radiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 517108, China
| | - W Yang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Y Lei
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - B Huang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
11
|
Ignatova TN, Chaitin HJ, Kukekov NV, Suslov ON, Dulatova GI, Hanafy KA, Vrionis FD. Gliomagenesis is orchestrated by the Oct3/4 regulatory network. J Neurosurg Sci 2024; 68:148-156. [PMID: 34342203 DOI: 10.23736/s0390-5616.21.05437-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a lethal brain tumor characterized by developmental hierarchical phenotypic heterogeneity, therapy resistance and recurrent growth. Neural stem cells (NSCs) from human central nervous system (CNS), and glioblastoma stem cells from patient-derived GBM (pdGSC) samples were cultured in both 2D well-plate and 3D monoclonal neurosphere culture system (pdMNCS). The pdMNCS model shows promise to establish a relevant 3D-tumor environment that maintains GBM cells in the stem cell phase within suspended neurospheres. METHODS Utilizing the pdMNCS, we examined GBM cell-lines for a wide spectrum of developmental cancer stem cell markers, including the early blastocyst inner-cell mass (ICM)-specific Nanog, Oct3/4,B, and CD133. RESULTS We observed that MNCS epigenotype is recapitulated using gliomasphere-derived cells. CD133, the marker of GSC is robustly expressed in 3D-gliomaspheres and localized within the plasma membrane compartment. Conversely, gliomasphere cultures grown in conventional 2D culture quickly lost CD133 expression, indicating its variable expression is dependent on cell-culture conditions. Incomplete differentiation of cytoskeleton microtubules and intermediate filaments (IFs) of patient derived cells, similar to commercially available GBM cell lines, was seen. Subsequently, in order to determine whether Oct3/4 it was necessary for CD133 expression and cancer stemness, we transfected 2D and 3D culture with siRNA against Oct3/4 and found a significant reduction in gliomasphere formation. CONCLUSIONS These results suggest that expression of Oct3/4,A- and CD133 suppress differentiation of GSCs.
Collapse
Affiliation(s)
- Tatyana N Ignatova
- Department of Neurosurgery, University of Tennessee, Health Science Center, Memphis, TN, USA
- Marcus Neuroscience Institute, Boca Raton Regional Hospital and Florida Atlantic University, Boca Raton, FL, USA
| | - Hersh J Chaitin
- College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Nickolay V Kukekov
- Department of Pathology and Center for Neurobiology and Behavior, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Oleg N Suslov
- McKnight Brain Institute, Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Galina I Dulatova
- Department of Neurosurgery, University of Tennessee, Health Science Center, Memphis, TN, USA
| | - Khalid A Hanafy
- Marcus Neuroscience Institute, Boca Raton Regional Hospital and Florida Atlantic University, Boca Raton, FL, USA
| | - Frank D Vrionis
- Marcus Neuroscience Institute, Boca Raton Regional Hospital and Florida Atlantic University, Boca Raton, FL, USA -
| |
Collapse
|
12
|
Lee JO, Ahn SS, Choi KS, Lee J, Jang J, Park JH, Hwang I, Park CK, Park SH, Chung JW, Choi SH. Added prognostic value of 3D deep learning-derived features from preoperative MRI for adult-type diffuse gliomas. Neuro Oncol 2024; 26:571-580. [PMID: 37855826 PMCID: PMC10912011 DOI: 10.1093/neuonc/noad202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND To investigate the prognostic value of spatial features from whole-brain MRI using a three-dimensional (3D) convolutional neural network for adult-type diffuse gliomas. METHODS In a retrospective, multicenter study, 1925 diffuse glioma patients were enrolled from 5 datasets: SNUH (n = 708), UPenn (n = 425), UCSF (n = 500), TCGA (n = 160), and Severance (n = 132). The SNUH and Severance datasets served as external test sets. Precontrast and postcontrast 3D T1-weighted, T2-weighted, and T2-FLAIR images were processed as multichannel 3D images. A 3D-adapted SE-ResNeXt model was trained to predict overall survival. The prognostic value of the deep learning-based prognostic index (DPI), a spatial feature-derived quantitative score, and established prognostic markers were evaluated using Cox regression. Model evaluation was performed using the concordance index (C-index) and Brier score. RESULTS The MRI-only median DPI survival prediction model achieved C-indices of 0.709 and 0.677 (BS = 0.142 and 0.215) and survival differences (P < 0.001 and P = 0.002; log-rank test) for the SNUH and Severance datasets, respectively. Multivariate Cox analysis revealed DPI as a significant prognostic factor, independent of clinical and molecular genetic variables: hazard ratio = 0.032 and 0.036 (P < 0.001 and P = 0.004) for the SNUH and Severance datasets, respectively. Multimodal prediction models achieved higher C-indices than models using only clinical and molecular genetic variables: 0.783 vs. 0.774, P = 0.001, SNUH; 0.766 vs. 0.748, P = 0.023, Severance. CONCLUSIONS The global morphologic feature derived from 3D CNN models using whole-brain MRI has independent prognostic value for diffuse gliomas. Combining clinical, molecular genetic, and imaging data yields the best performance.
Collapse
Affiliation(s)
- Jung Oh Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Artificial Intelligence Collaborative Network, Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Soo Ahn
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyu Sung Choi
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Artificial Intelligence Collaborative Network, Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Junhyeok Lee
- Interdisciplinary Programs in Cancer Biology Major, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Joon Jang
- Department of Biomedical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jung Hyun Park
- Department of Radiology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Inpyeong Hwang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Artificial Intelligence Collaborative Network, Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Hye Park
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jin Wook Chung
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Artificial Intelligence Collaborative Network, Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Innovate Biomedical Technology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Artificial Intelligence Collaborative Network, Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea
| |
Collapse
|
13
|
Lai Y, Wu Y, Chen X, Gu W, Zhou G, Weng M. MRI-based Machine Learning Radiomics Can Predict CSF1R Expression Level and Prognosis in High-grade Gliomas. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:209-229. [PMID: 38343263 PMCID: PMC10976932 DOI: 10.1007/s10278-023-00905-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 03/02/2024]
Abstract
The purpose of this study is to predict the mRNA expression of CSF1R in HGG non-invasively using MRI (magnetic resonance imaging) omics technology and to evaluate the correlation between the established radiomics model and prognosis. We investigated the predictive value of CSF1R in the Cancer Genome Atlas (TCGA) and The Cancer Imaging Archive (TCIA) database. The Support vector machine (SVM) and the Logistic regression (LR) algorithms were used to create a radiomics_score (Rad_score), respectively. The effectiveness and performance of the radiomics model was assessed in the training (n = 89) and tenfold cross-validation sets. We further analyzed the correlation between Rad_score and macrophage-related genes using Spearman correlation analysis. A radiomics nomogram combining the clinical factors and Rad_score was constructed to validate the radiomic signatures for individualized survival estimation and risk stratification. The results showed that CSF1R expression was markedly elevated in HGG tissues, which was related to worse prognosis. CSF1R expression was closely related to the abundance of infiltrating immune cells, such as macrophages. We identified nine features for establishing a radiomics model. The radiomics model predicting CSF1R achieved high AUC in training (0.768 in SVM and 0.792 in LR) and tenfold cross-validation sets (0.706 in SVM and 0.717 in LR). Rad_score was highly associated with tumor-related macrophage genes. A radiomics nomogram combining the Rad_score and clinical factors was constructed and revealed satisfactory performance. MRI-based Rad_score is a novel way to predict CSF1R expression and prognosis in high-grade glioma patients. The radiomics nomogram could optimize individualized survival estimation for HGG patients.
Collapse
Affiliation(s)
- Yuling Lai
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yiyang Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiangyuan Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenchao Gu
- Department of Diagnostic and Interventional Radiology, University of Tsukuba, Ibaraki, Japan.
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan.
| | - Guoxia Zhou
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| | - Meilin Weng
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
14
|
Huo X, Wang Y, Ma S, Zhu S, Wang K, Ji Q, Chen F, Wang L, Wu Z, Li W. Multimodal MRI-based radiomic nomogram for predicting telomerase reverse transcriptase promoter mutation in IDH-wildtype histological lower-grade gliomas. Medicine (Baltimore) 2023; 102:e36581. [PMID: 38134061 PMCID: PMC10735121 DOI: 10.1097/md.0000000000036581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
The presence of TERTp mutation in isocitrate dehydrogenase-wildtype (IDHwt) histologically lower-grade glioma (LGA) has been linked to a poor prognosis. In this study, we aimed to develop and validate a radiomic nomogram based on multimodal MRI for predicting TERTp mutations in IDHwt LGA. One hundred and nine IDH wildtype glioma patients (TERTp-mutant, 78; TERTp-wildtype, 31) with clinical, radiomic, and molecular information were collected and randomly divided into training and validation set. Clinical model, fusion radiomic model, and combined radiomic nomogram were constructed for the discrimination. Radiomic features were screened with 3 algorithms (Wilcoxon rank sum test, elastic net, and the recursive feature elimination) and the clinical characteristics of combined radiomic nomogram were screened by the Akaike information criterion. Finally, receiver operating characteristic curve, calibration curve, Hosmer-Lemeshow test, and decision curve analysis were utilized to assess these models. Fusion radiomic model with 4 radiomic features achieved an area under the curve value of 0.876 and 0.845 in the training and validation set. And, the combined radiomic nomogram achieved area under the curve value of 0.897 (training set) and 0.882 (validation set). Above that, calibration curve and Hosmer-Lemeshow test showed that the radiomic model and combined radiomic nomogram had good agreement between observations and predictions in the training set and the validation set. Finally, the decision curve analysis revealed that the 2 models had good clinical usefulness for the prediction of TERTp mutation status in IDHwt LGA. The combined radiomics nomogram performed great performance and high sensitivity in prediction of TERTp mutation status in IDHwt LGA, and has good clinical application.
Collapse
Affiliation(s)
- Xulei Huo
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yali Wang
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Sihan Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Sipeng Zhu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ke Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qiang Ji
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Feng Chen
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Dou Y, Li X, Tao J, Dong Y, Xu N, Wang S. Prediction of high-grade soft-tissue sarcoma using a combined intratumoural and peritumoural MRI-based radiomics nomogram. Clin Radiol 2023; 78:e1032-e1040. [PMID: 37748959 DOI: 10.1016/j.crad.2023.08.020] [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: 11/20/2022] [Revised: 08/03/2023] [Accepted: 08/29/2023] [Indexed: 09/27/2023]
Abstract
AIM To develop an intratumoural and peritumoural magnetic resonance imaging (MRI)-based radiomics nomogram for predicting tumour grade to improve clinical treatment and long-term prognosis. MATERIALS AND METHODS MRI (3 T) features and T2-weighted imaging with fat-saturation (T2WI-FS)-based radiomics features of 57 patients with soft-tissue sarcoma (STS) were analysed retrospectively. Tumour size, ratio of width and length, relative depth to the peripheral fascia, peritumoural oedema, heterogeneity on T2WI, necrosis signal, enhancement model, and peritumoural enhancement were obtained. Independent risk factors were screened to construct an MRI feature nomogram. Radiomics features were obtained from intratumoural and peritumoural images on T2WI-FS. The optimal radiomics model was selected by the four-step dimensionality reduction method of minimum and maximum normalisation, optimal feature selection, selection based on support vector machine with L1-norm regularisation model, and iterative feature selection. MRI features and optimal radiomics features were used to construct a radiomics nomogram. The MRI feature nomogram model, the radiomics model, and the radiomics nomogram model were assessed by receiver operating characteristic (ROC) curves and calibration curves of the training and validation sets. RESULTS Heterogeneity on T2WI and peritumoural enhancement were independent risk factors for predicting high-grade STS. The areas under the curves of the training set and verification set of the three models were as follows: MRI feature nomogram, 0.86 and 0.83, respectively; intratumoural and peritumoural combined radiomics model, 0.99 and 0.86, respectively; and radiomics nomogram model, 0.98 and 0.96, respectively. CONCLUSION The radiomics nomogram model based on MRI features and combined intratumoural and peritumoural radiomic features was best able to predict high-grade STS.
Collapse
Affiliation(s)
- Y Dou
- Department of Ultrasound, The First Affiliated Hospital, Dalian Medical University, Dalian, China; Department of Radiology, The Second Hospital, Dalian Medical University, Dalian, China
| | - X Li
- Department of Radiology, Huashan Hospital Fudan University, Shanghai, China
| | - J Tao
- Department of Pathology, The Second Hospital, Dalian Medical University, Dalian, China
| | - Y Dong
- Department of Radiology, The Second Hospital, Dalian Medical University, Dalian, China
| | - N Xu
- Department of Radiology, The Second Hospital, Dalian Medical University, Dalian, China
| | - S Wang
- Department of Radiology, The Second Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
16
|
Le VH, Minh TNT, Kha QH, Le NQK. A transfer learning approach on MRI-based radiomics signature for overall survival prediction of low-grade and high-grade gliomas. Med Biol Eng Comput 2023; 61:2699-2712. [PMID: 37432527 DOI: 10.1007/s11517-023-02875-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/20/2023] [Indexed: 07/12/2023]
Abstract
Lower-grade gliomas (LGG) can eventually progress to glioblastoma (GBM) and death. In the context of the transfer learning approach, we aimed to train and test an MRI-based radiomics model for predicting survival in GBM patients and validate it in LGG patients. From each patient's 704 MRI-based radiomics features, we chose seventeen optimal radiomics signatures in the GBM training set (n = 71) and used these features in both the GBM testing set (n = 31) and LGG validation set (n = 107) for further analysis. Each patient's risk score, calculated based on those optimal radiomics signatures, was chosen to represent the radiomics model. We compared the radiomics model with clinical, gene status models, and combined model integrating radiomics, clinical, and gene status in predicting survival. The average iAUCs of combined models in training, testing, and validation sets were respectively 0.804, 0.878, and 0.802, and those of radiomics models were 0.798, 0.867, and 0.717. The average iAUCs of gene status and clinical models ranged from 0.522 to 0.735 in all three sets. The radiomics model trained in GBM patients can effectively predict the overall survival of GBM and LGG patients, and the combined model improved this ability.
Collapse
Affiliation(s)
- Viet Huan Le
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Department of Thoracic Surgery, Khanh Hoa General Hospital, Nha Trang City, 65000, Vietnam
| | - Tran Nguyen Tuan Minh
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Quang Hien Kha
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei, 110, Taiwan.
- AIBioMed Research Group, Taipei Medical University, Taipei, 110, Taiwan.
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, 110, Taiwan.
| |
Collapse
|
17
|
Hung KC, Sun CK, Chang YP, Wu JY, Huang PY, Liu TH, Lin CH, Cheng WJ, Chen IW. Association of prognostic nutritional index with prognostic outcomes in patients with glioma: a meta-analysis and systematic review. Front Oncol 2023; 13:1188292. [PMID: 37564929 PMCID: PMC10411533 DOI: 10.3389/fonc.2023.1188292] [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: 03/17/2023] [Accepted: 07/04/2023] [Indexed: 08/12/2023] Open
Abstract
Background The potential link between Prognostic Nutritional Index (PNI) and prognosis in patients with glioma remains uncertain. This meta-analysis was conducted to assess the clinical value of PNI in glioma patients by integrating all available evidence to enhance statistical power. Method A systematic search of databases including Medline, EMBASE, Google Scholar, and Cochrane Library was conducted from inception to January 8, 2023 to retrieve all pertinent peer-reviewed articles. The primary outcome of the study was to examine the association between a high PNI value and overall survival, while secondary outcome included the relationship between a high PNI and progression-free survival. Results In this meta-analysis, we included 13 retrospective studies published from 2016 to 2022, which analyzed a total of 2,712 patients. Across all studies, surgery was the primary treatment modality, with or without chemotherapy and radiotherapy as adjunct therapies. A high PNI was linked to improved overall survival (Hazard Ratio (HR) = 0.61, 95% CI: 0.52 to 0.72, p < 0.00001, I2 = 25%), and this finding remained consistent even after conducting sensitivity analysis. Subgroup analyses based on ethnicity (Asian vs. non-Asian), sample size (<200 vs. >200), and source of hazard ratio (univariate vs. multivariate) yielded consistent outcomes. Furthermore, patients with a high PNI had better progression-free survival than those with a low PNI (HR=0.71, 95% CI: 0.58 to 0.88, p=0.001, I2 = 0%). Conclusion Our meta-analysis suggested that a high PNI was associated with better overall survival and progression-free survival in patients with glioma. These findings may have important implications in the treatment of patients with glioma. Additional studies on a larger scale are necessary to investigate if integrating the index into the treatment protocol leads to improved clinical outcomes in individuals with glioma. Systematic review registration [https://www.crd.york.ac.uk/prospero/], identifier [CRD42023389951].
Collapse
Affiliation(s)
- Kuo-Chuan Hung
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
| | - Cheuk-Kwan Sun
- Department of Emergency Medicine, E-Da Dachang Hospital, I-Shou University, Kaohsiung, Taiwan
- School of Medicine for International, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yang-Pei Chang
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jheng-Yan Wu
- Department of Nutrition, Chi Mei Medical Center, Tainan, Taiwan
| | - Po-Yu Huang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Ting-Hui Liu
- Department of General Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Chien-Hung Lin
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
| | - Wan-Jung Cheng
- Department of Anesthesiology, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - I-Wen Chen
- Department of Anesthesiology, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| |
Collapse
|
18
|
Tewarie IA, Senko AW, Jessurun CAC, Zhang AT, Hulsbergen AFC, Rendon L, McNulty J, Broekman MLD, Peng LC, Smith TR, Phillips JG. Predicting leptomeningeal disease spread after resection of brain metastases using machine learning. J Neurosurg 2023; 138:1561-1569. [PMID: 36272119 DOI: 10.3171/2022.8.jns22744] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/25/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The incidence of leptomeningeal disease (LMD) has increased as treatments for brain metastases (BMs) have improved and patients with metastatic disease are living longer. Sample sizes of individual studies investigating LMD after surgery for BMs and its risk factors have been limited, ranging from 200 to 400 patients at risk for LMD, which only allows the use of conventional biostatistics. Here, the authors used machine learning techniques to enhance LMD prediction in a cohort of surgically treated BMs. METHODS A conditional survival forest, a Cox proportional hazards model, an extreme gradient boosting (XGBoost) classifier, an extra trees classifier, and logistic regression were trained. A synthetic minority oversampling technique (SMOTE) was used to train the models and handle the inherent class imbalance. Patients were divided into an 80:20 training and test set. Fivefold cross-validation was used on the training set for hyperparameter optimization. Patients eligible for study inclusion were adults who had consecutively undergone neurosurgical BM treatment, had been admitted to Brigham and Women's Hospital from January 2007 through December 2019, and had a minimum of 1 month of follow-up after neurosurgical treatment. RESULTS A total of 1054 surgically treated BM patients were included in this analysis. LMD occurred in 168 patients (15.9%) at a median of 7.05 months after BM diagnosis. The discrimination of LMD occurrence was optimal using an XGboost algorithm (area under the curve = 0.83), and the time to LMD was prognosticated evenly by the random forest algorithm and the Cox proportional hazards model (C-index = 0.76). The most important feature for both LMD classification and regression was the BM proximity to the CSF space, followed by a cerebellar BM location. Lymph node metastasis of the primary tumor at BM diagnosis and a cerebellar BM location were the strongest risk factors for both LMD occurrence and time to LMD. CONCLUSIONS The outcomes of LMD patients in the BM population are predictable using SMOTE and machine learning. Lymph node metastasis of the primary tumor at BM diagnosis and a cerebellar BM location were the strongest LMD risk factors.
Collapse
Affiliation(s)
- Ishaan Ashwini Tewarie
- 1Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- 4Department of Neurosurgery, Leiden Medical Center, Leiden, The Netherlands; and
| | - Alexander W Senko
- 1Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Charissa A C Jessurun
- 1Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- 3Department of Neurosurgery, Haaglanden Medical Center, The Hague
- 4Department of Neurosurgery, Leiden Medical Center, Leiden, The Netherlands; and
| | - Abigail Tianai Zhang
- 1Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alexander F C Hulsbergen
- 1Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- 3Department of Neurosurgery, Haaglanden Medical Center, The Hague
- 4Department of Neurosurgery, Leiden Medical Center, Leiden, The Netherlands; and
| | - Luis Rendon
- 1Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jack McNulty
- 1Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marike L D Broekman
- 1Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- 3Department of Neurosurgery, Haaglanden Medical Center, The Hague
- 4Department of Neurosurgery, Leiden Medical Center, Leiden, The Netherlands; and
| | - Luke C Peng
- 1Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Timothy R Smith
- 1Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - John G Phillips
- 1Department of Neurosurgery, Computational Neuroscience Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- 5Department of Radiation Oncology, Tennessee Oncology, Nashville, Tennessee
| |
Collapse
|
19
|
Li L, Hou M, Fang S. Application of colony-stimulating factor 3 in determining the prognosis of high-grade gliomas based on magnetic resonance imaging radiomics. Heliyon 2023; 9:e15325. [PMID: 37095939 PMCID: PMC10122032 DOI: 10.1016/j.heliyon.2023.e15325] [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: 11/10/2022] [Revised: 04/02/2023] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Rationale and objectives Radiomics is a promising, non-invasive method for determining the prognosis of high-grade glioma (HGG). The connection between radiomics and the HGG prognostic biomarker is still insufficient. Materials and methods In this study, we collected the pathological, clinical, RNA-sequencing, and enhanced MRI data of HGG from TCIA and TCGA databases. We characterized the prognostic value of CSF3. Kaplan-Meier (KM) analysis, univariate and multivariate Cox regression, subgroup analysis, Spearman analysis, and gene set variation analysis enrichment were used to elucidate the prognostic value of the CSF3 gene and the correlation between CSF3 and tumor features. We used CIBERSORT to analyze the correlation between CSF3 and cancer immune infiltrates. Logistic regression (LR) and support vector machine methods (SVM) were used to build the radiomics models for the prognosis prediction of HGG based on the expression of CSF3. Results Based on the radiomics score calculated from LR model, 182 patients with HGG from TCGA database were divided into radiomics score (RS) high and low groups. CSF3 expression varied between tumor and normal group tissues. CSF3 expression was found to be a significant risk factor for survival outcomes. A positive association was found between CSF3 expression and immune infiltration. The radiomics model based on both LR and SVM methods showed high clinical practicability. Conclusion The results showed that CSF3 has a prognostic value in HGG. The developed radiomics models can predict the expression of CSF3, and further validate the predictions of the radiomics models for HGG.
Collapse
Affiliation(s)
- Leina Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Laboratory Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
- Corresponding author. Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| | - Meidan Hou
- Department of Radiology, The Second Affiliated Hospital of Dalian Medical University Dalian, Liaoning, China
| | - Shaobo Fang
- Department of Medical Imaging, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
20
|
Prediction of O-6-methylguanine-DNA methyltransferase and overall survival of the patients suffering from glioblastoma using MRI-based hybrid radiomics signatures in machine and deep learning framework. Neural Comput Appl 2023. [DOI: 10.1007/s00521-023-08405-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
|
21
|
Luo J, Pan M, Mo K, Mao Y, Zou D. Emerging role of artificial intelligence in diagnosis, classification and clinical management of glioma. Semin Cancer Biol 2023; 91:110-123. [PMID: 36907387 DOI: 10.1016/j.semcancer.2023.03.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 03/05/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023]
Abstract
Glioma represents a dominant primary intracranial malignancy in the central nervous system. Artificial intelligence that mainly includes machine learning, and deep learning computational approaches, presents a unique opportunity to enhance clinical management of glioma through improving tumor segmentation, diagnosis, differentiation, grading, treatment, prediction of clinical outcomes (prognosis, and recurrence), molecular features, clinical classification, characterization of the tumor microenvironment, and drug discovery. A growing body of recent studies apply artificial intelligence-based models to disparate data sources of glioma, covering imaging modalities, digital pathology, high-throughput multi-omics data (especially emerging single-cell RNA sequencing and spatial transcriptome), etc. While these early findings are promising, future studies are required to normalize artificial intelligence-based models to improve the generalizability and interpretability of the results. Despite prominent issues, targeted clinical application of artificial intelligence approaches in glioma will facilitate the development of precision medicine of this field. If these challenges can be overcome, artificial intelligence has the potential to profoundly change the way patients with or at risk of glioma are provided with more rational care.
Collapse
Affiliation(s)
- Jiefeng Luo
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Mika Pan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Ke Mo
- Clinical Research Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Yingwei Mao
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA.
| | - Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China; Clinical Research Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China.
| |
Collapse
|
22
|
Salome P, Sforazzini F, Grugnara G, Kudak A, Dostal M, Herold-Mende C, Heiland S, Debus J, Abdollahi A, Knoll M. MR Intensity Normalization Methods Impact Sequence Specific Radiomics Prognostic Model Performance in Primary and Recurrent High-Grade Glioma. Cancers (Basel) 2023; 15:cancers15030965. [PMID: 36765922 PMCID: PMC9913466 DOI: 10.3390/cancers15030965] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
PURPOSE This study investigates the impact of different intensity normalization (IN) methods on the overall survival (OS) radiomics models' performance of MR sequences in primary (pHGG) and recurrent high-grade glioma (rHGG). METHODS MR scans acquired before radiotherapy were retrieved from two independent cohorts (rHGG C1: 197, pHGG C2: 141) from multiple scanners (15, 14). The sequences are T1 weighted (w), contrast-enhanced T1w (T1wce), T2w, and T2w-FLAIR. Sequence-specific significant features (SF) associated with OS, extracted from the tumour volume, were derived after applying 15 different IN methods. Survival analyses were conducted using Cox proportional hazard (CPH) and Poisson regression (POI) models. A ranking score was assigned based on the 10-fold cross-validated (CV) concordance index (C-I), mean square error (MSE), and the Akaike information criterion (AICs), to evaluate the methods' performance. RESULTS Scatter plots of the 10-CV C-I and MSE against the AIC showed an impact on the survival predictions between the IN methods and MR sequences (C1/C2 C-I range: 0.62-0.71/0.61-0.72, MSE range: 0.20-0.42/0.13-0.22). White stripe showed stable results for T1wce (C1/C2 C-I: 0.71/0.65, MSE: 0.21/0.14). Combat (0.68/0.62, 0.22/0.15) and histogram matching (HM, 0.67/0.64, 0.22/0.15) showed consistent prediction results for T2w models. They were also the top-performing methods for T1w in C2 (Combat: 0.67, 0.13; HM: 0.67, 0.13); however, only HM achieved high predictions in C1 (0.66, 0.22). After eliminating IN impacted SF using Spearman's rank-order correlation coefficient, a mean decrease in the C-I and MSE of 0.05 and 0.03 was observed in all four sequences. CONCLUSION The IN method impacted the predictive power of survival models; thus, performance is sequence-dependent.
Collapse
Affiliation(s)
- Patrick Salome
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- Heidelberg Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Correspondence: (P.S.); (M.K.)
| | - Francesco Sforazzini
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- Heidelberg Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
| | - Gianluca Grugnara
- Department of Neuroradiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Andreas Kudak
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- CCU Radiation Therapy, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
| | - Matthias Dostal
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- CCU Radiation Therapy, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
| | - Christel Herold-Mende
- Brain Tumour Group, European Organization for Research and Treatment of Cancer, 1200 Brussels, Belgium
- Division of Neurosurgical Research, Department of Neurosurgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Sabine Heiland
- Department of Neuroradiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Jürgen Debus
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
| | - Amir Abdollahi
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
| | - Maximilian Knoll
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- Correspondence: (P.S.); (M.K.)
| |
Collapse
|
23
|
Nomogram Model for Predicting the Prognosis of High-Grade Glioma in Adults Receiving Standard Treatment: A Retrospective Cohort Study. J Clin Med 2022; 12:jcm12010196. [PMID: 36614997 PMCID: PMC9821755 DOI: 10.3390/jcm12010196] [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: 11/14/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES To identify the critical factors associated with the progression-free survival (PFS) and overall survival (OS) of high-grade glioma (HGG) in adults who have received standard treatment and establish a novel graphical nomogram and an online dynamic nomogram. PATIENTS AND METHODS This is a retrospective study of adult HGG patients receiving standard treatment (surgery, postoperative radiotherapy, and temozolomide (TMZ) chemotherapy) at Huashan Hospital, Fudan University between January 2017 and December 2019. We used uni- and multi-variable COX models to identify the significant prognostic factors for PFS and OS. Based on the significant predictors, graphical and online nomograms were established. RESULTS A total of 246 patients were enrolled in the study based on the inclusion criteria. The average PFS and OS were 22.99 ± 11.43 and 30.51 ± 13.73 months, respectively. According to the multi-variable COX model, age, extent of resection (EOR), and IDH mutation were associated with PFS and OS, while edema index (EI) was relevant to PFS. In addition, patients with IDH and TERT promoter co-mutations had longer PFSs and OSs, and no apparent survival benefit was found in the long-cycle TMZ adjuvant chemotherapy compared with the standard Stupp protocol. Based on these critical factors, a graphical nomogram and online nomogram were developed for predicting PFS and OS, respectively. The calibration curve showed favorable consistency between the predicted and actual survival rates. C-index and time-dependent AUC showed good discrimination abilities. CONCLUSIONS We identified the significant predictors for the PFS and OS of HGG adults receiving standard treatment and established user-friendly nomogram models to assist neurosurgeons in optimizing clinical management and treatment strategies.
Collapse
|
24
|
Yang Z, Chen M, Kazemimoghadam M, Ma L, Stojadinovic S, Wardak Z, Timmerman R, Dan T, Lu W, Gu X. Ensemble learning for glioma patients overall survival prediction using pre-operative MRIs. Phys Med Biol 2022; 67:10.1088/1361-6560/aca375. [PMID: 36384039 PMCID: PMC9990877 DOI: 10.1088/1361-6560/aca375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/16/2022] [Indexed: 11/18/2022]
Abstract
Objective: Gliomas are the most common primary brain tumors. Approximately 70% of the glioma patients diagnosed with glioblastoma have an averaged overall survival (OS) of only ∼16 months. Early survival prediction is essential for treatment decision-making in glioma patients. Here we proposed an ensemble learning approach to predict the post-operative OS of glioma patients using only pre-operative MRIs.Approach: Our dataset was from the Medical Image Computing and Computer Assisted Intervention Brain Tumor Segmentation challenge 2020, which consists of multimodal pre-operative MRI scans of 235 glioma patients with survival days recorded. The backbone of our approach was a Siamese network consisting of twinned ResNet-based feature extractors followed by a 3-layer classifier. During training, the feature extractors explored traits of intra and inter-class by minimizing contrastive loss of randomly paired 2D pre-operative MRIs, and the classifier utilized the extracted features to generate labels with cost defined by cross-entropy loss. During testing, the extracted features were also utilized to define distance between the test sample and the reference composed of training data, to generate an additional predictor via K-NN classification. The final label was the ensemble classification from both the Siamese model and the K-NN model.Main results: Our approach classifies the glioma patients into 3 OS classes: long-survivors (>15 months), mid-survivors (between 10 and 15 months) and short-survivors (<10 months). The performance is assessed by the accuracy (ACC) and the area under the curve (AUC) of 3-class classification. The final result achieved an ACC of 65.22% and AUC of 0.81.Significance: Our Siamese network based ensemble learning approach demonstrated promising ability in mining discriminative features with minimal manual processing and generalization requirement. This prediction strategy can be potentially applied to assist timely clinical decision-making.
Collapse
Affiliation(s)
- Zi Yang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mingli Chen
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mahdieh Kazemimoghadam
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lin Ma
- Department of Radiation Oncology, 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
| | - 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
| | - Tu Dan
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Weiguo Lu
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xuejun Gu
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Radiation Oncology, Stanford University, Palo Alto, CA 94305, USA
| |
Collapse
|
25
|
García-García S, García-Galindo M, Arrese I, Sarabia R, Cepeda S. Current Evidence, Limitations and Future Challenges of Survival Prediction for Glioblastoma Based on Advanced Noninvasive Methods: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58121746. [PMID: 36556948 PMCID: PMC9786785 DOI: 10.3390/medicina58121746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/16/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
Background and Objectives: Survival estimation for patients diagnosed with Glioblastoma (GBM) is an important information to consider in patient management and communication. Despite some known risk factors, survival estimation remains a major challenge. Novel non-invasive technologies such as radiomics and artificial intelligence (AI) have been implemented to increase the accuracy of these predictions. In this article, we reviewed and discussed the most significant available research on survival estimation for GBM through advanced non-invasive methods. Materials and Methods: PubMed database was queried for articles reporting on survival prognosis for GBM through advanced image and data management methods. Articles including in their title or abstract the following terms were initially screened: ((glioma) AND (survival)) AND ((artificial intelligence) OR (radiomics)). Exclusively English full-text articles, reporting on humans, published as of 1 September 2022 were considered. Articles not reporting on overall survival, evaluating the effects of new therapies or including other tumors were excluded. Research with a radiomics-based methodology were evaluated using the radiomics quality score (RQS). Results: 382 articles were identified. After applying the inclusion criteria, 46 articles remained for further analysis. These articles were thoroughly assessed, summarized and discussed. The results of the RQS revealed some of the limitations of current radiomics investigation on this field. Limitations of analyzed studies included data availability, patient selection and heterogeneity of methodologies. Future challenges on this field are increasing data availability, improving the general understanding of how AI handles data and establishing solid correlations between image features and tumor's biology. Conclusions: Radiomics and AI methods of data processing offer a new paradigm of possibilities to tackle the question of survival prognosis in GBM.
Collapse
Affiliation(s)
- Sergio García-García
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
- Correspondence:
| | - Manuel García-Galindo
- Faculty of Medicine, University of Valladolid, Avenida Ramón y Cajal 7, 47003 Valladolid, Spain
| | - Ignacio Arrese
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
| | - Rosario Sarabia
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
| | - Santiago Cepeda
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
| |
Collapse
|
26
|
Cost Matrix of Molecular Pathology in Glioma-Towards AI-Driven Rational Molecular Testing and Precision Care for the Future. Biomedicines 2022; 10:biomedicines10123029. [PMID: 36551786 PMCID: PMC9775648 DOI: 10.3390/biomedicines10123029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/09/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022] Open
Abstract
Gliomas are the most common and aggressive primary brain tumors. Gliomas carry a poor prognosis because of the tumor's resistance to radiation and chemotherapy leading to nearly universal recurrence. Recent advances in large-scale genomic research have allowed for the development of more targeted therapies to treat glioma. While precision medicine can target specific molecular features in glioma, targeted therapies are often not feasible due to the lack of actionable markers and the high cost of molecular testing. This review summarizes the clinically relevant molecular features in glioma and the current cost of care for glioma patients, focusing on the molecular markers and meaningful clinical features that are linked to clinical outcomes and have a realistic possibility of being measured, which is a promising direction for precision medicine using artificial intelligence approaches.
Collapse
|
27
|
Piao S, Luo X, Bao Y, Hu B, Liu X, Zhu Y, Yang L, Geng D, Li Y. An MRI-based joint model of radiomics and spatial distribution differentiates autoimmune encephalitis from low-grade diffuse astrocytoma. Front Neurol 2022; 13:998279. [PMID: 36408523 PMCID: PMC9669344 DOI: 10.3389/fneur.2022.998279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/12/2022] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND The differential diagnosis between autoimmune encephalitis and low-grade diffuse astrocytoma remains challenging. We aim to develop a quantitative model integrating radiomics and spatial distribution features derived from MRI for discriminating these two conditions. METHODS In our study, we included 188 patients with confirmed autoimmune encephalitis (n = 81) and WHO grade II diffuse astrocytoma (n = 107). Patients with autoimmune encephalitis (AE, n = 59) and WHO grade II diffuse astrocytoma (AS, n = 79) were divided into training and test sets, using stratified sampling according to MRI scanners. We further included an independent validation set (22 patients with AE and 28 patients with AS). Hyperintensity fluid-attenuated inversion recovery (FLAIR) lesions were segmented for each subject. Ten radiomics and eight spatial distribution features were selected via the least absolute shrinkage and selection operator (LASSO), and joint models were constructed by logistic regression for disease classification. Model performance was measured in the test set using the area under the receiver operating characteristic (ROC) curve (AUC). The discrimination performance of the joint model was compared with neuroradiologists. RESULTS The joint model achieved better performance (AUC 0.957/0.908, accuracy 0.914/0.840 for test and independent validation sets, respectively) than the radiomics and spatial distribution models. The joint model achieved lower performance than a senior neuroradiologist (AUC 0.917/0.875) but higher performance than a junior neuroradiologist (AUC 0.692/0.745) in the test and independent validation sets. CONCLUSION The joint model of radiomics and spatial distribution from a single FLAIR could effectively classify AE and AS, providing clinical decision support for the differential diagnosis between the two conditions.
Collapse
Affiliation(s)
- Sirong Piao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Xiao Luo
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Yifang Bao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Bin Hu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Xueling Liu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Yuqi Zhu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Liqin Yang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Daoying Geng
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Yuxin Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Radiomic and Volumetric Measurements as Clinical Trial Endpoints—A Comprehensive Review. Cancers (Basel) 2022; 14:cancers14205076. [PMID: 36291865 PMCID: PMC9599928 DOI: 10.3390/cancers14205076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary The extraction of quantitative data from standard-of-care imaging modalities offers opportunities to improve the relevance and salience of imaging biomarkers used in drug development. This review aims to identify the challenges and opportunities for discovering new imaging-based biomarkers based on radiomic and volumetric assessment in the single-site solid tumor sites: breast cancer, rectal cancer, lung cancer and glioblastoma. Developing approaches to harmonize three essential areas: segmentation, validation and data sharing may expedite regulatory approval and adoption of novel cancer imaging biomarkers. Abstract Clinical trials for oncology drug development have long relied on surrogate outcome biomarkers that assess changes in tumor burden to accelerate drug registration (i.e., Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1) criteria). Drug-induced reduction in tumor size represents an imperfect surrogate marker for drug activity and yet a radiologically determined objective response rate is a widely used endpoint for Phase 2 trials. With the addition of therapies targeting complex biological systems such as immune system and DNA damage repair pathways, incorporation of integrative response and outcome biomarkers may add more predictive value. We performed a review of the relevant literature in four representative tumor types (breast cancer, rectal cancer, lung cancer and glioblastoma) to assess the preparedness of volumetric and radiomics metrics as clinical trial endpoints. We identified three key areas—segmentation, validation and data sharing strategies—where concerted efforts are required to enable progress of volumetric- and radiomics-based clinical trial endpoints for wider clinical implementation.
Collapse
|
29
|
Zhao R, Zhuge Y, Camphausen K, Krauze AV. Machine learning based survival prediction in Glioma using large-scale registry data. Health Informatics J 2022; 28:14604582221135427. [PMID: 36264067 PMCID: PMC10673681 DOI: 10.1177/14604582221135427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2023]
Abstract
Gliomas are the most common central nervous system tumors exhibiting poor clinical outcomes. The ability to estimate prognosis is crucial for both patients and providers in order to select the most appropriate treatment. Machine learning (ML) allows for sophisticated approaches to survival prediction using real world clinical parameters needed to achieve superior predictive accuracy. We employed Cox Proportional hazards (CPH) model, Support Vector Machine (SVM) model, Random Forest (RF) model in a large glioma dataset (3462 patients, diagnosed 2000-2018) to explore the most optimal approach to survival prediction. Features employed were age, sex, surgical resection status, tumor histology and tumor site, administration of radiation therapy (RT) and chemotherapy status. Concordance index (c-index) was employed to assess the accuracy of survival time prediction. All three models performed well with prediction accuracy (CI 0.767, 0.771, 0.57 for CPH, SVM, RF models respectively) with the best performance achieved when incorporating RT and chemotherapy administration status which emerged as key predictive features. Within the subset of glioblastoma patients, similar prediction accuracy was achieved. These findings should prompt stricter clinician oversight over registry data accuracy through quality assurance as we move towards meaningful predictive ability using ML approaches in glioma.
Collapse
Affiliation(s)
| | | | | | - Andra V Krauze
- 3421National Cancer Institute, NIH, USA; 184934BC Cancer Surrey, Canada
| |
Collapse
|
30
|
Jing H, Yang F, Peng K, Qin D, He Y, Yang G, Zhang H. Multimodal MRI-Based Radiomic Nomogram for the Early Differentiation of Recurrence and Pseudoprogression of High-Grade Glioma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4667117. [PMID: 36246986 PMCID: PMC9553483 DOI: 10.1155/2022/4667117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/18/2022]
Abstract
Objective To evaluate the diagnostic value of multimodal MRI radiomics based on T2-weighted fluid attenuated inversion recovery imaging (T2WI-FLAIR) combined with T1-weighted contrast enhanced imaging (T1WI-CE) in the early differentiation of high-grade glioma recurrence from pseudoprogression. Methods A total of one hundred eighteen patients with brain gliomas who were diagnosed from March 2014 to April 2020 were retrospectively analyzed. According to the clinical characteristics, the patients were randomly split into a training group (n = 83) and a test group (n = 35) at a 7 : 3 ratio. The region of interest (ROI) was delineated, and 2632 radiomic features were extracted. We used multiple logistic regression to establish a classification model, including the T1 model, T2 model, and T1 + T2 model, to differentiate recurrence from pseudoprogression. The diagnostic efficiency of the model was evaluated by calculating the area under the receiver operating characteristic curve (AUC) and accuracy (ACC) and by analyzing the calibration curve of the nomogram and decision curve. Results There were 75 cases of recurrence and 43 cases of pseudoprogression. The diagnostic efficacies of the multimodal MRI-based radiomic model were relatively high. The AUC values and ACC of the training group were 0.831 and 77.11%, respectively, and the AUC values and ACC of the test group were 0.829 and 88.57%, respectively. The calibration curve of the nomogram showed that the discrimination probability was consistent with the actual occurrence in the training group, and the discrimination probability was roughly the same as the actual occurrence in the test group. In the decision curve analysis, the T1 + T2 model showed greater overall net efficiency. Conclusion The multimodal MRI radiomic model has relatively high efficiency in the early differentiation of recurrence from pseudoprogression, and it could be helpful for clinicians in devising correct treatment plans so that patients can be treated in a timely and accurate manner.
Collapse
Affiliation(s)
- Hui Jing
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Radiology, The Sixth Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Fan Yang
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Kun Peng
- Department of Radiology, The Sixth Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Danlei Qin
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yexin He
- Department of Radiology, Shanxi Provincial People's Hospital, Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
| | - Guoqiang Yang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hui Zhang
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi Province, China
- Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
31
|
di Noia C, Grist JT, Riemer F, Lyasheva M, Fabozzi M, Castelli M, Lodi R, Tonon C, Rundo L, Zaccagna F. Predicting Survival in Patients with Brain Tumors: Current State-of-the-Art of AI Methods Applied to MRI. Diagnostics (Basel) 2022; 12:diagnostics12092125. [PMID: 36140526 PMCID: PMC9497964 DOI: 10.3390/diagnostics12092125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Given growing clinical needs, in recent years Artificial Intelligence (AI) techniques have increasingly been used to define the best approaches for survival assessment and prediction in patients with brain tumors. Advances in computational resources, and the collection of (mainly) public databases, have promoted this rapid development. This narrative review of the current state-of-the-art aimed to survey current applications of AI in predicting survival in patients with brain tumors, with a focus on Magnetic Resonance Imaging (MRI). An extensive search was performed on PubMed and Google Scholar using a Boolean research query based on MeSH terms and restricting the search to the period between 2012 and 2022. Fifty studies were selected, mainly based on Machine Learning (ML), Deep Learning (DL), radiomics-based methods, and methods that exploit traditional imaging techniques for survival assessment. In addition, we focused on two distinct tasks related to survival assessment: the first on the classification of subjects into survival classes (short and long-term or eventually short, mid and long-term) to stratify patients in distinct groups. The second focused on quantification, in days or months, of the individual survival interval. Our survey showed excellent state-of-the-art methods for the first, with accuracy up to ∼98%. The latter task appears to be the most challenging, but state-of-the-art techniques showed promising results, albeit with limitations, with C-Index up to ∼0.91. In conclusion, according to the specific task, the available computational methods perform differently, and the choice of the best one to use is non-univocal and dependent on many aspects. Unequivocally, the use of features derived from quantitative imaging has been shown to be advantageous for AI applications, including survival prediction. This evidence from the literature motivates further research in the field of AI-powered methods for survival prediction in patients with brain tumors, in particular, using the wealth of information provided by quantitative MRI techniques.
Collapse
Affiliation(s)
- Christian di Noia
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
| | - James T. Grist
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Oxford Centre for Clinical Magnetic Research Imaging, University of Oxford, Oxford OX3 9DU, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK
| | - Frank Riemer
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Maria Lyasheva
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Miriana Fabozzi
- Centro Medico Polispecialistico (CMO), 80058 Torre Annunziata, Italy
| | - Mauro Castelli
- NOVA Information Management School (NOVA IMS), Universidade NOVA de Lisboa, Campus de Campolide, 1070-312 Lisboa, Portugal
| | - Raffaele Lodi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Caterina Tonon
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Leonardo Rundo
- Department of Information and Electrical Engineering and Applied Mathematics, University of Salerno, 84084 Fisciano, Italy
| | - Fulvio Zaccagna
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
- Correspondence: ; Tel.: +39-0514969951
| |
Collapse
|
32
|
Integrating Radiomics with Genomics for Non-Small Cell Lung Cancer Survival Analysis. JOURNAL OF ONCOLOGY 2022; 2022:5131170. [PMID: 36065309 PMCID: PMC9440821 DOI: 10.1155/2022/5131170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/14/2022] [Accepted: 07/11/2022] [Indexed: 11/18/2022]
Abstract
Purpose The objectives of our study were to assess the association of radiological imaging and gene expression with patient outcomes in non-small cell lung cancer (NSCLC) and construct a nomogram by combining selected radiomic, genomic, and clinical risk factors to improve the performance of the risk model. Methods A total of 116 cases of NSCLC with CT images, gene expression, and clinical factors were studied, wherein 87 patients were used as the training cohort, and 29 patients were used as an independent testing cohort. Handcrafted radiomic features and deep-learning genomic features were extracted and selected from CT images and gene expression analysis, respectively. Two risk scores were calculated through Cox regression models for each patient based on radiomic features and genomic features to predict overall survival (OS). Finally, a fusion survival model was constructed by incorporating these two risk scores and clinical factors. Results The fusion model that combined CT images, gene expression data, and clinical factors effectively stratified patients into low- and high-risk groups. The C-indexes for OS prediction were 0.85 and 0.736 in the training and testing cohorts, respectively, which was better than that based on unimodal data. Conclusions Combining radiomics and genomics can effectively improve OS prediction for NSCLC patients.
Collapse
|
33
|
Qin D, Yang G, Jing H, Tan Y, Zhao B, Zhang H. Tumor Progression and Treatment-Related Changes: Radiological Diagnosis Challenges for the Evaluation of Post Treated Glioma. Cancers (Basel) 2022; 14:cancers14153771. [PMID: 35954435 PMCID: PMC9367286 DOI: 10.3390/cancers14153771] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Glioma is the most common primary malignant tumor of the adult central nervous system. Despite aggressive multimodal treatment, its prognosis remains poor. During follow-up, it remains challenging to distinguish treatment-related changes from tumor progression in treated patients with gliomas due to both share clinical symptoms and morphological imaging characteristics (with new and/or increasing enhancing mass lesions). The early effective identification of tumor progression and treatment-related changes is of great significance for the prognosis and treatment of gliomas. We believe that advanced neuroimaging techniques can provide additional information for distinguishing both at an early stage. In this article, we focus on the research of magnetic resonance imaging technology and artificial intelligence in tumor progression and treatment-related changes. Finally, it provides new ideas and insights for clinical diagnosis. Abstract As the most common neuro-epithelial tumors of the central nervous system in adults, gliomas are highly malignant and easy to recurrence, with a dismal prognosis. Imaging studies are indispensable for tracking tumor progression (TP) or treatment-related changes (TRCs). During follow-up, distinguishing TRCs from TP in treated patients with gliomas remains challenging as both share similar clinical symptoms and morphological imaging characteristics (with new and/or increasing enhancing mass lesions) and fulfill criteria for progression. Thus, the early identification of TP and TRCs is of great significance for determining the prognosis and treatment. Histopathological biopsy is currently the gold standard for TP and TRC diagnosis. However, the invasive nature of this technique limits its clinical application. Advanced imaging methods (e.g., diffusion magnetic resonance imaging (MRI), perfusion MRI, magnetic resonance spectroscopy (MRS), positron emission tomography (PET), amide proton transfer (APT) and artificial intelligence (AI)) provide a non-invasive and feasible technical means for identifying of TP and TRCs at an early stage, which have recently become research hotspots. This paper reviews the current research on using the abovementioned advanced imaging methods to identify TP and TRCs of gliomas. First, the review focuses on the pathological changes of the two entities to establish a theoretical basis for imaging identification. Then, it elaborates on the application of different imaging techniques and AI in identifying the two entities. Finally, the current challenges and future prospects of these techniques and methods are discussed.
Collapse
Affiliation(s)
- Danlei Qin
- College of Medical Imaging, Shanxi Medical University, Taiyuan 030001, China;
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School, Hospital of Stomatology, Taiyuan 030001, China
| | - Guoqiang Yang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan 030001, China; (G.Y.); (Y.T.)
| | - Hui Jing
- Department of MRI, The Six Hospital, Shanxi Medical University, Taiyuan 030008, China;
| | - Yan Tan
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan 030001, China; (G.Y.); (Y.T.)
| | - Bin Zhao
- College of Medical Imaging, Shanxi Medical University, Taiyuan 030001, China;
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School, Hospital of Stomatology, Taiyuan 030001, China
- Correspondence: (B.Z.); (H.Z.)
| | - Hui Zhang
- College of Medical Imaging, Shanxi Medical University, Taiyuan 030001, China;
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan 030001, China; (G.Y.); (Y.T.)
- Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, Taiyuan 030001, China
- Correspondence: (B.Z.); (H.Z.)
| |
Collapse
|
34
|
Liu D, Chen J, Ge H, Hu X, Yang K, Liu Y, Hu G, Luo B, Yan Z, Song K, Xiao C, Zou Y, Zhang W, Liu H. Differentiation of malignant brain tumor types using intratumoral and peritumoral radiomic features. Front Oncol 2022; 12:848846. [PMID: 35965511 PMCID: PMC9366472 DOI: 10.3389/fonc.2022.848846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/28/2022] [Indexed: 12/14/2022] Open
Abstract
Tumor infiltration of central nervous system (CNS) malignant tumors may extend beyond visible contrast enhancement. This study explored tumor habitat characteristics in the intratumoral and peritumoral regions to distinguish common malignant brain tumors such as glioblastoma, primary central nervous system lymphoma, and brain metastases. The preoperative MRI data of 200 patients with solitary malignant brain tumors were included from two datasets for training. Quantitative radiomic features from the intratumoral and peritumoral regions were extracted for model training. The performance of the model was evaluated using data (n = 50) from the third clinical center. When combining the intratumoral and peritumoral features, the Adaboost model achieved the best area under the curve (AUC) of 0.91 and accuracy of 76.9% in the test cohort. Based on the optimal features and classifier, the model in the binary classification diagnosis achieves AUC of 0.98 (glioblastoma and lymphoma), 0.86 (lymphoma and metastases), and 0.70 (glioblastoma and metastases) in the test cohort, respectively. In conclusion, quantitative features from non-enhanced peritumoral regions (especially features from the 10-mm margin around the tumor) can provide additional information for the characterization of regional tumoral heterogeneity, which may offer potential value for future individualized assessment of patients with CNS tumors.
Collapse
Affiliation(s)
- Dongming Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jiu Chen
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Institute of Brain Sciences, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Honglin Ge
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Xinhua Hu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Institute of Brain Sciences, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Kun Yang
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Guanjie Hu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Bei Luo
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Zhen Yan
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Kun Song
- Department of Pathology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Chaoyong Xiao
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yuanjie Zou
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wenbin Zhang
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Institute of Brain Sciences, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Wenbin Zhang, ; Hongyi Liu,
| | - Hongyi Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Institute of Brain Sciences, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Wenbin Zhang, ; Hongyi Liu,
| |
Collapse
|
35
|
Mirón Mombiela R, Arildskov AR, Bruun FJ, Hasselbalch LH, Holst KB, Rasmussen SH, Borrás C. What Genetics Can Do for Oncological Imaging: A Systematic Review of the Genetic Validation Data Used in Radiomics Studies. Int J Mol Sci 2022; 23:6504. [PMID: 35742947 PMCID: PMC9224495 DOI: 10.3390/ijms23126504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 02/04/2023] Open
Abstract
(1) Background: Radiogenomics is motivated by the concept that biomedical images contain information that reflects underlying pathophysiology. This review focused on papers that used genetics to validate their radiomics models and outcomes and assess their contribution to this emerging field. (2) Methods: All original research with the words radiomics and genomics in English and performed in humans up to 31 January 2022, were identified on Medline and Embase. The quality of the studies was assessed with Radiomic Quality Score (RQS) and the Cochrane recommendation for diagnostic accuracy study Quality Assessment 2. (3) Results: 45 studies were included in our systematic review, and more than 50% were published in the last two years. The studies had a mean RQS of 12, and the studied tumors were very diverse. Up to 83% investigated the prognosis as the main outcome, with the rest focusing on response to treatment and risk assessment. Most applied either transcriptomics (54%) and/or genetics (35%) for genetic validation. (4) Conclusions: There is enough evidence to state that new science has emerged, focusing on establishing an association between radiological features and genomic/molecular expression to explain underlying disease mechanisms and enhance prognostic, risk assessment, and treatment response radiomics models in cancer patients.
Collapse
Affiliation(s)
- Rebeca Mirón Mombiela
- Radiology Derpartment, Herlev og Gentofte Hospital, Borgmester Ib Juuls Vej 17, Opgang 4, 4.Etage, E2, 2730 Herlev, Denmark; (A.R.A.); (F.J.B.); (L.H.H.); (K.B.H.); (S.H.R.)
| | - Anne Rix Arildskov
- Radiology Derpartment, Herlev og Gentofte Hospital, Borgmester Ib Juuls Vej 17, Opgang 4, 4.Etage, E2, 2730 Herlev, Denmark; (A.R.A.); (F.J.B.); (L.H.H.); (K.B.H.); (S.H.R.)
| | - Frederik Jager Bruun
- Radiology Derpartment, Herlev og Gentofte Hospital, Borgmester Ib Juuls Vej 17, Opgang 4, 4.Etage, E2, 2730 Herlev, Denmark; (A.R.A.); (F.J.B.); (L.H.H.); (K.B.H.); (S.H.R.)
| | - Lotte Harries Hasselbalch
- Radiology Derpartment, Herlev og Gentofte Hospital, Borgmester Ib Juuls Vej 17, Opgang 4, 4.Etage, E2, 2730 Herlev, Denmark; (A.R.A.); (F.J.B.); (L.H.H.); (K.B.H.); (S.H.R.)
| | - Kristine Bærentz Holst
- Radiology Derpartment, Herlev og Gentofte Hospital, Borgmester Ib Juuls Vej 17, Opgang 4, 4.Etage, E2, 2730 Herlev, Denmark; (A.R.A.); (F.J.B.); (L.H.H.); (K.B.H.); (S.H.R.)
| | - Sine Hvid Rasmussen
- Radiology Derpartment, Herlev og Gentofte Hospital, Borgmester Ib Juuls Vej 17, Opgang 4, 4.Etage, E2, 2730 Herlev, Denmark; (A.R.A.); (F.J.B.); (L.H.H.); (K.B.H.); (S.H.R.)
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBERFES, Institute of Health Research-INCLIVA, 46010 Valencia, Spain
| |
Collapse
|
36
|
Qing Z, Xiaoai K, Caiqiang X, Shenglin L, Xiaoyu H, Bin Z, Junlin Z. Nomogram for predicting early recurrence in patients with high-grade gliomas. World Neurosurg 2022; 164:e619-e628. [PMID: 35589036 DOI: 10.1016/j.wneu.2022.05.039] [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: 02/19/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To develop a nomogram to predict early recurrence of high-grade glioma (HGG) based on clinical pathology, genetic factors and MRI parameters. METHODS 154 patients with HGG were classified into recurrence and non-recurrence groups based on the pathological diagnosis and RANO criteria. Clinical pathology information included age, sex, preoperative Karnofsky performance status (KPS) scores,grade, and cell proliferation index (Ki-67). Gene information included P53, IDH1, MGMT, and TERT expression status. All patients underwent baseline MRIs before treatment, including T1WI, T2WI, T1C, Flair, and DWI examinations. Tumor location, single/multiple tumors, tumor diameter, peritumoral edema, necrotic cyst, hemorrhage, average apparent diffusion coefficient(ADC) value, and minimum ADC values were evaluated. Univariate and multivariate logistic regression analyses were used to determine the predictors of early recurrence and build nomogram. RESULTS Univariate analysis showed that the number of tumors (OR, 0.258; 95% CI: 0.104, 0.639; P = 0.003) and peritumoral edema (OR, 0.965; 95% CI 0.942, 0.988; P = 0.003; mean in the recurrence group 22.04±17.21 mm; mean in the non-recurrence group 14.22±12.84 mm) were statistically significantly different in patients with early recurrence. Genetic factors associated with early recurrence included IDH1 (OR, 4.405; 95% CI 1.874, 10.353; P= 0.001), and MGMT (OR, 2.389; 95% CI 1.234, 4.628; P= 0.010). Multivariate logistic regression analysis revealed that the number of tumors (OR, 0.227; 95% CI 0.084, 0.616; P = 0.004), peritumoral edema (OR, 0.969; 95% CI 0.945, 0.993; P = 0.013), and IDH1 (OR, 4.200; 95% CI 1.602, 10.013; P= 0.004) were independent risk factors for early recurrence. The nomogram showed the highest net benefit when the threshold probability was less than 60%. CONCLUSION A nomogram prediction model can effectively aid in clinical treatment decisions for patients with newly diagnosed HGG .
Collapse
Affiliation(s)
- Zhou Qing
- Department of Radiology, Lanzhou University Second Hospital, Gansu, China; Second Clinical School,Lanzhou University, China; Key Laboratory of Medical Imaging of Gansu Province, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence,China
| | - Ke Xiaoai
- Department of Radiology, Lanzhou University Second Hospital, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence,China
| | - Xue Caiqiang
- Department of Radiology, Lanzhou University Second Hospital, Gansu, China; Second Clinical School,Lanzhou University, China; Key Laboratory of Medical Imaging of Gansu Province, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence,China
| | - Li Shenglin
- Department of Radiology, Lanzhou University Second Hospital, Gansu, China; Second Clinical School,Lanzhou University, China; Key Laboratory of Medical Imaging of Gansu Province, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence,China
| | - Huang Xiaoyu
- Department of Radiology, Lanzhou University Second Hospital, Gansu, China; Second Clinical School,Lanzhou University, China; Key Laboratory of Medical Imaging of Gansu Province, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence,China
| | - Zhang Bin
- Department of Radiology, Lanzhou University Second Hospital, Gansu, China; Second Clinical School,Lanzhou University, China; Key Laboratory of Medical Imaging of Gansu Province, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence,China
| | - Zhou Junlin
- Department of Radiology, Lanzhou University Second Hospital, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence,China.
| |
Collapse
|
37
|
Pseudoprogression prediction in high grade primary CNS tumors by use of radiomics. Sci Rep 2022; 12:5915. [PMID: 35396525 PMCID: PMC8993885 DOI: 10.1038/s41598-022-09945-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/24/2022] [Indexed: 11/09/2022] Open
Abstract
Our aim is to define the capabilities of radiomics and machine learning in predicting pseudoprogression development from pre-treatment MR images in a patient cohort diagnosed with high grade gliomas. In this retrospective analysis, we analysed 131 patients with high grade gliomas. Segmentation of the contrast enhancing parts of the tumor before administration of radio-chemotherapy was semi-automatically performed using the 3D Slicer open-source software platform (version 4.10) on T1 post contrast MR images. Imaging data was split into training data, test data and an independent validation sample at random. We extracted a total of 107 radiomic features by hand-delineated regions of interest (ROI). Feature selection and model construction were performed using Generalized Boosted Regression Models (GBM). 131 patients were included, of which 64 patients had a histopathologically proven progressive disease and 67 were diagnosed with mixed or pure pseudoprogression after initial treatment. Our Radiomics approach is able to predict the occurrence of pseudoprogression with an AUC, mean sensitivity, mean specificity and mean accuracy of 91.49% [86.27%, 95.89%], 79.92% [73.08%, 87.55%], 88.61% [85.19%, 94.44%] and 84.35% [80.19%, 90.57%] in the full development group, 78.51% [75.27%, 82.46%], 66.26% [57.95%, 73.02%], 78.31% [70.48%, 84.19%] and 72.40% [68.06%, 76.85%] in the testing group and finally 72.87% [70.18%, 76.28%], 71.75% [62.29%, 75.00%], 80.00% [69.23%, 84.62%] and 76.04% [69.90%, 80.00%] in the independent validation sample, respectively. Our results indicate that radiomics is a promising tool to predict pseudo-progression, thus potentially allowing to reduce the use of biopsies and invasive histopathology.
Collapse
|
38
|
Zhou Q, Xue C, Ke X, Zhou J. Treatment Response and Prognosis Evaluation in High-Grade Glioma: An Imaging Review Based on MRI. J Magn Reson Imaging 2022; 56:325-340. [PMID: 35129845 DOI: 10.1002/jmri.28103] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/19/2022] Open
Abstract
In recent years, the development of advanced magnetic resonance imaging (MRI) technology and machine learning (ML) have created new tools for evaluating treatment response and prognosis of patients with high-grade gliomas (HGG); however, patient prognosis has not improved significantly. This is mainly due to the heterogeneity between and within HGG tumors, resulting in standard treatment methods not benefitting all patients. Moreover, the survival of patients with HGG is not only related to tumor cells, but also to noncancer cells in the tumor microenvironment (TME). Therefore, during preoperative diagnosis and follow-up treatment of patients with HGG, noninvasive imaging markers are needed to characterize intratumoral heterogeneity, and then to evaluate treatment response and predict prognosis, timeously adjust treatment strategies, and achieve individualized diagnosis and treatment. In this review, we summarize the research progress of conventional MRI, advanced MRI technology, and ML in evaluation of treatment response and prognosis of patients with HGG. We further discuss the significance of the TME in the prognosis of HGG patients, associate imaging features with the TME, indirectly reflecting the heterogeneity within the tumor, and shifting treatment strategies from tumor cells alone to systemic therapy of the TME, which may be a major development direction in the future. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY STAGE: 4.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| |
Collapse
|
39
|
Wang S, Xiao F, Sun W, Yang C, Ma C, Huang Y, Xu D, Li L, Chen J, Li H, Xu H. Radiomics Analysis Based on Magnetic Resonance Imaging for Preoperative Overall Survival Prediction in Isocitrate Dehydrogenase Wild-Type Glioblastoma. Front Neurosci 2022; 15:791776. [PMID: 35153659 PMCID: PMC8833841 DOI: 10.3389/fnins.2021.791776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/15/2021] [Indexed: 01/24/2023] Open
Abstract
PurposeThis study aimed to develop a radiomics signature for the preoperative prognosis prediction of isocitrate dehydrogenase (IDH)-wild-type glioblastoma (GBM) patients and to provide personalized assistance in the clinical decision-making for different patients.Materials and MethodsA total of 142 IDH-wild-type GBM patients classified using the new classification criteria of WHO 2021 from two centers were included in the study and randomly divided into a training set and a test set. Firstly, their clinical characteristics were screened using univariate Cox regression. Then, the radiomics features were extracted from the tumor and peritumoral edema areas on their contrast-enhanced T1-weighted image (CE-T1WI), T2-weighted image (T2WI), and T2-weighted fluid-attenuated inversion recovery (T2-FLAIR) magnetic resonance imaging (MRI) images. Subsequently, inter- and intra-class correlation coefficient (ICC) analysis, Spearman’s correlation analysis, univariate Cox, and the least absolute shrinkage and selection operator (LASSO) Cox regression were used step by step for feature selection and the construction of a radiomics signature. The combined model was established by integrating the selected clinical factors. Kaplan–Meier analysis was performed for the validation of the discrimination ability of the model, and the C-index was used to evaluate consistency in the prediction. Finally, a Radiomics + Clinical nomogram was generated for personalized prognosis analysis and then validated using the calibration curve.ResultsAnalysis of the clinical characteristics resulted in the screening of four risk factors. The combination of ICC, Spearman’s correlation, and univariate and LASSO Cox resulted in the selection of eight radiomics features, which made up the radiomics signature. Both the radiomics and combined models can significantly stratify high- and low-risk patients (p < 0.001 and p < 0.05 for the training and test sets, respectively) and obtained good prediction consistency (C-index = 0.74–0.86). The calibration plots exhibited good agreement in both 1- and 2-year survival between the prediction of the model and the actual observation.ConclusionRadiomics is an independent preoperative non-invasive prognostic tool for patients who were newly classified as having IDH-wild-type GBM. The constructed nomogram, which combined radiomics features with clinical factors, can predict the overall survival (OS) of IDH-wild-type GBM patients and could be a new supplement to treatment guidelines.
Collapse
Affiliation(s)
- Shouchao Wang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Feng Xiao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenbo Sun
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chao Yang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chao Ma
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yong Huang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dan Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lanqing Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Chen
- Precision Health Institute, GE Healthcare, Shanghai, China
| | - Huan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Huan Li,
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Haibo Xu,
| |
Collapse
|
40
|
Rapalino O. Neuro-Oncology: Imaging Diagnosis. HYBRID PET/MR NEUROIMAGING 2022:527-537. [DOI: 10.1007/978-3-030-82367-2_46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
41
|
Bracci S, Dolciami M, Trobiani C, Izzo A, Pernazza A, D'Amati G, Manganaro L, Ricci P. Quantitative CT texture analysis in predicting PD-L1 expression in locally advanced or metastatic NSCLC patients. Radiol Med 2021; 126:1425-1433. [PMID: 34373989 PMCID: PMC8558266 DOI: 10.1007/s11547-021-01399-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 07/06/2021] [Indexed: 12/18/2022]
Abstract
Purpose The assessment of Programmed death-ligand 1 (PD-L1) expression has become a game changer in the treatment of patients with advanced non-small cell lung cancer (NSCLC). We aimed to investigate the ability of Radiomics applied to computed tomography (CT) in predicting PD-L1 expression in patients with advanced NSCLC. Methods By applying texture analysis, we retrospectively analyzed 72 patients with advanced NSCLC. The datasets were randomly split into a training cohort (2/3) and a validation cohort (1/3). Forty radiomic features were extracted by manually drawing tumor volumes of interest (VOIs) on baseline contrast-enhanced CT. After selecting features on the training cohort, two predictive models were created using binary logistic regression, one for PD-L1 values ≥ 50% and the other for values between 1 and 49%. The two models were analyzed with ROC curves and tested in the validation cohort. Results The Radiomic Score (Rad-Score) for PD-L1 values ≥ 50%, which consisted of Skewness and Low Gray-Level Zone Emphasis (GLZLM_LGZE), presented a cut-off value of − 0.745 with an area under the curve (AUC) of 0.811 and 0.789 in the training and validation cohort, respectively. The Rad-Score for PD-L1 values between 1 and 49% consisted of Sphericity, Skewness, Conv_Q3 and Gray Level Non-Uniformity (GLZLM_GLNU), showing a cut-off value of 0.111 with AUC of 0.763 and 0.806 in the two population, respectively. Conclusion Rad-Scores obtained from CT texture analysis could be useful for predicting PD-L1 expression and guiding the therapeutic choice in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Stefano Bracci
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Miriam Dolciami
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Claudio Trobiani
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Antonella Izzo
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Angelina Pernazza
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Giulia D'Amati
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Lucia Manganaro
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Paolo Ricci
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
42
|
Yan J, Zhang B, Zhang S, Cheng J, Liu X, Wang W, Dong Y, Zhang L, Mo X, Chen Q, Fang J, Wang F, Tian J, Zhang S, Zhang Z. Quantitative MRI-based radiomics for noninvasively predicting molecular subtypes and survival in glioma patients. NPJ Precis Oncol 2021; 5:72. [PMID: 34312469 PMCID: PMC8313682 DOI: 10.1038/s41698-021-00205-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 05/13/2021] [Indexed: 12/24/2022] Open
Abstract
Gliomas can be classified into five molecular groups based on the status of IDH mutation, 1p/19q codeletion, and TERT promoter mutation, whereas they need to be obtained by biopsy or surgery. Thus, we aimed to use MRI-based radiomics to noninvasively predict the molecular groups and assess their prognostic value. We retrospectively identified 357 patients with gliomas and extracted radiomic features from their preoperative MRI images. Single-layered radiomic signatures were generated using a single MR sequence using Bayesian-regularization neural networks. Image fusion models were built by combing the significant radiomic signatures. By separately predicting the molecular markers, the predictive molecular groups were obtained. Prognostic nomograms were developed based on the predictive molecular groups and clinicopathologic data to predict progression-free survival (PFS) and overall survival (OS). The results showed that the image fusion model incorporating radiomic signatures from contrast-enhanced T1-weighted imaging (cT1WI) and apparent diffusion coefficient (ADC) achieved an AUC of 0.884 and 0.669 for predicting IDH and TERT status, respectively. cT1WI-based radiomic signature alone yielded favorable performance in predicting 1p/19q status (AUC = 0.815). The predictive molecular groups were comparable to actual ones in predicting PFS (C-index: 0.709 vs. 0.722, P = 0.241) and OS (C-index: 0.703 vs. 0.751, P = 0.359). Subgroup analyses by grades showed similar findings. The prognostic nomograms based on grades and the predictive molecular groups yielded a C-index of 0.736 and 0.735 in predicting PFS and OS, respectively. Accordingly, MRI-based radiomics may be useful for noninvasively detecting molecular groups and predicting survival in gliomas regardless of grades.
Collapse
Affiliation(s)
- Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bin Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shuaitong Zhang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China.,CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,School of Engineering Medicine, Beihang University, Beijing, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuhao Dong
- Department of Catheterization Lab, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Lu Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiaokai Mo
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Qiuying Chen
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jin Fang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Fei Wang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jie Tian
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China. .,CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China. .,School of Engineering Medicine, Beihang University, Beijing, China. .,Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shanxi, China.
| | - Shuixing Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
43
|
Pak E, Choi KS, Choi SH, Park CK, Kim TM, Park SH, Lee JH, Lee ST, Hwang I, Yoo RE, Kang KM, Yun TJ, Kim JH, Sohn CH. Prediction of Prognosis in Glioblastoma Using Radiomics Features of Dynamic Contrast-Enhanced MRI. Korean J Radiol 2021; 22:1514-1524. [PMID: 34269536 PMCID: PMC8390822 DOI: 10.3348/kjr.2020.1433] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/22/2021] [Accepted: 04/07/2021] [Indexed: 01/08/2023] Open
Abstract
Objective To develop a radiomics risk score based on dynamic contrast-enhanced (DCE) MRI for prognosis prediction in patients with glioblastoma. Materials and Methods One hundred and fifty patients (92 male [61.3%]; mean age ± standard deviation, 60.5 ± 13.5 years) with glioblastoma who underwent preoperative MRI were enrolled in the study. Six hundred and forty-two radiomic features were extracted from volume transfer constant (Ktrans), fractional volume of vascular plasma space (Vp), and fractional volume of extravascular extracellular space (Ve) maps of DCE MRI, wherein the regions of interest were based on both T1-weighted contrast-enhancing areas and non-enhancing T2 hyperintense areas. Using feature selection algorithms, salient radiomic features were selected from the 642 features. Next, a radiomics risk score was developed using a weighted combination of the selected features in the discovery set (n = 105); the risk score was validated in the validation set (n = 45) by investigating the difference in prognosis between the “radiomics risk score” groups. Finally, multivariable Cox regression analysis for progression-free survival was performed using the radiomics risk score and clinical variables as covariates. Results 16 radiomic features obtained from non-enhancing T2 hyperintense areas were selected among the 642 features identified. The radiomics risk score was used to stratify high- and low-risk groups in both the discovery and validation sets (both p < 0.001 by the log-rank test). The radiomics risk score and presence of isocitrate dehydrogenase (IDH) mutation showed independent associations with progression-free survival in opposite directions (hazard ratio, 3.56; p = 0.004 and hazard ratio, 0.34; p = 0.022, respectively). Conclusion We developed and validated the “radiomics risk score” from the features of DCE MRI based on non-enhancing T2 hyperintense areas for risk stratification of patients with glioblastoma. It was associated with progression-free survival independently of IDH mutation status.
Collapse
Affiliation(s)
- Elena Pak
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Kyu Sung Choi
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Center for Nanoparticle Research, Institute for Basic Science, and School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea.
| | - Chul-Kee Park
- Department of Neurosurgery and Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Tae Min Kim
- Department of Internal Medicine, Cancer Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Joo Ho Lee
- Department of Radiation Oncology, Cancer Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Soon-Tae Lee
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - Inpyeong Hwang
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Roh-Eul Yoo
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Tae Jin Yun
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Ji-Hoon Kim
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
44
|
Chen H, Li C, Zheng L, Lu W, Li Y, Wei Q. A machine learning-based survival prediction model of high grade glioma by integration of clinical and dose-volume histogram parameters. Cancer Med 2021; 10:2774-2786. [PMID: 33760360 PMCID: PMC8026951 DOI: 10.1002/cam4.3838] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/02/2020] [Accepted: 02/23/2021] [Indexed: 01/03/2023] Open
Abstract
PURPOSE Glioma is the most common type of primary brain tumor in adults, and it causes significant morbidity and mortality, especially in high-grade glioma (HGG) patients. The accurate prognostic prediction of HGG is vital and helpful for clinicians when developing therapeutic strategies. Therefore, we propose a machine learning-based survival prediction model by analyzing clinical and dose-volume histogram (DVH) parameters, to improve the performance of the risk model in HGG patients. METHODS Eight clinical variables and 39 DVH parameters were extracted for each patient, who received radiotherapy for HGG with active follow-up. Ninety-five patients were randomly divided into training and testing cohorts, and we employed random survival forest (RSF), support vector machine (SVM), and Cox proportional hazards (CPHs) models to predict survival. Calibration plots, concordance indexes, and decision curve analyses were used to evaluate the calibration, discrimination, and clinical utility of these three models. RESULTS The RSF model showed the best performance among the three models, with concordance indexes of 0.824 and 0.847 in the training and testing sets, respectively, followed by the SVM (0.792/0.823) and CPH (0.821/0.811) models. Specifically, in the RSF model, we identified age, gross tumor volume (GTV), grade, Karnofsky performance status (KPS), isocitrate dehydrogenase (IDH), and D99 as important variables associated with survival. The AUCs of the testing set were 92.4%, 87.7%, and 84.0% for 1-, 2-, and 3-year survival, respectively. According to this model, HGG patients can be divided into high- and low-risk groups. CONCLUSION The machine learning-based RSF model integrating both clinical and DVH variables is an improved and useful tool for predicting the survival of HGG patients.
Collapse
Affiliation(s)
- Haiyan Chen
- Department of Radiation OncologyKey Laboratory of Cancer Prevention and InterventionMinistry of EducationThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang University Cancer CenterHangzhouZhejiangChina
| | - Chao Li
- Department of Radiation OncologyKey Laboratory of Cancer Prevention and InterventionMinistry of EducationThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Lin Zheng
- Department of Radiation OncologyKey Laboratory of Cancer Prevention and InterventionMinistry of EducationThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Department of Radiation OncologyTaizhou Tumor HospitalTaizhouZhejiangChina
| | - Wei Lu
- Zhejiang University Cancer CenterHangzhouZhejiangChina
- Department of Colorectal Surgery and OncologyKey Laboratory of Cancer Prevention and InterventionMinistry of EducationThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Yanlin Li
- College of ScienceHangzhou Normal UniversityHangzhouZhejiangChina
| | - Qichun Wei
- Department of Radiation OncologyKey Laboratory of Cancer Prevention and InterventionMinistry of EducationThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang University Cancer CenterHangzhouZhejiangChina
| |
Collapse
|
45
|
He HL, Wang Q, Liu L, Luo NB, Su DK, Jin GQ. Peritumoral edema in preoperative magnetic resonance imaging is an independent prognostic factor for hepatocellular carcinoma. Clin Imaging 2021; 75:143-149. [PMID: 33556644 DOI: 10.1016/j.clinimag.2021.01.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 01/02/2021] [Accepted: 01/17/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Peritumoral edema is an independent prognostic risk factor for malignant tumors. Therefore, assessment of peritumoral edema in preoperative magnetic resonance imaging (MRI) may provide better prognostic information in patients with hepatocellular carcinoma (HCC). AIM To determine whether peritumoral edema in preoperative MRI is a prognostic factor for HCC. METHODS A retrospective analysis of 90 patients with HCC confirmed by surgical pathology was performed. All patients' peritumoral edema in preoperative MRI was reviewed by two radiologists. The association of disease recurrence with peritumoral edema and clinicopathological features was assessed using the Cox proportional hazards model. Interobserver agreement for evaluating peritumoral edema was determined using Cohen's κ coefficient. RESULTS Recurrence and non-recurrence after an average 20.8 month follow-up was 25.6% (23/90) and 74.4% (67/90), respectively. The ratio of peritumoral edema of 90 patients with HCC in preoperative MRI was 35.6% (32/90). In univariate Cox regression analysis, peritumoral edema [hazard ratio (HR) 11.08, P < 0.001], tumor diameter (HR 4.12, P = 0.001), microvascular invasion (HR 2.78, P = 0.020), gender (HR 0.29, P = 0.006), cirrhosis (HR 2.45, P = 0.049), ascites syndrome (HR 2.83, P = 0.022), aspartate aminotransferase(AST)/alanine aminotransferase(ALT) (HR 5.07, P = 0.003) were indicators for HCC recurrence. In multivariate Cox regression analysis, the tumor diameter (HR 2.53, P = 0.032) and peritumoral edema (HR 8.71, P < 0.001) were independent prognostic factors of HCC. The sensitivity, specificity, positive predictive value and negative predictive value of peritumoral edema and tumor diameter were 82.6%&60.9%, 80.6%&77.6%, 59.4%&48.3%, and 93.1%&85.3%, respectively. CONCLUSION Peritumoral edema in preoperative MRI may be considered as a biomarker of prognostic information for patients with HCC.
Collapse
Affiliation(s)
- Hai-Lu He
- Department of Radiology, Tumor Hospital, Guangxi Medical University, No. 71 Hedi Road, Nanning 530021, Guangxi Zhuang Autonomous Region, China.
| | - Qiang Wang
- Department of Anesthesia, Tumor Hospital, Guangxi Medical University, No. 71 Hedi Road, Nanning 530021, Guangxi Zhuang Autonomous Region, China.
| | - Lu Liu
- Department of Radiology, Tumor Hospital, Guangxi Medical University, No. 71 Hedi Road, Nanning 530021, Guangxi Zhuang Autonomous Region, China.
| | - Ning-Bin Luo
- Department of Radiology, Tumor Hospital, Guangxi Medical University, No. 71 Hedi Road, Nanning 530021, Guangxi Zhuang Autonomous Region, China.
| | - Dan-Ke Su
- Department of Radiology, Tumor Hospital, Guangxi Medical University, No. 71 Hedi Road, Nanning 530021, Guangxi Zhuang Autonomous Region, China.
| | - Guan-Qiao Jin
- Department of Radiology, Tumor Hospital, Guangxi Medical University, No. 71 Hedi Road, Nanning 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
46
|
Favorable role of IDH1/2 mutations aided with MGMT promoter gene methylation in the outcome of patients with malignant glioma. Future Sci OA 2020; 7:FSO663. [PMID: 33552543 PMCID: PMC7849969 DOI: 10.2144/fsoa-2020-0057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim The implications of molecular biomarkers IDH1/2 mutations and MGMT gene promoter methylation were evaluated for prognostic outcome of glioma patients. Materials & methods Glioma cases were analyzed for IDH1/2 mutations and MGMT promoter methylation by DNA sequencing and methylation-specific PCR, respectively. Results Mutations found in IDH1/2 genes totaled 63.4% (N = 40) wherein IDH1 mutations were significantly associated with oligidendrioglioma (p = 0.005) and astrocytoma (p = 0.0002). IDH1 mutants presented more, 60.5% in MGMT promoter-methylated cases (p = 0.03). IDH1 mutant cases had better survival for glioblastoma and oligodendrioglioma (log-rank p = 0.01). Multivariate analysis confirmed better survival in MGMT methylation carriers (hazard ratio [HR]: 0.59; p = 0.031). Combination of both biomarkers showed better prognosis on temozolomide (p < 0.05). Conclusion IDH1/2 mutations proved independent prognostic factors in glioma and associated with MGMT methylation for better survival.
Collapse
|
47
|
Qiu X, Gao J, Yang J, Hu J, Hu W, Kong L, Lu JJ. A Comparison Study of Machine Learning (Random Survival Forest) and Classic Statistic (Cox Proportional Hazards) for Predicting Progression in High-Grade Glioma after Proton and Carbon Ion Radiotherapy. Front Oncol 2020; 10:551420. [PMID: 33194609 PMCID: PMC7662123 DOI: 10.3389/fonc.2020.551420] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 09/29/2020] [Indexed: 12/30/2022] Open
Abstract
Background Machine learning (ML) algorithms are increasingly explored in glioma prognostication. Random survival forest (RSF) is a common ML approach in analyzing time-to-event survival data. However, it is controversial which method between RSF and traditional cornerstone method Cox proportional hazards (CPH) is better fitted. The purpose of this study was to compare RSF and CPH in predicting tumor progression of high-grade glioma (HGG) after particle beam radiotherapy (PBRT). Methods The study enrolled 82 consecutive HGG patients who were treated with PBRT at Shanghai Proton and Heavy Ion Center between 6/2015 and 11/2019. The entire cohort was split into the training and testing set in an 80/20 ratio. Ten variables from patient-related, tumor-related and treatment-related information were utilized for developing CPH and RSF for predicting progression-free survival (PFS). The model performance was compared in concordance index (C-index) for discrimination (accuracy), brier score (BS) for calibration (precision) and variable importance for interpretability. Results The CPH model demonstrated a better performance in terms of integrated C-index (62.9%) and BS (0.159) compared to RSF model (C-index = 61.1%, BS = 0.174). In the context of variable importance, CPH model indicated that age (P = 0.024), WHO grade (P = 0.020), IDH gene (P = 0.019), and MGMT promoter status (P = 0.040) were significantly correlated with PFS in the univariate analysis; multivariate analysis showed that age (P = 0.041), surgical completeness (P = 0.084), IDH gene (P = 0.057), and MGMT promoter (P = 0.092) had a significant or trend toward the relation with PFS. RSF showed that merely IDH and age were of positive importance for predicting PFS. A final nomogram was developed to predict tumor progression at the individual level based on CPH model. Conclusions In a relatively small dataset with HGG patients treated with PBRT, CPH outperformed RSF for predicting tumor progression. A comprehensive criterion with accuracy, precision, and interpretability is recommended in evaluating ML prognostication approaches for clinical deployment.
Collapse
Affiliation(s)
- Xianxin Qiu
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Jing Gao
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Jing Yang
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Jiyi Hu
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Weixu Hu
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Lin Kong
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Center, Shanghai, China
| | - Jiade J Lu
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| |
Collapse
|
48
|
Immune-Related lncRNA Correlated with Transcription Factors Provide Strong Prognostic Prediction in Gliomas. JOURNAL OF ONCOLOGY 2020; 2020:2319194. [PMID: 33178271 PMCID: PMC7647786 DOI: 10.1155/2020/2319194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 10/07/2020] [Indexed: 12/27/2022]
Abstract
Glioma is the most common and deadly tumor in central nervous system. According to previous studies, long noncoding RNAs (lncRNA) and transcription factors were significant factors of gliomas progression by regulating gliomas immune microenvironment. In our study, we built two independent cohorts from CGGA and TCGA. And we extracted 253 immune-related lncRNA correlated with prognosis. After LASSO analysis and multivariate Cox regression analysis, 8 immune-related lncRNA were used to construct classifier. The effectiveness of classifier was confirmed in both CGGA (AUC = 0.869) and TCGA (AUC = 0.902) cohorts. The correlation between transcription factors and immune-related lncRNA was calculated by WCGNA. Eventually, we built a network between 8 lncRNA and transcription factors. The function of core immune-related lncRNA in gliomas immune microenvironment was also investigated by CIBERTSORT. Our research provided a strong classifier of immune-related lncRNA to predict gliomas patient outcome. We also found the correlation between core immune-related lncRNA and transcription factors. These results may stimulate new strategy of immunotherapy in gliomas patients.
Collapse
|
49
|
Li G, Wu F, Zeng F, Zhai Y, Feng Y, Chang Y, Wang D, Jiang T, Zhang W. A novel DNA repair-related nomogram predicts survival in low-grade gliomas. CNS Neurosci Ther 2020; 27:186-195. [PMID: 33063446 PMCID: PMC7816205 DOI: 10.1111/cns.13464] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/20/2020] [Accepted: 09/26/2020] [Indexed: 12/17/2022] Open
Abstract
Aims We aimed to create a tumor recurrent‐based prediction model to predict recurrence and survival in patients with low‐grade glioma. Methods This study enrolled 291 patients (188 in the training group and 103 in the validation group) with clinicopathological information and transcriptome sequencing data. LASSO‐COX algorithm was applied to shrink predictive factor size and build a predictive recurrent signature. GO, KEGG, and GSVA analyses were performed for function annotations of the recurrent signature. The calibration curves and C‐Index were assessed to evaluate the nomogram's performance. Results This study found that DNA repair functions of tumor cells were significantly enriched in recurrent low‐grade gliomas. A predictive recurrent signature, built by the LASSO‐COX algorithm, was significantly associated with overall survival and progression‐free survival in low‐grade gliomas. Moreover, function annotations analysis of the predictive recurrent signature exhibited that the signature was associated with DNA repair functions. The nomogram, combining the predictive recurrent signature and clinical prognostic predictors, showed powerful prognostic ability in the training and validation groups. Conclusion An individualized prediction model was created to predict 1‐, 2‐, 3‐, 5‐, and 10‐year survival and recurrent rate of patients with low‐grade glioma, which may serve as a potential tool to guide postoperative individualized care.
Collapse
Affiliation(s)
- Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fan Wu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - You Zhai
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yuemei Feng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yuanhao Chang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Di Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Asian Glioma Genome Atlas Network (AGGA)
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Asian Glioma Genome Atlas Network (AGGA)
| |
Collapse
|
50
|
Xu C, Yuan B, He T, Ding B, Li S. Prognostic values of YTHDF1 regulated negatively by mir-3436 in Glioma. J Cell Mol Med 2020; 24:7538-7549. [PMID: 32449290 PMCID: PMC7339155 DOI: 10.1111/jcmm.15382] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/16/2020] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
M6A methylation is likely to be closely associated with the occurrence and development of tumours. In this study, we demonstrated that the transcription levels of the m6A RNA methylation regulators are closely related to the prognosis of glioma. Univariate Cox analysis was performed on the expression levels of methylation regulators and selected three hub genes in glioma. Next, we systematically compared the expression of these m6A RNA methylation regulators in gliomas with different clinicopathological features. The overall survival (OS) curve of the hub genes was initially established based on TCGA database information. YTHDF1 was selected from the hub genes following survival and prognosis analysis. A nomogram was developed to predict the survival probability. We further performed cell function and in vivo xenograft tumour experiments to further verify its role in tumour progression. Next, based on the miRanda and miRDB databases, we predicted one microRNA, hsa-mir-346, that might regulate and bind to 3'UTR of YTHDF1, which was confirmed by our fluorescent enzyme reporter gene experiment. In summary, m6A RNA methylation regulators play a potential role in the progression of gliomas. YTHDF1 may have an essential function in glioma diagnosis, treatment and prognosis.
Collapse
Affiliation(s)
- Chenyang Xu
- Henan University, Kaifeng, Henan, P.R. China.,Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, Henan, P.R. China
| | - Bingjian Yuan
- Henan University, Kaifeng, Henan, P.R. China.,Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, Henan, P.R. China
| | - Tao He
- Henan University, Kaifeng, Henan, P.R. China.,Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, Henan, P.R. China
| | - Bingqian Ding
- Henan University, Kaifeng, Henan, P.R. China.,Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, Henan, P.R. China
| | - Song Li
- Henan University, Kaifeng, Henan, P.R. China.,Department of Urology, Huaihe Hospital of Henan University, Kaifeng, Henan, P.R. China
| |
Collapse
|