1
|
Zhao K, Chen C, Zhang Y, Huang Z, Zhao Y, Yue Q, Xu J. Preoperative Assessment of Ki-67 Labeling Index in Pituitary Adenomas Using Delta-Radiomics Based on Dynamic Contrast-Enhanced MRI. J Magn Reson Imaging 2025. [PMID: 40091561 DOI: 10.1002/jmri.29764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Ki-67 labeling index (Ki-67 LI) is a proliferation marker that is correlated with aggressive behavior and prognosis of pituitary adenomas (PAs). Dynamic contrast-enhanced MRI (DCE-MRI) may potentially contribute to the preoperative assessment of Ki-67 LI. PURPOSE To investigate the feasibility of assessing Ki-67 LI of PAs preoperatively using delta-radiomics based on DCE-MRI. STUDY TYPE Retrospective. POPULATION 605 PA patients (female = 47.1%, average age = 52.2) from two centers (high Ki-67 LI (≥ 3%) = 229; low Ki-67 LI (< 3%) = 376), divided into a training set (n = 313), an internal validation set (n = 196), and an external validation set (n = 96). FIELD STRENGTH/SEQUENCE 1.5-T and 3-T, DCE-MRI. ASSESSMENT This study developed a non-delta-radiomics model based on the non-delta-radiomic features directly extracted from four phases, a delta-radiomics model based on the delta-radiomic features, and a combined model integrating clinical parameters (Knosp grade and tumor diameter) with delta-radiomic features. U test, recursive feature elimination (RFE), and least absolute shrinkage and selection operator (LASSO) regression were utilized to select important radiomic features. Support vector machine (SVM), XGBoost (XGB), logistic regression (LR), and Gaussian naive Bayes (GNB) were utilized to develop the models. STATISTICAL TESTS Receiver operating characteristic (ROC) curve. Calibration curve. Decision curve analysis (DCA). Intraclass correlation coefficients (ICC). DeLong test for ROC curves. U test or t test for numerical variables. Fisher's test or Chi-squared test for categorical variables. A p-value < 0.05 was considered statistically significant. RESULTS The combined model demonstrated the best performance in preoperatively assessing the Ki-67 LI of PAs, achieving AUCs of 0.937 and 0.897 in the internal and external validation sets, respectively. The models based on delta-radiomic features outperformed the non-delta-radiomic model. DATA CONCLUSION A delta-radiomics-based model using DCE-MRI may show high diagnostic performance for preoperatively assessing the Ki-67 LI status of PAs. EVIDENCE LEVEL 3 Technical Efficacy: Stage 2.
Collapse
Affiliation(s)
- Kaiyang Zhao
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Chaoyue Chen
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yang Zhang
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhouyang Huang
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yanjie Zhao
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qiang Yue
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Zang Y, Zheng F, Feng L, Shi X, Chen X. Preoperatively Predicting PIT1 Expression in Pituitary Adenomas Using Habitat, Intra-tumoral and Peri-tumoral Radiomics Based on MRI. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025:10.1007/s10278-024-01376-4. [PMID: 39904941 DOI: 10.1007/s10278-024-01376-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/10/2024] [Accepted: 12/08/2024] [Indexed: 02/06/2025]
Abstract
The study aimed to predict expression of pituitary transcription factor 1 (PIT1) in pituitary adenomas using habitat, intra-tumoral and peri-tumoral radiomics models. A total of 129 patients with pituitary adenoma (training set, n = 103; test set, n = 26) were retrospectively enrolled. A total of 12, 18, 14, 13, and 14 radiomics features were selected from the ROIintra, ROIintra+peri (ROIintra+2mm, ROIintra+4mm, ROIintra+6mm), and ROIhabitat, respectively. Then, three machine learning algorithms were employed to develop radiomic models, including logistic regression (LR), support vector machines (SVM), and multilayer perceptron (MLP). The performances of the intra-tumoral, combined intra-tumoral and peri-tumoral, and habitat models were evaluated. The peritumoral region (ROI2mm, ROI4mm, ROI6mm) of the combined model with the highest performance was individually selected for further peritumoral analysis. Moreover, a deep learning radiomics nomogram (DLRN) was constructed incorporating clinical characteristics and the peri-tumoral and habitat models for individual prediction. The combined modelintra+2mm based on ROIintra+2mm achieved a better performance (AUC, 0.800) than that of the intra-tumoral model alone (AUC, 0.731). And the habitat model showed a higher performance (AUC, 0.806) than that of the intra-tumoral model. In addition, the performance of the peri-tumoral model based on ROI2mm was 0.694 in the testing set. Furthermore, the DLRN achieved the highest performance of 0.900 in the test set. The DLRN showed the best performance for PIT1 expression in pituitary adenomas, followed by the habitat, combined modelintra+2mm, intra-tumoral model, and peri-tumoral model based on ROI2mm, respectively. These different models are helpful for the model choice in clinical work.
Collapse
Affiliation(s)
- Yuying Zang
- Department of Radiology, The Affiliated Children's Hospital, Capital Institute of Pediatrics, Beijing, China
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fei Zheng
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Limei Feng
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xinyao Shi
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xuzhu Chen
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Liu F, Zang Y, Feng L, Shi X, Wu W, Liu X, Song Y, Xu J, Gui S, Chen X. Concomitant Prediction of the Ki67 and PIT-1 Expression in Pituitary Adenoma Using Different Radiomics Models. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025; 38:394-409. [PMID: 38750186 PMCID: PMC11810862 DOI: 10.1007/s10278-024-01121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 02/12/2025]
Abstract
OBJECTIVES To preoperatively predict the high expression of Ki67 and positive pituitary transcription factor 1 (PIT-1) simultaneously in pituitary adenoma (PA) using three different radiomics models. METHODS A total of 247 patients with PA (training set: n = 198; test set: n = 49) were included in this retrospective study. The imaging features were extracted from preoperative contrast-enhanced T1WI (T1CE), T1-weighted imaging (T1WI), and T2-weighted imaging (T2WI). Feature selection was performed using Spearman's rank correlation coefficient and least absolute shrinkage and selection operator (LASSO). The classic machine learning (CML), deep learning (DL), and deep learning radiomics (DLR) models were constructed using logistic regression (LR), support vector machine (SVM), and multi-layer perceptron (MLP) algorithms. The area under the receiver operating characteristic (ROC) curve (AUC), sensitivity, specificity, accuracy, negative predictive value (NPV) and positive predictive value (PPV) were calculated for the training and test sets. In addition, combined with clinical characteristics, the best CML and the best DL models (SVM classifier), the DL radiomics nomogram (DLRN) was constructed to aid clinical decision-making. RESULTS Seven CML features, 96 DL features, and 107 DLR features were selected to construct CML, DL and DLR models. Compared to CML and DL model, the DLR model had the best performance. The AUC, sensitivity, specificity, accuracy, NPV and PPV were 0.827, 0.792, 0.800, 0.796, 0.800 and 0.792 in the test set, respectively. CONCLUSIONS Compared with CML and DL models, the DLR model shows the best performance in predicting the Ki67 and PIT-1 expression in PAs simultaneously.
Collapse
Affiliation(s)
- Fangzheng Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, 100070, Beijing, China
| | - Yuying Zang
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, 100070, Beijing, China
| | - Limei Feng
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, 100070, Beijing, China
| | - Xinyao Shi
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, 100070, Beijing, China
| | - Wentao Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, 100070, Beijing, China
| | - Xin Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, 100070, Beijing, China
| | - Yifan Song
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, 100070, Beijing, China
| | - Jintian Xu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, 100070, Beijing, China
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, 100070, Beijing, China.
| | - Xuzhu Chen
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, 100070, Beijing, China.
| |
Collapse
|
4
|
Patel K, Sanghvi H, Gill GS, Agarwal O, Pandya AS, Agarwal A, Gupta M. Differentiating Cystic Lesions in the Sellar Region of the Brain Using Artificial Intelligence and Machine Learning for Early Diagnosis: A Prospective Review of the Novel Diagnostic Modalities. Cureus 2024; 16:e75476. [PMID: 39791061 PMCID: PMC11717160 DOI: 10.7759/cureus.75476] [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] [Accepted: 12/09/2024] [Indexed: 01/12/2025] Open
Abstract
This paper investigates the potential of artificial intelligence (AI) and machine learning (ML) to enhance the differentiation of cystic lesions in the sellar region, such as pituitary adenomas, Rathke cleft cysts (RCCs) and craniopharyngiomas (CP), through the use of advanced neuroimaging techniques, particularly magnetic resonance imaging (MRI). The goal is to explore how AI-driven models, including convolutional neural networks (CNNs), deep learning, and ensemble methods, can overcome the limitations of traditional diagnostic approaches, providing more accurate and early differentiation of these lesions. The review incorporates findings from critical studies, such as using the Open Access Series of Imaging Studies (OASIS) dataset (Kaggle, San Francisco, USA) for MRI-based brain research, highlighting the significance of statistical rigor and automated segmentation in developing reliable AI models. By drawing on these insights and addressing the challenges posed by small, single-institutional datasets, the paper aims to demonstrate how AI applications can improve diagnostic precision, enhance clinical decision-making, and ultimately lead to better patient outcomes in managing sellar region cystic lesions.
Collapse
Affiliation(s)
- Kaivan Patel
- Department of Internal Medicine, Broward Health North, Deerfield Beach, USA
| | - Harshal Sanghvi
- Department of Technology and Clinical Trials, Advanced Research, Deerfield Beach, USA
| | - Gurnoor S Gill
- Department of Medicine, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, USA
| | - Ojas Agarwal
- Department of Medicine, New York University, New York City, USA
| | - Abhijit S Pandya
- College of Electrical Engineering and Computer Science (CEECS), Florida Atlantic University, Boca Raton, USA
| | - Ankur Agarwal
- College of Electrical Engineering and Computer Science (CEECS), Florida Atlantic University, Boca Raton, USA
| | - Manish Gupta
- Department of Technology and Clinical Trials, Advanced Research, Deerfield Beach, USA
| |
Collapse
|
5
|
Zilka T, Benesova W. Radiomics of pituitary adenoma using computer vision: a review. Med Biol Eng Comput 2024; 62:3581-3597. [PMID: 39012416 PMCID: PMC11568991 DOI: 10.1007/s11517-024-03163-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 07/01/2024] [Indexed: 07/17/2024]
Abstract
Pituitary adenomas (PA) represent the most common type of sellar neoplasm. Extracting relevant information from radiological images is essential for decision support in addressing various objectives related to PA. Given the critical need for an accurate assessment of the natural progression of PA, computer vision (CV) and artificial intelligence (AI) play a pivotal role in automatically extracting features from radiological images. The field of "Radiomics" involves the extraction of high-dimensional features, often referred to as "Radiomic features," from digital radiological images. This survey offers an analysis of the current state of research in PA radiomics. Our work comprises a systematic review of 34 publications focused on PA radiomics and other automated information mining pertaining to PA through the analysis of radiological data using computer vision methods. We begin with a theoretical exploration essential for understanding the theoretical background of radionmics, encompassing traditional approaches from computer vision and machine learning, as well as the latest methodologies in deep radiomics utilizing deep learning (DL). Thirty-four research works under examination are comprehensively compared and evaluated. The overall results achieved in the analyzed papers are high, e.g., the best accuracy is up to 96% and the best achieved AUC is up to 0.99, which establishes optimism for the successful use of radiomic features. Methods based on deep learning seem to be the most promising for the future. In relation to this perspective DL methods, several challenges are remarkable: It is important to create high-quality and sufficiently extensive datasets necessary for training deep neural networks. Interpretability of deep radiomics is also a big open challenge. It is necessary to develop and verify methods that will explain to us how deep radiomic features reflect various physics-explainable aspects.
Collapse
Affiliation(s)
- Tomas Zilka
- Saint Michal's Hospital, Bratislava, Slovakia
- Masaryk University, Brno, Czech Republic
| | - Wanda Benesova
- Slovak University of Technology in Bratislava, Bratislava, Slovakia.
| |
Collapse
|
6
|
Bini F, Missori E, Pucci G, Pasini G, Marinozzi F, Forte GI, Russo G, Stefano A. Preclinical Implementation of matRadiomics: A Case Study for Early Malformation Prediction in Zebrafish Model. J Imaging 2024; 10:290. [PMID: 39590754 PMCID: PMC11595506 DOI: 10.3390/jimaging10110290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Radiomics provides a structured approach to support clinical decision-making through key steps; however, users often face difficulties when switching between various software platforms to complete the workflow. To streamline this process, matRadiomics integrates the entire radiomics workflow within a single platform. This study extends matRadiomics to preclinical settings and validates it through a case study focused on early malformation differentiation in a zebrafish model. The proposed plugin incorporates Pyradiomics and streamlines feature extraction, selection, and classification using machine learning models (linear discriminant analysis-LDA; k-nearest neighbors-KNNs; and support vector machines-SVMs) with k-fold cross-validation for model validation. Classifier performances are evaluated using area under the ROC curve (AUC) and accuracy. The case study indicated the criticality of the long time required to extract features from preclinical images, generally of higher resolution than clinical images. To address this, a feature analysis was conducted to optimize settings, reducing extraction time while maintaining similarity to the original features. As a result, SVM exhibited the best performance for early malformation differentiation in zebrafish (AUC = 0.723; accuracy of 0.72). This case study underscores the plugin's versatility and effectiveness in early biological outcome prediction, emphasizing its applicability across biomedical research fields.
Collapse
Affiliation(s)
- Fabiano Bini
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Eudossiana 18, 00184 Rome, Italy; (F.B.); (E.M.); (F.M.)
| | - Elisa Missori
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Eudossiana 18, 00184 Rome, Italy; (F.B.); (E.M.); (F.M.)
| | - Gaia Pucci
- Institute of Bioimaging and Complex Biological Systems—National Research Council (IBSBC—CNR), Contrada Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (G.I.F.); (G.R.); (A.S.)
| | - Giovanni Pasini
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Eudossiana 18, 00184 Rome, Italy; (F.B.); (E.M.); (F.M.)
- Institute of Bioimaging and Complex Biological Systems—National Research Council (IBSBC—CNR), Contrada Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (G.I.F.); (G.R.); (A.S.)
| | - Franco Marinozzi
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Eudossiana 18, 00184 Rome, Italy; (F.B.); (E.M.); (F.M.)
| | - Giusi Irma Forte
- Institute of Bioimaging and Complex Biological Systems—National Research Council (IBSBC—CNR), Contrada Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (G.I.F.); (G.R.); (A.S.)
| | - Giorgio Russo
- Institute of Bioimaging and Complex Biological Systems—National Research Council (IBSBC—CNR), Contrada Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (G.I.F.); (G.R.); (A.S.)
| | - Alessandro Stefano
- Institute of Bioimaging and Complex Biological Systems—National Research Council (IBSBC—CNR), Contrada Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (G.I.F.); (G.R.); (A.S.)
| |
Collapse
|
7
|
Ayoub NF, Glicksman JT. Artificial Intelligence in Rhinology. Otolaryngol Clin North Am 2024; 57:831-842. [PMID: 38821734 DOI: 10.1016/j.otc.2024.04.010] [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: 06/02/2024]
Abstract
Rhinology, allergy, and skull base surgery are fields primed for the integration and implementation of artificial intelligence (AI). The heterogeneity of the disease processes within these fields highlights the opportunity for AI to augment clinical care and promote personalized medicine. Numerous research studies have been published demonstrating the development and clinical potential of AI models within the field. Most describe in silico evaluation models without direct clinical implementation. The major themes of existing studies include diagnostic or clinical decisions support, clustering patients into specific phenotypes or endotypes, predicting post-treatment outcomes, and surgical planning.
Collapse
Affiliation(s)
- Noel F Ayoub
- Department of Otolaryngology-Head & Neck Surgery, Mass Eye and Ear/Harvard Medical School, Boston, MA, USA.
| | - Jordan T Glicksman
- Department of Otolaryngology-Head & Neck Surgery, Mass Eye and Ear/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Zheng B, Zhao Z, Zheng P, Liu Q, Li S, Jiang X, Huang X, Ye Y, Wang H. The current state of MRI-based radiomics in pituitary adenoma: promising but challenging. Front Endocrinol (Lausanne) 2024; 15:1426781. [PMID: 39371931 PMCID: PMC11449739 DOI: 10.3389/fendo.2024.1426781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/30/2024] [Indexed: 10/08/2024] Open
Abstract
In the clinical diagnosis and treatment of pituitary adenomas, MRI plays a crucial role. However, traditional manual interpretations are plagued by inter-observer variability and limitations in recognizing details. Radiomics, based on MRI, facilitates quantitative analysis by extracting high-throughput data from images. This approach elucidates correlations between imaging features and pituitary tumor characteristics, thereby establishing imaging biomarkers. Recent studies have demonstrated the extensive application of radiomics in differential diagnosis, subtype identification, consistency evaluation, invasiveness assessment, and treatment response in pituitary adenomas. This review succinctly presents the general workflow of radiomics, reviews pertinent literature with a summary table, and provides a comparative analysis with traditional methods. We further elucidate the connections between radiological features and biological findings in the field of pituitary adenoma. While promising, the clinical application of radiomics still has a considerable distance to traverse, considering the issues with reproducibility of imaging features and the significant heterogeneity in pituitary adenoma patients.
Collapse
Affiliation(s)
- Baoping Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pingping Zheng
- Department of Neurosurgery, People’s Hospital of Biyang County, Zhumadian, China
| | - Qiang Liu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Li
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Huang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Youfan Ye
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haijun Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Akade E, Aslani F, Verdi K, Bahadoram M, Kaydani GA. Diagnosis of choroid plexus papilloma: Current perspectives and future directions. CANCER PATHOGENESIS AND THERAPY 2024; 2:173-179. [PMID: 39027146 PMCID: PMC11252511 DOI: 10.1016/j.cpt.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/17/2023] [Accepted: 09/22/2023] [Indexed: 07/20/2024]
Abstract
Choroid plexus papilloma (CPP) is a rare, slow-growing, and typically benign brain tumor that predominantly affects children. CPP is characterized by well-defined circular or lobulated masses in the ventricles, leading to symptoms related to increased intracranial pressure and hydrocephalus. CPP diagnosis relies on a combination of clinical presentation, imaging findings, and histological examination. The World Health Organization (WHO) classification categorizes choroid plexus tumors into CPP (Grade І), atypical CPP (aCPP, Grade II), and choroid plexus carcinoma (CPC, Grade III). This article reviewed current diagnostics modalities and explored the emergence of new diagnostic methods for CPP. Research on molecular markers and genetic alterations associated with CPP is ongoing, and some potential markers have been identified. These results offered insights into potential therapeutic targets and personalized treatment approaches for CPP. Advancements in radiomics and liquid biopsy hold promise for improving diagnostic accuracy and monitoring treatment outcomes for choroid plexus tumors. Radiomics can provide quantitative data from imaging studies, whereas liquid biopsy can analyze tumor-derived genetic material and molecular markers from body fluids, such as cerebrospinal fluid (CSF) and blood. The rapidly evolving fields of molecular and genetic research and novel diagnostic methods require continuous updates and advancements before their application in clinical practice. We hope that these advancements will lead to earlier and more precise diagnoses, better treatment options, and improved outcomes in patients with CPP and other brain tumors.
Collapse
Affiliation(s)
- Esma'il Akade
- Department of Medical Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 1579461357, Iran
| | - Fereshteh Aslani
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 1579461357, Iran
| | - Kimia Verdi
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 1579461357, Iran
| | - Mohammad Bahadoram
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 1579461357, Iran
| | - Gholam Abbas Kaydani
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 1579461357, Iran
| |
Collapse
|
10
|
Gitto S, Cuocolo R, Giannetta V, Badalyan J, Di Luca F, Fusco S, Zantonelli G, Albano D, Messina C, Sconfienza LM. Effects of Interobserver Segmentation Variability and Intensity Discretization on MRI-Based Radiomic Feature Reproducibility of Lipoma and Atypical Lipomatous Tumor. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1187-1200. [PMID: 38332405 PMCID: PMC11169199 DOI: 10.1007/s10278-024-00999-x] [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: 09/16/2023] [Revised: 12/17/2023] [Accepted: 12/21/2023] [Indexed: 02/10/2024]
Abstract
Segmentation and image intensity discretization impact on radiomics workflow. The aim of this study is to investigate the influence of interobserver segmentation variability and intensity discretization methods on the reproducibility of MRI-based radiomic features in lipoma and atypical lipomatous tumor (ALT). Thirty patients with lipoma or ALT were retrospectively included. Three readers independently performed manual contour-focused segmentation on T1-weighted and T2-weighted sequences, including the whole tumor volume. Additionally, a marginal erosion was applied to segmentations to evaluate its influence on feature reproducibility. After image pre-processing, with included intensity discretization employing both fixed bin number and width approaches, 1106 radiomic features were extracted from each sequence. Intraclass correlation coefficient (ICC) 95% confidence interval lower bound ≥ 0.75 defined feature stability. In contour-focused vs. margin shrinkage segmentation, the rates of stable features extracted from T1-weighted and T2-weighted images ranged from 92.68 to 95.21% vs. 90.69 to 95.66% after fixed bin number discretization and from 95.75 to 97.65% vs. 95.39 to 96.47% after fixed bin width discretization, respectively, with no difference between the two segmentation approaches (p ≥ 0.175). Higher stable feature rates and higher feature ICC values were found when implementing discretization with fixed bin width compared to fixed bin number, regardless of the segmentation approach (p < 0.001). In conclusion, MRI radiomic features of lipoma and ALT are reproducible regardless of the segmentation approach and intensity discretization method, although a certain degree of interobserver variability highlights the need for a preliminary reliability analysis in future studies.
Collapse
Affiliation(s)
- Salvatore Gitto
- IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157, Milan, Italy
- Dipartimento Di Scienze Biomediche Per La Salute, Università Degli Studi Di Milano, Milan, Italy
| | - Renato Cuocolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Vincenzo Giannetta
- Diagnostic and Interventional Radiology Department, IRCCS Ospedale San Raffaele-Turro, Università Vita-Salute San Raffaele, Milan, Italy
| | - Julietta Badalyan
- Scuola Di Specializzazione in Statistica Sanitaria E Biometria, Università Degli Studi Di Milano, Milan, Italy
| | - Filippo Di Luca
- Scuola Di Specializzazione in Radiodiagnostica, Università Degli Studi Di Milano, Milan, Italy
| | - Stefano Fusco
- Dipartimento Di Scienze Biomediche Per La Salute, Università Degli Studi Di Milano, Milan, Italy
| | - Giulia Zantonelli
- Dipartimento Di Scienze Biomediche Per La Salute, Università Degli Studi Di Milano, Milan, Italy
| | - Domenico Albano
- IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157, Milan, Italy
- Dipartimento Di Scienze Biomediche, Chirurgiche Ed Odontoiatriche, Università Degli Studi Di Milano, Milan, Italy
| | - Carmelo Messina
- IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157, Milan, Italy
- Dipartimento Di Scienze Biomediche Per La Salute, Università Degli Studi Di Milano, Milan, Italy
| | - Luca Maria Sconfienza
- IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157, Milan, Italy.
- Dipartimento Di Scienze Biomediche Per La Salute, Università Degli Studi Di Milano, Milan, Italy.
| |
Collapse
|
11
|
Maroufi SF, Doğruel Y, Pour-Rashidi A, Kohli GS, Parker CT, Uchida T, Asfour MZ, Martin C, Nizzola M, De Bonis A, Tawfik-Helika M, Tavallai A, Cohen-Gadol AA, Palmisciano P. Current status of artificial intelligence technologies in pituitary adenoma surgery: a scoping review. Pituitary 2024; 27:91-128. [PMID: 38183582 DOI: 10.1007/s11102-023-01369-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2023] [Indexed: 01/08/2024]
Abstract
PURPOSE Pituitary adenoma surgery is a complex procedure due to critical adjacent neurovascular structures, variations in size and extensions of the lesions, and potential hormonal imbalances. The integration of artificial intelligence (AI) and machine learning (ML) has demonstrated considerable potential in assisting neurosurgeons in decision-making, optimizing surgical outcomes, and providing real-time feedback. This scoping review comprehensively summarizes the current status of AI/ML technologies in pituitary adenoma surgery, highlighting their strengths and limitations. METHODS PubMed, Embase, Web of Science, and Scopus were searched following the PRISMA-ScR guidelines. Studies discussing the use of AI/ML in pituitary adenoma surgery were included. Eligible studies were grouped to analyze the different outcomes of interest of current AI/ML technologies. RESULTS Among the 2438 identified articles, 44 studies met the inclusion criteria, with a total of seventeen different algorithms utilized across all studies. Studies were divided into two groups based on their input type: clinicopathological and imaging input. The four main outcome variables evaluated in the studies included: outcome (remission, recurrence or progression, gross-total resection, vision improvement, and hormonal recovery), complications (CSF leak, readmission, hyponatremia, and hypopituitarism), cost, and adenoma-related factors (aggressiveness, consistency, and Ki-67 labeling) prediction. Three studies focusing on workflow analysis and real-time navigation were discussed separately. CONCLUSION AI/ML modeling holds promise for improving pituitary adenoma surgery by enhancing preoperative planning and optimizing surgical strategies. However, addressing challenges such as algorithm selection, performance evaluation, data heterogeneity, and ethics is essential to establish robust and reliable ML models that can revolutionize neurosurgical practice and benefit patients.
Collapse
Affiliation(s)
- Seyed Farzad Maroufi
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Neurosurgical Research Network (NRN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Yücel Doğruel
- Department of Neurosurgery, Yeditepe University School of Medicine, Istanbul, Turkey
| | - Ahmad Pour-Rashidi
- Department of Neurosurgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Gurkirat S Kohli
- Department of Neurosurgery, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | | | - Tatsuya Uchida
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Mohamed Z Asfour
- Department of Neurosurgery, Nasser Institute for Research and Treatment Hospital, Cairo, Egypt
| | - Clara Martin
- Department of Neurosurgery, Hospital de Alta Complejidad en Red "El Cruce", Florencio Varela, Buenos Aires, Argentina
| | | | - Alessandro De Bonis
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Amin Tavallai
- Department of Pediatric Neurosurgery, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Paolo Palmisciano
- Department of Neurological Surgery, University of California, Davis, 4860 Y Street, Suite 3740, Sacramento, CA, 95817, USA.
| |
Collapse
|
12
|
Wu J, Zhang F, Huang Y, Wei L, Mei T, Wang S, Zeng Z, Wang W. Predictive value of cyst/tumor volume ratio of pituitary adenoma for tumor cell proliferation. BMC Med Imaging 2024; 24:69. [PMID: 38515047 PMCID: PMC10958862 DOI: 10.1186/s12880-024-01246-z] [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: 07/31/2023] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND MRI has been widely used to predict the preoperative proliferative potential of pituitary adenoma (PA). However, the relationship between the cyst/tumor volume ratio (C/T ratio) and the proliferative potential of PA has not been reported. Herein, we determined the predictive value of the C/T ratio of PA for tumor cell proliferation. METHODS The clinical data of 72 patients with PA and cystic change on MRI were retrospectively analyzed. PA volume, cyst volume, and C/T ratio were calculated. The corresponding intraoperative specimens were collected. Immunohistochemistry and hematoxylin-eosin staining were performed to evaluate the Ki67 index and nuclear atypia. Patients were categorized according to the Ki67 index (< 3% and ≥ 3%) and nuclear atypia (absence and presence). Univariate and multivariate analyses were used to identify the significant predictors of the Ki67 index and nuclear atypia. The receiver operating characteristic curve assessed the prediction ability of the significant predictors. RESULTS Larger tumor volumes, smaller cyst volumes, and lower C/T ratios were found in patients with higher Ki67 indexes and those with nuclear atypia (P < 0.05). C/T ratio was an independent predictor of the Ki67 index (odds ratio = 0.010, 95% confidence interval = 0.000-0.462) and nuclear atypia (odds ratio = 0.010, 95% confidence interval = 0.000-0.250). The predictive value of the C/T ratio did not differ significantly from that of tumor volume (P > 0.05) but was better than that of cyst volume (P < 0.05). The area under the curve of the C/T ratio for predicting the Ki67 index and nuclear atypia was larger than that for predicting cyst volume and tumor volume. CONCLUSIONS C/T ratios can be used to predict PA tumor proliferation preoperatively. Our findings may facilitate the selection of surgery timing and the efficacy evaluation of surgery.
Collapse
Affiliation(s)
- Jianwu Wu
- Department of Neurosurgery, 900 Hospital of the Joint Logistics Team, No. 156 Xi'erhuanbei Road, Fuzhou, 350025, P. R. China
| | - Fangfang Zhang
- Department of Endocrinology, the Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, 350009, P. R. China
| | - Yinxing Huang
- Department of Neurosurgery, 900 Hospital of the Joint Logistics Team, No. 156 Xi'erhuanbei Road, Fuzhou, 350025, P. R. China
| | - Liangfeng Wei
- Department of Neurosurgery, 900 Hospital of the Joint Logistics Team, No. 156 Xi'erhuanbei Road, Fuzhou, 350025, P. R. China
| | - Tao Mei
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, P. R. China
| | - Shousen Wang
- Department of Neurosurgery, 900 Hospital of the Joint Logistics Team, No. 156 Xi'erhuanbei Road, Fuzhou, 350025, P. R. China.
| | - Zihuan Zeng
- Department of Neurosurgery, 900 Hospital of the Joint Logistics Team, No. 156 Xi'erhuanbei Road, Fuzhou, 350025, P. R. China.
| | - Wei Wang
- Department of Neurosurgery, the First Affiliated Hospital of Wenzhou Medical University, No. 2, Fuxue Lane, Wuma Street, Lucheng District, Wenzhou, 325000, P. R. China.
| |
Collapse
|
13
|
Bioletto F, Prencipe N, Berton AM, Aversa LS, Cuboni D, Varaldo E, Gasco V, Ghigo E, Grottoli S. Radiomic Analysis in Pituitary Tumors: Current Knowledge and Future Perspectives. J Clin Med 2024; 13:336. [PMID: 38256471 PMCID: PMC10816809 DOI: 10.3390/jcm13020336] [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: 11/27/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Radiomic analysis has emerged as a valuable tool for extracting quantitative features from medical imaging data, providing in-depth insights into various contexts and diseases. By employing methods derived from advanced computational techniques, radiomics quantifies textural information through the evaluation of the spatial distribution of signal intensities and inter-voxel relationships. In recent years, these techniques have gained considerable attention also in the field of pituitary tumors, with promising results. Indeed, the extraction of radiomic features from pituitary magnetic resonance imaging (MRI) images has been shown to provide useful information on various relevant aspects of these diseases. Some of the key topics that have been explored in the existing literature include the association of radiomic parameters with histopathological and clinical data and their correlation with tumor invasiveness and aggressive behavior. Their prognostic value has also been evaluated, assessing their role in the prediction of post-surgical recurrence, response to medical treatments, and long-term outcomes. This review provides a comprehensive overview of the current knowledge and application of radiomics in pituitary tumors. It also examines the current limitations and future directions of radiomic analysis, highlighting the major challenges that need to be addressed before a consistent integration of these techniques into routine clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Silvia Grottoli
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (F.B.); (N.P.); (A.M.B.); (L.S.A.); (D.C.); (E.V.); (V.G.); (E.G.)
| |
Collapse
|
14
|
Patel RV, Groff KJ, Bi WL. Applications and Integration of Radiomics for Skull Base Oncology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1462:285-305. [PMID: 39523272 DOI: 10.1007/978-3-031-64892-2_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Radiomics, a quantitative approach to extracting features from medical images, represents a new frontier in skull base oncology. Novel image analysis approaches have enabled us to capture patterns from images imperceptible by the human eye. This rich source of data can be combined with a range of clinical features, holding the potential to be a noninvasive source of biomarkers. Applications of radiomics in skull base pathologies have centered around three common tumor classes: meningioma, sellar/parasellar tumors, and vestibular schwannomas. Radiomic investigations can be categorized into five domains: tumor detection/segmentation, classification between tumor types, tumor grading, detection of tumor features, and prognostication. Various computational architectures have been employed across these domains, with deep-learning methods becoming more common versus machine learning. Across radiomic applications, contrast-enhanced T1-weighted MRI images remain the most utilized sequence for model development. Efforts to standardize and connect radiomic features to tumor biology have facilitated more clinically applicable radiomic models. Despite the advancement in model performance, several challenges continue to hinder translatability, including small sample sizes and model training on homogenous single institution data. To recognize the potential of radiomics for skull base oncology, prospective, multi-institutional collaboration will be the cornerstone for a validated radiomic technology.
Collapse
Affiliation(s)
- Ruchit V Patel
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Karenna J Groff
- New York University Grossman School of Medicine, New York, NY, USA
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Khan DZ, Hanrahan JG, Baldeweg SE, Dorward NL, Stoyanov D, Marcus HJ. Current and Future Advances in Surgical Therapy for Pituitary Adenoma. Endocr Rev 2023; 44:947-959. [PMID: 37207359 PMCID: PMC10502574 DOI: 10.1210/endrev/bnad014] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/14/2023] [Accepted: 05/17/2023] [Indexed: 05/21/2023]
Abstract
The vital physiological role of the pituitary gland, alongside its proximity to critical neurovascular structures, means that pituitary adenomas can cause significant morbidity or mortality. While enormous advancements have been made in the surgical care of pituitary adenomas, numerous challenges remain, such as treatment failure and recurrence. To meet these clinical challenges, there has been an enormous expansion of novel medical technologies (eg, endoscopy, advanced imaging, artificial intelligence). These innovations have the potential to benefit each step of the patient's journey, and ultimately, drive improved outcomes. Earlier and more accurate diagnosis addresses this in part. Analysis of novel patient data sets, such as automated facial analysis or natural language processing of medical records holds potential in achieving an earlier diagnosis. After diagnosis, treatment decision-making and planning will benefit from radiomics and multimodal machine learning models. Surgical safety and effectiveness will be transformed by smart simulation methods for trainees. Next-generation imaging techniques and augmented reality will enhance surgical planning and intraoperative navigation. Similarly, surgical abilities will be augmented by the future operative armamentarium, including advanced optical devices, smart instruments, and surgical robotics. Intraoperative support to surgical team members will benefit from a data science approach, utilizing machine learning analysis of operative videos to improve patient safety and orientate team members to a common workflow. Postoperatively, neural networks leveraging multimodal datasets will allow early detection of individuals at risk of complications and assist in the prediction of treatment failure, thus supporting patient-specific discharge and monitoring protocols. While these advancements in pituitary surgery hold promise to enhance the quality of care, clinicians must be the gatekeepers of the translation of such technologies, ensuring systematic assessment of risk and benefit prior to clinical implementation. In doing so, the synergy between these innovations can be leveraged to drive improved outcomes for patients of the future.
Collapse
Affiliation(s)
- Danyal Z Khan
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences, University College London, London W1W 7TY, UK
| | - John G Hanrahan
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences, University College London, London W1W 7TY, UK
| | - Stephanie E Baldeweg
- Department of Diabetes & Endocrinology, University College London Hospitals NHS Foundation Trust, London NW1 2BU, UK
- Centre for Obesity and Metabolism, Department of Experimental and Translational Medicine, Division of Medicine, University College London, London WC1E 6BT, UK
| | - Neil L Dorward
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Danail Stoyanov
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences, University College London, London W1W 7TY, UK
- Digital Surgery Ltd, Medtronic, London WD18 8WW, UK
| | - Hani J Marcus
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences, University College London, London W1W 7TY, UK
| |
Collapse
|
16
|
Dumitriu-Stan RI, Burcea IF, Salmen T, Poiana C. Prognostic Models in Growth-Hormone- and Prolactin-Secreting Pituitary Neuroendocrine Tumors: A Systematic Review. Diagnostics (Basel) 2023; 13:2118. [PMID: 37371013 DOI: 10.3390/diagnostics13122118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/02/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Growth-hormone (GH)- and prolactin (PRL)-secreting PitNETs (pituitary neuroendocrine tumors) are divided into multiple histological subtypes, which determine their clinical and biological variable behavior. Proliferation markers alone have a questionable degree of prediction, so we try to identify validated prognostic models as accurately as possible. (1) Background: The data available so far show that the use of staging and clinical-pathological classification of PitNETs, along with imaging, are useful in predicting the evolution of these tumors. So far, there is no consensus for certain markers that could predict tumor evolution. The application of the WHO (World Health Organisation) classification in practice needs to be further evaluated and validated. (2) Methods: We performed the CRD42023401959 protocol in Prospero with a systematic literature search in PubMed and Web of Science databases and included original full-text articles (randomized control trials and clinical trials) from the last 10 years, published in English, and the search used the following keywords: (i) pituitary adenoma AND (prognosis OR outcome OR prediction), (ii) growth hormone pituitary adenoma AND (prognosis OR outcome OR prediction), (iii) prolactin pituitary adenoma AND (prognosis OR outcome OR prediction); (iv) mammosomatotroph adenoma AND (prognosis OR outcome OR prediction). (3) Results: Two researchers extracted the articles of interest and if any disagreements occurred in the selection process, these were settled by a third reviewer. The articles were then assessed using the ROBIS bias assessment and 75 articles were included. (4) Conclusions: the clinical-pathological classification along with factors such as GH, IGF-1, prolactin levels both preoperatively and postoperatively offer valuable information.
Collapse
Affiliation(s)
- Roxana-Ioana Dumitriu-Stan
- Department of Endocrinology, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Doctoral School of 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Iulia-Florentina Burcea
- Department of Endocrinology, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
- 'C. I. Parhon' National Institute of Endocrinology, 011863 Bucharest, Romania
| | - Teodor Salmen
- Doctoral School of 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Catalina Poiana
- Department of Endocrinology, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
- 'C. I. Parhon' National Institute of Endocrinology, 011863 Bucharest, Romania
| |
Collapse
|
17
|
Koechli C, Zwahlen DR, Schucht P, Windisch P. Radiomics and machine learning for predicting the consistency of benign tumors of the central nervous system: A systematic review. Eur J Radiol 2023; 164:110866. [PMID: 37207398 DOI: 10.1016/j.ejrad.2023.110866] [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: 03/14/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/21/2023]
Abstract
PURPOSE Predicting the consistency of benign central nervous system (CNS) tumors prior to surgery helps to improve surgical outcomes. This review summarizes and analyzes the literature on using radiomics and/or machine learning (ML) for consistency prediction. METHOD The Medical Literature Analysis and Retrieval System Online (MEDLINE) database was screened for studies published in English from January 1st 2000. Data was extracted according to the PRISMA guidelines and quality of the studies was assessed in compliance with the Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2). RESULTS Eight publications were included focusing on pituitary macroadenomas (n = 5), pituitary adenomas (n = 1), and meningiomas (n = 2) using a retrospective (n = 6), prospective (n = 1), and unknown (n = 1) study design with a total of 763 patients for the consistency prediction. The studies reported an area under the curve (AUC) of 0.71-0.99 for their respective best performing model regarding the consistency prediction. Of all studies, four articles validated their models internally whereas none validated their models externally. Two articles stated making data available on request with the remaining publications lacking information with regard to data availability. CONCLUSIONS The research on consistency prediction of CNS tumors is still at an early stage regarding the use of radiomics and different ML techniques. Best-practice procedures regarding radiomics and ML need to be followed more rigorously to facilitate the comparison between publications and, accordingly, the possible implementation into clinical practice in the future.
Collapse
Affiliation(s)
- Carole Koechli
- Department of Radiation Oncology, Kantonsspital Winterthur, 8401 Winterthur, Switzerland; Universitätsklinik für Neurochirurgie, Bern University Hospital, 3010 Bern, Switzerland.
| | - Daniel R Zwahlen
- Department of Radiation Oncology, Kantonsspital Winterthur, 8401 Winterthur, Switzerland
| | - Philippe Schucht
- Universitätsklinik für Neurochirurgie, Bern University Hospital, 3010 Bern, Switzerland
| | - Paul Windisch
- Department of Radiation Oncology, Kantonsspital Winterthur, 8401 Winterthur, Switzerland
| |
Collapse
|
18
|
Wijethilake N, MacCormac O, Vercauteren T, Shapey J. Imaging biomarkers associated with extra-axial intracranial tumors: a systematic review. Front Oncol 2023; 13:1131013. [PMID: 37182138 PMCID: PMC10167010 DOI: 10.3389/fonc.2023.1131013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/27/2023] [Indexed: 05/16/2023] Open
Abstract
Extra-axial brain tumors are extra-cerebral tumors and are usually benign. The choice of treatment for extra-axial tumors is often dependent on the growth of the tumor, and imaging plays a significant role in monitoring growth and clinical decision-making. This motivates the investigation of imaging biomarkers for these tumors that may be incorporated into clinical workflows to inform treatment decisions. The databases from Pubmed, Web of Science, Embase, and Medline were searched from 1 January 2000 to 7 March 2022, to systematically identify relevant publications in this area. All studies that used an imaging tool and found an association with a growth-related factor, including molecular markers, grade, survival, growth/progression, recurrence, and treatment outcomes, were included in this review. We included 42 studies, comprising 22 studies (50%) of patients with meningioma; 17 studies (38.6%) of patients with pituitary tumors; three studies (6.8%) of patients with vestibular schwannomas; and two studies (4.5%) of patients with solitary fibrous tumors. The included studies were explicitly and narratively analyzed according to tumor type and imaging tool. The risk of bias and concerns regarding applicability were assessed using QUADAS-2. Most studies (41/44) used statistics-based analysis methods, and a small number of studies (3/44) used machine learning. Our review highlights an opportunity for future work to focus on machine learning-based deep feature identification as biomarkers, combining various feature classes such as size, shape, and intensity. Systematic Review Registration: PROSPERO, CRD42022306922.
Collapse
Affiliation(s)
- Navodini Wijethilake
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Oscar MacCormac
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
- Department of Neurosurgery, King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | - Tom Vercauteren
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Jonathan Shapey
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
- Department of Neurosurgery, King’s College Hospital NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
19
|
Herkenhoff CGB, Trarbach EB, Batista RL, Soares IC, Frassetto FP, do Nascimento FBP, Grande IPP, Silva PPB, Duarte FHG, Bronstein MD, Jallad RS. Survivin: A Potential Marker of Resistance to Somatostatin Receptor Ligands. J Clin Endocrinol Metab 2023; 108:876-887. [PMID: 36273993 DOI: 10.1210/clinem/dgac610] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 09/19/2022] [Indexed: 02/13/2023]
Abstract
CONTEXT Invasive and somatostatin receptor ligand (SRL)-resistant pituitary tumors represent a challenge in the clinical practice of endocrinologists. Efforts have been made to elucidate reliable makers for both. Survivin and eukaryotic translation initiation factor-binding protein 1 (4EBP1) are upregulated in several cancers and involved in apoptosis and cell proliferation. OBJECTIVE We explored the role of these markers in somatotropinomas. METHODS Immunostains for survivin and 4EBP1, and also for somatostatin receptor type 2 (SSTR2), Ki-67, and cytokeratin 18, were analyzed in tissue microarrays containing 52 somatotropinoma samples. Tumor invasiveness was evaluated in all samples while drug resistance was evaluated in 34 patients who received SRL treatment. All these parameters were correlated with first-generation SRL (fg-SRL) responsiveness and tumor invasiveness. RESULTS Low survivin expression (P = 0.04), hyperintense signal on T2 weighted image (T2WI) (P = 0.01), younger age (P = 0.01), sparsely granular adenomas (SGA) (P = 0.04), high postoperative growth hormone (GH) and insulin-like growth factor-1 (IGF-1) levels (P = 0.049 and P < 0.001, respectively), and large postoperative tumor size (P = 0.02) were associated with resistance to fg-SRL. Low survivin and SSTR2 expression and high 4EBP1 expression were associated with SGA (P = 0.04, P = 0.01, and P = 0.001, respectively). Younger age (P = 0.03), large tumor pre- and postoperative (P = 0.04 and P = 0.006, respectively), low SSTR2 expression (P = 0.03), and high baseline GH and IGF-1 (P = 0.01 and P = 0.02, respectively) were associated with tumor invasiveness. However, survivin, 4EBP1, Ki-67, and granulation patterns were not associated with tumor invasion. CONCLUSION This study suggests that low survivin expression is predictive of resistance to fg-SRL in somatotropinomas, but not of tumor invasiveness.
Collapse
Affiliation(s)
- Clarissa G Borba Herkenhoff
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
| | - Ericka B Trarbach
- Laboratory of Cellular and Molecular Endocrinology/LIM25 Division of Endocrinology and Metabology, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
| | - Rafael Loch Batista
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
- Service of Endocrine Oncology, Cancer Institute of the State of São Paulo (ICESP), Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
| | - Iberê Cauduro Soares
- Department of Pathology, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
| | - Fernando Pereira Frassetto
- Department of Pathology, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
| | | | - Isabella Pacetti Pajaro Grande
- Laboratory of Cellular and Molecular Endocrinology/LIM25 Division of Endocrinology and Metabology, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
| | - Paula P B Silva
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
| | - Felipe H G Duarte
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
| | - Marcello D Bronstein
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
- Laboratory of Cellular and Molecular Endocrinology/LIM25 Division of Endocrinology and Metabology, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
| | - Raquel S Jallad
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
- Laboratory of Cellular and Molecular Endocrinology/LIM25 Division of Endocrinology and Metabology, Clinics Hospital, University of São Paulo Medical School, São Paulo, CEP 05403-010, Brazil
| |
Collapse
|
20
|
Wang X, Dai Y, Lin H, Cheng J, Zhang Y, Cao M, Zhou Y. Shape and texture analyses based on conventional MRI for the preoperative prediction of the aggressiveness of pituitary adenomas. Eur Radiol 2023; 33:3312-3321. [PMID: 36738323 DOI: 10.1007/s00330-023-09412-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/21/2022] [Accepted: 12/29/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Pituitary adenomas can exhibit aggressive behavior, characterized by rapid growth, resistance to conventional treatment, and early recurrence. This study aims to evaluate the clinical value of shape-related features combined with textural features based on conventional MRI in evaluating the aggressiveness of pituitary adenomas and develop the best diagnostic model. METHODS Two hundred forty-six pituitary adenoma patients (84 aggressive, 162 non-aggressive) who underwent preoperative MRI were retrospectively reviewed. The patients were divided into training (n = 193) and testing (n = 53) sets. Clinical information, shape-related, and textural features extracted from the tumor volume on contrast-enhanced T1-weighted images (CE-T1WI), were compared between aggressive and non-aggressive groups. Variables with significant differences were enrolled into Pearson's correlation analysis to weaken multicollinearity. Logistic regression models based on the selected features were constructed to predict tumor aggressiveness under fivefold cross-validation. RESULTS Sixty-five imaging features, including five shape-related and sixty textural features, were extracted from volumetric CE-T1WI. Forty-seven features were significantly different between aggressive and non-aggressive groups (all p values < 0.05). After feature selection, four features (SHAPE_Sphericity, SHAPE_Compacity, DISCRETIZED_Q3, and DISCRETIZED_Kurtosis) were put into logistic regression analysis. Based on the combination of these features and Knosp grade, the model yielded an area under the curve value of 0.935, with a sensitivity of 94.4% and a specificity of 82.9%, to discriminate between aggressive and non-aggressive pituitary adenomas in the testing set. CONCLUSION The radiomic model based on tumor shape and textural features study from CE-T1WI might potentially assist in the preoperative aggressiveness diagnosis of pituitary adenomas. KEY POINTS • Pituitary adenomas with aggressive behavior exhibit rapid growth, resistance to conventional treatment, and early recurrence despite gross resection and may require multiline treatments. • Shape-related features and texture features based on CE-T1WI were significantly correlated with the Ki-67 labeling index, mitotic count, and p53 expression, and the proposed model achieved a favorable prediction of the aggressiveness of PAs with an AUC value of 0.935. • The prediction model might provide valuable guidance for individualized treatment in patients with PAs.
Collapse
Affiliation(s)
- Xiaoqing Wang
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongming Dai
- Central Research Institute, United Imaging Healthcare, Shanghai, China
| | - Hai Lin
- Central Research Institute, United Imaging Healthcare, Shanghai, China
| | - Jiahui Cheng
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiming Zhang
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengqiu Cao
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Yan Zhou
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
21
|
Deep learning reconstruction in pediatric brain MRI: comparison of image quality with conventional T2-weighted MRI. Neuroradiology 2023; 65:207-214. [PMID: 36156109 DOI: 10.1007/s00234-022-03053-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/09/2022] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Deep learning-based MRI reconstruction has recently been introduced to improve image quality. This study aimed to evaluate the performance of deep learning reconstruction in pediatric brain MRI. METHODS A total of 107 consecutive children who underwent 3.0 T brain MRI were included in this study. T2-weighted brain MRI was reconstructed using the three different reconstruction modes: deep learning reconstruction, conventional reconstruction with an intensity filter, and original T2 image without a filter. Two pediatric radiologists independently evaluated the following image quality parameters of three reconstructed images on a 5-point scale: overall image quality, image noisiness, sharpness of gray-white matter differentiation, truncation artifact, motion artifact, cerebrospinal fluid and vascular pulsation artifacts, and lesion conspicuity. The subjective image quality parameters were compared among the three reconstruction modes. Quantitative analysis of the signal uniformity using the coefficient of variation was performed for each reconstruction. RESULTS The overall image quality, noisiness, and gray-white matter sharpness were significantly better with deep learning reconstruction than with conventional or original reconstruction (all P < 0.001). Deep learning reconstruction had significantly fewer truncation artifacts than the other two reconstructions (all P < 0.001). Motion and pulsation artifacts showed no significant differences among the three reconstruction modes. For 36 lesions in 107 patients, lesion conspicuity was better with deep learning reconstruction than original reconstruction. Deep learning reconstruction showed lower signal variation compared to conventional and original reconstructions. CONCLUSION Deep learning reconstruction can reduce noise and truncation artifacts and improve lesion conspicuity and overall image quality in pediatric T2-weighted brain MRI.
Collapse
|
22
|
Maiseyeu I, Güresir Á, Vatter H, Herrlinger U, Becker A, Wach J, Güresir E. Preoperative Risk Stratification of Increased MIB-1 Labeling Index in Pituitary Adenoma: A Newly Proposed Prognostic Scoring System. J Clin Med 2022; 11:jcm11237151. [PMID: 36498723 PMCID: PMC9738462 DOI: 10.3390/jcm11237151] [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: 10/19/2022] [Revised: 11/20/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
The MIB-1 index is an important risk factor for progression-free survival (PFS) in pituitary adenoma (PA). Preoperatively, the MIB-1 index is not available in the decision-making process. A preoperative method regarding MIB-1 index estimation in PA has not been evaluated so far. Between 2011 and 2021, 109 patients with tumor morphology data, MIB-1 index data, and inflammatory and pituitary hormone laboratory values underwent surgery for PA. An MIB-1 index cutoff point (≥4/<4%) determines the probability of PFS in completely resected PA. An elevated MIB-1 index (≥4%) was present in 32 cases (29.4%) and was significantly associated with increased IGF-1, age ≤ 60, increased ACTH, and increased fibrinogen levels in the multivariable analysis. A scoring system (“FATE”) using preoperative IGF-1, age, ACTH, and plasma fibrinogen level enables the estimation of the MIB-1 index (sensitivity 72%, specificity 68%). The FATE score is also significantly associated with the time to PA progression after the complete resection of the PA. We propose the FATE score to preoperatively estimate the risk of an elevated MIB-1 index (≥4%), which might enable tailoring to medical decision-making, and follow-up interval scheduling, as well as inform future studies analyzing proliferative activities.
Collapse
Affiliation(s)
- Ivan Maiseyeu
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
- Correspondence: ; Tel.: +49-228-287-16521
| | - Ági Güresir
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Hartmut Vatter
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Ulrich Herrlinger
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, 53127 Bonn, Germany
| | - Albert Becker
- Department of Neuropathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Johannes Wach
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Erdem Güresir
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
23
|
Li N, Desiderio DM, Zhan X. The use of mass spectrometry in a proteome-centered multiomics study of human pituitary adenomas. MASS SPECTROMETRY REVIEWS 2022; 41:964-1013. [PMID: 34109661 DOI: 10.1002/mas.21710] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 06/12/2023]
Abstract
A pituitary adenoma (PA) is a common intracranial neoplasm, and is a complex, chronic, and whole-body disease with multicausing factors, multiprocesses, and multiconsequences. It is very difficult to clarify molecular mechanism and treat PAs from the single-factor strategy model. The rapid development of multiomics and systems biology changed the paradigms from a traditional single-factor strategy to a multiparameter systematic strategy for effective management of PAs. A series of molecular alterations at the genome, transcriptome, proteome, peptidome, metabolome, and radiome levels are involved in pituitary tumorigenesis, and mutually associate into a complex molecular network system. Also, the center of multiomics is moving from structural genomics to phenomics, including proteomics and metabolomics in the medical sciences. Mass spectrometry (MS) has been extensively used in phenomics studies of human PAs to clarify molecular mechanisms, and to discover biomarkers and therapeutic targets/drugs. MS-based proteomics and proteoform studies play central roles in the multiomics strategy of PAs. This article reviews the status of multiomics, multiomics-based molecular pathway networks, molecular pathway network-based pattern biomarkers and therapeutic targets/drugs, and future perspectives for personalized, predeictive, and preventive (3P) medicine in PAs.
Collapse
Affiliation(s)
- Na Li
- Shandong Key Laboratory of Radiation Oncology, Cancer Hospital of Shandong First Medical University, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong, China
| | - Dominic M Desiderio
- The Charles B. Stout Neuroscience Mass Spectrometry Laboratory, Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Xianquan Zhan
- Shandong Key Laboratory of Radiation Oncology, Cancer Hospital of Shandong First Medical University, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
24
|
Won SY, Lee N, Park YW, Ahn SS, Ku CR, Kim EH, Lee SK. Quality reporting of radiomics analysis in pituitary adenomas: promoting clinical translation. Br J Radiol 2022; 95:20220401. [PMID: 36018049 PMCID: PMC9793472 DOI: 10.1259/bjr.20220401] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/15/2022] [Accepted: 07/27/2022] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE To evaluate the quality of radiomics studies on pituitary adenoma according to the radiomics quality score (RQS) and Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis (TRIPOD). METHODS PubMed MEDLINE and EMBASE were searched to identify radiomics studies on pituitary adenomas. From 138 articles, 20 relevant original research articles were included. Studies were scored based on RQS and TRIPOD guidelines. RESULTS Most included studies did not perform pre-processing; isovoxel resampling, signal intensity normalization, and N4 bias field correction were performed in only five (25%), eight (40%), and four (20%) studies, respectively. Only two (10%) studies performed external validation. The mean RQS and basic adherence rate were 2.8 (7.6%) and 26.6%, respectively. There was a low adherence rate for conducting comparison to "gold-standard" (20%), multiple segmentation (25%), and stating potential clinical utility (25%). No study stated the biological correlation, conducted a test-retest or phantom study, was a prospective study, conducted cost-effectiveness analysis, or provided open-source code and data, which resulted in low-level evidence. The overall adherence rate for TRIPOD was 54.6%, and it was low for reporting the title (5%), abstract (0%), explaining the sample size (10%), and suggesting a full prediction model (5%). CONCLUSION The radiomics reporting quality for pituitary adenoma is insufficient. Pre-processing is required for feature reproducibility and external validation is necessary. Feature reproducibility, clinical utility demonstration, higher evidence levels, and open science are required. Titles, abstracts, and full prediction model suggestions should be improved for transparent reporting. ADVANCES IN KNOWLEDGE Despite the rapidly increasing number of radiomics researches on pituitary adenoma, the quality of science in these researches is unknown. Our study indicates that the overall quality needs to be significantly improved in radiomics studies on pituitary adenoma, and since the concept of RQS and IBSI is still unfamiliar to clinicians and radiologist researchers, our study may help to reach higher technical and clinical impact in the future study.
Collapse
Affiliation(s)
| | - Narae Lee
- Department of Nuclear Medicine, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Yae Won Park
- Department of Radiology and Research, Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Soo Ahn
- Department of Radiology and Research, Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Cheol Ryong Ku
- Department of Endocrinology, Yonsei University College of Medicine, Seoul, Korea
| | - Eui Hyun Kim
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Koo Lee
- Department of Radiology and Research, Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Wang X, Li M, Jiang X, Wang F, Ling S, Niu C. Prediction of Higher Ki-67 Index in Pituitary Adenomas by Pre- and Intra-Operative Clinical Characteristics. Brain Sci 2022; 12:brainsci12081002. [PMID: 36009065 PMCID: PMC9405805 DOI: 10.3390/brainsci12081002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/08/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Objective: The Ki-67 index is an indicator of the active proliferation and aggressive behavior of pituitary adenomas (PAs). Appropriate pre- and intra-operatives of the Ki-67 index can help surgeons develop better and more personalized treatment strategies for patients with PAs. This study aimed to investigate the influence factors for predicting the Ki-67 index in PAs. Methods: Data of 178 patients with PAs confirmed by pathology were retrospectively analyzed. According to the Ki-67 index, the patients were divided into the Ki-67 < 3% and Ki-67 ≥ 3% cohorts. Patient data, including age, sex, postoperative immunohistochemical pituitary hormone positive index, Knosp grade, tumor breaking through the sellar floor, rich blood supply to the tumor, tumor located inside the sella, erosion of the dorsum sellae bone, and pituitary-specific transcription factor, were collected. A univariate logistic analysis was used to evaluate the influence factors for a high Ki-67 index. Multiple regression and receiver operating characteristic (ROC) curve were used to analyze the factors with p < 0.05. The mutant status of Ki-67 index was predicted by nomogram. Results: Multivariate regression analysis showed that rich blood supply to the tumor and erosion of the dorsum sellae bone were independent risk factors for the Ki-67 proliferation index. The ROC curves demonstrated that age, rich blood supply to the tumor, and erosion of the dorsum sellae bone can predict the occurrence of a high Ki-67 index. Together, the three risk factors provide a stronger ability to predict the Ki-67 index. The nomogram was developed and validated. Conclusion: Age, rich blood supply to the tumor, and erosion of the dorsum sellae bone are influencing factors for predicting the Ki-67 index. Suitable nomogram prediction models were developed and validated, and there is potential for personalized treatment for PA patients.
Collapse
Affiliation(s)
- Xuanzhi Wang
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (X.W.); (M.L.); (X.J.); (F.W.); (S.L.)
| | - Mingwu Li
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (X.W.); (M.L.); (X.J.); (F.W.); (S.L.)
| | - Xiaofeng Jiang
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (X.W.); (M.L.); (X.J.); (F.W.); (S.L.)
| | - Fei Wang
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (X.W.); (M.L.); (X.J.); (F.W.); (S.L.)
| | - Shiying Ling
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (X.W.); (M.L.); (X.J.); (F.W.); (S.L.)
| | - Chaoshi Niu
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (X.W.); (M.L.); (X.J.); (F.W.); (S.L.)
- Anhui Province Key Laboratory of Brain Function and Brain Disease, Hefei 230001, China
- Anhui Provincial Clinical Research Center for Neurosurgical Disease, Hefei 230001, China
- Correspondence:
| |
Collapse
|
26
|
Machine learning in neuro-oncology: toward novel development fields. J Neurooncol 2022; 159:333-346. [PMID: 35761160 DOI: 10.1007/s11060-022-04068-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/11/2022] [Indexed: 10/17/2022]
Abstract
PURPOSE Artificial Intelligence (AI) involves several and different techniques able to elaborate a large amount of data responding to a specific planned outcome. There are several possible applications of this technology in neuro-oncology. METHODS We reviewed, according to PRISMA guidelines, available studies adopting AI in different fields of neuro-oncology including neuro-radiology, pathology, surgery, radiation therapy, and systemic treatments. RESULTS Neuro-radiology presented the major number of studies assessing AI. However, this technology is being successfully tested also in other operative settings including surgery and radiation therapy. In this context, AI shows to significantly reduce resources and costs maintaining an elevated qualitative standard. Pathological diagnosis and development of novel systemic treatments are other two fields in which AI showed promising preliminary data. CONCLUSION It is likely that AI will be quickly included in some aspects of daily clinical practice. Possible applications of these techniques are impressive and cover all aspects of neuro-oncology.
Collapse
|
27
|
Sahin S, Yildiz G, Oguz SH, Civan O, Cicek E, Durcan E, Comunoglu N, Ozkaya HM, Oz AB, Soylemezoglu F, Oguz KK, Dagdelen S, Erbas T, Kizilkilic O, Kadioglu P. Discrimination between non-functioning pituitary adenomas and hypophysitis using machine learning methods based on magnetic resonance imaging‑derived texture features. Pituitary 2022; 25:474-479. [PMID: 35334029 DOI: 10.1007/s11102-022-01213-3] [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] [Accepted: 02/27/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Hypophysitis is a heterogeneous condition that includes inflammation of the pituitary gland and infundibulum, and it can cause symptoms related to mass effects and hormonal deficiencies. We aimed to evaluate the potential role of machine learning methods in differentiating hypophysitis from non-functioning pituitary adenomas. METHODS The radiomic parameters obtained from T1A-C images were used. Among the radiomic parameters, parameters capable of distinguishing between hypophysitis and non-functioning pituitary adenomas were selected. In order to avoid the effects of confounding factors and to improve the performance of the classifiers, parameters with high correlation with each other were eliminated. Machine learning algorithms were performed with the combination of gray-level run-length matrix-low gray level run emphasis, gray-level co-occurrence matrix-correlation, and gray-level co-occurrence entropy. RESULTS A total of 34 patients were included, 17 of whom had hypophysitis and 17 had non-functioning pituitary adenomas. Among the 38 radiomics parameters obtained from post-contrast T1-weighted images, 10 tissue features that could differentiate the lesions were selected. Machine learning algorithms were performed using three selected parameters; gray level run length matrix-low gray level run emphasis, gray-level co-occurrence matrix-correlation, and gray level co-occurrence entropy. Error matrices were calculated by using the machine learning algorithm and it was seen that support vector machines showed the best performance in distinguishing the two lesion types. CONCLUSIONS Our analysis reported that support vector machines showed the best performance in distinguishing hypophysitis from non-functioning pituitary adenomas, emphasizing the importance of machine learning in differentiating the two lesions.
Collapse
Affiliation(s)
- Serdar Sahin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Gokcen Yildiz
- Department of Radiology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Seda Hanife Oguz
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Orkun Civan
- Department of Radiology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Ebru Cicek
- Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University- Cerrahpasa, Istanbul, Turkey
| | - Emre Durcan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Nil Comunoglu
- Department of Pathology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Hande Mefkure Ozkaya
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Aysim Buge Oz
- Department of Pathology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Figen Soylemezoglu
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Kader Karli Oguz
- Department of Radiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Selçuk Dagdelen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Tomris Erbas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Osman Kizilkilic
- Department of Radiology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Pinar Kadioglu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
- Cerrahpasa Medical Faculty, Department of Internal Medicine, Division of Endocrinology and Metabolism, Istanbul University-Cerrahpasa, Kocamustafapasa Street No: 53, 34098, Fatih, Istanbul, Turkey.
| |
Collapse
|
28
|
Machine Learning for the Detection and Segmentation of Benign Tumors of the Central Nervous System: A Systematic Review. Cancers (Basel) 2022; 14:cancers14112676. [PMID: 35681655 PMCID: PMC9179850 DOI: 10.3390/cancers14112676] [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: 04/26/2022] [Revised: 05/18/2022] [Accepted: 05/26/2022] [Indexed: 11/20/2022] Open
Abstract
Simple Summary Machine learning in radiology of the central nervous system has seen many interesting publications in the past few years. Since the focus has largely been on malignant tumors such as brain metastases and high-grade gliomas, we conducted a systematic review on benign tumors to summarize what has been published and where there might be gaps in the research. We found several studies that report good results, but the descriptions of methodologies could be improved to enable better comparisons and assessment of biases. Abstract Objectives: To summarize the available literature on using machine learning (ML) for the detection and segmentation of benign tumors of the central nervous system (CNS) and to assess the adherence of published ML/diagnostic accuracy studies to best practice. Methods: The MEDLINE database was searched for the use of ML in patients with any benign tumor of the CNS, and the records were screened according to PRISMA guidelines. Results: Eleven retrospective studies focusing on meningioma (n = 4), vestibular schwannoma (n = 4), pituitary adenoma (n = 2) and spinal schwannoma (n = 1) were included. The majority of studies attempted segmentation. Links to repositories containing code were provided in two manuscripts, and no manuscripts shared imaging data. Only one study used an external test set, which raises the question as to whether some of the good performances that have been reported were caused by overfitting and may not generalize to data from other institutions. Conclusions: Using ML for detecting and segmenting benign brain tumors is still in its infancy. Stronger adherence to ML best practices could facilitate easier comparisons between studies and contribute to the development of models that are more likely to one day be used in clinical practice.
Collapse
|
29
|
Shu XJ, Chang H, Wang Q, Chen WG, Zhao K, Li BY, Sun GC, Chen SB, Xu BN. Deep Learning model-based approach for Preoperative prediction of Ki67 labeling index status in a noninvasive way using magnetic resonance images: a single-center study. Clin Neurol Neurosurg 2022; 219:107301. [DOI: 10.1016/j.clineuro.2022.107301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/01/2022] [Accepted: 05/15/2022] [Indexed: 11/30/2022]
|
30
|
Radiomics of Musculoskeletal Sarcomas: A Narrative Review. J Imaging 2022; 8:jimaging8020045. [PMID: 35200747 PMCID: PMC8876222 DOI: 10.3390/jimaging8020045] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/31/2022] [Accepted: 02/10/2022] [Indexed: 12/23/2022] Open
Abstract
Bone and soft-tissue primary malignant tumors or sarcomas are a large, diverse group of mesenchymal-derived malignancies. They represent a model for intra- and intertumoral heterogeneities, making them particularly suitable for radiomics analyses. Radiomic features offer information on cancer phenotype as well as the tumor microenvironment which, combined with other pertinent data such as genomics and proteomics and correlated with outcomes data, can produce accurate, robust, evidence-based, clinical-decision support systems. Our purpose in this narrative review is to offer an overview of radiomics studies dealing with Magnetic Resonance Imaging (MRI)-based radiomics models of bone and soft-tissue sarcomas that could help distinguish different histotypes, low-grade from high-grade sarcomas, predict response to multimodality therapy, and thus better tailor patients’ treatments and finally improve their survivals. Although showing promising results, interobserver segmentation variability, feature reproducibility, and model validation are three main challenges of radiomics that need to be addressed in order to translate radiomics studies to clinical applications. These efforts, together with a better knowledge and application of the “Radiomics Quality Score” and Image Biomarker Standardization Initiative reporting guidelines, could improve the quality of sarcoma radiomics studies and facilitate radiomics towards clinical translation.
Collapse
|
31
|
Kalasauskas D, Kosterhon M, Keric N, Korczynski O, Kronfeld A, Ringel F, Othman A, Brockmann MA. Beyond Glioma: The Utility of Radiomic Analysis for Non-Glial Intracranial Tumors. Cancers (Basel) 2022; 14:cancers14030836. [PMID: 35159103 PMCID: PMC8834271 DOI: 10.3390/cancers14030836] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/30/2022] [Accepted: 02/04/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Tumor qualities, such as growth rate, firmness, and intrusion into healthy tissue, can be very important for operation planning and further treatment. Radiomics is a promising new method that allows the determination of some of these qualities on images performed before surgery. In this article, we provide a review of the use of radiomics in various tumors of the central nervous system, such as metastases, lymphoma, meningioma, medulloblastoma, and pituitary tumors. Abstract The field of radiomics is rapidly expanding and gaining a valuable role in neuro-oncology. The possibilities related to the use of radiomic analysis, such as distinguishing types of malignancies, predicting tumor grade, determining the presence of particular molecular markers, consistency, therapy response, and prognosis, can considerably influence decision-making in medicine in the near future. Even though the main focus of radiomic analyses has been on glial CNS tumors, studies on other intracranial tumors have shown encouraging results. Therefore, as the main focus of this review, we performed an analysis of publications on PubMed and Web of Science databases, focusing on radiomics in CNS metastases, lymphoma, meningioma, medulloblastoma, and pituitary tumors.
Collapse
Affiliation(s)
- Darius Kalasauskas
- Department of Neurosurgery, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.K.); (M.K.); (N.K.); (F.R.)
| | - Michael Kosterhon
- Department of Neurosurgery, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.K.); (M.K.); (N.K.); (F.R.)
| | - Naureen Keric
- Department of Neurosurgery, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.K.); (M.K.); (N.K.); (F.R.)
| | - Oliver Korczynski
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.K.); (A.K.); (A.O.)
| | - Andrea Kronfeld
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.K.); (A.K.); (A.O.)
| | - Florian Ringel
- Department of Neurosurgery, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.K.); (M.K.); (N.K.); (F.R.)
| | - Ahmed Othman
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.K.); (A.K.); (A.O.)
| | - Marc A. Brockmann
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.K.); (A.K.); (A.O.)
- Correspondence:
| |
Collapse
|
32
|
Zhang Y, Luo Y, Kong X, Wan T, Long Y, Ma J. A Preoperative MRI-Based Radiomics-Clinicopathological Classifier to Predict the Recurrence of Pituitary Macroadenoma Within 5 Years. Front Neurol 2022; 12:780628. [PMID: 35069413 PMCID: PMC8767054 DOI: 10.3389/fneur.2021.780628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/02/2021] [Indexed: 11/23/2022] Open
Abstract
Objective: To investigate the ability of a MRI-based radiomics-clinicopathological model to predict pituitary macroadenoma (PMA) recurrence within 5 years. Materials and Methods: We recruited 74 recurrent and 94 non-recurrent subjects, following first surgery with 5-year follow-up data. Univariate and multivariate analyses were conducted to identify independent clinicopathological risk factors. Two independent and blinded neuroradiologists used 3D-Slicer software to manually delineate whole tumors using preoperative axial contrast-enhanced T1WI (CE-T1WI) images. 3D-Slicer was then used to extract radiomics features from segmented tumors. Dimensionality reduction was carried out by the least absolute shrinkage and selection operator (LASSO). Two multilayer perceptron (MLP) models were established, including independent clinicopathological risk factors (Model 1) and a combination of screened radiomics features and independent clinicopathological markers (Model 2). The predictive performance of these models was evaluated by receiver operator characteristic (ROC) curve analysis. Results: In total, 1,130 features were identified, and 4 of these were selected by LASSO. In the test set, the area under the curve (AUC) of Model 2 was superior to Model 1 {0.783, [95% confidence interval (CI): 0.718—.860] vs. 0.739, (95% CI: 0.665–0.818)}. Model 2 also yielded the higher accuracy (0.808 vs. 0.692), sensitivity (0.826 vs. 0.652), and specificity (0.793 vs. 0.724) than Model 1. Conclusions: The integrated classifier was superior to a clinical classifier and may facilitate the prediction of individualized prognosis and therapy.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuqi Luo
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xin Kong
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Wan
- School of Biomedical Science and Medical Engineering, Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, China
| | - Yunling Long
- Department of Biomedical Engineering, School of Biomedical Engineering, Capital Medical University, Beijing, China.,Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Beijing, China
| | - Jun Ma
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
33
|
Xue C, Liu S, Deng J, Liu X, Li S, Zhang P, Zhou J. Apparent Diffusion Coefficient Histogram Analysis for the Preoperative Evaluation of Ki-67 Expression in Pituitary Macroadenoma. Clin Neuroradiol 2022; 32:269-276. [PMID: 35029726 DOI: 10.1007/s00062-021-01134-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 12/21/2021] [Indexed: 11/03/2022]
Abstract
PURPOSE To explore the value of an apparent diffusion coefficient (ADC) histogram in predicting the Ki-67 proliferation index in pituitary macroadenomas. MATERIAL AND METHODS This retrospective study analyzed the pathological and imaging data of 102 patients with pathologically confirmed pituitary macroadenoma. Immunohistochemistry staining was used to assess Ki-67 expression in tumor tissue samples, and a high Ki-67 labeling index was defined as 3%. The ADC images of the maximum slice of tumors were selected and the region of interest (ROI) of each slice was delineated using the MaZda software (version 4.7, Technical University of Lodz, Institute of Electronics, Łódź, Poland) and analyzed by ADC histogram. Histogram characteristic parameters were compared between the high Ki-67 group (n = 42) and the low Ki-67 group (n = 60). The important parameters were further analyzed by receiver operating characteristic (ROC). RESULTS The mean value, and the 1st, 10th, 50th, 90th, and 99th percentiles were found to be negatively correlated with Ki-67 expression (all P < 0.05), with correlation coefficients of -0.292, -0.352, -0.344, -0.289, -0.253 and -0.267, respectively. The mean ADC and the 1st, 10th, 50th, 90th, and 99th quantiles extracted from the histogram were significantly lower in the high Ki-67 group than in the low Ki-67 group (all P < 0.05). The area under the ROC curve was 0.699-0.720; however, there were no significant between-group differences in variance, skewness and kurtosis (all P > 0.05). CONCLUSION An ADC histogram can be a reliable tool to predict the Ki-67 proliferation status in patients with pituitary macroadenomas.
Collapse
Affiliation(s)
- Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, 730030, Chengguan District, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Suwei Liu
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, 730030, Chengguan District, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Juan Deng
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, 730030, Chengguan District, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Xianwang Liu
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, 730030, Chengguan District, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Shenglin Li
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, 730030, Chengguan District, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Peng Zhang
- Department of Pathology, Lanzhou University Second Hospital, Cuiyingmen No. 82, 730030, Chengguan District, Lanzhou, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, 730030, Chengguan District, Lanzhou, China. .,Second Clinical School, Lanzhou University, Lanzhou, China. .,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China. .,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China.
| |
Collapse
|
34
|
Dai C, Sun B, Wang R, Kang J. The Application of Artificial Intelligence and Machine Learning in Pituitary Adenomas. Front Oncol 2022; 11:784819. [PMID: 35004306 PMCID: PMC8733587 DOI: 10.3389/fonc.2021.784819] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/02/2021] [Indexed: 12/28/2022] Open
Abstract
Pituitary adenomas (PAs) are a group of tumors with complex and heterogeneous clinical manifestations. Early accurate diagnosis, individualized management, and precise prediction of the treatment response and prognosis of patients with PA are urgently needed. Artificial intelligence (AI) and machine learning (ML) have garnered increasing attention to quantitatively analyze complex medical data to improve individualized care for patients with PAs. Therefore, we critically examined the current use of AI and ML in the management of patients with PAs, and we propose improvements for future uses of AI and ML in patients with PAs. AI and ML can automatically extract many quantitative features based on massive medical data; moreover, related diagnosis and prediction models can be developed through quantitative analysis. Previous studies have suggested that AI and ML have wide applications in early accurate diagnosis; individualized treatment; predicting the response to treatments, including surgery, medications, and radiotherapy; and predicting the outcomes of patients with PAs. In addition, facial imaging-based AI and ML, pathological picture-based AI and ML, and surgical microscopic video-based AI and ML have also been reported to be useful in assisting the management of patients with PAs. In conclusion, the current use of AI and ML models has the potential to assist doctors and patients in making crucial surgical decisions by providing an accurate diagnosis, response to treatment, and prognosis of PAs. These AI and ML models can improve the quality and safety of medical services for patients with PAs and reduce the complication rates of neurosurgery. Further work is needed to obtain more reliable algorithms with high accuracy, sensitivity, and specificity for the management of PA patients.
Collapse
Affiliation(s)
- Congxin Dai
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Bowen Sun
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Kang
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
35
|
AIM in Endocrinology. Artif Intell Med 2022. [DOI: 10.1007/978-3-030-64573-1_328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
|
36
|
Stumpo V, Staartjes VE, Regli L, Serra C. Machine Learning in Pituitary Surgery. ACTA NEUROCHIRURGICA. SUPPLEMENT 2021; 134:291-301. [PMID: 34862553 DOI: 10.1007/978-3-030-85292-4_33] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Machine learning applications in neurosurgery are increasingly reported for diverse tasks such as faster and more accurate preoperative diagnosis, enhanced lesion characterization, as well as surgical outcome, complications and healthcare cost prediction. Even though the pertinent literature in pituitary surgery is less extensive with respect to other neurosurgical diseases, past research attempted to answer clinically relevant questions to better assist surgeons and clinicians. In the present chapter we review reported ML applications in pituitary surgery including differential diagnosis, preoperative lesion characterization (immunohistochemistry, cavernous sinus invasion, tumor consistency), surgical outcome and complication predictions (gross total resection, tumor recurrence, and endocrinological remission, cerebrospinal fluid leak, postoperative hyponatremia). Moreover, we briefly discuss from a practical standpoint the current barriers to clinical translation of machine learning research. On the topic of pituitary surgery, published reports can be considered mostly preliminary, requiring larger training populations and strong external validation. Thoughtful selection of clinically relevant outcomes of interest and transversal application of model development pipeline-together with accurate methodological planning and multicenter collaborations-have the potential to overcome current limitations and ultimately provide additional tools for more informed patient management.
Collapse
Affiliation(s)
- Vittorio Stumpo
- Machine Intelligence in Clinical Neuroscience (MICN) Laboratory, Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Victor E Staartjes
- Machine Intelligence in Clinical Neuroscience (MICN) Laboratory, Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Luca Regli
- Machine Intelligence in Clinical Neuroscience (MICN) Laboratory, Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Carlo Serra
- Machine Intelligence in Clinical Neuroscience (MICN) Laboratory, Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
Li Q, Zhu Y, Chen M, Guo R, Hu Q, Lu Y, Deng Z, Deng S, Zhang T, Wen H, Gao R, Nie Y, Li H, Chen J, Shi G, Shen J, Cheung WW, Liu Z, Guo Y, Chen Y. Development and Validation of a Deep Learning Algorithm to Automatic Detection of Pituitary Microadenoma From MRI. Front Med (Lausanne) 2021; 8:758690. [PMID: 34912820 PMCID: PMC8666533 DOI: 10.3389/fmed.2021.758690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/28/2021] [Indexed: 11/13/2022] Open
Abstract
Background: It is often difficult to diagnose pituitary microadenoma (PM) by MRI alone, due to its relatively small size, variable anatomical structure, complex clinical symptoms, and signs among individuals. We develop and validate a deep learning -based system to diagnose PM from MRI. Methods: A total of 11,935 infertility participants were initially recruited for this project. After applying the exclusion criteria, 1,520 participants (556 PM patients and 964 controls subjects) were included for further stratified into 3 non-overlapping cohorts. The data used for the training set were derived from a retrospective study, and in the validation dataset, prospective temporal and geographical validation set were adopted. A total of 780 participants were used for training, 195 participants for testing, and 545 participants were used to validate the diagnosis performance. The PM-computer-aided diagnosis (PM-CAD) system consists of two parts: pituitary region detection and PM diagnosis. The diagnosis performance of the PM-CAD system was measured using the receiver operating characteristics (ROC) curve and area under the ROC curve (AUC), calibration curve, accuracy, sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and F1-score. Results: Pituitary microadenoma-computer-aided diagnosis system showed 94.36% diagnostic accuracy and 98.13% AUC score in the testing dataset. We confirm the robustness and generalization of our PM-CAD system, the diagnostic accuracy in the internal dataset was 96.50% and in the external dataset was 92.26 and 92.36%, the AUC was 95.5, 94.7, and 93.7%, respectively. In human-computer competition, the diagnosis performance of our PM-CAD system was comparable to radiologists with >10 years of professional expertise (diagnosis accuracy of 94.0% vs. 95.0%, AUC of 95.6% vs. 95.0%). For the misdiagnosis cases from radiologists, our system showed a 100% accurate diagnosis. A browser-based software was designed to assist the PM diagnosis. Conclusions: This is the first report showing that the PM-CAD system is a viable tool for detecting PM. Our results suggest that the PM-CAD system is applicable to radiology departments, especially in primary health care institutions.
Collapse
Affiliation(s)
- Qingling Li
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of VIP Medical Service Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanhua Zhu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minglin Chen
- School of Electronics and Communication Engineering, Sun Yat-sen University, Guangzhou, China
| | - Ruomi Guo
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qingyong Hu
- Department of Computer Science, University of Oxford, Oxfordshire, United Kingdom
| | - Yaxin Lu
- Department of Medical Artificial Intelligence Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhenghui Deng
- School of Electronics and Communication Engineering, Sun Yat-sen University, Guangzhou, China
| | - Songqing Deng
- School of Electronics and Communication Engineering, Sun Yat-sen University, Guangzhou, China
| | - Tiecheng Zhang
- Department of Magnetic Resonance, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Huiquan Wen
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rong Gao
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuanpeng Nie
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haicheng Li
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianning Chen
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guojun Shi
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Shen
- Department of Radiology, The Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wai Wilson Cheung
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
| | - Zifeng Liu
- Department of Medical Artificial Intelligence Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yulan Guo
- School of Electronics and Communication Engineering, Sun Yat-sen University, Guangzhou, China
| | - Yanming Chen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
38
|
Application of artificial intelligence and radiomics in pituitary neuroendocrine and sellar tumors: a quantitative and qualitative synthesis. Neuroradiology 2021; 64:647-668. [PMID: 34839380 DOI: 10.1007/s00234-021-02845-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/21/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE To systematically review the literature regarding the application of machine learning (ML) of magnetic resonance imaging (MRI) radiomics in common sellar tumors. To identify future directions for application of ML in sellar tumor MRI. METHODS PubMed, Medline, Embase, Google Scholar, Scopus, ArxIV, and bioRxiv were searched to identify relevant studies published between 2010 and September 2021. Studies were included if they specifically involved ML of MRI radiomics in the analysis of sellar masses. Risk of bias assessment was performed using the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) Tool. RESULTS Fifty-eight articles were identified for review. All papers utilized retrospective data, and a quantitative systematic review was performed for thirty-one studies utilizing a public dataset which compared pituitary adenomas, meningiomas, and gliomas. One of the analyzed architectures yielded the highest classification accuracy of 0.996. The remaining twenty-seven articles were qualitatively reviewed and showed promising findings in predicting specific tumor characteristics such as tumor consistency, Ki-67 proliferative index, and post-surgical recurrence. CONCLUSION This review highlights the potential clinical application of ML using MRI radiomic data of the sellar region in diagnosis and predicting treatment outcomes. We describe future directions for practical application in the clinical care of patients with pituitary neuroendocrine and other sellar tumors.
Collapse
|
39
|
Rui W, Qiao N, Wu Y, Zhang Y, Aili A, Zhang Z, Ye H, Wang Y, Zhao Y, Yao Z. Radiomics analysis allows for precise prediction of silent corticotroph adenoma among non-functioning pituitary adenomas. Eur Radiol 2021; 32:1570-1578. [PMID: 34837512 DOI: 10.1007/s00330-021-08361-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 09/01/2021] [Accepted: 09/25/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To predict silent corticotroph adenomas (SCAs) among non-functioning pituitary adenomas preoperatively using noninvasive radiomics. METHODS A total of 302 patients including 146 patients diagnosed with SCAs and 156 patients with non-SCAs were enrolled (training set: n = 242; test set: n = 60). Tumor segmentation was manually generated using ITK-SNAP. From T1-weighted imaging (T1WI), T2-weighted imaging (T2WI), and contrast-enhanced T1WI, 2550 radiomics features were extracted using Pyradiomics. Pearson's correlation coefficient values were calculated to exclude redundant features. Several machine learning algorithms were developed to predict SCAs incorporating the radiomics and semantic features including clinical, laboratory, and radiology-associated features. The performance of models was evaluated by AUC. RESULTS Patients in the SCA group were younger (49.5 vs 55.2 years old) and more female (85.6% vs 37.2%) than those in the non-SCA group (p < 0.001). More invasiveness (p = 0.011) and cystic and microcystic change (p < 0.001) were observed in patients with SCAs. The ensemble algorithm presented the largest AUC of 0.927 among all the algorithms trained in the test set, and the accuracy, specificity, and sensitivity of predicting SCAs were all 0.867 (at cut-off 0.5). The overall model performed better than that only using semantic features available in the clinic. Radiomics prediction was the most important feature, with gender ranking second and age ranking third. Radiomics features on T2WI were superior to those on other MR modalities in SCA prediction. CONCLUSION Our ensemble learning model outperformed current clinical practice in differentiating patients with SCAs and non-SCAs using radiomics, which might help make appropriate treatment strategies. KEY POINTS • Radiomics might improve the preoperative diagnosis of SCAs by MR images. • T2WI was superior to T1WI and CE-T1WI in the preoperative diagnosis of SCAs. • The ensemble machine learning model outperformed current clinical practice in SCAs diagnosis and treatment decision-making could be more individualised using the nomogram.
Collapse
Affiliation(s)
- Wenting Rui
- Department of Radiology, Huashan Hospital, Fudan University, Mid Wulumuqi Road, Shanghai, 200040, People's Republic of China
| | - Nidan Qiao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Mid Wulumuqi Road, Shanghai, 200040, People's Republic of China.,Neurosurgical Institute of Fudan University, Shanghai, People's Republic of China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, People's Republic of China.,National Center for Neurological Disorders, Shanghai, People's Republic of China
| | - Yue Wu
- Department of Radiology, Huashan Hospital, Fudan University, Mid Wulumuqi Road, Shanghai, 200040, People's Republic of China
| | - Yong Zhang
- GE Healthcare, MR Research, Huatuo Road, Shanghai, 201203, People's Republic of China
| | - Ababikere Aili
- Department of Radiology, Kuqa County People's Hospital, Aksu, 842000, Xinjiang, People's Republic of China
| | - Zhaoyun Zhang
- Department of Endocrinology, Huashan Hospital, Fudan University, Mid Wulumuqi Road, Shanghai, 200040, People's Republic of China
| | - Hongying Ye
- Department of Endocrinology, Huashan Hospital, Fudan University, Mid Wulumuqi Road, Shanghai, 200040, People's Republic of China
| | - Yongfei Wang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Mid Wulumuqi Road, Shanghai, 200040, People's Republic of China.,Neurosurgical Institute of Fudan University, Shanghai, People's Republic of China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, People's Republic of China.,National Center for Neurological Disorders, Shanghai, People's Republic of China
| | - Yao Zhao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Mid Wulumuqi Road, Shanghai, 200040, People's Republic of China. .,Neurosurgical Institute of Fudan University, Shanghai, People's Republic of China. .,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, People's Republic of China. .,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, People's Republic of China. .,National Center for Neurological Disorders, Shanghai, People's Republic of China.
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Mid Wulumuqi Road, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
40
|
Ng S, Messerer M, Engelhardt J, Bruneau M, Cornelius JF, Cavallo LM, Cossu G, Froelich S, Meling TR, Paraskevopoulos D, Schroeder HWS, Tatagiba M, Zazpe I, Berhouma M, Daniel RT, Laws ER, Knosp E, Buchfelder M, Dufour H, Gaillard S, Jacquesson T, Jouanneau E. Aggressive pituitary neuroendocrine tumors: current practices, controversies, and perspectives, on behalf of the EANS skull base section. Acta Neurochir (Wien) 2021; 163:3131-3142. [PMID: 34365544 DOI: 10.1007/s00701-021-04953-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/25/2021] [Indexed: 12/14/2022]
Abstract
Aggressive pituitary neuroendocrine tumors (APT) account for 10% of pituitary tumors. Their management is a rapidly evolving field of clinical research and has led pituitary teams to shift toward a neuro-oncological-like approach. The new terminology "Pituitary neuroendocrine tumors" (PitNet) that was recently proposed to replace "pituitary adenomas" reflects this change of paradigm. In this narrative review, we aim to provide a state of the art of actual knowledge, controversies, and recommendations in the management of APT. We propose an overview of current prognostic markers, including the recent five-tiered clinicopathological classification. We further establish and discuss the following recommendations from a neurosurgical perspective: (i) surgery and multi-staged surgeries (without or with parasellar resection in symptomatic patients) should be discussed at each stage of the disease, because it may potentialize adjuvant medical therapies; (ii) temozolomide is effective in most patients, although 30% of patients are non-responders and the optimal timeline to initiate and interrupt this treatment remains questionable; (iii) some patients with selected clinicopathological profiles may benefit from an earlier local radiotherapy and/or chemotherapy; (iv) novel therapies such as VEGF-targeted therapies and anti-CTLA-4/anti-PD1 immunotherapies are promising and should be discussed as 2nd or 3rd line of treatment. Finally, whether neurosurgeons have to operate on "pituitary adenomas" or "PitNets," their role and expertise remain crucial at each stage of the disease, prompting our community to deal with evolving concepts and therapeutic resources.
Collapse
|
41
|
Machine Learning: Applications and Advanced Progresses of Radiomics in Endocrine Neoplasms. JOURNAL OF ONCOLOGY 2021; 2021:8615450. [PMID: 34671399 PMCID: PMC8523238 DOI: 10.1155/2021/8615450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/13/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022]
Abstract
Endocrine neoplasms remain a great threat to human health. It is extremely important to make a clear diagnosis and timely treatment of endocrine tumors. Machine learning includes radiomics, which has long been utilized in clinical cancer research. Radiomics refers to the extraction of valuable information by analyzing a large amount of standard data with high-throughput medical images mainly including computed tomography, positron emission tomography, magnetic resonance imaging, and ultrasound. With the quantitative imaging analysis and model building, radiomics can reflect specific underlying characteristics of a disease that otherwise could not be evaluated visually. More and more promising results of radiomics in oncological practice have been seen in recent years. Radiomics may have the potential to supplement traditional imaging analysis and assist in providing precision medicine for patients. Radiomics had developed rapidly in endocrine neoplasms practice in the past decade. In this review, we would introduce the general workflow of radiomics and summarize the applications and developments of radiomics in endocrine neoplasms in recent years. The limitations of current radiomic research studies and future development directions would also be discussed.
Collapse
|
42
|
Abdel Razek AAK, Alksas A, Shehata M, AbdelKhalek A, Abdel Baky K, El-Baz A, Helmy E. Clinical applications of artificial intelligence and radiomics in neuro-oncology imaging. Insights Imaging 2021; 12:152. [PMID: 34676470 PMCID: PMC8531173 DOI: 10.1186/s13244-021-01102-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/26/2021] [Indexed: 12/15/2022] Open
Abstract
This article is a comprehensive review of the basic background, technique, and clinical applications of artificial intelligence (AI) and radiomics in the field of neuro-oncology. A variety of AI and radiomics utilized conventional and advanced techniques to differentiate brain tumors from non-neoplastic lesions such as inflammatory and demyelinating brain lesions. It is used in the diagnosis of gliomas and discrimination of gliomas from lymphomas and metastasis. Also, semiautomated and automated tumor segmentation has been developed for radiotherapy planning and follow-up. It has a role in the grading, prediction of treatment response, and prognosis of gliomas. Radiogenomics allowed the connection of the imaging phenotype of the tumor to its molecular environment. In addition, AI is applied for the assessment of extra-axial brain tumors and pediatric tumors with high performance in tumor detection, classification, and stratification of patient's prognoses.
Collapse
Affiliation(s)
| | - Ahmed Alksas
- Biomaging Lab, Department of Bioengineering, University of Louisville, Louisville, KY, 40292, USA
| | - Mohamed Shehata
- Biomaging Lab, Department of Bioengineering, University of Louisville, Louisville, KY, 40292, USA
| | - Amr AbdelKhalek
- Internship at Mansoura University Hospital, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Khaled Abdel Baky
- Department of Diagnostic Radiology, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Ayman El-Baz
- Biomaging Lab, Department of Bioengineering, University of Louisville, Louisville, KY, 40292, USA
| | - Eman Helmy
- Department of Diagnostic Radiology, Faculty of Medicine, Mansoura University, Elgomheryia Street, Mansoura, 3512, Egypt.
| |
Collapse
|
43
|
Yi Z, Long L, Zeng Y, Liu Z. Current Advances and Challenges in Radiomics of Brain Tumors. Front Oncol 2021; 11:732196. [PMID: 34722274 PMCID: PMC8551958 DOI: 10.3389/fonc.2021.732196] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Imaging diagnosis is crucial for early detection and monitoring of brain tumors. Radiomics enable the extraction of a large mass of quantitative features from complex clinical imaging arrays, and then transform them into high-dimensional data which can subsequently be mined to find their relevance with the tumor's histological features, which reflect underlying genetic mutations and malignancy, along with grade, progression, therapeutic effect, or even overall survival (OS). Compared to traditional brain imaging, radiomics provides quantitative information linked to meaningful biologic characteristics and application of deep learning which sheds light on the full automation of imaging diagnosis. Recent studies have shown that radiomics' application is broad in identifying primary tumor, differential diagnosis, grading, evaluation of mutation status and aggression, prediction of treatment response and recurrence in pituitary tumors, gliomas, and brain metastases. In this descriptive review, besides establishing a general understanding among protocols, results, and clinical significance of these studies, we further discuss the current limitations along with future development of radiomics.
Collapse
Affiliation(s)
- Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- XiangYa School of Medicine, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lifu Long
- XiangYa School of Medicine, Central South University, Changsha, China
| | - Yu Zeng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
44
|
Gitto S, Cuocolo R, Emili I, Tofanelli L, Chianca V, Albano D, Messina C, Imbriaco M, Sconfienza LM. Effects of Interobserver Variability on 2D and 3D CT- and MRI-Based Texture Feature Reproducibility of Cartilaginous Bone Tumors. J Digit Imaging 2021; 34:820-832. [PMID: 34405298 PMCID: PMC8455795 DOI: 10.1007/s10278-021-00498-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 05/27/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
This study aims to investigate the influence of interobserver manual segmentation variability on the reproducibility of 2D and 3D unenhanced computed tomography (CT)- and magnetic resonance imaging (MRI)-based texture analysis. Thirty patients with cartilaginous bone tumors (10 enchondromas, 10 atypical cartilaginous tumors, 10 chondrosarcomas) were retrospectively included. Three radiologists independently performed manual contour-focused segmentation on unenhanced CT and T1-weighted and T2-weighted MRI by drawing both a 2D region of interest (ROI) on the slice showing the largest tumor area and a 3D ROI including the whole tumor volume. Additionally, a marginal erosion was applied to both 2D and 3D segmentations to evaluate the influence of segmentation margins. A total of 783 and 1132 features were extracted from original and filtered 2D and 3D images, respectively. Intraclass correlation coefficient ≥ 0.75 defined feature stability. In 2D vs. 3D contour-focused segmentation, the rates of stable features were 74.71% vs. 86.57% (p < 0.001), 77.14% vs. 80.04% (p = 0.142), and 95.66% vs. 94.97% (p = 0.554) for CT and T1-weighted and T2-weighted images, respectively. Margin shrinkage did not improve 2D (p = 0.343) and performed worse than 3D (p < 0.001) contour-focused segmentation in terms of feature stability. In 2D vs. 3D contour-focused segmentation, matching stable features derived from CT and MRI were 65.8% vs. 68.7% (p = 0.191), and those derived from T1-weighted and T2-weighted images were 76.0% vs. 78.2% (p = 0.285). 2D and 3D radiomic features of cartilaginous bone tumors extracted from unenhanced CT and MRI are reproducible, although some degree of interobserver segmentation variability highlights the need for reliability analysis in future studies.
Collapse
Affiliation(s)
- Salvatore Gitto
- Dipartimento Di Scienze Biomediche Per La Salute, Università Degli Studi Di Milano, Via Luigi Mangiagalli 31, 20133, Milan, Italy.
| | - Renato Cuocolo
- Dipartimento Di Medicina Clinica E Chirurgia, Università Degli Studi Di Napoli "Federico II", Naples, Italy.,Laboratory of Augmented Reality for Health Monitoring (ARHeMLab), Dipartimento Di Ingegneria Elettrica E Delle Tecnologie Dell'Informazione, Università Degli Studi Di Napoli "Federico II", Naples, Italy
| | - Ilaria Emili
- Unità di Radiodiagnostica, Presidio CTO, ASST Pini-CTO, Milan, Italy
| | - Laura Tofanelli
- Dipartimento di Radiologia Diagnostica ed Interventistica, Università degli Studi di Milano, Ospedale San Paolo, Milan, Italy
| | - Vito Chianca
- Ospedale Evangelico Betania, Naples, Italy.,Clinica Di Radiologia, Istituto Imaging Della Svizzera Italiana - Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Domenico Albano
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Sezione Di Scienze Radiologiche, Dipartimento Di Biomedicina, Neuroscienze E Diagnostica Avanzata, Università Degli Studi Di Palermo, Palermo, Italy
| | | | - Massimo Imbriaco
- Dipartimento Di Scienze Biomediche Avanzate, Università Degli Studi Di Napoli "Federico II", Naples, Italy
| | - Luca Maria Sconfienza
- Dipartimento Di Scienze Biomediche Per La Salute, Università Degli Studi Di Milano, Via Luigi Mangiagalli 31, 20133, Milan, Italy.,IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|
45
|
Johns H, Bernhardt J, Churilov L. Distance-based Classification and Regression Trees for the analysis of complex predictors in health and medical research. Stat Methods Med Res 2021; 30:2085-2104. [PMID: 34319834 DOI: 10.1177/09622802211032712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Predicting patient outcomes based on patient characteristics and care processes is a common task in medical research. Such predictive features are often multifaceted and complex, and are usually simplified into one or more scalar variables to facilitate statistical analysis. This process, while necessary, results in a loss of important clinical detail. While this loss may be prevented by using distance-based predictive methods which better represent complex healthcare features, the statistical literature on such methods is limited, and the range of tools facilitating distance-based analysis is substantially smaller than those of other methods. Consequently, medical researchers must choose to either reduce complex predictive features to scalar variables to facilitate analysis, or instead use a limited number of distance-based predictive methods which may not fulfil the needs of the analysis problem at hand. We address this limitation by developing a Distance-Based extension of Classification and Regression Trees (DB-CART) capable of making distance-based predictions of categorical, ordinal and numeric patient outcomes. We also demonstrate how this extension is compatible with other extensions to CART, including a recently published method for predicting care trajectories in chronic disease. We demonstrate DB-CART by using it to expand upon previously published dose-response analysis of stroke rehabilitation data. Our method identified additional detail not captured by the previously published analysis, reinforcing previous conclusions. We also demonstrate how by combining DB-CART with other extensions to CART, the method is capable of making predictions about complex, multifaceted outcome data based on complex, multifaceted predictive features.
Collapse
Affiliation(s)
- Hannah Johns
- Center for Research Excellence in Stroke Rehabilitation and Brain Recovery, Heidelberg, VIC, Australia.,Florey Institute of Neuroscience and Mental Health, Heidelberg, VIC, Australia.,Melbourne Medical School, University of Melbourne, Parkville, VIC, Australia
| | - Julie Bernhardt
- Center for Research Excellence in Stroke Rehabilitation and Brain Recovery, Heidelberg, VIC, Australia.,Florey Institute of Neuroscience and Mental Health, Heidelberg, VIC, Australia
| | - Leonid Churilov
- Florey Institute of Neuroscience and Mental Health, Heidelberg, VIC, Australia.,Melbourne Medical School, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
46
|
Park YW, Eom J, Kim S, Kim H, Ahn SS, Ku CR, Kim EH, Lee EJ, Kim SH, Lee SK. Radiomics With Ensemble Machine Learning Predicts Dopamine Agonist Response in Patients With Prolactinoma. J Clin Endocrinol Metab 2021; 106:e3069-e3077. [PMID: 33713414 DOI: 10.1210/clinem/dgab159] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Indexed: 11/19/2022]
Abstract
CONTEXT Early identification of the response of prolactinoma patients to dopamine agonists (DA) is crucial in treatment planning. OBJECTIVE To develop a radiomics model using an ensemble machine learning classifier with conventional magnetic resonance images (MRIs) to predict the DA response in prolactinoma patients. DESIGN Retrospective study. SETTING Severance Hospital, Seoul, Korea. PATIENTS A total of 177 prolactinoma patients who underwent baseline MRI (109 DA responders and 68 DA nonresponders) were allocated to the training (n = 141) and test (n = 36) sets. Radiomic features (n = 107) were extracted from coronal T2-weighed MRIs. After feature selection, single models (random forest, light gradient boosting machine, extra-trees, quadratic discrimination analysis, and linear discrimination analysis) with oversampling methods were trained to predict the DA response. A soft voting ensemble classifier was used to achieve the final performance. The performance of the classifier was validated in the test set. RESULTS The ensemble classifier showed an area under the curve (AUC) of 0.81 [95% confidence interval (CI), 0.74-0.87] in the training set. In the test set, the ensemble classifier showed an AUC, accuracy, sensitivity, and specificity of 0.81 (95% CI, 0.67-0.96), 77.8%, 78.6%, and 77.3%, respectively. The ensemble classifier achieved the highest performance among all the individual models in the test set. CONCLUSIONS Radiomic features may be useful biomarkers to predict the DA response in prolactinoma patients.
Collapse
Affiliation(s)
- Yae Won Park
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
- Pituitary Tumor Center, Severance Hospital, Seoul, Korea
| | - Jihwan Eom
- Department of Computer Science, Yonsei University, Seoul, Korea
| | - Sooyon Kim
- Department of Statistics and Data Science, Yonsei University, Seoul, Korea
| | - Hwiyoung Kim
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Soo Ahn
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
- Pituitary Tumor Center, Severance Hospital, Seoul, Korea
| | - Cheol Ryong Ku
- Pituitary Tumor Center, Severance Hospital, Seoul, Korea
- Department of Endocrinology, Yonsei University College of Medicine, Seoul, Korea
| | - Eui Hyun Kim
- Pituitary Tumor Center, Severance Hospital, Seoul, Korea
- Department of Endocrinology, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Jig Lee
- Pituitary Tumor Center, Severance Hospital, Seoul, Korea
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Sun Ho Kim
- Department of Neurosurgery, Ewha Womans University College of Medicine, Seoul, Korea
| | - Seung-Koo Lee
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Korea
- Pituitary Tumor Center, Severance Hospital, Seoul, Korea
| |
Collapse
|
47
|
La Greca Saint-Esteven A, Vuong D, Tschanz F, van Timmeren JE, Dal Bello R, Waller V, Pruschy M, Guckenberger M, Tanadini-Lang S. Systematic Review on the Association of Radiomics with Tumor Biological Endpoints. Cancers (Basel) 2021; 13:cancers13123015. [PMID: 34208595 PMCID: PMC8234501 DOI: 10.3390/cancers13123015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022] Open
Abstract
Radiomics supposes an alternative non-invasive tumor characterization tool, which has experienced increased interest with the advent of more powerful computers and more sophisticated machine learning algorithms. Nonetheless, the incorporation of radiomics in cancer clinical-decision support systems still necessitates a thorough analysis of its relationship with tumor biology. Herein, we present a systematic review focusing on the clinical evidence of radiomics as a surrogate method for tumor molecular profile characterization. An extensive literature review was conducted in PubMed, including papers on radiomics and a selected set of clinically relevant and commonly used tumor molecular markers. We summarized our findings based on different cancer entities, additionally evaluating the effect of different modalities for the prediction of biomarkers at each tumor site. Results suggest the existence of an association between the studied biomarkers and radiomics from different modalities and different tumor sites, even though a larger number of multi-center studies are required to further validate the reported outcomes.
Collapse
Affiliation(s)
- Agustina La Greca Saint-Esteven
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
- Correspondence:
| | - Diem Vuong
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Fabienne Tschanz
- Laboratory of Applied Radiobiology, Department of Radiation Oncology, University of Zurich, 8091 Zurich, Switzerland; (F.T.); (V.W.); (M.P.)
| | - Janita E. van Timmeren
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Riccardo Dal Bello
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Verena Waller
- Laboratory of Applied Radiobiology, Department of Radiation Oncology, University of Zurich, 8091 Zurich, Switzerland; (F.T.); (V.W.); (M.P.)
| | - Martin Pruschy
- Laboratory of Applied Radiobiology, Department of Radiation Oncology, University of Zurich, 8091 Zurich, Switzerland; (F.T.); (V.W.); (M.P.)
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| |
Collapse
|
48
|
Cai X, Zhu J, Yang J, Tang C, Yuan F, Cong Z, Ma C. A Nomogram for Preoperatively Predicting the Ki-67 Index of a Pituitary Tumor: A Retrospective Cohort Study. Front Oncol 2021; 11:687333. [PMID: 34136412 PMCID: PMC8200848 DOI: 10.3389/fonc.2021.687333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/10/2021] [Indexed: 12/23/2022] Open
Abstract
Background The Ki-67 index is an indicator of proliferation and aggressive behavior in pituitary adenomas (PAs). This study aims to develop and validate a predictive nomogram for forecasting Ki-67 index levels preoperatively in PAs. Methods A total of 439 patients with PAs underwent PA resection at the Department of Neurosurgery in Jinling Hospital between January 2018 and October 2020; they were enrolled in this retrospective study and were classified randomly into a training cohort (n = 300) and a validation cohort (n = 139). A range of clinical, radiological, and laboratory characteristics were collected. The Ki-67 index was classified into the low Ki-67 index (<3%) and the high Ki-67 index (≥3%). Least absolute shrinkage and selection operator algorithm and uni- and multivariate logistic regression analyses were applied to identify independent risk factors associated with Ki-67. A nomogram was constructed to visualize these risk factors. The receiver operation characteristic curve and calibration curve were computed to evaluate the predictive performance of the nomogram model. Results Age, primary-recurrence subtype, maximum dimension, and prolactin were included in the nomogram model. The areas under the curve (AUCs) of the nomogram model were 0.694 in the training cohort and 0.658 in the validation cohort. A well-fitted calibration curve was also generated for the nomogram model. A subgroup analysis revealed stable predictive performance for the nomogram model. A correlation analysis revealed that age (R = −0.23; p < 0.01), maximum dimension (R = 0.17; p < 0.01), and prolactin (R = 0.16; p < 0.01) were all significantly correlated with the Ki-67 index level. Conclusions Age, primary-recurrence subtype, maximum dimension, and prolactin are independent predictors for the Ki-67 index level. The current study provides a novel and feasible nomogram, which can further assist neurosurgeons to develop better, more individualized treatment strategies for patients with PAs by predicting the Ki-67 index level preoperatively.
Collapse
Affiliation(s)
- Xiangming Cai
- School of Medicine, Southeast University, Nanjing, China
| | - Junhao Zhu
- School of Medicine, Nanjing Medical University, Nanjing, China
| | - Jin Yang
- Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Chao Tang
- Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Feng Yuan
- Department of Neurosurgery, Jinling Hospital, Nanjing, China.,School of Medicine, Nanjing University, Nanjing, China
| | - Zixiang Cong
- Department of Neurosurgery, Jinling Hospital, Nanjing, China.,School of Medicine, Nanjing University, Nanjing, China
| | - Chiyuan Ma
- School of Medicine, Southeast University, Nanjing, China.,School of Medicine, Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Jinling Hospital, Nanjing, China.,School of Medicine, Nanjing University, Nanjing, China.,School of Nanjing Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
49
|
Deep Myometrial Infiltration of Endometrial Cancer on MRI: A Radiomics-Powered Machine Learning Pilot Study. Acad Radiol 2021; 28:737-744. [PMID: 32229081 DOI: 10.1016/j.acra.2020.02.028] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 02/08/2023]
Abstract
RATIONALE AND OBJECTIVES To evaluate an MRI radiomics-powered machine learning (ML) model's performance for the identification of deep myometrial invasion (DMI) in endometrial cancer (EC) patients and explore its clinical applicability. MATERIALS AND METHODS Preoperative MRI scans of EC patients were retrospectively selected. Three radiologists performed whole-lesion segmentation on T2-weighted images for feature extraction. Feature robustness was tested before randomly splitting the population in training and test sets (80/20% proportion). A multistep feature selection was applied to the first, excluding noninformative, low variance features and redundant, highly-intercorrelated ones. A Random Forest wrapper was used to identify the most informative among the remaining. An ensemble of J48 decision trees was tuned and finalized in the training set using 10-fold cross-validation, and then assessed on the test set. A radiologist evaluated all MRI scans without and with the aid of ML to detect the presence of DMI. McNemars's test was employed to compare the two readings. RESULTS Of the 54 patients included, 17 had DMI. In all, 1132 features were extracted. After feature selection, the Random Forest wrapper identified the three most informative which were used for ML training. The classifier reached an accuracy of 86% and 91% and areas under the Receiver Operating Characteristic curve of 0.92 and 0.94 in the cross-validation and final testing, respectively. The radiologist performance increased from 82% to 100% when using ML (p = 0.48). CONCLUSION We proved the feasibility of a radiomics-powered ML model for DMI detection on MR T2-w images that might help radiologists to increase their performance.
Collapse
|
50
|
Artificial intelligence applications in medical imaging: A review of the medical physics research in Italy. Phys Med 2021; 83:221-241. [DOI: 10.1016/j.ejmp.2021.04.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 02/06/2023] Open
|