1
|
Wang X, Song J, Qiu Q, Su Y, Wang L, Cao X. A Stacked Multimodality Model Based on Functional MRI Features and Deep Learning Radiomics for Predicting the Early Response to Radiotherapy in Nasopharyngeal Carcinoma. Acad Radiol 2024:S1076-6332(24)00771-2. [PMID: 39496536 DOI: 10.1016/j.acra.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND This study aimed to construct and assess a comprehensive model that integrates MRI-derived deep learning radiomics, functional imaging (fMRI), and clinical indicators to predict early efficacy of radiotherapy in nasopharyngeal carcinoma (NPC). METHODS This retrospective study recruited NPC patients with radiotherapy from two Chinese hospitals between October 2018 and July 2022, divided into a training set (hospital I, 194 cases), an internal validation set (hospital I, 82 cases), and an external validation set (hospital II, 40 cases). We extracted 3404 radiomic features and 2048 deep learning features from multi-sequence MRI includes T1WI, CE-T1WI, T2WI and T2WI/FS. Additionally, both the Apparent diffusion coefficient (ADC), its maximum (ADCmax) and Tumor blood flow (TBF), its maximum (TBFmax) were obtained by Diffusion-weighted imaging (DWI) and Arterial spin labeling (ASL) respectively. We used four classifiers (LR, XGBoost, SVM and KNN) and stacked algorithm as model construction methods. The area under the receiver operating characteristic curve (AUC) and decision curve analysis was used to assess models. RESULTS The manual radiomics model based on XGBoost and the deep learning model based on KNN (the AUCs in the training set: 0.909, 0.823, respectively) showed better predictive efficacy than other machine learning algorithms. The stacked model that integrated MRI-based deep learning radiomics, fMRI, and hematological indicators, has the strongest efficacy prediction ability of AUC in the training set [0.984 (95%CI: 0.972-0.996)], the internal validation set [0.936 (95%CI: 0.885-0.987)], and the external validation set [0.959 (95%CI: 0.901-1.000)]. CONCLUSION Our research has developed a clinical-radiomics integrated model based on MRI which can predict early radiotherapy response in NPC and provide guidance for personalized treatment.
Collapse
Affiliation(s)
- Xiaowen Wang
- Shandong University Cancer Center, Jinan, Shandong, China (X.W.); Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (X.W., X.C.)
| | - Jian Song
- Medical Imageology, Shandong Medical College, Jinan, China (J.S.)
| | - Qingtao Qiu
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Q.Q., Y.S., L.W.)
| | - Ya Su
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Q.Q., Y.S., L.W.)
| | - Lizhen Wang
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Q.Q., Y.S., L.W.)
| | - Xiujuan Cao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (X.W., X.C.).
| |
Collapse
|
2
|
Kwon J, Kim J, Park H. Leveraging segmentation-guided spatial feature embedding for overall survival prediction in glioblastoma with multimodal magnetic resonance imaging. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 255:108338. [PMID: 39042996 DOI: 10.1016/j.cmpb.2024.108338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND AND OBJECTIVE Patients with glioblastoma have a five-year relative survival rate of less than 5 %. Thus, accurately predicting the overall survival (OS) of patients with glioblastoma is crucial for effective treatment planning. METHODS To fully leverage the imaging characteristics of glioblastomas, we propose a segmentation-guided regression method for predicting OS of patients with brain tumors using multimodal magnetic resonance imaging. Specifically, a brain tumor segmentation network was first pre-trained without leveraging survival information. Subsequently, the survival regression network was jointly trained with the guidance of brain tumor segmentation, focusing on tumor voxels and suppressing irrelevant backgrounds. RESULTS Our proposed framework, based on the well-known backbone of UNETR++, achieved a Dice score of 0.7910, Spearman correlation of 0.4112, and Harrell's concordance index of 0.6488. The model consistently showed promising results compared with baseline methods on two different datasets (BraTS and UCSF-PDGM). Furthermore, ablation studies on our training configurations demonstrated that both the pre-training segmentation network and contrastive loss significantly improved all metrics for OS prediction. CONCLUSIONS In this study, we propose a joint learning framework based on a pre-trained segmentation backbone for OS prediction by leveraging a brain tumor segmentation map. By utilizing a spatial feature map, our model can operate using a sliding-window approach, which can be adopted by varying the matrix sizes and resolutions of the input images.
Collapse
Affiliation(s)
- Junmo Kwon
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, 16419, South Korea; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, 16419, South Korea
| | - Jonghun Kim
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Hyunjin Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, 16419, South Korea; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, 16419, South Korea.
| |
Collapse
|
3
|
Liang HX, Wang ZY, Li Y, Ren AN, Chen ZF, Wang XZ, Wang XM, Yuan ZG. The application value of support vector machine model based on multimodal MRI in predicting IDH-1mutation and Ki-67 expression in glioma. BMC Med Imaging 2024; 24:244. [PMID: 39285364 PMCID: PMC11403938 DOI: 10.1186/s12880-024-01414-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
PURPOSE To investigate the application value of support vector machine (SVM) model based on diffusion-weighted imaging (DWI), dynamic contrast-enhanced (DCE) and amide proton transfer- weighted (APTW) imaging in predicting isocitrate dehydrogenase 1(IDH-1) mutation and Ki-67 expression in glioma. METHODS The DWI, DCE and APTW images of 309 patients with glioma confirmed by pathology were retrospectively analyzed and divided into the IDH-1 group (IDH-1(+) group and IDH-1(-) group) and Ki-67 group (low expression group (Ki-67 ≤ 10%) and high expression group (Ki-67 > 10%)). All cases were divided into the training set, and validation set according to the ratio of 7:3. The training set was used to select features and establish machine learning models. The SVM model was established with the data after feature selection. Four single sequence models and one combined model were established in IDH-1 group and Ki-67 group. The receiver operator characteristic (ROC) curve was used to evaluate the diagnostic performance of the model. Validation set data was used for further validation. RESULTS Both in the IDH-1 group and Ki-67 group, the combined model had better predictive efficiency than single sequence model, although the single sequence model had a better predictive efficiency. In the Ki-67 group, the combined model was built from six selected radiomics features, and the AUC were 0.965 and 0.931 in the training and validation sets, respectively. In the IDH-1 group, the combined model was built from four selected radiomics features, and the AUC were 0.997 and 0.967 in the training and validation sets, respectively. CONCLUSION The radiomics model established by DWI, DCE and APTW images could be used to detect IDH-1 mutation and Ki-67 expression in glioma patients before surgery. The prediction performance of the radiomics model based on the combination sequence was better than that of the single sequence model.
Collapse
Affiliation(s)
- He-Xin Liang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Hospital Affiliated to Cheeloo College of Medicine of Shandong University, Jinan, China
- Cancer Hospital, Affiliated to Shandong First Medical University, Jinan, China
| | - Zong-Ying Wang
- Department of Radiology, Weifang People's Hospital, Weifang, China
| | - Yao Li
- Zhucheng People's Hospital, Weifang, China
| | - An-Ning Ren
- Medical Imaging Center of the Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zhi-Feng Chen
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
- Department of Data Science & AI, Faculty of IT, Monash University, Clayton, VIC, Australia
| | - Xi-Zhen Wang
- Medical Imaging Center of the Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xi-Ming Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Hospital Affiliated to Cheeloo College of Medicine of Shandong University, Jinan, China
| | - Zhen-Guo Yuan
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
- Shandong Provincial Hospital Affiliated to Cheeloo College of Medicine of Shandong University, Jinan, China.
| |
Collapse
|
4
|
Jiang C, Sun C, Wang X, Ma S, Jia W, Zhang D. BTK Expression Level Prediction and the High-Grade Glioma Prognosis Using Radiomic Machine Learning Models. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1359-1374. [PMID: 38381384 PMCID: PMC11300408 DOI: 10.1007/s10278-024-01026-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/22/2024]
Abstract
We aimed to study whether the Bruton's tyrosine kinase (BTK) expression is correlated with the prognosis of patients with high-grade gliomas (HGGs) and predict its expression level prior to surgery, by constructing radiomic models. Clinical and gene expression data of 310 patients from The Cancer Genome Atlas (TCGA) were included for gene-based prognostic analysis. Among them, contrast-enhanced T1-weighted imaging (T1WI + C) from The Cancer Imaging Archive (TCIA) with genomic data was selected from 82 patients for radiomic models, including support vector machine (SVM) and logistic regression (LR) models. Furthermore, the nomogram incorporating radiomic signatures was constructed to evaluate its clinical efficacy. BTK was identified as an independent risk factor for HGGs through univariate and multivariate Cox regression analyses. Three radiomic features were selected to construct the SVM and LR models, and the validation set showed area under curve (AUCs) values of 0.711 (95% CI, 0.598-0.824) and 0.736 (95% CI, 0.627-0.844), respectively. The median survival times of the high Rad_score and low-Rad_score groups based on LR model were 15.53 and 23.03 months, respectively. In addition, the total risk score of each patient was used to construct a predictive nomogram, and the AUCs calculated from the corresponding time-dependent ROC curves were 0.533, 0.659, and 0.767 for 1, 3, and 5 years, respectively. BTK is an independent risk factor associated with poor prognosis in patients, and the radiomic model constructed in this study can effectively and non-invasively predict preoperative BTK expression levels and patient prognosis based on T1WI + C.
Collapse
Affiliation(s)
- Chenggang Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Chen Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Xi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Dainan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China.
| |
Collapse
|
5
|
Joyce T, Tasci E, Jagasia S, Shephard J, Chappidi S, Zhuge Y, Zhang L, Cooley Zgela T, Sproull M, Mackey M, Camphausen K, Krauze AV. Serum CD133-Associated Proteins Identified by Machine Learning Are Connected to Neural Development, Cancer Pathways, and 12-Month Survival in Glioblastoma. Cancers (Basel) 2024; 16:2740. [PMID: 39123468 PMCID: PMC11311306 DOI: 10.3390/cancers16152740] [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: 06/17/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Glioma is the most prevalent type of primary central nervous system cancer, while glioblastoma (GBM) is its most aggressive variant, with a median survival of only 15 months when treated with maximal surgical resection followed by chemoradiation therapy (CRT). CD133 is a potentially significant GBM biomarker. However, current clinical biomarker studies rely on invasive tissue samples. These make prolonged data acquisition impossible, resulting in increased interest in the use of liquid biopsies. Our study, analyzed 7289 serum proteins from 109 patients with pathology-proven GBM obtained prior to CRT using the aptamer-based SOMAScan® proteomic assay technology. We developed a novel methodology that identified 24 proteins linked to both serum CD133 and 12-month overall survival (OS) through a multi-step machine learning (ML) analysis. These identified proteins were subsequently subjected to survival and clustering evaluations, categorizing patients into five risk groups that accurately predicted 12-month OS based on their protein profiles. Most of these proteins are involved in brain function, neural development, and/or cancer biology signaling, highlighting their significance and potential predictive value. Identifying these proteins provides a valuable foundation for future serum investigations as validation of clinically applicable GBM biomarkers can unlock immense potential for diagnostics and treatment monitoring.
Collapse
Affiliation(s)
- Thomas Joyce
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Erdal Tasci
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Sarisha Jagasia
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Jason Shephard
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Shreya Chappidi
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
- Department of Computer Science and Technology, University of Cambridge, 15 JJ Thomson Ave, Cambridge CB3 0FD, UK
| | - Ying Zhuge
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Longze Zhang
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Theresa Cooley Zgela
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Mary Sproull
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Megan Mackey
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Kevin Camphausen
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| | - Andra V. Krauze
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA; (T.J.); (S.J.); (J.S.); (S.C.); (Y.Z.); (L.Z.); (T.C.Z.); (M.S.); (M.M.); (K.C.)
| |
Collapse
|
6
|
Lorenzo G, Ahmed SR, Hormuth DA, Vaughn B, Kalpathy-Cramer J, Solorio L, Yankeelov TE, Gomez H. Patient-Specific, Mechanistic Models of Tumor Growth Incorporating Artificial Intelligence and Big Data. Annu Rev Biomed Eng 2024; 26:529-560. [PMID: 38594947 DOI: 10.1146/annurev-bioeng-081623-025834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Despite the remarkable advances in cancer diagnosis, treatment, and management over the past decade, malignant tumors remain a major public health problem. Further progress in combating cancer may be enabled by personalizing the delivery of therapies according to the predicted response for each individual patient. The design of personalized therapies requires the integration of patient-specific information with an appropriate mathematical model of tumor response. A fundamental barrier to realizing this paradigm is the current lack of a rigorous yet practical mathematical theory of tumor initiation, development, invasion, and response to therapy. We begin this review with an overview of different approaches to modeling tumor growth and treatment, including mechanistic as well as data-driven models based on big data and artificial intelligence. We then present illustrative examples of mathematical models manifesting their utility and discuss the limitations of stand-alone mechanistic and data-driven models. We then discuss the potential of mechanistic models for not only predicting but also optimizing response to therapy on a patient-specific basis. We describe current efforts and future possibilities to integrate mechanistic and data-driven models. We conclude by proposing five fundamental challenges that must be addressed to fully realize personalized care for cancer patients driven by computational models.
Collapse
Affiliation(s)
- Guillermo Lorenzo
- Oden Institute for Computational Engineering and Sciences, University of Texas, Austin, Texas, USA
- Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy
| | - Syed Rakin Ahmed
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
- Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Harvard Graduate Program in Biophysics, Harvard Medical School, Harvard University, Cambridge, Massachusetts, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David A Hormuth
- Livestrong Cancer Institutes, University of Texas, Austin, Texas, USA
- Oden Institute for Computational Engineering and Sciences, University of Texas, Austin, Texas, USA
| | - Brenna Vaughn
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| | | | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| | - Thomas E Yankeelov
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Biomedical Engineering, Department of Oncology, and Department of Diagnostic Medicine, University of Texas, Austin, Texas, USA
- Livestrong Cancer Institutes, University of Texas, Austin, Texas, USA
- Oden Institute for Computational Engineering and Sciences, University of Texas, Austin, Texas, USA
| | - Hector Gomez
- School of Mechanical Engineering and Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| |
Collapse
|
7
|
Gao J, Liu Z, Pan H, Cao X, Kan Y, Wen Z, Chen S, Wen M, Zhang L. Preoperative Discrimination of CDKN2A/B Homozygous Deletion Status in Isocitrate Dehydrogenase-Mutant Astrocytoma: A Deep Learning-Based Radiomics Model Using MRI. J Magn Reson Imaging 2024; 59:1655-1664. [PMID: 37555723 DOI: 10.1002/jmri.28945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B) homozygous deletion has been verified as an independent and critical biomarker of negative prognosis and short survival in isocitrate dehydrogenase (IDH)-mutant astrocytoma. Therefore, noninvasive and accurate discrimination of CDKN2A/B homozygous deletion status is essential for the clinical management of IDH-mutant astrocytoma patients. PURPOSE To develop a noninvasive, robust preoperative model based on MR image features for discriminating CDKN2A/B homozygous deletion status of IDH-mutant astrocytoma. STUDY TYPE Retrospective. POPULATION Two hundred fifty-one patients: 107 patients with CDKN2A/B homozygous deletion and 144 patients without CDKN2A/B homozygous deletion. FIELD STRENGTH/SEQUENCE 3.0 T/1.5 T: Contrast-enhanced T1-weighted spin-echo inversion recovery sequence (CE-T1WI) and T2-weighted fluid-attenuation spin-echo inversion recovery sequence (T2FLAIR). ASSESSMENT A total of 1106 radiomics and 1000 deep learning features extracted from CE-T1WI and T2FLAIR were used to develop models to discriminate the CDKN2A/B homozygous deletion status. Radiomics models, deep learning-based radiomics (DLR) models and the final integrated model combining radiomics features with deep learning features were developed and compared their preoperative discrimination performance. STATISTICAL TESTING Pearson chi-square test and Mann Whitney U test were used for assessing the statistical differences in patients' clinical characteristics. The Delong test compared the statistical differences of receiver operating characteristic (ROC) curves and area under the curve (AUC) of different models. The significance threshold is P < 0.05. RESULTS The final combined model (training AUC = 0.966; validation AUC = 0.935; test group: AUC = 0.943) outperformed the optimal models based on only radiomics or DLR features (training: AUC = 0.916 and 0.952; validation: AUC = 0.886 and 0.912; test group: AUC = 0.862 and 0.902). DATA CONCLUSION Whether based on a single sequence or a combination of two sequences, radiomics and DLR models have achieved promising performance in assessing CDKN2A/B homozygous deletion status. However, the final model combining both deep learning and radiomics features from CE-T1WI and T2FLAIR outperformed the optimal radiomics or DLR model. EVIDENCE LEVEL 4 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jueni Gao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi Liu
- Department of Nuclear Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Hongyu Pan
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Xu Cao
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yubo Kan
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhipeng Wen
- Department of Radiology, Sichuan Cancer Hospital, Chengdu, China
| | - Shanxiong Chen
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Ming Wen
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liqiang Zhang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Du Y, Guo W, Xiao Y, Chen H, Yao J, Wu J. Ultrasound-based deep learning radiomics model for differentiating benign, borderline, and malignant ovarian tumours: a multi-class classification exploratory study. BMC Med Imaging 2024; 24:89. [PMID: 38622546 PMCID: PMC11020982 DOI: 10.1186/s12880-024-01251-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 03/18/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Accurate preoperative identification of ovarian tumour subtypes is imperative for patients as it enables physicians to custom-tailor precise and individualized management strategies. So, we have developed an ultrasound (US)-based multiclass prediction algorithm for differentiating between benign, borderline, and malignant ovarian tumours. METHODS We randomised data from 849 patients with ovarian tumours into training and testing sets in a ratio of 8:2. The regions of interest on the US images were segmented and handcrafted radiomics features were extracted and screened. We applied the one-versus-rest method in multiclass classification. We inputted the best features into machine learning (ML) models and constructed a radiomic signature (Rad_Sig). US images of the maximum trimmed ovarian tumour sections were inputted into a pre-trained convolutional neural network (CNN) model. After internal enhancement and complex algorithms, each sample's predicted probability, known as the deep transfer learning signature (DTL_Sig), was generated. Clinical baseline data were analysed. Statistically significant clinical parameters and US semantic features in the training set were used to construct clinical signatures (Clinic_Sig). The prediction results of Rad_Sig, DTL_Sig, and Clinic_Sig for each sample were fused as new feature sets, to build the combined model, namely, the deep learning radiomic signature (DLR_Sig). We used the receiver operating characteristic (ROC) curve and the area under the ROC curve (AUC) to estimate the performance of the multiclass classification model. RESULTS The training set included 440 benign, 44 borderline, and 196 malignant ovarian tumours. The testing set included 109 benign, 11 borderline, and 49 malignant ovarian tumours. DLR_Sig three-class prediction model had the best overall and class-specific classification performance, with micro- and macro-average AUC of 0.90 and 0.84, respectively, on the testing set. Categories of identification AUC were 0.84, 0.85, and 0.83 for benign, borderline, and malignant ovarian tumours, respectively. In the confusion matrix, the classifier models of Clinic_Sig and Rad_Sig could not recognise borderline ovarian tumours. However, the proportions of borderline and malignant ovarian tumours identified by DLR_Sig were the highest at 54.55% and 63.27%, respectively. CONCLUSIONS The three-class prediction model of US-based DLR_Sig can discriminate between benign, borderline, and malignant ovarian tumours. Therefore, it may guide clinicians in determining the differential management of patients with ovarian tumours.
Collapse
Affiliation(s)
- Yangchun Du
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, 530021, Nanning, China
- Department of Ultrasound, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, No.6 Taoyuan Road, Qingxiu District, 530021, Nanning, China
| | - Wenwen Guo
- Department of Pathology, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, No.6 Taoyuan Road, Qingxiu District, 530021, Nanning, China
| | - Yanju Xiao
- Department of Ultrasound, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, No.6 Taoyuan Road, Qingxiu District, 530021, Nanning, China
| | - Haining Chen
- Department of Ultrasound, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, No.6 Taoyuan Road, Qingxiu District, 530021, Nanning, China
| | - Jinxiu Yao
- Department of Ultrasound, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, No.6 Taoyuan Road, Qingxiu District, 530021, Nanning, China
| | - Ji Wu
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, 530021, Nanning, China.
| |
Collapse
|
9
|
Fan H, Luo Y, Gu F, Tian B, Xiong Y, Wu G, Nie X, Yu J, Tong J, Liao X. Artificial intelligence-based MRI radiomics and radiogenomics in glioma. Cancer Imaging 2024; 24:36. [PMID: 38486342 PMCID: PMC10938723 DOI: 10.1186/s40644-024-00682-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/03/2024] [Indexed: 03/18/2024] Open
Abstract
The specific genetic subtypes that gliomas exhibit result in variable clinical courses and the need to involve multidisciplinary teams of neurologists, epileptologists, neurooncologists and neurosurgeons. Currently, the diagnosis of gliomas pivots mainly around the preliminary radiological findings and the subsequent definitive surgical diagnosis (via surgical sampling). Radiomics and radiogenomics present a potential to precisely diagnose and predict survival and treatment responses, via morphological, textural, and functional features derived from MRI data, as well as genomic data. In spite of their advantages, it is still lacking standardized processes of feature extraction and analysis methodology among different research groups, which have made external validations infeasible. Radiomics and radiogenomics can be used to better understand the genomic basis of gliomas, such as tumor spatial heterogeneity, treatment response, molecular classifications and tumor microenvironment immune infiltration. These novel techniques have also been used to predict histological features, grade or even overall survival in gliomas. In this review, workflows of radiomics and radiogenomics are elucidated, with recent research on machine learning or artificial intelligence in glioma.
Collapse
Affiliation(s)
- Haiqing Fan
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Yilin Luo
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Fang Gu
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Bin Tian
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Yongqin Xiong
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Guipeng Wu
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Xin Nie
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Jing Yu
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Juan Tong
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China
| | - Xin Liao
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, 550000, Guizhou, Guiyang, China.
| |
Collapse
|
10
|
Zhang H, Zhou B, Zhang H, Zhang Y, Lei Y, Huang B. Peritumoral Radiomics for Identification of Telomerase Reverse Transcriptase Promoter Mutation in Patients With Glioblastoma Based on Preoperative MRI. Can Assoc Radiol J 2024; 75:143-152. [PMID: 37552107 DOI: 10.1177/08465371231183309] [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: 08/09/2023] Open
Abstract
Purpose: To evaluate the value of intra- and peritumoral deep learning (DL) features based on multi-parametric magnetic resonance imaging (MRI) for identifying telomerase reverse transcriptase (TERT) promoter mutation in glioblastoma (GBM). Methods: In this study, we included 229 patients with GBM who underwent preoperative MRI in two hospitals between November 2016 and September 2022. We used four 2D Convolutional Neural Networks (GoogLeNet, DenseNet121, VGG16, and MobileNetV3-Large) to extract intra- and peritumoral DL features. The Mann-Whitney U test, Pearson correlation analysis, least absolute shrinkage and selection operator, and logistic regression analysis were used for feature selection and construction of DL radiomics (DLR) signatures in different regions. These multi-parametric and multi-region signatures were combined to identify TERT promoter mutation. The area under the receiver operating characteristic curve (AUC) was used to evaluate the effects of the signatures. Results: The signatures based on the DL features from the peritumoral regions with expansion distances of 2 mm, 8 mm, and 10 mm using the GoogLeNet architecture correlated with the optimal AUC values (test set: .823, .753, and .768) in the T2-weighted, T1-weighted contrast-enhanced, and T1-weighted images. Using the stacking fusion method, DLR with multi-parameter and multi-region fusion achieved the best discrimination with AUC values of .948 and .902 in the training and test sets, respectively. Conclusions: The radiomics model based on the fusion of multi-parameter MRI intra- and peritumoral DLR signatures may help to identify TERT promoter mutation in patients with GBM.
Collapse
Affiliation(s)
- Hongbo Zhang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Beibei Zhou
- Department of Radiology, The Seventh Affiliated Hospital Sun Yat-sen University, Shenzhen, China
| | - Hanwen Zhang
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yuze Zhang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yi Lei
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| | - Biao Huang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Herr J, Stoyanova R, Mellon EA. Convolutional Neural Networks for Glioma Segmentation and Prognosis: A Systematic Review. Crit Rev Oncog 2024; 29:33-65. [PMID: 38683153 DOI: 10.1615/critrevoncog.2023050852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Deep learning (DL) is poised to redefine the way medical images are processed and analyzed. Convolutional neural networks (CNNs), a specific type of DL architecture, are exceptional for high-throughput processing, allowing for the effective extraction of relevant diagnostic patterns from large volumes of complex visual data. This technology has garnered substantial interest in the field of neuro-oncology as a promising tool to enhance medical imaging throughput and analysis. A multitude of methods harnessing MRI-based CNNs have been proposed for brain tumor segmentation, classification, and prognosis prediction. They are often applied to gliomas, the most common primary brain cancer, to classify subtypes with the goal of guiding therapy decisions. Additionally, the difficulty of repeating brain biopsies to evaluate treatment response in the setting of often confusing imaging findings provides a unique niche for CNNs to help distinguish the treatment response to gliomas. For example, glioblastoma, the most aggressive type of brain cancer, can grow due to poor treatment response, can appear to grow acutely due to treatment-related inflammation as the tumor dies (pseudo-progression), or falsely appear to be regrowing after treatment as a result of brain damage from radiation (radiation necrosis). CNNs are being applied to separate this diagnostic dilemma. This review provides a detailed synthesis of recent DL methods and applications for intratumor segmentation, glioma classification, and prognosis prediction. Furthermore, this review discusses the future direction of MRI-based CNN in the field of neuro-oncology and challenges in model interpretability, data availability, and computation efficiency.
Collapse
Affiliation(s)
| | - Radka Stoyanova
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, Fl 33136, USA
| | - Eric Albert Mellon
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, Fl 33136, USA
| |
Collapse
|
12
|
Lu T, Ma J, Zou J, Jiang C, Li Y, Han J. CT-based intratumoral and peritumoral deep transfer learning features prediction of lymph node metastasis in non-small cell lung cancer. JOURNAL OF X-RAY SCIENCE AND TECHNOLOGY 2024; 32:597-609. [PMID: 38578874 DOI: 10.3233/xst-230326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
BACKGROUND The main metastatic route for lung cancer is lymph node metastasis, and studies have shown that non-small cell lung cancer (NSCLC) has a high risk of lymph node infiltration. OBJECTIVE This study aimed to compare the performance of handcrafted radiomics (HR) features and deep transfer learning (DTL) features in Computed Tomography (CT) of intratumoral and peritumoral regions in predicting the metastatic status of NSCLC lymph nodes in different machine learning classifier models. METHODS We retrospectively collected data of 199 patients with pathologically confirmed NSCLC. All patients were divided into training (n = 159) and validation (n = 40) cohorts, respectively. The best HR and DTL features in the intratumoral and peritumoral regions were extracted and selected, respectively. Support Vector Machine (SVM), k-Nearest Neighbors (KNN), Light Gradient Boosting Machine (Light GBM), Multilayer Perceptron (MLP), and Logistic Regression (LR) models were constructed, and the performance of the models was evaluated. RESULTS Among the five models in the training and validation cohorts, the LR classifier model performed best in terms of HR and DTL features. The AUCs of the training cohort were 0.841 (95% CI: 0.776-0.907) and 0.955 (95% CI: 0.926-0.983), and the AUCs of the validation cohort were 0.812 (95% CI: 0.677-0.948) and 0.893 (95% CI: 0.795-0.991), respectively. The DTL signature was superior to the handcrafted radiomics signature. CONCLUSIONS Compared with the radiomics signature, the DTL signature constructed based on intratumoral and peritumoral areas in CT can better predict NSCLC lymph node metastasis.
Collapse
Affiliation(s)
- Tianyu Lu
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jianbing Ma
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jiajun Zou
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chenxu Jiang
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yangyang Li
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jun Han
- Department of Radiology, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
13
|
Bajaj S, Khunte M, Moily NS, Payabvash S, Wintermark M, Gandhi D, Malhotra A. Value Proposition of FDA-Approved Artificial Intelligence Algorithms for Neuroimaging. J Am Coll Radiol 2023; 20:1241-1249. [PMID: 37574094 DOI: 10.1016/j.jacr.2023.06.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 08/15/2023]
Abstract
PURPOSE The number of FDA-cleared artificial intelligence (AI) algorithms for neuroimaging has grown in the past decade. The adoption of these algorithms into clinical practice depends largely on whether this technology provides value in the clinical setting. The objective of this study was to analyze trends in FDA-cleared AI algorithms for neuroimaging and understand their value proposition as advertised by the AI developers and vendors. METHODS A list of AI algorithms cleared by the FDA for neuroimaging between May 2008 and August 2022 was extracted from the ACR Data Science Institute AI Central database. Product information for each device was collected from the database. For each device, information on the advertised value as presented on the developer's website was collected. RESULTS A total of 59 AI neuroimaging algorithms were cleared by the FDA between May 2008 and August 2022. Most of these algorithms (24 of 59) were compatible with noncontrast CT, 21 with MRI, 9 with CT perfusion, 8 with CT angiography, 3 with MR perfusion, and 2 with PET. Six algorithms were compatible with multiple imaging techniques. Of the 59 algorithms, websites were located that discussed the product value for 55 algorithms. The most widely advertised value proposition was improved quality of care (38 of 55 [69.1%]). A total of 24 algorithms (43.6%) proposed saving user time, 9 (15.7%) advertised decreased costs, and 6 (10.9%) described increased revenue. Product websites for 26 algorithms (43.6%) showed user testimonials advertising the value of the technology. CONCLUSIONS The results of this study indicate a wide range of value propositions advertised by developers and vendors of AI algorithms for neuroimaging. Most vendors advertised that their products would improve patient care. Further research is necessary to determine whether the value claimed by developers is actually demonstrated in clinical practice.
Collapse
Affiliation(s)
- Suryansh Bajaj
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Mihir Khunte
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | | | - Seyedmehdi Payabvash
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Max Wintermark
- Chair, Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dheeraj Gandhi
- Director, Interventional Neuroradiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ajay Malhotra
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut.
| |
Collapse
|
14
|
Zhang H, Zhang H, Zhang Y, Zhou B, Wu L, Lei Y, Huang B. Deep Learning Radiomics for the Assessment of Telomerase Reverse Transcriptase Promoter Mutation Status in Patients With Glioblastoma Using Multiparametric MRI. J Magn Reson Imaging 2023; 58:1441-1451. [PMID: 36896953 DOI: 10.1002/jmri.28671] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Studies have shown that magnetic resonance imaging (MRI)-based deep learning radiomics (DLR) has the potential to assess glioma grade; however, its role in predicting telomerase reverse transcriptase (TERT) promoter mutation status in patients with glioblastoma (GBM) remains unclear. PURPOSE To evaluate the value of deep learning (DL) in multiparametric MRI-based radiomics in identifying TERT promoter mutations in patients with GBM preoperatively. STUDY TYPE Retrospective. POPULATION A total of 274 patients with isocitrate dehydrogenase-wildtype GBM were included in the study. The training and external validation cohorts included 156 (54.3 ± 12.7 years; 96 males) and 118 (54 .2 ± 13.4 years; 73 males) patients, respectively. FIELD STRENGTH/SEQUENCE Axial contrast-enhanced T1-weighted spin-echo inversion recovery sequence (T1CE), T1-weighted spin-echo inversion recovery sequence (T1WI), and T2-weighted spin-echo inversion recovery sequence (T2WI) on 1.5-T and 3.0-T scanners were used in this study. ASSESSMENT Overall tumor area regions (the tumor core and edema) were segmented, and the radiomics and DL features were extracted from preprocessed multiparameter preoperative brain MRI images-T1WI, T1CE, and T2WI. A model based on the DLR signature, clinical signature, and clinical DLR (CDLR) nomogram was developed and validated to identify TERT promoter mutation status. STATISTICAL TESTS The Mann-Whitney U test, Pearson test, least absolute shrinkage and selection operator, and logistic regression analysis were applied for feature selection and construction of radiomics and DL signatures. Results were considered statistically significant at P-value <0.05. RESULTS The DLR signature showed the best discriminative power for predicting TERT promoter mutations, yielding an AUC of 0.990 and 0.890 in the training and external validation cohorts, respectively. Furthermore, the DLR signature outperformed CDLR nomogram (P = 0.670) and significantly outperformed clinical models in the validation cohort. DATA CONCLUSION The multiparameter MRI-based DLR signature exhibited a promising performance for the assessment of TERT promoter mutations in patients with GBM, which could provide information for individualized treatment. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Hongbo Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hanwen Zhang
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yuze Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Beibei Zhou
- Department of Radiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Lei Wu
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yi Lei
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| | - Biao Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
15
|
Choi Y, Jang J, Kim BS, Ahn KJ. Pretreatment MR-based radiomics in patients with glioblastoma: A systematic review and meta-analysis of prognostic endpoints. Eur J Radiol 2023; 168:111130. [PMID: 37827087 DOI: 10.1016/j.ejrad.2023.111130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/23/2023] [Accepted: 09/30/2023] [Indexed: 10/14/2023]
Abstract
PURPOSE Recent studies have shown promise of MR-based radiomics in predicting the survival of patients with untreated glioblastoma. This study aimed to comprehensively collate evidence to assess the prognostic value of radiomics in glioblastoma. METHODS PubMed-MEDLINE, Embase, and Web of Science were searched to find original articles investigating the prognostic value of MR-based radiomics in glioblastoma published up to July 14, 2023. Concordance indexes (C-indexes) and Cox proportional hazards ratios (HRs) of overall survival (OS) and progression-free survival (PFS) were pooled via random-effects modeling. For studies aimed at classifying long-term and short-term PFS, a hierarchical regression model was used to calculate pooled sensitivity and specificity. Between-study heterogeneity was assessed using the Higgin inconsistency index (I2). Subgroup regression analysis was performed to find potential factors contributing to heterogeneity. Publication bias was assessed via funnel plots and the Egger test. RESULTS Among 1371 abstracts, 18 and 17 studies were included for qualitative and quantitative data synthesis, respectively. Respective pooled C-indexes and HRs for OS were 0.65 (95 % confidence interval [CI], 0.58-0.72) and 2.88 (95 % CI, 2.28-3.64), whereas those for PFS were 0.61 (95 % CI, 0.55-0.66) and 2.78 (95 % CI, 1.91-4.03). Among 4 studies that predicted short-term PFS, the pooled sensitivity and specificity were 0.77 (95 % CI, 0.58-0.89) and 0.60 (95 % CI, 0.45-0.73), respectively. There was a substantial between-study heterogeneity among studies with the survival endpoint of OS C-index (n = 9, I2 = 83.8 %). Publication bias was not observed overall. CONCLUSION Pretreatment MR-based radiomics provided modest prognostic value in both OS and PFS in patients with glioblastoma.
Collapse
Affiliation(s)
- Yangsean Choi
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Republic of Korea; Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, Republic of Korea
| | - Jinhee Jang
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Bum-Soo Kim
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Kook-Jin Ahn
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Republic of Korea.
| |
Collapse
|
16
|
Papi Z, Fathi S, Dalvand F, Vali M, Yousefi A, Tabatabaei MH, Amouheidari A, Abedi I. Auto-Segmentation and Classification of Glioma Tumors with the Goals of Treatment Response Assessment Using Deep Learning Based on Magnetic Resonance Imaging. Neuroinformatics 2023; 21:641-650. [PMID: 37458971 DOI: 10.1007/s12021-023-09640-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 10/18/2023]
Abstract
Glioma is the most common primary intracranial neoplasm in adults. Radiotherapy is a treatment approach in glioma patients, and Magnetic Resonance Imaging (MRI) is a beneficial diagnostic tool in treatment planning. Treatment response assessment in glioma patients is usually based on the Response Assessment in Neuro Oncology (RANO) criteria. The limitation of assessment based on RANO is two-dimensional (2D) manual measurements. Deep learning (DL) has great potential in neuro-oncology to improve the accuracy of response assessment. In the current research, firstly, the BraTS 2018 Challenge dataset included 210 HGG and 75 LGG were applied to train a designed U-Net network for automatic tumor and intra-tumoral segmentation, followed by training of the designed classifier with transfer learning for determining grading HGG and LGG. Then, designed networks were employed for the segmentation and classification of local MRI images of 49 glioma patients pre and post-radiotherapy. The results of tumor segmentation and its intra-tumoral regions were utilized to determine the volume of different regions and treatment response assessment. Treatment response assessment demonstrated that radiotherapy is effective on the whole tumor and enhancing region with p-value ≤ 0.05 with a 95% confidence level, while it did not affect necrosis and peri-tumoral edema regions. This work demonstrated the potential of using deep learning in MRI images to provide a beneficial tool in the automated treatment response assessment so that the patient can obtain the best treatment.
Collapse
Affiliation(s)
- Zahra Papi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sina Fathi
- Department of Health Information Management, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Dalvand
- Department of Medical Radiation, Shahid Beheshti University, Tehran, Iran
| | - Mahsa Vali
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Ali Yousefi
- Department of Management- Operations Research, University of Isfahan, Isfahan, Iran
| | | | | | - Iraj Abedi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
17
|
Wang Y, Li L, Li C, Xi Y, Lin Y, Wang S. Expert knowledge guided manifold representation learning for magnetic resonance imaging-based glioma grading. Biomed Signal Process Control 2023. [DOI: 10.1016/j.bspc.2023.104876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
18
|
Capobianco E, Dominietto M. Assessment of brain cancer atlas maps with multimodal imaging features. J Transl Med 2023; 21:385. [PMID: 37308956 PMCID: PMC10262565 DOI: 10.1186/s12967-023-04222-3] [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: 04/18/2023] [Accepted: 05/22/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Glioblastoma Multiforme (GBM) is a fast-growing and highly aggressive brain tumor that invades the nearby brain tissue and presents secondary nodular lesions across the whole brain but generally does not spread to distant organs. Without treatment, GBM can result in death in about 6 months. The challenges are known to depend on multiple factors: brain localization, resistance to conventional therapy, disrupted tumor blood supply inhibiting effective drug delivery, complications from peritumoral edema, intracranial hypertension, seizures, and neurotoxicity. MAIN TEXT Imaging techniques are routinely used to obtain accurate detections of lesions that localize brain tumors. Especially magnetic resonance imaging (MRI) delivers multimodal images both before and after the administration of contrast, which results in displaying enhancement and describing physiological features as hemodynamic processes. This review considers one possible extension of the use of radiomics in GBM studies, one that recalibrates the analysis of targeted segmentations to the whole organ scale. After identifying critical areas of research, the focus is on illustrating the potential utility of an integrated approach with multimodal imaging, radiomic data processing and brain atlases as the main components. The templates associated with the outcome of straightforward analyses represent promising inference tools able to spatio-temporally inform on the GBM evolution while being generalizable also to other cancers. CONCLUSIONS The focus on novel inference strategies applicable to complex cancer systems and based on building radiomic models from multimodal imaging data can be well supported by machine learning and other computational tools potentially able to translate suitably processed information into more accurate patient stratifications and evaluations of treatment efficacy.
Collapse
Affiliation(s)
- Enrico Capobianco
- The Jackson Laboratory, 10 Discovery Drive, Farmington, CT, 06032, USA.
| | - Marco Dominietto
- Paul Scherrer Institute (PSI), Forschungsstrasse 111, 5232, Villigen, Switzerland
- Gate To Brain SA, Via Livio 7, 6830, Chiasso, Switzerland
| |
Collapse
|
19
|
Hense K, Deuter D, Greenlee MW, Wendl C, Schmidt NO, Stroszczynski C, Doenitz C, Ott C, Rosengarth K. Analysis of Functional Neuroplastic Changes in the Cortical Language System in Relation to Different Growth Patterns of Glioblastoma. Brain Sci 2023; 13:867. [PMID: 37371347 DOI: 10.3390/brainsci13060867] [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: 05/15/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
The interpretation of fMRI data in glioblastoma (GB) is challenging as these tumors exhibit specific hemodynamic processes which, together with malignancy, tumor volume and proximity to eloquent cortex areas, may lead to misinterpretations of fMRI signals. The aim of this study was to investigate if different radiologically defined GB tumor growth patterns may also influence the fMRI signal, activation pattern and functional connectivity differently. Sixty-four patients with left-hemispheric glioblastoma were included and stratified according to their radiologically defined tumor growth pattern into groups with a uniform (U-TGP) or diffuse tumor growth pattern (D-TGP). Task-based fMRI data were analyzed using SPM12 with the marsbar, LI and CONN toolboxes. The percent signal change and the laterality index were analyzed, as well as functional connectivity between 23 selected ROIs. Comparisons of both patient groups showed only minor non-significant differences, indicating that the tumor growth pattern is not a relevant influencing factor for fMRI signal. In addition to these results, signal reductions were found in areas that were not affected by the tumor underlining that a GB is not a localized but rather a systemic disease affecting the entire brain.
Collapse
Affiliation(s)
- Katharina Hense
- Department of Neurosurgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Daniel Deuter
- Department of Neurosurgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Mark W Greenlee
- Institute for Experimental Psychology, University of Regensburg, 93053 Regensburg, Germany
| | - Christina Wendl
- Department of Radiology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Nils Ole Schmidt
- Department of Neurosurgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | | | - Christian Doenitz
- Department of Neurosurgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christian Ott
- Department of Neurosurgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Katharina Rosengarth
- Department of Neurosurgery, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
20
|
Qi D, Li J, Quarles CC, Fonkem E, Wu E. Assessment and prediction of glioblastoma therapy response: challenges and opportunities. Brain 2023; 146:1281-1298. [PMID: 36445396 PMCID: PMC10319779 DOI: 10.1093/brain/awac450] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/30/2022] Open
Abstract
Glioblastoma is the most aggressive type of primary adult brain tumour. The median survival of patients with glioblastoma remains approximately 15 months, and the 5-year survival rate is <10%. Current treatment options are limited, and the standard of care has remained relatively constant since 2011. Over the last decade, a range of different treatment regimens have been investigated with very limited success. Tumour recurrence is almost inevitable with the current treatment strategies, as glioblastoma tumours are highly heterogeneous and invasive. Additionally, another challenging issue facing patients with glioblastoma is how to distinguish between tumour progression and treatment effects, especially when relying on routine diagnostic imaging techniques in the clinic. The specificity of routine imaging for identifying tumour progression early or in a timely manner is poor due to the appearance similarity of post-treatment effects. Here, we concisely describe the current status and challenges in the assessment and early prediction of therapy response and the early detection of tumour progression or recurrence. We also summarize and discuss studies of advanced approaches such as quantitative imaging, liquid biomarker discovery and machine intelligence that hold exceptional potential to aid in the therapy monitoring of this malignancy and early prediction of therapy response, which may decisively transform the conventional detection methods in the era of precision medicine.
Collapse
Affiliation(s)
- Dan Qi
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
| | - Jing Li
- School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - C Chad Quarles
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Ekokobe Fonkem
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
- Department of Medical Education, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Erxi Wu
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
- Department of Medical Education, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX 77843, USA
- Department of Oncology and LIVESTRONG Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
21
|
Deep learning-based harmonization of CT reconstruction kernels towards improved clinical task performance. Eur Radiol 2023; 33:2426-2438. [PMID: 36355196 DOI: 10.1007/s00330-022-09229-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/11/2022] [Accepted: 10/09/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVES To develop a deep learning-based harmonization framework, assessing whether it can improve performance of radiomics models given different kernels in different clinical tasks and additionally generalize to mitigate the effects of new/unobserved kernels on radiomics features. METHODS Patient data with 2 reconstruction kernels and phantom data with 22 reconstruction kernels were included. Eighty-five patients were studied for lymph node metastasis (LNM) prediction, and 164 patients for differential diagnosis between lung cancer (LC) and pulmonary tuberculosis (TB). Two convolutional neural network (CNN) models were developed to convert images (i) from B70f to B30f (CNNa) and (ii) from B30f to B70f (CNNb). Model performance between the two kernels was evaluated using AUC and compared with other well-known harmonization methods. Patient-normalized feature difference (PNFD) was used to identify the incompatible kernels (i.e., kernel with median PNFD > 1) with baseline (B30f/B70f), and measure the ability of the CNN models to convert the non-comparable kernels. RESULTS For LC versus pulmonary TB diagnosis, AUCs of CNNa vs. others were 0.85 vs. 0.54-0.74 (p = 0.0001-0.0003), and for CNNb vs. others: 0.87 vs. 0.54-0.86 (p = 0.0001-0.55). For LNM prediction, AUCs of CNNa vs. others were 0.68 vs. 0.56-0.61 (p = 0.10-0.39), and for CNNb vs. others: 0.78 vs. 0.70-0.73 (p = 0.07-0.40). After CNN harmonization, 17 of 20 (85%) of investigated unknown kernels produced comparable radiomics feature values relative to baseline (median PNFD from 1.10-2.31 to 0.23-1.13). CONCLUSION The CNN harmonization effectively improved performance of radiomics models between reconstruction kernels in different clinical tasks, and reduced feature differences between unknown kernels vs. baseline. KEY POINTS • The soft (B30f) and sharp (B70f) kernels strongly affect radiomics reproducibility and generalizability. • The convolutional neural network (CNN) harmonization methods performed better than location-scale (ComBat and centering-scaling) and matrix factorization harmonization methods (based on singular value decomposition (SVD) and independent component analysis (ICA)) in both clinical tasks. • The CNN harmonization methods improve feature reproducibility not only between specific kernels (B30f and B70f) from the same scanner, but also between unobserved kernels from different scanners of different vendors.
Collapse
|
22
|
Zhang Y, Wu C, Xiao Z, Lv F, Liu Y. A Deep Learning Radiomics Nomogram to Predict Response to Neoadjuvant Chemotherapy for Locally Advanced Cervical Cancer: A Two-Center Study. Diagnostics (Basel) 2023; 13:diagnostics13061073. [PMID: 36980381 PMCID: PMC10047639 DOI: 10.3390/diagnostics13061073] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Purpose: This study aimed to establish a deep learning radiomics nomogram (DLRN) based on multiparametric MR images for predicting the response to neoadjuvant chemotherapy (NACT) in patients with locally advanced cervical cancer (LACC). Methods: Patients with LACC (FIGO stage IB-IIIB) who underwent preoperative NACT were enrolled from center 1 (220 cases) and center 2 (independent external validation dataset, 65 cases). Handcrafted and deep learning-based radiomics features were extracted from T2WI, DWI and contrast-enhanced (CE)-T1WI, and radiomics signatures were built based on the optimal features. Two types of radiomics signatures and clinical features were integrated into the DLRN for prediction. The AUC, calibration curve and decision curve analysis (DCA) were employed to illustrate the performance of these models and their clinical utility. In addition, disease-free survival (DFS) was assessed by Kaplan–Meier survival curves based on the DLRN. Results: The DLRN showed favorable predictive values in differentiating responders from nonresponders to NACT with AUCs of 0.963, 0.940 and 0.910 in the three datasets, with good calibration (all p > 0.05). Furthermore, the DLRN performed better than the clinical model and handcrafted radiomics signature in all datasets (all p < 0.05) and slightly higher than the DL-based radiomics signature in the internal validation dataset (p = 0.251). DCA indicated that the DLRN has potential in clinical applications. Furthermore, the DLRN was strongly correlated with the DFS of LACC patients (HR = 0.223; p = 0.004). Conclusion: The DLRN performed well in preoperatively predicting the therapeutic response in LACC and could provide valuable information for individualized treatment.
Collapse
Affiliation(s)
- Yajiao Zhang
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China;
| | - Chao Wu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhibo Xiao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Furong Lv
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yanbing Liu
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China;
- Correspondence:
| |
Collapse
|
23
|
Luo J, Pan M, Mo K, Mao Y, Zou D. Emerging role of artificial intelligence in diagnosis, classification and clinical management of glioma. Semin Cancer Biol 2023; 91:110-123. [PMID: 36907387 DOI: 10.1016/j.semcancer.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 03/05/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023]
Abstract
Glioma represents a dominant primary intracranial malignancy in the central nervous system. Artificial intelligence that mainly includes machine learning, and deep learning computational approaches, presents a unique opportunity to enhance clinical management of glioma through improving tumor segmentation, diagnosis, differentiation, grading, treatment, prediction of clinical outcomes (prognosis, and recurrence), molecular features, clinical classification, characterization of the tumor microenvironment, and drug discovery. A growing body of recent studies apply artificial intelligence-based models to disparate data sources of glioma, covering imaging modalities, digital pathology, high-throughput multi-omics data (especially emerging single-cell RNA sequencing and spatial transcriptome), etc. While these early findings are promising, future studies are required to normalize artificial intelligence-based models to improve the generalizability and interpretability of the results. Despite prominent issues, targeted clinical application of artificial intelligence approaches in glioma will facilitate the development of precision medicine of this field. If these challenges can be overcome, artificial intelligence has the potential to profoundly change the way patients with or at risk of glioma are provided with more rational care.
Collapse
Affiliation(s)
- Jiefeng Luo
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Mika Pan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Ke Mo
- Clinical Research Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Yingwei Mao
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA.
| | - Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China; Clinical Research Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China.
| |
Collapse
|
24
|
Zhang J, Liu J, Liang Z, Xia L, Zhang W, Xing Y, Zhang X, Tang G. Differentiation of acute and chronic vertebral compression fractures using conventional CT based on deep transfer learning features and hand-crafted radiomics features. BMC Musculoskelet Disord 2023; 24:165. [PMID: 36879285 PMCID: PMC9987077 DOI: 10.1186/s12891-023-06281-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND We evaluated the diagnostic efficacy of deep learning radiomics (DLR) and hand-crafted radiomics (HCR) features in differentiating acute and chronic vertebral compression fractures (VCFs). METHODS A total of 365 patients with VCFs were retrospectively analysed based on their computed tomography (CT) scan data. All patients completed MRI examination within 2 weeks. There were 315 acute VCFs and 205 chronic VCFs. Deep transfer learning (DTL) features and HCR features were extracted from CT images of patients with VCFs using DLR and traditional radiomics, respectively, and feature fusion was performed to establish the least absolute shrinkage and selection operator. The MRI display of vertebral bone marrow oedema was used as the gold standard for acute VCF, and the model performance was evaluated using the receiver operating characteristic (ROC).To separately evaluate the effectiveness of DLR, traditional radiomics and feature fusion in the differential diagnosis of acute and chronic VCFs, we constructed a nomogram based on the clinical baseline data to visualize the classification evaluation. The predictive power of each model was compared using the Delong test, and the clinical value of the nomogram was evaluated using decision curve analysis (DCA). RESULTS Fifty DTL features were obtained from DLR, 41 HCR features were obtained from traditional radiomics, and 77 features fusion were obtained after feature screening and fusion of the two. The area under the curve (AUC) of the DLR model in the training cohort and test cohort were 0.992 (95% confidence interval (CI), 0.983-0.999) and 0.871 (95% CI, 0.805-0.938), respectively. While the AUCs of the conventional radiomics model in the training cohort and test cohort were 0.973 (95% CI, 0.955-0.990) and 0.854 (95% CI, 0.773-0.934), respectively. The AUCs of the features fusion model in the training cohort and test cohort were 0.997 (95% CI, 0.994-0.999) and 0.915 (95% CI, 0.855-0.974), respectively. The AUCs of nomogram constructed by the features fusion in combination with clinical baseline data were 0.998 (95% CI, 0.996-0.999) and 0.946 (95% CI, 0.906-0.987) in the training cohort and test cohort, respectively. The Delong test showed that the differences between the features fusion model and the nomogram in the training cohort and the test cohort were not statistically significant (P values were 0.794 and 0.668, respectively), and the differences in the other prediction models in the training cohort and the test cohort were statistically significant (P < 0.05). DCA showed that the nomogram had high clinical value. CONCLUSION The features fusion model can be used for the differential diagnosis of acute and chronic VCFs, and its differential diagnosis ability is improved when compared with that when either radiomics is used alone. At the same time, the nomogram has a high predictive value for acute and chronic VCFs and can be a potential decision-making tool to assist clinicians, especially when a patient is unable to undergo spinal MRI examination.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Radiology, Clinical Medical College of Shanghai Tenth People's Hospital of Nanjing Medical University, 301 Middle Yanchang Road, Shanghai, 200072, P.R. China.,Department of Radiology, Sir RunRun Hospital affiliated to Nanjing Medical University, 109 Longmian Road, Nanjing, Jiangsu, 211002, P.R. China
| | - Jiayi Liu
- Department of Radiology, Sir RunRun Hospital affiliated to Nanjing Medical University, 109 Longmian Road, Nanjing, Jiangsu, 211002, P.R. China
| | - Zhipeng Liang
- Department of Radiology, Sir RunRun Hospital affiliated to Nanjing Medical University, 109 Longmian Road, Nanjing, Jiangsu, 211002, P.R. China
| | - Liang Xia
- Department of Radiology, Sir RunRun Hospital affiliated to Nanjing Medical University, 109 Longmian Road, Nanjing, Jiangsu, 211002, P.R. China
| | - Weixiao Zhang
- Department of Radiology, Sir RunRun Hospital affiliated to Nanjing Medical University, 109 Longmian Road, Nanjing, Jiangsu, 211002, P.R. China
| | - Yanfen Xing
- Department of Radiology, Sir RunRun Hospital affiliated to Nanjing Medical University, 109 Longmian Road, Nanjing, Jiangsu, 211002, P.R. China
| | - Xueli Zhang
- Department of Radiology, Shanghai TenthPeople's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, P.R. China
| | - Guangyu Tang
- Department of Radiology, Clinical Medical College of Shanghai Tenth People's Hospital of Nanjing Medical University, 301 Middle Yanchang Road, Shanghai, 200072, P.R. China. .,Department of Radiology, Shanghai TenthPeople's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, P.R. China.
| |
Collapse
|
25
|
Salome P, Sforazzini F, Grugnara G, Kudak A, Dostal M, Herold-Mende C, Heiland S, Debus J, Abdollahi A, Knoll M. MR Intensity Normalization Methods Impact Sequence Specific Radiomics Prognostic Model Performance in Primary and Recurrent High-Grade Glioma. Cancers (Basel) 2023; 15:cancers15030965. [PMID: 36765922 PMCID: PMC9913466 DOI: 10.3390/cancers15030965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
PURPOSE This study investigates the impact of different intensity normalization (IN) methods on the overall survival (OS) radiomics models' performance of MR sequences in primary (pHGG) and recurrent high-grade glioma (rHGG). METHODS MR scans acquired before radiotherapy were retrieved from two independent cohorts (rHGG C1: 197, pHGG C2: 141) from multiple scanners (15, 14). The sequences are T1 weighted (w), contrast-enhanced T1w (T1wce), T2w, and T2w-FLAIR. Sequence-specific significant features (SF) associated with OS, extracted from the tumour volume, were derived after applying 15 different IN methods. Survival analyses were conducted using Cox proportional hazard (CPH) and Poisson regression (POI) models. A ranking score was assigned based on the 10-fold cross-validated (CV) concordance index (C-I), mean square error (MSE), and the Akaike information criterion (AICs), to evaluate the methods' performance. RESULTS Scatter plots of the 10-CV C-I and MSE against the AIC showed an impact on the survival predictions between the IN methods and MR sequences (C1/C2 C-I range: 0.62-0.71/0.61-0.72, MSE range: 0.20-0.42/0.13-0.22). White stripe showed stable results for T1wce (C1/C2 C-I: 0.71/0.65, MSE: 0.21/0.14). Combat (0.68/0.62, 0.22/0.15) and histogram matching (HM, 0.67/0.64, 0.22/0.15) showed consistent prediction results for T2w models. They were also the top-performing methods for T1w in C2 (Combat: 0.67, 0.13; HM: 0.67, 0.13); however, only HM achieved high predictions in C1 (0.66, 0.22). After eliminating IN impacted SF using Spearman's rank-order correlation coefficient, a mean decrease in the C-I and MSE of 0.05 and 0.03 was observed in all four sequences. CONCLUSION The IN method impacted the predictive power of survival models; thus, performance is sequence-dependent.
Collapse
Affiliation(s)
- Patrick Salome
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- Heidelberg Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Correspondence: (P.S.); (M.K.)
| | - Francesco Sforazzini
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- Heidelberg Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
| | - Gianluca Grugnara
- Department of Neuroradiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Andreas Kudak
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- CCU Radiation Therapy, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
| | - Matthias Dostal
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- CCU Radiation Therapy, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
| | - Christel Herold-Mende
- Brain Tumour Group, European Organization for Research and Treatment of Cancer, 1200 Brussels, Belgium
- Division of Neurosurgical Research, Department of Neurosurgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Sabine Heiland
- Department of Neuroradiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Jürgen Debus
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
| | - Amir Abdollahi
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
| | - Maximilian Knoll
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- Correspondence: (P.S.); (M.K.)
| |
Collapse
|
26
|
Yan J, Sun Q, Tan X, Liang C, Bai H, Duan W, Mu T, Guo Y, Qiu Y, Wang W, Yao Q, Pei D, Zhao Y, Liu D, Duan J, Chen S, Sun C, Wang W, Liu Z, Hong X, Wang X, Guo Y, Xu Y, Liu X, Cheng J, Li ZC, Zhang Z. Image-based deep learning identifies glioblastoma risk groups with genomic and transcriptomic heterogeneity: a multi-center study. Eur Radiol 2023; 33:904-914. [PMID: 36001125 DOI: 10.1007/s00330-022-09066-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVES To develop and validate a deep learning imaging signature (DLIS) for risk stratification in patients with multiforme (GBM), and to investigate the biological pathways and genetic alterations underlying the DLIS. METHODS The DLIS was developed from multi-parametric MRI based on a training set (n = 600) and validated on an internal validation set (n = 164), an external test set 1 (n = 100), an external test set 2 (n = 161), and a public TCIA set (n = 88). A co-profiling framework based on a radiogenomics analysis dataset (n = 127) using multiscale high-dimensional data, including imaging, transcriptome, and genome, was established to uncover the biological pathways and genetic alterations underpinning the DLIS. RESULTS The DLIS was associated with survival (log-rank p < 0.001) and was an independent predictor (p < 0.001). The integrated nomogram incorporating the DLIS achieved improved C indices than the clinicomolecular nomogram (net reclassification improvement 0.39, p < 0.001). DLIS significantly correlated with core pathways of GBM (apoptosis and cell cycle-related P53 and RB pathways, and cell proliferation-related RTK pathway), as well as key genetic alterations (del_CDNK2A). The prognostic value of DLIS-correlated genes was externally confirmed on TCGA/CGGA sets (p < 0.01). CONCLUSIONS Our study offers a biologically interpretable deep learning predictor of survival outcomes in patients with GBM, which is crucial for better understanding GBM patient's prognosis and guiding individualized treatment. KEY POINTS • MRI-based deep learning imaging signature (DLIS) stratifies GBM into risk groups with distinct molecular characteristics. • DLIS is associated with P53, RB, and RTK pathways and del_CDNK2A mutation. • The prognostic value of DLIS-correlated pathway genes is externally demonstrated.
Collapse
Affiliation(s)
- Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Qiuchang Sun
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiangliang Tan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaofeng Liang
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Hongmin Bai
- Department of Neurosurgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, China
| | - Wenchao Duan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Tianhao Mu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.,HaploX Biotechnology, Shenzhen, Guangdong, China
| | - Yang Guo
- Department of Neurosurgery, Henan Provincial Hospital, Zhengzhou, 450052, Henan Province, China
| | - Yuning Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan Province, China
| | - Qiaoli Yao
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Dongling Pei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Yuanshen Zhao
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Danni Liu
- HaploX Biotechnology, Shenzhen, Guangdong, China
| | - Jingxian Duan
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Shifu Chen
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.,HaploX Biotechnology, Shenzhen, Guangdong, China
| | - Chen Sun
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Wenqing Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Zhen Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Xuanke Hong
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Xiangxiang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Yu Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China.
| | - Zhi-Cheng Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Shenzhen United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, 518045, China.
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China.
| |
Collapse
|
27
|
Feng D, Chen X, Wang X, Mou X, Bai L, Zhang S, Zhou Z. Predicting effectiveness of anti-VEGF injection through self-supervised learning in OCT images. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:2439-2458. [PMID: 36899541 DOI: 10.3934/mbe.2023114] [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: 06/18/2023]
Abstract
Anti-vascular endothelial growth factor (Anti-VEGF) therapy has become a standard way for choroidal neovascularization (CNV) and cystoid macular edema (CME) treatment. However, anti-VEGF injection is a long-term therapy with expensive cost and may be not effective for some patients. Therefore, predicting the effectiveness of anti-VEGF injection before the therapy is necessary. In this study, a new optical coherence tomography (OCT) images based self-supervised learning (OCT-SSL) model for predicting the effectiveness of anti-VEGF injection is developed. In OCT-SSL, we pre-train a deep encoder-decoder network through self-supervised learning to learn the general features using a public OCT image dataset. Then, model fine-tuning is performed on our own OCT dataset to learn the discriminative features to predict the effectiveness of anti-VEGF. Finally, classifier trained by the features from fine-tuned encoder as a feature extractor is built to predict the response. Experimental results on our private OCT dataset demonstrated that the proposed OCT-SSL can achieve an average accuracy, area under the curve (AUC), sensitivity and specificity of 0.93, 0.98, 0.94 and 0.91, respectively. Meanwhile, it is found that not only the lesion region but also the normal region in OCT image is related to the effectiveness of anti-VEGF.
Collapse
Affiliation(s)
- Dehua Feng
- School of Information and Communications Engineering, Xi'an Jiaotong University, Shaanxi 710049, China
| | - Xi Chen
- School of Information and Communications Engineering, Xi'an Jiaotong University, Shaanxi 710049, China
| | - Xiaoyu Wang
- School of Information and Communications Engineering, Xi'an Jiaotong University, Shaanxi 710049, China
| | - Xuanqin Mou
- School of Information and Communications Engineering, Xi'an Jiaotong University, Shaanxi 710049, China
| | - Ling Bai
- Department of Ophthalmology, the Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi 710004, China
| | - Shu Zhang
- Department of Geriatric Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi 710004, China
| | - Zhiguo Zhou
- Department of Biostatistics and Data Science, University of Kansas Medical Center, KS 66160, USA
| |
Collapse
|
28
|
Zhai Y, Bai J, Xue Y, Li M, Mao W, Zhang X, Zhang Y. Development and validation of a preoperative MRI-based radiomics nomogram to predict progression-free survival in patients with clival chordomas. Front Oncol 2022; 12:996262. [PMID: 36591445 PMCID: PMC9800789 DOI: 10.3389/fonc.2022.996262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Objectives The aim of this study was to establish and validate a MRI-based radiomics nomogram to predict progression-free survival (PFS) of clival chordoma. Methods A total of 174 patients were enrolled in the study (train cohort: 121 cases, test cohort: 53 cases). Radiomic features were extracted from multiparametric MRIs. Intraclass correlation coefficient analysis and a Lasso and Elastic-Net regularized generalized linear model were used for feature selection. Then, a nomogram was established via univariate and multivariate Cox regression analysis in the train cohort. The performance of this nomogram was assessed by area under curve (AUC) and calibration curve. Results A total of 3318 radiomic features were extracted from each patient, of which 2563 radiomic features were stable features. After feature selection, seven radiomic features were selected. Cox regression analysis revealed that 2 clinical factors (degree of resection, and presence or absence of primary chordoma) and 4 radiomic features were independent prognostic factors. The AUC of the established nomogram was 0.747, 0.807, and 0.904 for PFS prediction at 1, 3, and 5 years in the train cohort, respectively, compared with 0.582, 0.852, and 0.914 in the test cohort. Calibration and risk score stratified survival curves were satisfactory in the train and test cohort. Conclusions The presented nomogram demonstrated a favorable predictive accuracy of PFS, which provided a novel tool to predict prognosis and risk stratification. Our results suggest that radiomic analysis can effectively help neurosurgeons perform individualized evaluations of patients with clival chordomas.
Collapse
Affiliation(s)
- Yixuan Zhai
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiwei Bai
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yake Xue
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingxuan Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Wenbin Mao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuezhi Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,Center of Brain Tumor, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,*Correspondence: Yazhuo Zhang,
| |
Collapse
|
29
|
Pati S, Baid U, Edwards B, Sheller M, Wang SH, Reina GA, Foley P, Gruzdev A, Karkada D, Davatzikos C, Sako C, Ghodasara S, Bilello M, Mohan S, Vollmuth P, Brugnara G, Preetha CJ, Sahm F, Maier-Hein K, Zenk M, Bendszus M, Wick W, Calabrese E, Rudie J, Villanueva-Meyer J, Cha S, Ingalhalikar M, Jadhav M, Pandey U, Saini J, Garrett J, Larson M, Jeraj R, Currie S, Frood R, Fatania K, Huang RY, Chang K, Balaña C, Capellades J, Puig J, Trenkler J, Pichler J, Necker G, Haunschmidt A, Meckel S, Shukla G, Liem S, Alexander GS, Lombardo J, Palmer JD, Flanders AE, Dicker AP, Sair HI, Jones CK, Venkataraman A, Jiang M, So TY, Chen C, Heng PA, Dou Q, Kozubek M, Lux F, Michálek J, Matula P, Keřkovský M, Kopřivová T, Dostál M, Vybíhal V, Vogelbaum MA, Mitchell JR, Farinhas J, Maldjian JA, Yogananda CGB, Pinho MC, Reddy D, Holcomb J, Wagner BC, Ellingson BM, Cloughesy TF, Raymond C, Oughourlian T, Hagiwara A, Wang C, To MS, Bhardwaj S, Chong C, Agzarian M, Falcão AX, Martins SB, Teixeira BCA, Sprenger F, Menotti D, Lucio DR, LaMontagne P, Marcus D, Wiestler B, Kofler F, Ezhov I, Metz M, Jain R, Lee M, Lui YW, McKinley R, Slotboom J, Radojewski P, Meier R, Wiest R, Murcia D, Fu E, Haas R, Thompson J, Ormond DR, Badve C, Sloan AE, Vadmal V, Waite K, Colen RR, Pei L, Ak M, Srinivasan A, Bapuraj JR, Rao A, Wang N, Yoshiaki O, Moritani T, Turk S, Lee J, Prabhudesai S, Morón F, Mandel J, Kamnitsas K, Glocker B, Dixon LVM, Williams M, Zampakis P, Panagiotopoulos V, Tsiganos P, Alexiou S, Haliassos I, Zacharaki EI, Moustakas K, Kalogeropoulou C, Kardamakis DM, Choi YS, Lee SK, Chang JH, Ahn SS, Luo B, Poisson L, Wen N, Tiwari P, Verma R, Bareja R, Yadav I, Chen J, Kumar N, Smits M, van der Voort SR, Alafandi A, Incekara F, Wijnenga MMJ, Kapsas G, Gahrmann R, Schouten JW, Dubbink HJ, Vincent AJPE, van den Bent MJ, French PJ, Klein S, Yuan Y, Sharma S, Tseng TC, Adabi S, Niclou SP, Keunen O, Hau AC, Vallières M, Fortin D, Lepage M, Landman B, Ramadass K, Xu K, Chotai S, Chambless LB, Mistry A, Thompson RC, Gusev Y, Bhuvaneshwar K, Sayah A, Bencheqroun C, Belouali A, Madhavan S, Booth TC, Chelliah A, Modat M, Shuaib H, Dragos C, Abayazeed A, Kolodziej K, Hill M, Abbassy A, Gamal S, Mekhaimar M, Qayati M, Reyes M, Park JE, Yun J, Kim HS, Mahajan A, Muzi M, Benson S, Beets-Tan RGH, Teuwen J, Herrera-Trujillo A, Trujillo M, Escobar W, Abello A, Bernal J, Gómez J, Choi J, Baek S, Kim Y, Ismael H, Allen B, Buatti JM, Kotrotsou A, Li H, Weiss T, Weller M, Bink A, Pouymayou B, Shaykh HF, Saltz J, Prasanna P, Shrestha S, Mani KM, Payne D, Kurc T, Pelaez E, Franco-Maldonado H, Loayza F, Quevedo S, Guevara P, Torche E, Mendoza C, Vera F, Ríos E, López E, Velastin SA, Ogbole G, Soneye M, Oyekunle D, Odafe-Oyibotha O, Osobu B, Shu'aibu M, Dorcas A, Dako F, Simpson AL, Hamghalam M, Peoples JJ, Hu R, Tran A, Cutler D, Moraes FY, Boss MA, Gimpel J, Veettil DK, Schmidt K, Bialecki B, Marella S, Price C, Cimino L, Apgar C, Shah P, Menze B, Barnholtz-Sloan JS, Martin J, Bakas S. Federated learning enables big data for rare cancer boundary detection. Nat Commun 2022; 13:7346. [PMID: 36470898 PMCID: PMC9722782 DOI: 10.1038/s41467-022-33407-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/16/2022] [Indexed: 12/12/2022] Open
Abstract
Although machine learning (ML) has shown promise across disciplines, out-of-sample generalizability is concerning. This is currently addressed by sharing multi-site data, but such centralization is challenging/infeasible to scale due to various limitations. Federated ML (FL) provides an alternative paradigm for accurate and generalizable ML, by only sharing numerical model updates. Here we present the largest FL study to-date, involving data from 71 sites across 6 continents, to generate an automatic tumor boundary detector for the rare disease of glioblastoma, reporting the largest such dataset in the literature (n = 6, 314). We demonstrate a 33% delineation improvement for the surgically targetable tumor, and 23% for the complete tumor extent, over a publicly trained model. We anticipate our study to: 1) enable more healthcare studies informed by large diverse data, ensuring meaningful results for rare diseases and underrepresented populations, 2) facilitate further analyses for glioblastoma by releasing our consensus model, and 3) demonstrate the FL effectiveness at such scale and task-complexity as a paradigm shift for multi-site collaborations, alleviating the need for data-sharing.
Collapse
Affiliation(s)
- Sarthak Pati
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Informatics, Technical University of Munich, Munich, Bavaria, Germany
| | - Ujjwal Baid
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chiharu Sako
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Satyam Ghodasara
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michel Bilello
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Suyash Mohan
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Philipp Vollmuth
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Gianluca Brugnara
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Felix Sahm
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Klaus Maier-Hein
- Division of Medical Image Computing, German Cancer Research Center, Heidelberg, Germany
- Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Maximilian Zenk
- Division of Medical Image Computing, German Cancer Research Center, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Wolfgang Wick
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany
| | - Evan Calabrese
- Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Jeffrey Rudie
- Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Javier Villanueva-Meyer
- Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Soonmee Cha
- Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Madhura Ingalhalikar
- Symbiosis Center for Medical Image Analysis, Symbiosis International University, Pune, Maharashtra, India
| | - Manali Jadhav
- Symbiosis Center for Medical Image Analysis, Symbiosis International University, Pune, Maharashtra, India
| | - Umang Pandey
- Symbiosis Center for Medical Image Analysis, Symbiosis International University, Pune, Maharashtra, India
| | - Jitender Saini
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - John Garrett
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Matthew Larson
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Robert Jeraj
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Stuart Currie
- Leeds Teaching Hospitals Trust, Department of Radiology, Leeds, UK
| | - Russell Frood
- Leeds Teaching Hospitals Trust, Department of Radiology, Leeds, UK
| | - Kavi Fatania
- Leeds Teaching Hospitals Trust, Department of Radiology, Leeds, UK
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Chang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | | | | | - Josep Puig
- Department of Radiology (IDI), Girona Biomedical Research Institute (IdIBGi), Josep Trueta University Hospital, Girona, Spain
| | - Johannes Trenkler
- Institute of Neuroradiology, Neuromed Campus (NMC), Kepler University Hospital Linz, Linz, Austria
| | - Josef Pichler
- Department of Neurooncology, Neuromed Campus (NMC), Kepler University Hospital Linz, Linz, Austria
| | - Georg Necker
- Institute of Neuroradiology, Neuromed Campus (NMC), Kepler University Hospital Linz, Linz, Austria
| | - Andreas Haunschmidt
- Institute of Neuroradiology, Neuromed Campus (NMC), Kepler University Hospital Linz, Linz, Austria
| | - Stephan Meckel
- Institute of Neuroradiology, Neuromed Campus (NMC), Kepler University Hospital Linz, Linz, Austria
- Institute of Diagnostic and Interventional Neuroradiology, RKH Klinikum Ludwigsburg, Ludwigsburg, Germany
| | - Gaurav Shukla
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiation Oncology, Christiana Care Health System, Philadelphia, PA, USA
| | - Spencer Liem
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gregory S Alexander
- Department of Radiation Oncology, University of Maryland, Baltimore, MD, USA
| | - Joseph Lombardo
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Joshua D Palmer
- Department of Radiation Oncology, The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Adam E Flanders
- Department of Radiology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam P Dicker
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Haris I Sair
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Malone Center for Engineering in Healthcare, The Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Craig K Jones
- The Malone Center for Engineering in Healthcare, The Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Archana Venkataraman
- Department of Electrical and Computer Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Meirui Jiang
- The Chinese University of Hong Kong, Hong Kong, China
| | - Tiffany Y So
- The Chinese University of Hong Kong, Hong Kong, China
| | - Cheng Chen
- The Chinese University of Hong Kong, Hong Kong, China
| | | | - Qi Dou
- The Chinese University of Hong Kong, Hong Kong, China
| | - Michal Kozubek
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Filip Lux
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Jan Michálek
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Petr Matula
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Miloš Keřkovský
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Masaryk University, Brno and University Hospital Brno, Brno, Czech Republic
| | - Tereza Kopřivová
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Masaryk University, Brno and University Hospital Brno, Brno, Czech Republic
| | - Marek Dostál
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Masaryk University, Brno and University Hospital Brno, Brno, Czech Republic
- Department of Biophysics, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Václav Vybíhal
- Department of Neurosurgery, Faculty of Medicine, Masaryk University, Brno, and University Hospital and Czech Republic, Brno, Czech Republic
| | - Michael A Vogelbaum
- Department of Neuro Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - J Ross Mitchell
- University of Alberta, Edmonton, AB, Canada
- Alberta Machine Intelligence Institute, Edmonton, AB, Canada
| | - Joaquim Farinhas
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | | | - Marco C Pinho
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Divya Reddy
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - James Holcomb
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CaA, USA
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CaA, USA
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Talia Oughourlian
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Akifumi Hagiwara
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Chencai Wang
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Minh-Son To
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
- Division of Surgery and Perioperative Medicine, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Sargam Bhardwaj
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Chee Chong
- South Australia Medical Imaging, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Marc Agzarian
- South Australia Medical Imaging, Flinders Medical Centre, Bedford Park, SA, Australia
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Bernardo C A Teixeira
- Instituto de Neurologia de Curitiba, Curitiba, Paraná, Brazil
- Department of Radiology, Hospital de Clínicas da Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Flávia Sprenger
- Department of Radiology, Hospital de Clínicas da Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - David Menotti
- Department of Informatics, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Diego R Lucio
- Department of Informatics, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Pamela LaMontagne
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Daniel Marcus
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Benedikt Wiestler
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- TranslaTUM (Zentralinstitut für translationale Krebsforschung der Technischen Universität München), Klinikum rechts der Isar, Munich, Germany
| | - Florian Kofler
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- TranslaTUM (Zentralinstitut für translationale Krebsforschung der Technischen Universität München), Klinikum rechts der Isar, Munich, Germany
- Image-Based Biomedical Modeling, Department of Informatics, Technical University of Munich, Munich, Germany
| | - Ivan Ezhov
- Department of Informatics, Technical University of Munich, Munich, Bavaria, Germany
- TranslaTUM (Zentralinstitut für translationale Krebsforschung der Technischen Universität München), Klinikum rechts der Isar, Munich, Germany
- Image-Based Biomedical Modeling, Department of Informatics, Technical University of Munich, Munich, Germany
| | - Marie Metz
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Rajan Jain
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Matthew Lee
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Yvonne W Lui
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Richard McKinley
- Support Center for Advanced Neuroimaging, University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Johannes Slotboom
- Support Center for Advanced Neuroimaging, University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Piotr Radojewski
- Support Center for Advanced Neuroimaging, University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Raphael Meier
- Support Center for Advanced Neuroimaging, University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Roland Wiest
- Support Center for Advanced Neuroimaging, University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Derrick Murcia
- Department of Neurosurgery, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Eric Fu
- Department of Neurosurgery, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Rourke Haas
- Department of Neurosurgery, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - John Thompson
- Department of Neurosurgery, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - David Ryan Ormond
- Department of Neurosurgery, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Chaitra Badve
- Department of Radiology, University Hospitals Cleveland, Cleveland, OH, USA
| | - Andrew E Sloan
- Department of Neurological Surgery, University Hospitals-Seidman Cancer Center, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Vachan Vadmal
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kristin Waite
- National Cancer Institute, National Institute of Health, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - Rivka R Colen
- Department of Radiology, Neuroradiology Division, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linmin Pei
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Murat Ak
- Department of Radiology, Neuroradiology Division, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ashok Srinivasan
- Department of Neuroradiology, University of Michigan, Ann Arbor, MI, USA
| | - J Rajiv Bapuraj
- Department of Neuroradiology, University of Michigan, Ann Arbor, MI, USA
| | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas Wang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Ota Yoshiaki
- Department of Neuroradiology, University of Michigan, Ann Arbor, MI, USA
| | - Toshio Moritani
- Department of Neuroradiology, University of Michigan, Ann Arbor, MI, USA
| | - Sevcan Turk
- Department of Neuroradiology, University of Michigan, Ann Arbor, MI, USA
| | - Joonsang Lee
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Snehal Prabhudesai
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Fanny Morón
- Department of Radiology, Baylor College of Medicine, Houston, TX, USA
| | - Jacob Mandel
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Konstantinos Kamnitsas
- Department of Computing, Imperial College London, London, UK
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Ben Glocker
- Department of Computing, Imperial College London, London, UK
| | - Luke V M Dixon
- Department of Radiology, Imperial College NHS Healthcare Trust, London, UK
| | - Matthew Williams
- Computational Oncology Group, Institute for Global Health Innovation, Imperial College London, London, UK
| | - Peter Zampakis
- Department of NeuroRadiology, University of Patras, Patras, Greece
| | | | - Panagiotis Tsiganos
- Clinical Radiology Laboratory, Department of Medicine, University of Patras, Patras, Greece
| | - Sotiris Alexiou
- Department of Electrical and Computer Engineering, University of Patras, Patras, Greece
| | - Ilias Haliassos
- Department of Neuro-Oncology, University of Patras, Patras, Greece
| | - Evangelia I Zacharaki
- Department of Electrical and Computer Engineering, University of Patras, Patras, Greece
| | | | | | | | | | | | | | - Sung Soo Ahn
- Yonsei University College of Medicine, Seoul, Korea
| | - Bing Luo
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI, USA
| | - Laila Poisson
- Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - Ning Wen
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI, USA
- SJTU-Ruijin-UIH Institute for Medical Imaging Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | | | - Ruchika Verma
- Alberta Machine Intelligence Institute, Edmonton, AB, Canada
- Case Western Reserve University, Cleveland, OH, USA
| | - Rohan Bareja
- Case Western Reserve University, Cleveland, OH, USA
| | - Ipsa Yadav
- Case Western Reserve University, Cleveland, OH, USA
| | | | - Neeraj Kumar
- University of Alberta, Edmonton, AB, Canada
- Alberta Machine Intelligence Institute, Edmonton, AB, Canada
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Sebastian R van der Voort
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Ahmed Alafandi
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Fatih Incekara
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, Netherlands
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Maarten M J Wijnenga
- Department of Neurology, Brain Tumor Center, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Georgios Kapsas
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Renske Gahrmann
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Joost W Schouten
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Hendrikus J Dubbink
- Department of Pathology, Brain Tumor Center, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Arnaud J P E Vincent
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Martin J van den Bent
- Department of Neurology, Brain Tumor Center, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Pim J French
- Department of Neurology, Brain Tumor Center, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Stefan Klein
- Biomedical Imaging Group Rotterdam, Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Yading Yuan
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sonam Sharma
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tzu-Chi Tseng
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saba Adabi
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Simone P Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Olivier Keunen
- Translation Radiomics, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Ann-Christin Hau
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Luxembourg Center of Neuropathology, Laboratoire National De Santé, Luxembourg, Luxembourg
| | - Martin Vallières
- Department of Computer Science, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalière Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - David Fortin
- Centre de Recherche du Centre Hospitalière Universitaire de Sherbrooke, Sherbrooke, QC, Canada
- Division of Neurosurgery and Neuro-Oncology, Faculty of Medicine and Health Science, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Martin Lepage
- Centre de Recherche du Centre Hospitalière Universitaire de Sherbrooke, Sherbrooke, QC, Canada
- Department of Nuclear Medicine and Radiobiology, Sherbrooke Molecular Imaging Centre, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Bennett Landman
- Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, USA
| | - Karthik Ramadass
- Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kaiwen Xu
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Silky Chotai
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lola B Chambless
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Akshitkumar Mistry
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Reid C Thompson
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yuriy Gusev
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington, DC, USA
| | - Krithika Bhuvaneshwar
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington, DC, USA
| | - Anousheh Sayah
- Division of Neuroradiology & Neurointerventional Radiology, Department of Radiology, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Camelia Bencheqroun
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington, DC, USA
| | - Anas Belouali
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington, DC, USA
| | - Subha Madhavan
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University, Washington, DC, USA
| | - Thomas C Booth
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
- Department of Neuroradiology, Ruskin Wing, King's College Hospital NHS Foundation Trust, London, UK
| | - Alysha Chelliah
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Marc Modat
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Haris Shuaib
- Stoke Mandeville Hospital, Mandeville Road, Aylesbury, UK
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Carmen Dragos
- Stoke Mandeville Hospital, Mandeville Road, Aylesbury, UK
| | | | | | | | | | - Shady Gamal
- University of Cairo School of Medicine, Giza, Egypt
| | | | | | | | - Ji Eun Park
- Department of Radiology, Asan Medical Center, Seoul, South Korea
| | - Jihye Yun
- Department of Radiology, Asan Medical Center, Seoul, South Korea
| | - Ho Sung Kim
- Department of Radiology, Asan Medical Center, Seoul, South Korea
| | - Abhishek Mahajan
- The Clatterbridge Cancer Centre NHS Foundation Trust Pembroke Place, Liverpool, UK
| | - Mark Muzi
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Sean Benson
- Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Regina G H Beets-Tan
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, Netherlands
- GROW School of Oncology and Developmental Biology, Maastricht, Netherlands
| | - Jonas Teuwen
- Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | - William Escobar
- Clínica Imbanaco Grupo Quirón Salud, Cali, Colombia
- Universidad del Valle, Cali, Colombia
| | | | - Jose Bernal
- Universidad del Valle, Cali, Colombia
- The University of Edinburgh, Edinburgh, UK
| | | | - Joseph Choi
- Department of Industrial and Systems Engineering, University of Iowa, Iowa, USA
| | - Stephen Baek
- Department of Industrial and Systems Engineering, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - Yusung Kim
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - Heba Ismael
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - Bryan Allen
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - John M Buatti
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | | | - Hongwei Li
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Andrea Bink
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Bertrand Pouymayou
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | | | - Joel Saltz
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York, USA
| | - Prateek Prasanna
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York, USA
| | - Sampurna Shrestha
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York, USA
| | - Kartik M Mani
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York, USA
- Department of Radiation Oncology, Stony Brook University, Stony Brook, NY, USA
| | - David Payne
- Department of Radiology, Stony Brook University, Stony Brook, NY, USA
| | - Tahsin Kurc
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York, USA
- Scientific Data Group, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Enrique Pelaez
- Escuela Superior Politecnica del Litoral, Guayaquil, Guayas, Ecuador
| | | | - Francis Loayza
- Escuela Superior Politecnica del Litoral, Guayaquil, Guayas, Ecuador
| | | | | | | | | | - Franco Vera
- Universidad de Concepción, Concepción, Biobío, Chile
| | - Elvis Ríos
- Universidad de Concepción, Concepción, Biobío, Chile
| | - Eduardo López
- Universidad de Concepción, Concepción, Biobío, Chile
| | - Sergio A Velastin
- School of Electronic Engineering and Computer Science, Queen Mary University of London, London, UK
| | - Godwin Ogbole
- Department of Radiology, University College Hospital Ibadan, Oyo, Nigeria
| | - Mayowa Soneye
- Department of Radiology, University College Hospital Ibadan, Oyo, Nigeria
| | - Dotun Oyekunle
- Department of Radiology, University College Hospital Ibadan, Oyo, Nigeria
| | | | - Babatunde Osobu
- Department of Radiology, University College Hospital Ibadan, Oyo, Nigeria
| | - Mustapha Shu'aibu
- Department of Radiology, Muhammad Abdullahi Wase Teaching Hospital, Kano, Nigeria
| | - Adeleye Dorcas
- Department of Radiology, Obafemi Awolowo University Ile-Ife, Ile-Ife, Osun, Nigeria
| | - Farouk Dako
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amber L Simpson
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
- School of Computing, Queen's University, Kingston, ON, Canada
| | - Mohammad Hamghalam
- School of Computing, Queen's University, Kingston, ON, Canada
- Department of Electrical Engineering, Qazvin Branch, Islamic Azad University, Qazvin, Iran
| | - Jacob J Peoples
- School of Computing, Queen's University, Kingston, ON, Canada
| | - Ricky Hu
- School of Computing, Queen's University, Kingston, ON, Canada
| | - Anh Tran
- School of Computing, Queen's University, Kingston, ON, Canada
| | - Danielle Cutler
- The Faculty of Arts & Sciences, Queen's University, Kingston, ON, Canada
| | - Fabio Y Moraes
- Department of Oncology, Queen's University, Kingston, ON, Canada
| | - Michael A Boss
- Center for Research and Innovation, American College of Radiology, Philadelphia, PA, USA
| | - James Gimpel
- Center for Research and Innovation, American College of Radiology, Philadelphia, PA, USA
| | - Deepak Kattil Veettil
- Center for Research and Innovation, American College of Radiology, Philadelphia, PA, USA
| | - Kendall Schmidt
- Data Science Institute, American College of Radiology, Reston, VA, USA
| | - Brian Bialecki
- Data Science Institute, American College of Radiology, Reston, VA, USA
| | - Sailaja Marella
- Center for Research and Innovation, American College of Radiology, Philadelphia, PA, USA
| | - Cynthia Price
- Center for Research and Innovation, American College of Radiology, Philadelphia, PA, USA
| | - Lisa Cimino
- Center for Research and Innovation, American College of Radiology, Philadelphia, PA, USA
| | - Charles Apgar
- Center for Research and Innovation, American College of Radiology, Philadelphia, PA, USA
| | | | - Bjoern Menze
- Department of Informatics, Technical University of Munich, Munich, Bavaria, Germany
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Jill S Barnholtz-Sloan
- National Cancer Institute, National Institute of Health, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
- Center for Biomedical Informatics and Information Technology, National Cancer Institute (NCI), National Institute of Health, Bethesda, MD, USA
| | | | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
30
|
García-García S, García-Galindo M, Arrese I, Sarabia R, Cepeda S. Current Evidence, Limitations and Future Challenges of Survival Prediction for Glioblastoma Based on Advanced Noninvasive Methods: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58121746. [PMID: 36556948 PMCID: PMC9786785 DOI: 10.3390/medicina58121746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/16/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
Background and Objectives: Survival estimation for patients diagnosed with Glioblastoma (GBM) is an important information to consider in patient management and communication. Despite some known risk factors, survival estimation remains a major challenge. Novel non-invasive technologies such as radiomics and artificial intelligence (AI) have been implemented to increase the accuracy of these predictions. In this article, we reviewed and discussed the most significant available research on survival estimation for GBM through advanced non-invasive methods. Materials and Methods: PubMed database was queried for articles reporting on survival prognosis for GBM through advanced image and data management methods. Articles including in their title or abstract the following terms were initially screened: ((glioma) AND (survival)) AND ((artificial intelligence) OR (radiomics)). Exclusively English full-text articles, reporting on humans, published as of 1 September 2022 were considered. Articles not reporting on overall survival, evaluating the effects of new therapies or including other tumors were excluded. Research with a radiomics-based methodology were evaluated using the radiomics quality score (RQS). Results: 382 articles were identified. After applying the inclusion criteria, 46 articles remained for further analysis. These articles were thoroughly assessed, summarized and discussed. The results of the RQS revealed some of the limitations of current radiomics investigation on this field. Limitations of analyzed studies included data availability, patient selection and heterogeneity of methodologies. Future challenges on this field are increasing data availability, improving the general understanding of how AI handles data and establishing solid correlations between image features and tumor's biology. Conclusions: Radiomics and AI methods of data processing offer a new paradigm of possibilities to tackle the question of survival prognosis in GBM.
Collapse
Affiliation(s)
- Sergio García-García
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
- Correspondence:
| | - Manuel García-Galindo
- Faculty of Medicine, University of Valladolid, Avenida Ramón y Cajal 7, 47003 Valladolid, Spain
| | - Ignacio Arrese
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
| | - Rosario Sarabia
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
| | - Santiago Cepeda
- Department of Neurosurgery, University Hospital Río Hortega, Dulzaina 2, 47012 Valladolid, Spain
| |
Collapse
|
31
|
di Noia C, Grist JT, Riemer F, Lyasheva M, Fabozzi M, Castelli M, Lodi R, Tonon C, Rundo L, Zaccagna F. Predicting Survival in Patients with Brain Tumors: Current State-of-the-Art of AI Methods Applied to MRI. Diagnostics (Basel) 2022; 12:diagnostics12092125. [PMID: 36140526 PMCID: PMC9497964 DOI: 10.3390/diagnostics12092125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Given growing clinical needs, in recent years Artificial Intelligence (AI) techniques have increasingly been used to define the best approaches for survival assessment and prediction in patients with brain tumors. Advances in computational resources, and the collection of (mainly) public databases, have promoted this rapid development. This narrative review of the current state-of-the-art aimed to survey current applications of AI in predicting survival in patients with brain tumors, with a focus on Magnetic Resonance Imaging (MRI). An extensive search was performed on PubMed and Google Scholar using a Boolean research query based on MeSH terms and restricting the search to the period between 2012 and 2022. Fifty studies were selected, mainly based on Machine Learning (ML), Deep Learning (DL), radiomics-based methods, and methods that exploit traditional imaging techniques for survival assessment. In addition, we focused on two distinct tasks related to survival assessment: the first on the classification of subjects into survival classes (short and long-term or eventually short, mid and long-term) to stratify patients in distinct groups. The second focused on quantification, in days or months, of the individual survival interval. Our survey showed excellent state-of-the-art methods for the first, with accuracy up to ∼98%. The latter task appears to be the most challenging, but state-of-the-art techniques showed promising results, albeit with limitations, with C-Index up to ∼0.91. In conclusion, according to the specific task, the available computational methods perform differently, and the choice of the best one to use is non-univocal and dependent on many aspects. Unequivocally, the use of features derived from quantitative imaging has been shown to be advantageous for AI applications, including survival prediction. This evidence from the literature motivates further research in the field of AI-powered methods for survival prediction in patients with brain tumors, in particular, using the wealth of information provided by quantitative MRI techniques.
Collapse
Affiliation(s)
- Christian di Noia
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
| | - James T. Grist
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Oxford Centre for Clinical Magnetic Research Imaging, University of Oxford, Oxford OX3 9DU, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK
| | - Frank Riemer
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Maria Lyasheva
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Miriana Fabozzi
- Centro Medico Polispecialistico (CMO), 80058 Torre Annunziata, Italy
| | - Mauro Castelli
- NOVA Information Management School (NOVA IMS), Universidade NOVA de Lisboa, Campus de Campolide, 1070-312 Lisboa, Portugal
| | - Raffaele Lodi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Caterina Tonon
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Leonardo Rundo
- Department of Information and Electrical Engineering and Applied Mathematics, University of Salerno, 84084 Fisciano, Italy
| | - Fulvio Zaccagna
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, 40125 Bologna, Italy
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
- Correspondence: ; Tel.: +39-0514969951
| |
Collapse
|
32
|
Yan L, Gao N, Ai F, Zhao Y, Kang Y, Chen J, Weng Y. Deep learning models for predicting the survival of patients with chondrosarcoma based on a surveillance, epidemiology, and end results analysis. Front Oncol 2022; 12:967758. [PMID: 36072795 PMCID: PMC9442032 DOI: 10.3389/fonc.2022.967758] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAccurate prediction of prognosis is critical for therapeutic decisions in chondrosarcoma patients. Several prognostic models have been created utilizing multivariate Cox regression or binary classification-based machine learning approaches to predict the 3- and 5-year survival of patients with chondrosarcoma, but few studies have investigated the results of combining deep learning with time-to-event prediction. Compared with simplifying the prediction as a binary classification problem, modeling the probability of an event as a function of time by combining it with deep learning can provide better accuracy and flexibility.Materials and methodsPatients with the diagnosis of chondrosarcoma between 2000 and 2018 were extracted from the Surveillance, Epidemiology, and End Results (SEER) registry. Three algorithms—two based on neural networks (DeepSurv, neural multi-task logistic regression [NMTLR]) and one on ensemble learning (random survival forest [RSF])—were selected for training. Meanwhile, a multivariate Cox proportional hazards (CoxPH) model was also constructed for comparison. The dataset was randomly divided into training and testing datasets at a ratio of 7:3. Hyperparameter tuning was conducted through a 1000-repeated random search with 5-fold cross-validation on the training dataset. The model performance was assessed using the concordance index (C-index), Brier score, and Integrated Brier Score (IBS). The accuracy of predicting 1-, 3-, 5- and 10-year survival was evaluated using receiver operating characteristic curves (ROC), calibration curves, and the area under the ROC curves (AUC).ResultsA total of 3145 patients were finally enrolled in our study. The mean age at diagnosis was 52 ± 18 years, 1662 of the 3145 patients were male (53%), and mean survival time was 83 ± 67 months. Two deep learning models outperformed the RSF and classical CoxPH models, with the C-index on test datasets achieving values of 0.832 (DeepSurv) and 0.821 (NMTLR). The DeepSurv model produced better accuracy and calibrated survival estimates in predicting 1-, 3- 5- and 10-year survival (AUC:0.895-0.937). We deployed the DeepSurv model as a web application for use in clinical practice; it can be accessed through https://share.streamlit.io/whuh-ml/chondrosarcoma/Predict/app.py.ConclusionsTime-to-event prediction models based on deep learning algorithms are successful in predicting chondrosarcoma prognosis, with DeepSurv producing the best discriminative performance and calibration.
Collapse
Affiliation(s)
- Lizhao Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nan Gao
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangxing Ai
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingsong Zhao
- Department of Orthopaedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Kang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianghai Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jianghai Chen, ; Yuxiong Weng,
| | - Yuxiong Weng
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jianghai Chen, ; Yuxiong Weng,
| |
Collapse
|
33
|
Xu C, Peng Y, Zhu W, Chen Z, Li J, Tan W, Zhang Z, Chen X. An automated approach for predicting glioma grade and survival of LGG patients using CNN and radiomics. Front Oncol 2022; 12:969907. [PMID: 36033433 PMCID: PMC9413530 DOI: 10.3389/fonc.2022.969907] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/15/2022] [Indexed: 12/24/2022] Open
Abstract
Objectives To develop and validate an efficient and automatically computational approach for stratifying glioma grades and predicting survival of lower-grade glioma (LGG) patients using an integration of state-of-the-art convolutional neural network (CNN) and radiomics. Method This retrospective study reviewed 470 preoperative MR images of glioma from BraTs public dataset (n=269) and Jinling hospital (n=201). A fully automated pipeline incorporating tumor segmentation and grading was developed, which can avoid variability and subjectivity of manual segmentations. First, an integrated approach by fusing CNN features and radiomics features was employed to stratify glioma grades. Then, a deep-radiomics signature based on the integrated approach for predicting survival of LGG patients was developed and subsequently validated in an independent cohort. Results The performance of tumor segmentation achieved a Dice coefficient of 0.81. The intraclass correlation coefficients (ICCs) of the radiomics features between the segmentation network and physicians were all over 0.75. The performance of glioma grading based on integrated approach achieved the area under the curve (AUC) of 0.958, showing the effectiveness of the integrated approach. The multivariable Cox regression results demonstrated that the deep-radiomics signature remained an independent prognostic factor and the integrated nomogram showed significantly better performance than the clinical nomogram in predicting overall survival of LGG patients (C-index: 0.865 vs. 0.796, P=0.005). Conclusion The proposed integrated approach can be noninvasively and efficiently applied in prediction of gliomas grade and survival. Moreover, our fully automated pipeline successfully achieved computerized segmentation instead of manual segmentation, which shows the potential to be a reproducible approach in clinical practice.
Collapse
Affiliation(s)
- Chenan Xu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
| | - Yuanyuan Peng
- School of Electronics and Information Engineering and Medical Image Processing, Analysis and Visualization Lab, Soochow University, Suzhou, China
| | - Weifang Zhu
- School of Electronics and Information Engineering and Medical Image Processing, Analysis and Visualization Lab, Soochow University, Suzhou, China
| | - Zhongyue Chen
- School of Electronics and Information Engineering and Medical Image Processing, Analysis and Visualization Lab, Soochow University, Suzhou, China
| | - Jianrui Li
- Department of Diagnostic Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenhao Tan
- School of Electronics and Information Engineering and Medical Image Processing, Analysis and Visualization Lab, Soochow University, Suzhou, China
| | - Zhiqiang Zhang
- Department of Diagnostic Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- *Correspondence: Zhiqiang Zhang, ; Xinjian Chen,
| | - Xinjian Chen
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
- School of Electronics and Information Engineering and Medical Image Processing, Analysis and Visualization Lab, Soochow University, Suzhou, China
- *Correspondence: Zhiqiang Zhang, ; Xinjian Chen,
| |
Collapse
|
34
|
Guan X, Lu N, Zhang J. Evaluation of Epidermal Growth Factor Receptor 2 Status in Gastric Cancer by CT-Based Deep Learning Radiomics Nomogram. Front Oncol 2022; 12:905203. [PMID: 35898877 PMCID: PMC9309372 DOI: 10.3389/fonc.2022.905203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/21/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose To explore the role of computed tomography (CT)-based deep learning and radiomics in preoperative evaluation of epidermal growth factor receptor 2 (HER2) status in gastric cancer. Materials and methods The clinical data on gastric cancer patients were evaluated retrospectively, and 357 patients were chosen for this study (training cohort: 249; test cohort: 108). The preprocessed enhanced CT arterial phase images were selected for lesion segmentation, radiomics and deep learning feature extraction. We integrated deep learning features and radiomic features (Inte). Four methods were used for feature selection. We constructed models with support vector machine (SVM) or random forest (RF), respectively. The area under the receiver operating characteristics curve (AUC) was used to assess the performance of these models. We also constructed a nomogram including Inte-feature scores and clinical factors. Results The radiomics-SVM model showed good classification performance (AUC, training cohort: 0.8069; test cohort: 0.7869). The AUC of the ResNet50-SVM model and the Inte-SVM model in the test cohort were 0.8955 and 0.9055. The nomogram also showed excellent discrimination achieving greater AUC (training cohort, 0.9207; test cohort, 0.9224). Conclusion CT-based deep learning radiomics nomogram can accurately and effectively assess the HER2 status in patients with gastric cancer before surgery and it is expected to assist physicians in clinical decision-making and facilitates individualized treatment planning.
Collapse
Affiliation(s)
- Xiao Guan
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Na Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | | |
Collapse
|
35
|
A Survey of Radiomics in Precision Diagnosis and Treatment of Adult Gliomas. J Clin Med 2022; 11:jcm11133802. [PMID: 35807084 PMCID: PMC9267404 DOI: 10.3390/jcm11133802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/18/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
Glioma is the most common primary malignant tumor of the adult central nervous system (CNS), which mostly shows invasive growth. In most cases, surgery is often difficult to completely remove, and the recurrence rate and mortality of patients are high. With the continuous development of molecular genetics and the great progress of molecular biology technology, more and more molecular biomarkers have been proved to have important guiding significance in the individualized diagnosis, treatment, and prognosis evaluation of glioma. With the updates of the World Health Organization (WHO) classification of tumors of the CNS in 2021, the diagnosis and treatment of glioma has entered the era of precision medicine in the true sense. Due to its ability to non-invasively achieve accurate identification of glioma from other intracranial tumors, and to predict the grade, genotyping, treatment response, and prognosis of glioma, which provides a scientific basis for the clinical application of individualized diagnosis and treatment model of glioma, radiomics has become a research hotspot in the field of precision medicine. This paper reviewed the research related to radiomics of adult gliomas published in recent years and summarized the research proceedings of radiomics in differential diagnosis, preoperative grading and genotyping, treatment and efficacy evaluation, and survival prediction of adult gliomas.
Collapse
|
36
|
Mazaheri Y, Thakur SB, Bitencourt AGV, Lo Gullo R, Hötker AM, Bates DDB, Akin O. Evaluation of cancer outcome assessment using MRI: A review of deep-learning methods. BJR Open 2022; 4:20210072. [PMID: 36105425 PMCID: PMC9459949 DOI: 10.1259/bjro.20210072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Accurate evaluation of tumor response to treatment is critical to allow personalized treatment regimens according to the predicted response and to support clinical trials investigating new therapeutic agents by providing them with an accurate response indicator. Recent advances in medical imaging, computer hardware, and machine-learning algorithms have resulted in the increased use of these tools in the field of medicine as a whole and specifically in cancer imaging for detection and characterization of malignant lesions, prognosis, and assessment of treatment response. Among the currently available imaging techniques, magnetic resonance imaging (MRI) plays an important role in the evaluation of treatment assessment of many cancers, given its superior soft-tissue contrast and its ability to allow multiplanar imaging and functional evaluation. In recent years, deep learning (DL) has become an active area of research, paving the way for computer-assisted clinical and radiological decision support. DL can uncover associations between imaging features that cannot be visually identified by the naked eye and pertinent clinical outcomes. The aim of this review is to highlight the use of DL in the evaluation of tumor response assessed on MRI. In this review, we will first provide an overview of common DL architectures used in medical imaging research in general. Then, we will review the studies to date that have applied DL to magnetic resonance imaging for the task of treatment response assessment. Finally, we will discuss the challenges and opportunities of using DL within the clinical workflow.
Collapse
Affiliation(s)
| | | | | | - Roberto Lo Gullo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Andreas M. Hötker
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - David D B Bates
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Oguz Akin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, United States
| |
Collapse
|
37
|
Guo S, Wang L, Chen Q, Wang L, Zhang J, Zhu Y. Multimodal MRI Image Decision Fusion-Based Network for Glioma Classification. Front Oncol 2022; 12:819673. [PMID: 35280828 PMCID: PMC8907622 DOI: 10.3389/fonc.2022.819673] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose Glioma is the most common primary brain tumor, with varying degrees of aggressiveness and prognosis. Accurate glioma classification is very important for treatment planning and prognosis prediction. The main purpose of this study is to design a novel effective algorithm for further improving the performance of glioma subtype classification using multimodal MRI images. Method MRI images of four modalities for 221 glioma patients were collected from Computational Precision Medicine: Radiology-Pathology 2020 challenge, including T1, T2, T1ce, and fluid-attenuated inversion recovery (FLAIR) MRI images, to classify astrocytoma, oligodendroglioma, and glioblastoma. We proposed a multimodal MRI image decision fusion-based network for improving the glioma classification accuracy. First, the MRI images of each modality were input into a pre-trained tumor segmentation model to delineate the regions of tumor lesions. Then, the whole tumor regions were centrally clipped from original MRI images followed by max-min normalization. Subsequently, a deep learning-based network was designed based on a unified DenseNet structure, which extracts features through a series of dense blocks. After that, two fully connected layers were used to map the features into three glioma subtypes. During the training stage, we used the images of each modality after tumor segmentation to train the network to obtain its best accuracy on our testing set. During the inferring stage, a linear weighted module based on a decision fusion strategy was applied to assemble the predicted probabilities of the pre-trained models obtained in the training stage. Finally, the performance of our method was evaluated in terms of accuracy, area under the curve (AUC), sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), etc. Results The proposed method achieved an accuracy of 0.878, an AUC of 0.902, a sensitivity of 0.772, a specificity of 0.930, a PPV of 0.862, an NPV of 0.949, and a Cohen's Kappa of 0.773, which showed a significantly higher performance than existing state-of-the-art methods. Conclusion Compared with current studies, this study demonstrated the effectiveness and superiority in the overall performance of our proposed multimodal MRI image decision fusion-based network method for glioma subtype classification, which would be of enormous potential value in clinical practice.
Collapse
Affiliation(s)
- Shunchao Guo
- Key Laboratory of Intelligent Medical Image Analysis and Precise Diagnosis of Guizhou Province, College of Computer Science and Technology, Guizhou University, Guiyang, China.,College of Computer and Information, Qiannan Normal University for Nationalities, Duyun, China
| | - Lihui Wang
- Key Laboratory of Intelligent Medical Image Analysis and Precise Diagnosis of Guizhou Province, College of Computer Science and Technology, Guizhou University, Guiyang, China
| | - Qijian Chen
- Key Laboratory of Intelligent Medical Image Analysis and Precise Diagnosis of Guizhou Province, College of Computer Science and Technology, Guizhou University, Guiyang, China
| | - Li Wang
- Key Laboratory of Intelligent Medical Image Analysis and Precise Diagnosis of Guizhou Province, College of Computer Science and Technology, Guizhou University, Guiyang, China
| | - Jian Zhang
- Key Laboratory of Intelligent Medical Image Analysis and Precise Diagnosis of Guizhou Province, College of Computer Science and Technology, Guizhou University, Guiyang, China
| | - Yuemin Zhu
- CREATIS, CNRS UMR 5220, Inserm U1044, INSA Lyon, University of Lyon, Lyon, France
| |
Collapse
|
38
|
Cattell R, Ying J, Lei L, Ding J, Chen S, Serrano Sosa M, Huang C. Preoperative prediction of lymph node metastasis using deep learning-based features. Vis Comput Ind Biomed Art 2022; 5:8. [PMID: 35254557 PMCID: PMC8901808 DOI: 10.1186/s42492-022-00104-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/17/2022] [Indexed: 11/10/2022] Open
Abstract
Lymph node involvement increases the risk of breast cancer recurrence. An accurate non-invasive assessment of nodal involvement is valuable in cancer staging, surgical risk, and cost savings. Radiomics has been proposed to pre-operatively predict sentinel lymph node (SLN) status; however, radiomic models are known to be sensitive to acquisition parameters. The purpose of this study was to develop a prediction model for preoperative prediction of SLN metastasis using deep learning-based (DLB) features and compare its predictive performance to state-of-the-art radiomics. Specifically, this study aimed to compare the generalizability of radiomics vs DLB features in an independent test set with dissimilar resolution. Dynamic contrast-enhancement images from 198 patients (67 positive SLNs) were used in this study. Of these subjects, 163 had an in-plane resolution of 0.7 × 0.7 mm2, which were randomly divided into a training set (approximately 67%) and a validation set (approximately 33%). The remaining 35 subjects with a different in-plane resolution (0.78 × 0.78 mm2) were treated as independent testing set for generalizability. Two methods were employed: (1) conventional radiomics (CR), and (2) DLB features which replaced hand-curated features with pre-trained VGG-16 features. The threshold determined using the training set was applied to the independent validation and testing dataset. Same feature reduction, feature selection, model creation procedures were used for both approaches. In the validation set (same resolution as training), the DLB model outperformed the CR model (accuracy 83% vs 80%). Furthermore, in the independent testing set of the dissimilar resolution, the DLB model performed markedly better than the CR model (accuracy 77% vs 71%). The predictive performance of the DLB model outperformed the CR model for this task. More interestingly, these improvements were seen particularly in the independent testing set of dissimilar resolution. This could indicate that DLB features can ultimately result in a more generalizable model.
Collapse
Affiliation(s)
- Renee Cattell
- Department of Biomedical Engineering, Stony Brook University, NY, 11794, Stony Brook, USA.,Department of Radiation Oncology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Jia Ying
- Department of Biomedical Engineering, Stony Brook University, NY, 11794, Stony Brook, USA
| | - Lan Lei
- Program in Public Health, Stony Brook Medicine, Stony Brook, NY, 11794, USA.,Department of Medicine, Northside Hospital Gwinnett, GA, 30046, Lawrenceville, USA
| | - Jie Ding
- Department of Biomedical Engineering, Stony Brook University, NY, 11794, Stony Brook, USA.,Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Shenglan Chen
- Department of Biomedical Engineering, Stony Brook University, NY, 11794, Stony Brook, USA.,Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Mario Serrano Sosa
- Department of Biomedical Engineering, Stony Brook University, NY, 11794, Stony Brook, USA
| | - Chuan Huang
- Department of Biomedical Engineering, Stony Brook University, NY, 11794, Stony Brook, USA. .,Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
39
|
Zhou Q, Xue C, Ke X, Zhou J. Treatment Response and Prognosis Evaluation in High-Grade Glioma: An Imaging Review Based on MRI. J Magn Reson Imaging 2022; 56:325-340. [PMID: 35129845 DOI: 10.1002/jmri.28103] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/19/2022] Open
Abstract
In recent years, the development of advanced magnetic resonance imaging (MRI) technology and machine learning (ML) have created new tools for evaluating treatment response and prognosis of patients with high-grade gliomas (HGG); however, patient prognosis has not improved significantly. This is mainly due to the heterogeneity between and within HGG tumors, resulting in standard treatment methods not benefitting all patients. Moreover, the survival of patients with HGG is not only related to tumor cells, but also to noncancer cells in the tumor microenvironment (TME). Therefore, during preoperative diagnosis and follow-up treatment of patients with HGG, noninvasive imaging markers are needed to characterize intratumoral heterogeneity, and then to evaluate treatment response and predict prognosis, timeously adjust treatment strategies, and achieve individualized diagnosis and treatment. In this review, we summarize the research progress of conventional MRI, advanced MRI technology, and ML in evaluation of treatment response and prognosis of patients with HGG. We further discuss the significance of the TME in the prognosis of HGG patients, associate imaging features with the TME, indirectly reflecting the heterogeneity within the tumor, and shifting treatment strategies from tumor cells alone to systemic therapy of the TME, which may be a major development direction in the future. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY STAGE: 4.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.,Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| |
Collapse
|
40
|
Ding J, Zhao R, Qiu Q, Chen J, Duan J, Cao X, Yin Y. Developing and validating a deep learning and radiomic model for glioma grading using multiplanar reconstructed magnetic resonance contrast-enhanced T1-weighted imaging: a robust, multi-institutional study. Quant Imaging Med Surg 2022; 12:1517-1528. [PMID: 35111644 DOI: 10.21037/qims-21-722] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022]
Abstract
Background Although surgical pathology or biopsy are considered the gold standard for glioma grading, these procedures have limitations. This study set out to evaluate and validate the predictive performance of a deep learning radiomics model based on contrast-enhanced T1-weighted multiplanar reconstruction images for grading gliomas. Methods Patients from three institutions who diagnosed with gliomas by surgical specimen and multiplanar reconstructed (MPR) images were enrolled in this study. The training cohort included 101 patients from institution 1, including 43 high-grade glioma (HGG) patients and 58 low-grade glioma (LGG) patients, while the test cohorts consisted of 50 patients from institutions 2 and 3 (25 HGG patients, 25 LGG patients). We then extracted radiomics features and deep learning features using six pretrained models from the MPR images. The Spearman correlation test and the recursive elimination feature selection method were used to reduce the redundancy and select most predictive features. Subsequently, three classifiers were used to construct classification models. The performance of the grading models was evaluated using the area under the receiver operating curve, sensitivity, specificity, accuracy, precision, and negative predictive value. Finally, the prediction performances of the test cohort were compared to determine the optimal classification model. Results For the training cohort, 62% (13 out of 21) of the classification models constructed with MPR images from multiple planes outperformed those constructed with single-plane MPR images, and 61% (11 out of 18) of classification models constructed with both radiomics features and deep learning features had higher area under the curve (AUC) values than those constructed with only radiomics or deep learning features. The optimal model was a random forest model that combined radiomic features and VGG16 deep learning features derived from MPR images, which achieved AUC of 0.847 in the training cohort and 0.898 in the test cohort. In the test cohort, the sensitivity, specificity, and accuracy of the optimal model were 0.840, 0.760, and 0.800, respectively. Conclusions Multiplanar CE-T1W MPR imaging features are more effective than features from single planes when differentiating HGG and LGG. The combination of deep learning features and radiomics features can effectively grade glioma and assist clinical decision-making.
Collapse
Affiliation(s)
- Jialin Ding
- School of Physics and Electronics, Shandong Normal University, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Rubin Zhao
- Department of Radiation Oncology and Technology, Linyi People's Hospital, Linyi, China
| | - Qingtao Qiu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinhu Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinghao Duan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiujuan Cao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yong Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
41
|
Li Q, Feng QX, Qi L, Liu C, Zhang J, Yang G, Zhang YD, Liu XS. Prognostic aspects of lymphovascular invasion in localized gastric cancer: new insights into the radiomics and deep transfer learning from contrast-enhanced CT imaging. Abdom Radiol (NY) 2022; 47:496-507. [PMID: 34766197 DOI: 10.1007/s00261-021-03309-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/03/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Lymphovascular invasion (LVI) is a factor significantly impacting treatment and outcome of patients with gastric cancer (GC). We aimed to investigate prognostic aspects of a preoperative LVI prediction in GC using radiomics and deep transfer learning (DTL) from contrast-enhanced CT (CECT) imaging. METHODS A total of 1062 GC patients (728 training and 334 testing) between Jan 2014 and Dec 2018 undergoing gastrectomy were retrospectively included. Based on CECT imaging, we built two gastric imaging (GI) markers, GI-marker-1 from radiomics and GI-marker-2 from DTL features, to decode LVI status. We then integrated demographics, clinical data, GI markers, radiologic interpretation, and biopsies into a Gastric Cancer Risk (GRISK) model for predicting LVI. The performance of GRISK model was tested and applied to predict survival outcomes in GC patients. Furthermore, the prognosis between LVI (+) and LVI (-) patients was compared in chemotherapy and non-chemotherapy cohorts, respectively. RESULTS GI-marker-1 and GI-marker-2 yield similar performance in predicting LVI in training and testing dataset. The GRISK model yields the diagnostic performance with AUC of 0.755 (95% CI 0.719-0.790) and 0.725 (95% CI 0.669-0.781) in training and testing dataset. Patients with LVI (+) trend toward lower progression-free survival (PFS) and overall survival (OS). The difference of prognosis between LVI (+) and LVI (-) was more noticeable in non-chemotherapy than that in chemotherapy group. CONCLUSION Radiomics and deep transfer learning features on CECT demonstrate potential power for predicting LVI in GC patients. Prospective use of a GRISK model can help to optimize individualized treatment decisions and predict survival outcomes.
Collapse
Affiliation(s)
- Qiong Li
- Department of Radiology, the First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, Jiangsu, China
| | - Qiu-Xia Feng
- Department of Radiology, the First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, Jiangsu, China
| | - Liang Qi
- Department of Radiology, the First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, Jiangsu, China
| | - Chang Liu
- Department of Radiology, the First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, Jiangsu, China
| | - Jing Zhang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, 200062, China
| | - Guang Yang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, 200062, China
| | - Yu-Dong Zhang
- Department of Radiology, the First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, Jiangsu, China.
| | - Xi-Sheng Liu
- Department of Radiology, the First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, Jiangsu, China.
| |
Collapse
|
42
|
Ertl-Wagner B, Khalvati F. The data behind the image-Deep learning and its potential impact in neuro-oncological imaging. Neuro Oncol 2022; 24:300-301. [PMID: 34695189 PMCID: PMC8804883 DOI: 10.1093/neuonc/noab249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Birgit Ertl-Wagner
- Division of Neuroradiology, Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Farzad Khalvati
- Division of Neuroradiology, Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Zhang H, Zhang B, Pan W, Dong X, Li X, Chen J, Wang D, Ji W. Preoperative Contrast-Enhanced MRI in Differentiating Glioblastoma From Low-Grade Gliomas in The Cancer Imaging Archive Database: A Proof-of-Concept Study. Front Oncol 2022; 11:761359. [PMID: 35111665 PMCID: PMC8801812 DOI: 10.3389/fonc.2021.761359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
PurposeThis study aimed to develop a repeatable MRI-based machine learning model to differentiate between low-grade gliomas (LGGs) and glioblastoma (GBM) and provide more clinical information to improve treatment decision-making.MethodsPreoperative MRIs of gliomas from The Cancer Imaging Archive (TCIA)–GBM/LGG database were selected. The tumor on contrast-enhanced MRI was segmented. Quantitative image features were extracted from the segmentations. A random forest classification algorithm was used to establish a model in the training set. In the test phase, a random forest model was tested using an external test set. Three radiologists reviewed the images for the external test set. The area under the receiver operating characteristic curve (AUC) was calculated. The AUCs of the radiomics model and radiologists were compared.ResultsThe random forest model was fitted using a training set consisting of 142 patients [mean age, 52 years ± 16 (standard deviation); 78 men] comprising 88 cases of GBM. The external test set included 25 patients (14 with GBM). Random forest analysis yielded an AUC of 1.00 [95% confidence interval (CI): 0.86–1.00]. The AUCs for the three readers were 0.92 (95% CI 0.74–0.99), 0.70 (95% CI 0.49–0.87), and 0.59 (95% CI 0.38–0.78). Statistical differences were only found between AUC and Reader 1 (1.00 vs. 0.92, respectively; p = 0.16).ConclusionAn MRI radiomics-based random forest model was proven useful in differentiating GBM from LGG and showed better diagnostic performance than that of two inexperienced radiologists.
Collapse
Affiliation(s)
- Huangqi Zhang
- Department of Radiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Binhao Zhang
- Department of Radiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Wenting Pan
- Department of Radiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Xue Dong
- Department of Radiology, Taizhou Hospital, Zhejiang University, Taizhou, China
| | - Xin Li
- Department of Radiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Jinyao Chen
- Department of Radiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Dongnv Wang
- Department of Radiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Wenbin Ji
- Department of Radiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
- *Correspondence: Wenbin Ji,
| |
Collapse
|
44
|
Li Y, Ye M, Jia B, Chen L, Zhou Z. Practice of the new supervised machine learning predictive analytics for glioma patient survival after tumor resection: Experiences in a high-volume Chinese center. Front Surg 2022; 9:975022. [PMID: 36873808 PMCID: PMC9981970 DOI: 10.3389/fsurg.2022.975022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 12/28/2022] [Indexed: 02/19/2023] Open
Abstract
OBJECTIVE This study aims to assess the effectiveness of the Gradient Boosting (GB) algorithm on glioma prognosis prediction and to explore new predictive models for glioma patient survival after tumor resection. METHODS A cohort of 776 glioma cases (WHO grades II-IV) between 2010 and 2017 was obtained. Clinical characteristics and biomarker information were reviewed. Subsequently, we constructed the conventional Cox survival model and three different supervised machine learning models, including support vector machine (SVM), random survival forest (RSF), Tree GB, and Component GB. Then, the model performance was compared with each other. At last, we also assessed the feature importance of models. RESULTS The concordance indexes of the conventional survival model, SVM, RSF, Tree GB, and Component GB were 0.755, 0.787, 0.830, 0.837, and 0.840, respectively. All areas under the cumulative receiver operating characteristic curve of both GB models were above 0.800 at different survival times. Their calibration curves showed good calibration of survival prediction. Meanwhile, the analysis of feature importance revealed Karnofsky performance status, age, tumor subtype, extent of resection, and so on as crucial predictive factors. CONCLUSION Gradient Boosting models performed better in predicting glioma patient survival after tumor resection than other models.
Collapse
Affiliation(s)
- Yushan Li
- Department of Ultrasound, Gansu Provincial Hospital, Lanzhou, China
| | - Maodong Ye
- Medical Cosmetic Center, First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Baolong Jia
- Pingliang Second People's Hospital Neurosurgery Department, Pingliang, China
| | - Linwei Chen
- Neurosurgery Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Correspondence: Linwei Chen Zubang Zhou
| | - Zubang Zhou
- Department of Ultrasound, Gansu Provincial Hospital, Lanzhou, China
- Correspondence: Linwei Chen Zubang Zhou
| |
Collapse
|
45
|
Huang J, Shlobin NA, DeCuypere M, Lam SK. Deep Learning for Outcome Prediction in Neurosurgery: A Systematic Review of Design, Reporting, and Reproducibility. Neurosurgery 2022; 90:16-38. [PMID: 34982868 DOI: 10.1227/neu.0000000000001736] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Deep learning (DL) is a powerful machine learning technique that has increasingly been used to predict surgical outcomes. However, the large quantity of data required and lack of model interpretability represent substantial barriers to the validity and reproducibility of DL models. The objective of this study was to systematically review the characteristics of DL studies involving neurosurgical outcome prediction and to assess their bias and reporting quality. Literature search using the PubMed, Scopus, and Embase databases identified 1949 records of which 35 studies were included. Of these, 32 (91%) developed and validated a DL model while 3 (9%) validated a pre-existing model. The most commonly represented subspecialty areas were oncology (16 of 35, 46%), spine (8 of 35, 23%), and vascular (6 of 35, 17%). Risk of bias was low in 18 studies (51%), unclear in 5 (14%), and high in 12 (34%), most commonly because of data quality deficiencies. Adherence to transparent reporting of a multivariable prediction model for individual prognosis or diagnosis reporting standards was low, with a median of 12 transparent reporting of a multivariable prediction model for individual prognosis or diagnosis items (39%) per study not reported. Model transparency was severely limited because code was provided in only 3 studies (9%) and final models in 2 (6%). With the exception of public databases, no study data sets were readily available. No studies described DL models as ready for clinical use. The use of DL for neurosurgical outcome prediction remains nascent. Lack of appropriate data sets poses a major concern for bias. Although studies have demonstrated promising results, greater transparency in model development and reporting is needed to facilitate reproducibility and validation.
Collapse
Affiliation(s)
- Jonathan Huang
- Ann and Robert H. Lurie Children's Hospital, Division of Pediatric Neurosurgery, Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | | | | |
Collapse
|
46
|
Pettit RW, Fullem R, Cheng C, Amos CI. Artificial intelligence, machine learning, and deep learning for clinical outcome prediction. Emerg Top Life Sci 2021; 5:ETLS20210246. [PMID: 34927670 PMCID: PMC8786279 DOI: 10.1042/etls20210246] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022]
Abstract
AI is a broad concept, grouping initiatives that use a computer to perform tasks that would usually require a human to complete. AI methods are well suited to predict clinical outcomes. In practice, AI methods can be thought of as functions that learn the outcomes accompanying standardized input data to produce accurate outcome predictions when trialed with new data. Current methods for cleaning, creating, accessing, extracting, augmenting, and representing data for training AI clinical prediction models are well defined. The use of AI to predict clinical outcomes is a dynamic and rapidly evolving arena, with new methods and applications emerging. Extraction or accession of electronic health care records and combining these with patient genetic data is an area of present attention, with tremendous potential for future growth. Machine learning approaches, including decision tree methods of Random Forest and XGBoost, and deep learning techniques including deep multi-layer and recurrent neural networks, afford unique capabilities to accurately create predictions from high dimensional, multimodal data. Furthermore, AI methods are increasing our ability to accurately predict clinical outcomes that previously were difficult to model, including time-dependent and multi-class outcomes. Barriers to robust AI-based clinical outcome model deployment include changing AI product development interfaces, the specificity of regulation requirements, and limitations in ensuring model interpretability, generalizability, and adaptability over time.
Collapse
Affiliation(s)
- Rowland W. Pettit
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, U.S.A
| | - Robert Fullem
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, U.S.A
| | - Chao Cheng
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, U.S.A
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Christopher I. Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, U.S.A
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX, U.S.A
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, U.S.A
| |
Collapse
|
47
|
Bo L, Zhang Z, Jiang Z, Yang C, Huang P, Chen T, Wang Y, Yu G, Tan X, Cheng Q, Li D, Liu Z. Differentiation of Brain Abscess From Cystic Glioma Using Conventional MRI Based on Deep Transfer Learning Features and Hand-Crafted Radiomics Features. Front Med (Lausanne) 2021; 8:748144. [PMID: 34869438 PMCID: PMC8636043 DOI: 10.3389/fmed.2021.748144] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022] Open
Abstract
Objectives: To develop and validate the model for distinguishing brain abscess from cystic glioma by combining deep transfer learning (DTL) features and hand-crafted radiomics (HCR) features in conventional T1-weighted imaging (T1WI) and T2-weighted imaging (T2WI). Methods: This single-center retrospective analysis involved 188 patients with pathologically proven brain abscess (102) or cystic glioma (86). One thousand DTL and 105 HCR features were extracted from the T1WI and T2WI of the patients. Three feature selection methods and four classifiers, such as k-nearest neighbors (KNN), random forest classifier (RFC), logistic regression (LR), and support vector machine (SVM), for distinguishing brain abscess from cystic glioma were compared. The best feature combination and classifier were chosen according to the quantitative metrics including area under the curve (AUC), Youden Index, and accuracy. Results: In most cases, deep learning-based radiomics (DLR) features, i.e., DTL features combined with HCR features, contributed to a higher accuracy than HCR and DTL features alone for distinguishing brain abscesses from cystic gliomas. The AUC values of the model established, based on the DLR features in T2WI, were 0.86 (95% CI: 0.81, 0.91) in the training cohort and 0.85 (95% CI: 0.75, 0.95) in the test cohort, respectively. Conclusions: The model established with the DLR features can distinguish brain abscess from cystic glioma efficiently, providing a useful, inexpensive, convenient, and non-invasive method for differential diagnosis. This is the first time that conventional MRI radiomics is applied to identify these diseases. Also, the combination of HCR and DTL features can lead to get impressive performance.
Collapse
Affiliation(s)
- Linlin Bo
- Shandong Key Laboratory of Medical Physics and Image Processing, Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Zijian Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zekun Jiang
- Shandong Key Laboratory of Medical Physics and Image Processing, Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Chao Yang
- Shandong Key Laboratory of Medical Physics and Image Processing, Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Pu Huang
- Shandong Key Laboratory of Medical Physics and Image Processing, Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Tingyin Chen
- Department of Network Information Center, Xiangya Hospital, Centra South University, Changsha, China
| | - Yifan Wang
- Department of Network Information Center, Xiangya Hospital, Centra South University, Changsha, China
| | - Gang Yu
- Shandong Key Laboratory of Medical Physics and Image Processing, Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Xiao Tan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Dengwang Li
- Shandong Key Laboratory of Medical Physics and Image Processing, Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, School of Physics and Electronics, Shandong Normal University, Jinan, 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
|
48
|
Lin Z, Cai W, Hou W, Chen Y, Gao B, Mao R, Wang L, Li Z. CT-Guided Survival Prediction of Esophageal Cancer. IEEE J Biomed Health Inform 2021; 26:2660-2669. [PMID: 34855605 DOI: 10.1109/jbhi.2021.3132173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Survival prediction of esophageal cancer is an essential task for doctors to make personalized cancer treatment plans. However, handcrafted features from medical images need prior medical knowledge, which is usually limited and not complete, yielding unsatisfying survival predictions. To address these challenges, we propose a novel and efficient deep learning-based survival prediction framework for evaluating clinical outcomes before concurrent chemoradiotherapy. The proposed model consists of two key components: a 3D Coordinate Attention Convolutional Autoencoder (CACA) and an uncertainty-based jointly Optimizing Cox Model (UOCM). The CACA is built upon an autoencoder structure with 3D coordinate attention layers, capturing latent representations and encoding 3D spatial characteristics with precise positional information. Additionally, we designed an Uncertainty-based jointly Optimizing Cox Model, which jointly optimizes the CACA and survival prediction task. The survival prediction task models the interactions between a patient's feature signatures and clinical outcome to predict a reliable hazard ratio of patients. To verify the effectiveness of our model, we conducted extensive experiments on a dataset including computed tomography of 285 patients with esophageal cancer. Experimental results demonstrated that the proposed method achieved a C-index of 0.72, outperforming the state-of-the-art method.
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW In this paper, we discuss how recent advancements in image processing and machine learning (ML) are shaping a new and exciting era for the osteoporosis imaging field. With this paper, we want to give the reader a basic exposure to the ML concepts that are necessary to build effective solutions for image processing and interpretation, while presenting an overview of the state of the art in the application of machine learning techniques for the assessment of bone structure, osteoporosis diagnosis, fracture detection, and risk prediction. RECENT FINDINGS ML effort in the osteoporosis imaging field is largely characterized by "low-cost" bone quality estimation and osteoporosis diagnosis, fracture detection, and risk prediction, but also automatized and standardized large-scale data analysis and data-driven imaging biomarker discovery. Our effort is not intended to be a systematic review, but an opportunity to review key studies in the recent osteoporosis imaging research landscape with the ultimate goal of discussing specific design choices, giving the reader pointers to possible solutions of regression, segmentation, and classification tasks as well as discussing common mistakes.
Collapse
Affiliation(s)
- Valentina Pedoia
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 Fourth Street, Suite 201, QB3 Building, San Francisco, CA, 94158, USA.
| | - Francesco Caliva
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 Fourth Street, Suite 201, QB3 Building, San Francisco, CA, 94158, USA
| | - Galateia Kazakia
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 Fourth Street, Suite 201, QB3 Building, San Francisco, CA, 94158, USA
| | - Andrew J Burghardt
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 Fourth Street, Suite 201, QB3 Building, San Francisco, CA, 94158, USA
| | - Sharmila Majumdar
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 Fourth Street, Suite 201, QB3 Building, San Francisco, CA, 94158, USA
| |
Collapse
|
50
|
Yin P, Sun C, Wang S, Chen L, Hong N. Clinical-Deep Neural Network and Clinical-Radiomics Nomograms for Predicting the Intraoperative Massive Blood Loss of Pelvic and Sacral Tumors. Front Oncol 2021; 11:752672. [PMID: 34760700 PMCID: PMC8574215 DOI: 10.3389/fonc.2021.752672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/06/2021] [Indexed: 11/29/2022] Open
Abstract
Background Patients with pelvic and sacral tumors are prone to massive blood loss (MBL) during surgery, which may endanger their lives. Purposes This study aimed to determine the feasibility of using deep neural network (DNN) and radiomics nomogram (RN) based on 3D computed tomography (CT) features and clinical characteristics to predict the intraoperative MBL of pelvic and sacral tumors. Materials and Methods This single-center retrospective analysis included 810 patients with pelvic and sacral tumors. 1316 CT and CT enhanced radiomics features were extracted. RN1 and RN2 were constructed by random grouping and time node grouping, respectively. The DNN models were constructed for comparison with RN. Clinical factors associated with the MBL were also evaluated. The area under the receiver operating characteristic curve (AUC) and accuracy (ACC) were used to evaluate different models. Results Radscore, tumor type, tumor location, and sex were significant predictors of the MBL of pelvic and sacral tumors (P < 0.05), of which radscore (OR, ranging from 2.109 to 4.706, P < 0.001) was the most important. The clinical-DNN and clinical-RN performed better than DNN and RN. The best-performing clinical-DNN model based on CT features exhibited an AUC of 0.92 and an ACC of 0.97 in the training set, and an AUC of 0.92 and an ACC of 0.75 in the validation set. Conclusions The clinical-DNN and clinical-RN had good performance in predicting the MBL of pelvic and sacral tumors, which could be used for clinical decision-making.
Collapse
Affiliation(s)
- Ping Yin
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Chao Sun
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Sicong Wang
- Department of Pharmaceuticals Diagnosis, GE Healthcare (China), Shanghai, China
| | - Lei Chen
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, Beijing, China
| |
Collapse
|