1
|
Hu K, Bian C, Yu J, Jiang D, Chen Z, Zhao F, Li H. Construction of a combined prognostic model for pancreatic ductal adenocarcinoma based on deep learning and digital pathology images. BMC Gastroenterol 2024; 24:387. [PMID: 39482576 PMCID: PMC11528996 DOI: 10.1186/s12876-024-03469-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Deep learning has made significant advancements in the field of digital pathology, and the integration of multiple models has further improved accuracy. In this study, we aimed to construct a combined prognostic model using deep learning-extracted features from digital pathology images of pancreatic ductal adenocarcinoma (PDAC) alongside clinical predictive indicators and to explore its prognostic value. METHODS A retrospective analysis was conducted on 142 postoperative pathologically confirmed PDAC cases. These cases were divided into training (n = 114) and testing sets (n = 28) at an 8:2 ratio. Tumor whole-slide imaging features were extracted and screened to construct a pathological risk model based on a pre-trained deep learning model. Clinical and pathological data from the training set were used to select independent predictive factors for PDAC and establish a clinical risk model using LASSO, univariate, and multivariate Cox regression analyses. Based on the pathological and clinical risk models, a combined model was developed. The Harrell concordance index (C-index) was computed to assess the predictive performance of each model for PDAC survival prognosis. RESULTS For the training and testing sets, the C-index values for the clinical risk model were 0.76 and 0.75, respectively; for the pathological risk model, they were 0.82 and 0.73, respectively; and for the combined model, they were 0.86 and 0.77, respectively. The combined model exhibited appropriate calibration at 1-, 3-, and 5-year time points, as well as a superior area under the curve of the receiver operating characteristic curve and clinical net benefit compared to the single models. CONCLUSIONS Integrating the pathological and clinical risk models may provide a higher predictive value for survival prognosis.
Collapse
Affiliation(s)
- Kaixin Hu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Chenyang Bian
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jiayin Yu
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Dawei Jiang
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhangjun Chen
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Fengqing Zhao
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Huangbao Li
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| |
Collapse
|
2
|
Fotouhi M, Shahbandi A, Mehr FSK, Shahla MM, Nouredini SM, Kankam SB, Khorasanizadeh M, Chambless LB. Application of radiomics for diagnosis, subtyping, and prognostication of medulloblastomas: a systematic review. Neurosurg Rev 2024; 47:827. [PMID: 39467891 DOI: 10.1007/s10143-024-03060-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/20/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024]
Abstract
Applications of radiomics for clinical management of medulloblastomas are expanding. This systematic review aims to aggregate and overview the current role of radiomics in the diagnosis, subtyping, and prognostication of medulloblastomas. The present systematic review was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. PubMed/MEDLINE were searched using a standardized search term. Articles found within the database from the inception until November 2022 were considered for screening. Retrieved records were screened independently by two authors based on their titles and abstracts. The full text of selected articles was reviewed to finalize the eligibility. Due to the heterogeneity of included studies, no formal data synthesis was conducted. Of the 249 screened citations, 21 studies were included and analyzed. Radiomics demonstrated promising performance for discriminating medulloblastomas from other posterior fossa tumors, particularly ependymomas and pilocytic astrocytomas. It was also efficacious in determining the subtype (i.e., WNT+, SHH+, group 3, and group 4) of medulloblastomas non-invasively. Regarding prognostication, radiomics exhibited some ability to predict overall survival and progression-free survival of patients with medulloblastomas. Our systematic review revealed that radiomics represents a promising tool for diagnosis and prognostication of medulloblastomas. Further prospective research measuring the clinical value of radiomics in this setting is warranted.
Collapse
Affiliation(s)
- Maryam Fotouhi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | - Samuel B Kankam
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
- T. H. Chan School of Public Health, Harvard University, Cambridge, MA, USA
| | | | - Lola B Chambless
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
3
|
Chen X, Huang Y, Wee L, Zhao K, Mao Y, Li Z, Yao S, Li S, Liang Y, Huang X, Dekker A, Chen X, Liu Z. Integrated analysis of radiomics, RNA, and clinicopathologic phenotype reveals biological basis of prognostic risk stratification in colorectal cancer. Sci Bull (Beijing) 2024:S2095-9273(24)00743-6. [PMID: 39438165 DOI: 10.1016/j.scib.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/28/2024] [Accepted: 08/24/2024] [Indexed: 10/25/2024]
Affiliation(s)
- Xiaobo Chen
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China; Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht 6229 ER, the Netherlands
| | - Leonard Wee
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht 6229 ER, the Netherlands
| | - Ke Zhao
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
| | - Yun Mao
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming 650106, China
| | - Su Yao
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Suyun Li
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
| | - Yanting Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
| | - Xin Huang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht 6229 ER, the Netherlands
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China.
| |
Collapse
|
4
|
Yang L, Li M, Liu Y, Jiang Z, Xu S, Ding H, Gao X, Liu S, Qi L, Wang K. Draw on advantages and avoid disadvantages: CT-derived individualized radiomic signature for predicting chemo-radiotherapy sensitivity in unresectable advanced non-small cell lung cancer. J Cancer Res Clin Oncol 2024; 150:453. [PMID: 39387925 PMCID: PMC11467094 DOI: 10.1007/s00432-024-05971-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Presently, the options of concurrent chemo-radiotherapy (CCR) in patients with locally advanced non-small cell lung cancer (LA-NSCLC) are controversial and there is no reliable prediction tool to stratify poor- and good-responders. Although radiomic analysis has provided new opportunities for personalized medicine in oncological practice, the repeatability and reproducibility of radiomic features are critical challenges that hinder their widespread clinical adoption. This study aimed to develop a qualitative radiomic signature based on the within-sample rank of radiomics features, and to use this novel method to predict CCR sensitivity in LA-NSCLC, avoiding the variability of quantitative signatures to multicenter effect. METHODS We retrospectively analyzed 125 patients with stage III NSCLC who received treatment from our hospital. Radiomic features were extracted from pretreatment plain CT scans and constructed as feature pairs based on their within-sample rank. Fisher and univariate Cox analyses were performed to select feature pairs significantly associated with patients' overall survival (OS). NSCLC-Radiomic (R422) cohort including 104 NSCLC patients was used as an independent testing cohort. NSCLC-Radiogenomic (RG211) cohort with matched RNA sequencing profiles, was used for functional enrichment analysis to reveal the underlying biological mechanism reflected by the signature. RESULTS A qualitative signature, consisting of 15 radiomic feature pairs (termed as 15-RiFPS), was developed based on the Genetic Algorithm, which could optimally distinguish responder from non-responder with significantly improved OS if they received CCR treatment (log-rank P = 0.0009, HR = 13.79, 95% CIs 1.83-104.1). The performance of 15-RiFPS was validated in an independent public cohort (log-rank P = 0.0037, HR = 2.40, 95% CIs 1.30-4.40). Furthermore, the transcriptomic analyses provided biological pathways ('glutathione metabolic process', 'cellular oxidant detoxification') underlying the signature. CONCLUSIONS We developed a CT-derived 15-RiFPS, which could potentially help predict individualized therapeutic benefit of CCR in patients with LA-NSCLC. Additionally, we investigated the underlying intra-tumoral biological characteristics behind 15-RiFPS which would accelerate its clinical application. This approach could be applied to a wider range of treatments and cancer types.
Collapse
Affiliation(s)
- Liping Yang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, 150001, People's Republic of China
| | - Mengyue Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Yixin Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Zhiyun Jiang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, 150001, People's Republic of China
| | - Shichuan Xu
- Department of Equipment, The Second Hospital of Harbin, Harbin, People's Republic of China
| | - Hongchao Ding
- Department of Physical Diagnosis, Heilongjiang Provincial Hospital, Harbin, People's Republic of China
| | - Xing Gao
- Department of Physical Diagnosis, Heilongjiang Provincial Hospital, Harbin, People's Republic of China
| | - Shilong Liu
- Department of Thoracic Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, 150001, People's Republic of China.
| | - Lishuang Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150001, People's Republic of China.
| | - Kezheng Wang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, 150001, People's Republic of China.
| |
Collapse
|
5
|
Piffer S, Greto D, Ubaldi L, Mortilla M, Ciccarone A, Desideri I, Genitori L, Livi L, Marrazzo L, Pallotta S, Retico A, Sardi I, Talamonti C. Radiomic- and dosiomic-based clustering development for radio-induced neurotoxicity in pediatric medulloblastoma. Childs Nerv Syst 2024; 40:2301-2310. [PMID: 38642113 PMCID: PMC11269375 DOI: 10.1007/s00381-024-06416-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Texture analysis extracts many quantitative image features, offering a valuable, cost-effective, and non-invasive approach for individual medicine. Furthermore, multimodal machine learning could have a large impact for precision medicine, as texture biomarkers can underlie tissue microstructure. This study aims to investigate imaging-based biomarkers of radio-induced neurotoxicity in pediatric patients with metastatic medulloblastoma, using radiomic and dosiomic analysis. METHODS This single-center study retrospectively enrolled children diagnosed with metastatic medulloblastoma (MB) and treated with hyperfractionated craniospinal irradiation (CSI). Histological confirmation of medulloblastoma and baseline follow-up magnetic resonance imaging (MRI) were mandatory. Treatment involved helical tomotherapy (HT) delivering a dose of 39 Gray (Gy) to brain and spinal axis and a posterior fossa boost up to 60 Gy. Clinical outcomes, such as local and distant brain control and neurotoxicity, were recorded. Radiomic and dosiomic features were extracted from tumor regions on T1, T2, FLAIR (fluid-attenuated inversion recovery) MRI-maps, and radiotherapy dose distribution. Different machine learning feature selection and reduction approaches were performed for supervised and unsupervised clustering. RESULTS Forty-eight metastatic medulloblastoma patients (29 males and 19 females) with a mean age of 12 ± 6 years were enrolled. For each patient, 332 features were extracted. Greater level of abstraction of input data by combining selection of most performing features and dimensionality reduction returns the best performance. The resulting one-component radiomic signature yielded an accuracy of 0.73 with sensitivity, specificity, and precision of 0.83, 0.64, and 0.68, respectively. CONCLUSIONS Machine learning radiomic-dosiomic approach effectively stratified pediatric medulloblastoma patients who experienced radio-induced neurotoxicity. Strategy needs further validation in external dataset for its potential clinical use in ab initio management paradigms of medulloblastoma.
Collapse
Affiliation(s)
- Stefano Piffer
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
- National Institute for Nuclear Physics (INFN), Florence Division, Florence, Italy.
| | - Daniela Greto
- Radiation Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Leonardo Ubaldi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- National Institute for Nuclear Physics (INFN), Florence Division, Florence, Italy
| | - Marzia Mortilla
- Radiology Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Antonio Ciccarone
- Medical Physics Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Isacco Desideri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Lorenzo Genitori
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Lorenzo Livi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- Radiation Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Livia Marrazzo
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- National Institute for Nuclear Physics (INFN), Florence Division, Florence, Italy
| | - Stefania Pallotta
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- National Institute for Nuclear Physics (INFN), Florence Division, Florence, Italy
| | | | - Iacopo Sardi
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Cinzia Talamonti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- National Institute for Nuclear Physics (INFN), Florence Division, Florence, Italy
| |
Collapse
|
6
|
Xiao Y, Wu F, Hou K, Wang F, Zhou C, Huang P, Yang C, Zeng M. MR radiomics to predict microvascular invasion status and biological process in combined hepatocellular carcinoma-cholangiocarcinoma. Insights Imaging 2024; 15:172. [PMID: 38981992 PMCID: PMC11233482 DOI: 10.1186/s13244-024-01741-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/09/2024] [Indexed: 07/11/2024] Open
Abstract
OBJECTIVES To establish an MRI-based radiomics model for predicting the microvascular invasion (MVI) status of cHCC-CCA and to investigate biological processes underlying the radiomics model. METHODS The study consisted of a retrospective dataset (82 in the training set, 36 in the validation set) and a prospective dataset (25 patients in the test set) from two hospitals. Based on the training set, logistic regression analyses were employed to develop the clinical-imaging model, while radiomic features were extracted to construct a radiomics model. The diagnosis performance was further validated in the validation and test sets. Prognostic aspects of the radiomics model were investigated using the Kaplan-Meier method and log-rank test. Differential gene expression analysis and gene ontology (GO) analysis were conducted to explore biological processes underlying the radiomics model based on RNA sequencing data. RESULTS One hundred forty-three patients (mean age, 56.4 ± 10.5; 114 men) were enrolled, in which 73 (51.0%) were confirmed as MVI-positive. The radiomics model exhibited good performance in predicting MVI status, with the area under the curve of 0.935, 0.873, and 0.779 in training, validation, and test sets, respectively. Overall survival (OS) was significantly different between the predicted MVI-negative and MVI-positive groups (median OS: 25 vs 18 months, p = 0.008). Radiogenomic analysis revealed associations between the radiomics model and biological processes involved in regulating the immune response. CONCLUSION A robust MRI-based radiomics model was established for predicting MVI status in cHCC-CCA, in which potential prognostic value and underlying biological processes that regulate immune response were demonstrated. CRITICAL RELEVANCE STATEMENT MVI is a significant manifestation of tumor invasiveness, and the MR-based radiomics model established in our study will facilitate risk stratification. Furthermore, underlying biological processes demonstrated in the radiomics model will offer valuable insights for guiding immunotherapy strategies. KEY POINTS MVI is of prognostic significance in cHCC-CCA, but lacks reliable preoperative assessment. The MRI-based radiomics model predicts MVI status effectively in cHCC-CCA. The MRI-based radiomics model demonstrated prognostic value and underlying biological processes. The radiomics model could guide immunotherapy and risk stratification in cHCC-CCA.
Collapse
Affiliation(s)
- Yuyao Xiao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fei Wu
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kai Hou
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fang Wang
- Shanghai United Imaging Intelligence Co. Ltd, Shanghai, China
| | - Changwu Zhou
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Huang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chun Yang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Institute of Medical Imaging, Shanghai, China.
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Sun C, Jiang C, Wang X, Ma S, Zhang D, Jia W. MR-Based Radiomics Predicts CDK6 Expression and Prognostic Value in High-grade Glioma. Acad Radiol 2024:S1076-6332(24)00364-7. [PMID: 38964985 DOI: 10.1016/j.acra.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024]
Abstract
RATIONALE AND OBJECTIVES This study aims to assess the prognostic value of Cyclin-dependent kinases 6 (CDK6) expression levels and establish a machine learning-based radiomics model for predicting the expression levels of CDK6 in high-grade gliomas (HGG). MATERIALS AND METHODS Clinical parameters and genomic data were extracted from 310 HGG patients in the Cancer Genome Atlas (TCGA) database and 27 patients in the Repository of Molecular Brain Neoplasia Data (REMBRANDT) database. Univariate and multivariate Cox regression, as well as Kaplan-Meier analysis, were performed for prognosis analysis. The correlation between immune cell Infiltration with CDK6 was assessed using spearman correlation analysis. Radiomic features were extracted from contrast-enhanced magnetic resonance imaging (CE-MRI) in the Cancer Imaging Archive (TCIA) database (n = 82) and REMBRANDT database (n = 27). Logistic regression (LR) and support vector machine (SVM) were employed to establish the radiomics model for predicting CDK6 expression. Receiver operating characteristic curve (ROC), calibration curve, and decision curve analysis (DCA) were utilized to assess the predictive performance of the radiomics model. Generate radiomic scores (RS) based on the LR model. An RS-based nomogram was constructed to predict the prognosis of HGG. RESULTS CDK6 was significantly overexpressed in HGG tissues and was related to lower overall survival. A significant elevation in infiltrating M0 macrophages was observed in the CDK6 high group (P < 0.001). The LR radiomics model for the prediction of CDK6 expression levels (AUC=0.810 in the training cohort, AUC = 0.784 after cross-validation, AUC=0.750 in the testing cohort) was established utilizing three radiomic features. The predictive efficiencies of the RS-based nomogram, as measured by AUC, were 0.769 for 1-year, 0.815 for 3-year, and 0.780 for 5-year, respectively. CONCLUSION The expression level of CDK6 can impact the prognosis of patients with HGG. The expression level of HGG can be noninvasively prognosticated utilizing a radiomics model.
Collapse
Affiliation(s)
- Chen Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Chenggang Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Xi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Dainan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 West Road, South Fourth Ring Road, Beijing, China.
| |
Collapse
|
8
|
Zhou X, Lu Y, Wu Y, Yu Y, Liu Y, Wang C, Zhao Z, Wang C, Gao Z, Li Z, Zhao Y, Cao W. Construction and validation of a deep learning prognostic model based on digital pathology images of stage III colorectal cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108369. [PMID: 38703632 DOI: 10.1016/j.ejso.2024.108369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/09/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND TNM staging is the main reference standard for prognostic prediction of colorectal cancer (CRC), but the prognosis heterogeneity of patients with the same stage is still large. This study aimed to classify the tumor microenvironment of patients with stage III CRC and quantify the classified tumor tissues based on deep learning to explore the prognostic value of the developed tumor risk signature (TRS). METHODS A tissue classification model was developed to identify nine tissues (adipose, background, debris, lymphocytes, mucus, smooth muscle, normal mucosa, stroma, and tumor) in whole-slide images (WSIs) of stage III CRC patients. This model was used to extract tumor tissues from WSIs of 265 stage III CRC patients from The Cancer Genome Atlas and 70 stage III CRC patients from the Sixth Affiliated Hospital of Sun Yat-sen University. We used three different deep learning models for tumor feature extraction and applied a Cox model to establish the TRS. Survival analysis was conducted to explore the prognostic performance of TRS. RESULTS The tissue classification model achieved 94.4 % accuracy in identifying nine tissue types. The TRS showed a Harrell's concordance index of 0.736, 0.716, and 0.711 in the internal training, internal validation, and external validation sets. Survival analysis showed that TRS had significant predictive ability (hazard ratio: 3.632, p = 0.03) for prognostic prediction. CONCLUSION The TRS is an independent and significant prognostic factor for PFS of stage III CRC patients and it contributes to risk stratification of patients with different clinical stages.
Collapse
Affiliation(s)
- Xuezhi Zhou
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan, China; Engineering Technology Research Center of Neurosense and Control of Henan Province, Xinxiang, China; Henan International Joint Laboratory of Neural Information Analysis and Drug Intelligent Design, Xinxiang, China
| | - Yizhan Lu
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan, China; Engineering Technology Research Center of Neurosense and Control of Henan Province, Xinxiang, China; Henan International Joint Laboratory of Neural Information Analysis and Drug Intelligent Design, Xinxiang, China
| | - Yue Wu
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yi Yu
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan, China; Engineering Technology Research Center of Neurosense and Control of Henan Province, Xinxiang, China; Henan International Joint Laboratory of Neural Information Analysis and Drug Intelligent Design, Xinxiang, China
| | - Yong Liu
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan, China; Engineering Technology Research Center of Neurosense and Control of Henan Province, Xinxiang, China; Henan International Joint Laboratory of Neural Information Analysis and Drug Intelligent Design, Xinxiang, China
| | - Chang Wang
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan, China; Engineering Technology Research Center of Neurosense and Control of Henan Province, Xinxiang, China; Henan International Joint Laboratory of Neural Information Analysis and Drug Intelligent Design, Xinxiang, China
| | - Zongya Zhao
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan, China; Engineering Technology Research Center of Neurosense and Control of Henan Province, Xinxiang, China; Henan International Joint Laboratory of Neural Information Analysis and Drug Intelligent Design, Xinxiang, China
| | - Chong Wang
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan, China; Engineering Technology Research Center of Neurosense and Control of Henan Province, Xinxiang, China; Henan International Joint Laboratory of Neural Information Analysis and Drug Intelligent Design, Xinxiang, China
| | - Zhixian Gao
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan, China; Engineering Technology Research Center of Neurosense and Control of Henan Province, Xinxiang, China; Henan International Joint Laboratory of Neural Information Analysis and Drug Intelligent Design, Xinxiang, China
| | - Zhenxin Li
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan, China; Engineering Technology Research Center of Neurosense and Control of Henan Province, Xinxiang, China; Henan International Joint Laboratory of Neural Information Analysis and Drug Intelligent Design, Xinxiang, China.
| | - Yandong Zhao
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Wuteng Cao
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Ismail M, Um H, Salloum R, Hollnagel F, Ahmed R, de Blank P, Tiwari P. A Radiomic Approach for Evaluating Intra-Subgroup Heterogeneity in SHH and Group 4 Pediatric Medulloblastoma: A Preliminary Multi-Institutional Study. Cancers (Basel) 2024; 16:2248. [PMID: 38927953 PMCID: PMC11201623 DOI: 10.3390/cancers16122248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/15/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Medulloblastoma (MB) is the most frequent malignant brain tumor in children with extensive heterogeneity that results in varied clinical outcomes. Recently, MB was categorized into four molecular subgroups, WNT, SHH, Group 3, and Group 4. While SHH and Group 4 are known for their intermediate prognosis, studies have reported wide disparities in patient outcomes within these subgroups. This study aims to create a radiomic prognostic signature, medulloblastoma radiomics risk (mRRisk), to identify the risk levels within the SHH and Group 4 subgroups, individually, for reliable risk stratification. Our hypothesis is that this signature can comprehensively capture tumor characteristics that enable the accurate identification of the risk level. In total, 70 MB studies (48 Group 4, and 22 SHH) were retrospectively curated from three institutions. For each subgroup, 232 hand-crafted features that capture the entropy, surface changes, and contour characteristics of the tumor were extracted. Features were concatenated and fed into regression models for risk stratification. Contrasted with Chang stratification that did not yield any significant differences within subgroups, significant differences were observed between two risk groups in Group 4 (p = 0.04, Concordance Index (CI) = 0.82) on the cystic core and non-enhancing tumor, and SHH (p = 0.03, CI = 0.74) on the enhancing tumor. Our results indicate that radiomics may serve as a prognostic tool for refining MB risk stratification, towards improved patient care.
Collapse
Affiliation(s)
- Marwa Ismail
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53706, USA (P.T.)
| | - Hyemin Um
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53706, USA (P.T.)
| | - Ralph Salloum
- Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Fauzia Hollnagel
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Raheel Ahmed
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Peter de Blank
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Pallavi Tiwari
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53706, USA (P.T.)
- Departments of Medical Physics and Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53792, USA
| |
Collapse
|
10
|
Zhou L, Ji Q, Peng H, Chen F, Zheng Y, Jiao Z, Gong J, Li W. Automatic image segmentation and online survival prediction model of medulloblastoma based on machine learning. Eur Radiol 2024; 34:3644-3655. [PMID: 37994966 DOI: 10.1007/s00330-023-10316-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 08/19/2023] [Accepted: 08/26/2023] [Indexed: 11/24/2023]
Abstract
OBJECTIVES To develop a dynamic nomogram containing radiomics signature and clinical features for estimating the overall survival (OS) of patients with medulloblastoma (MB) and design an automatic image segmentation model to reduce labor and time costs. METHODS Data from 217 medulloblastoma (MB) patients over the past 4 years were collected and separated into a training set and a test set. Intraclass correlation coefficient (ICC), random survival forest (RSF), and least absolute shrinkage and selection operator (LASSO) regression methods were employed to select variables in the training set. Univariate and multivariate Cox proportional hazard models, as well as Kaplan-Meier analysis, were utilized to determine the relationship among the radiomics signature, clinical features, and overall survival. A dynamic nomogram was developed. Additionally, a 3D-Unet deep learning model was used to train the automatic tumor delineation model. RESULTS Higher Rad-scores were significantly associated with worse OS in both the training and validation sets (p < 0.001 and p = 0.047, respectively). The Cox model combined clinical and radiomics signatures ([IBS = 0.079], [C-index = 0.747, SE = 0.045]) outperformed either radiomics signatures alone ([IBS = 0.081], [C-index = 0.738, SE = 0.041]) or clinical features alone ([IBS = 0.085], [C-index = 0.565, SE = 0.041]). The segmentation model had mean Dice coefficients of 0.80, 0.82, and 0.78 in the training, validation, and test sets respectively. A deep learning-based tumor segmentation model was built with Dice coefficients of 0.8372, 0.8017, and 0.7673 on the training set, validation set, and test set, respectively. CONCLUSIONS A combination of radiomics features and clinical characteristics enhances the accuracy of OS prediction in medulloblastoma patients. Additionally, building an MRI image automatic segmentation model reduces labor and time costs. CLINICAL RELEVANCE STATEMENT A survival prognosis model based on radiomics and clinical characteristics could improve the accuracy of prognosis estimation for medulloblastoma patients, and an MRI-based automatic tumor segmentation model could reduce the cost of time. KEY POINTS • A model that combines radiomics and clinical features can predict the survival prognosis of patients with medulloblastoma. • Online nomogram and image automatic segmentation model can help doctors better judge the prognosis of medulloblastoma and save working time. • The developed AI system can help doctors judge the prognosis of diseases and promote the development of precision medicine.
Collapse
Affiliation(s)
- Lili Zhou
- Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No. 119, Nansihuan West Road, Fengtai District, Beijing, 100070, China
| | - Qiang Ji
- Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No. 119, Nansihuan West Road, Fengtai District, Beijing, 100070, China
| | - Hong Peng
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Huangpu District, Shanghai, 20011, China
| | - Feng Chen
- Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No. 119, Nansihuan West Road, Fengtai District, Beijing, 100070, China
| | - Yi Zheng
- Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No. 119, Nansihuan West Road, Fengtai District, Beijing, 100070, China
| | | | - Jian Gong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical Unversity, No. 119, Nansihuan West Road, Fengtai District, Beijing, 100070, China.
| | - Wenbin Li
- Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No. 119, Nansihuan West Road, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
11
|
Luo Y, Zhuang Y, Zhang S, Wang J, Teng S, Zeng H. Multiparametric MRI-Based Radiomics Signature with Machine Learning for Preoperative Prediction of Prognosis Stratification in Pediatric Medulloblastoma. Acad Radiol 2024; 31:1629-1642. [PMID: 37643930 DOI: 10.1016/j.acra.2023.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/24/2023] [Accepted: 06/24/2023] [Indexed: 08/31/2023]
Abstract
RATIONALE AND OBJECTIVES Despite advances in risk-stratified treatment strategies for children with medulloblastoma (MB), the prognosis for MB with short-term recurrence is extremely poor, and there is still a lack of evaluation of short-term recurrence risk or short-term survival. This study aimed to construct and validate a radiomics model for predicting the outcome of MB based on preoperative multiparametric magnetic resonance images (MRIs) and to provide an objective for clinical decision-making. MATERIALS AND METHODS The clinical and imaging data of 64 patients with MB admitted to Shenzhen Children's Hospital from December 2012 to December 2021 and confirmed by pathology were retrospectively collected. According to the 18-month progression-free survival, the cases were classified into a good prognosis group and a poor prognosis group, and all cases were divided into training group (70%) and validation group (30%) randomly. Radiomics features were extracted from MRI of each child. The consistency test, t-test, and the least absolute shrinkage and selection operator were used for feature selection. The support vector machine (SVM) and receiver operator characteristic were used to evaluate the distinguishing ability of the selected features to the prognostic groups. RAD score was calculated based on the selected features. The clinical characteristics and RAD score were included in the multivariate logistic regression, and prediction models were constructed by screening out independent influences. The radiomics nomogram was constructed, and its clinical significance was evaluated. RESULTS A total of 1930 radiomic features were extracted from the images of each patient, and 11 features were included in the construction of radiomics score after selected. The area under the curve (AUC) values of the SVM model in the training and validation groups were 0.946 and 0.797, respectively. The radiomics nomogram was constructed based on the training cohort, and the AUC values in the training group and the validation group were 0.926 and 0.835, respectively. The results of clinical decision curve analysis showed that a good net benefit could be obtained from the nomogram. CONCLUSION The radiomics nomogram established based on MRI can be used as a noninvasive predictive tool to evaluate the prognosis of children with MB, which is expected to help neurosurgeons better conduct preoperative planning and patient follow-up management.
Collapse
Affiliation(s)
- Yi Luo
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen 518038, China (Y.L., Y.Z., S.Z., H.Z.); Shantou University Medical College, Shantou 515041, China (Y.L., S.Z.)
| | - Yijiang Zhuang
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen 518038, China (Y.L., Y.Z., S.Z., H.Z.)
| | - Siqi Zhang
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen 518038, China (Y.L., Y.Z., S.Z., H.Z.); Shantou University Medical College, Shantou 515041, China (Y.L., S.Z.)
| | - Jingsheng Wang
- Department of Neurosurgery, Shenzhen Children's Hospital, Shenzhen 518038, China (J.W.)
| | - Songyu Teng
- Shenzhen Children's Hospital of China Medical University, Shenzhen 518038, China (S.T.)
| | - Hongwu Zeng
- Department of Radiology, Shenzhen Children's Hospital, Shenzhen 518038, China (Y.L., Y.Z., S.Z., H.Z.).
| |
Collapse
|
12
|
Wang Z, Guan F, Duan W, Guo Y, Pei D, Qiu Y, Wang M, Xing A, Liu Z, Yu B, Zheng H, Liu X, Yan D, Ji Y, Cheng J, Yan J, Zhang Z. Diffusion tensor imaging-based machine learning for IDH wild-type glioblastoma stratification to reveal the biological underpinning of radiomic features. CNS Neurosci Ther 2023; 29:3339-3350. [PMID: 37222229 PMCID: PMC10580329 DOI: 10.1111/cns.14263] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/09/2023] [Accepted: 05/03/2023] [Indexed: 05/25/2023] Open
Abstract
INTRODUCTION This study addresses the lack of systematic investigation into the prognostic value of hand-crafted radiomic features derived from diffusion tensor imaging (DTI) in isocitrate dehydrogenase (IDH) wild-type glioblastoma (GBM), as well as the limited understanding of the biological interpretation of individual DTI radiomic features and metrics. AIMS To develop and validate a DTI-based radiomic model for predicting prognosis in patients with IDH wild-type GBM and reveal the biological underpinning of individual DTI radiomic features and metrics. RESULTS The DTI-based radiomic signature was an independent prognostic factor (p < 0.001). Incorporating the radiomic signature into a clinical model resulted in a radiomic-clinical nomogram that predicted survival better than either the radiomic model or clinical model alone, with a better calibration and classification accuracy. Four categories of pathways (synapse, proliferation, DNA damage response, and complex cellular functions) were significantly correlated with the DTI-based radiomic features and DTI metrics. CONCLUSION The prognostic radiomic features derived from DTI are driven by distinct pathways involved in synapse, proliferation, DNA damage response, and complex cellular functions of GBM.
Collapse
Affiliation(s)
- Zilong Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Fangzhan Guan
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Wenchao Duan
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yu Guo
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Dongling Pei
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuning Qiu
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Minkai Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Aoqi Xing
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhongyi Liu
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Bin Yu
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Hongwei Zheng
- Department of MRIThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xianzhi Liu
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Dongming Yan
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuchen Ji
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jingliang Cheng
- Department of MRIThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jing Yan
- Department of MRIThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhenyu Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
13
|
Zhao Y, Li L, Han K, Li T, Duan J, Sun Q, Zhu C, Liang D, Chai N, Li ZC. A radio-pathologic integrated model for prediction of lymph node metastasis stage in patients with gastric cancer. Abdom Radiol (NY) 2023; 48:3332-3342. [PMID: 37716926 DOI: 10.1007/s00261-023-04037-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND Accurate prediction of lymph node metastasis stage (LNMs) facilitates precision therapy for gastric cancer. We aimed to develop and validate a deep learning-based radio-pathologic model to predict the LNM stage in patients with gastric cancer by integrating CT images and histopathological whole-slide images (WSIs). METHODS A total of 252 patients were enrolled and randomly divided into a training set (n = 202) and a testing set (n = 50). Both pretreatment contrast-enhanced abdominal CT and WSI of biopsy specimens were collected for each patient. The deep radiologic and pathologic features were extracted from CT and WSI using ResNet-50 and Vision Transformer (ViT) network, respectively. By fusing both radiologic and pathologic features, a radio-pathologic integrated model was constructed to predict the five LNM stages. For comparison, four single-modality models using CT images or WSIs were also constructed, respectively. All models were trained on the training set and validated on the testing set. RESULTS The radio-pathologic integrated mode achieved an overall accuracy of 84.0% and a kappa coefficient of 0.795 on the testing set. The areas under the curves (AUCs) of the integrated model in predicting the five LNM stages were 0.978 (95% Confidence Interval (CI 0.917-1.000), 0.946 (95% CI 0.867-1.000), 0.890 (95% CI 0.718-1.000), 0.971 (95% CI 0.920-1.000), and 0.982 (95% CI 0.911-1.000), respectively. Moreover, the integrated model achieved an AUC of 0.978 (95% CI 0.912-1.000) in predicting the binary status of nodal metastasis. CONCLUSION Our study suggests that radio-pathologic integrated model that combined both macroscale radiologic image and microscale pathologic image can better predict lymph node metastasis stage in patients with gastric cancer.
Collapse
Affiliation(s)
- Yuanshen Zhao
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Longsong Li
- Department of Gastroenterology, The First Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China
| | - Ke Han
- Department of Gastroenterology, The First Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China
| | - Tao Li
- Department of Radiology, The First Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Jingxian Duan
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qiuchang Sun
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Chaofan Zhu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Dong Liang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- National Innovation Center for Advanced Medical Devices, Shenzhen, China
- Shenzhen United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, China
| | - Ningli Chai
- Department of Gastroenterology, The First Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China.
| | - Zhi-Cheng Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- National Innovation Center for Advanced Medical Devices, Shenzhen, China.
- Shenzhen United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, China.
| |
Collapse
|
14
|
Ismail M, Craig S, Ahmed R, de Blank P, Tiwari P. Opportunities and Advances in Radiomics and Radiogenomics for Pediatric Medulloblastoma Tumors. Diagnostics (Basel) 2023; 13:2727. [PMID: 37685265 PMCID: PMC10487205 DOI: 10.3390/diagnostics13172727] [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: 07/24/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Recent advances in artificial intelligence have greatly impacted the field of medical imaging and vastly improved the development of computational algorithms for data analysis. In the field of pediatric neuro-oncology, radiomics, the process of obtaining high-dimensional data from radiographic images, has been recently utilized in applications including survival prognostication, molecular classification, and tumor type classification. Similarly, radiogenomics, or the integration of radiomic and genomic data, has allowed for building comprehensive computational models to better understand disease etiology. While there exist excellent review articles on radiomics and radiogenomic pipelines and their applications in adult solid tumors, in this review article, we specifically review these computational approaches in the context of pediatric medulloblastoma tumors. Based on our systematic literature research via PubMed and Google Scholar, we provide a detailed summary of a total of 15 articles that have utilized radiomic and radiogenomic analysis for survival prognostication, tumor segmentation, and molecular subgroup classification in the context of pediatric medulloblastoma. Lastly, we shed light on the current challenges with the existing approaches as well as future directions and opportunities with using these computational radiomic and radiogenomic approaches for pediatric medulloblastoma tumors.
Collapse
Affiliation(s)
- Marwa Ismail
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53706, USA; (S.C.); (P.T.)
| | - Stephen Craig
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53706, USA; (S.C.); (P.T.)
| | - Raheel Ahmed
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Peter de Blank
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA;
| | - Pallavi Tiwari
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53706, USA; (S.C.); (P.T.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
15
|
Zhao N, Weng S, Liu Z, Xu H, Ren Y, Guo C, Liu L, Zhang Z, Ji Y, Han X. CRISPR-Cas9 identifies growth-related subtypes of glioblastoma with therapeutical significance through cell line knockdown. BMC Cancer 2023; 23:749. [PMID: 37580710 PMCID: PMC10424363 DOI: 10.1186/s12885-023-11131-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 06/29/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a type of highly malignant brain tumor that is known for its significant intratumoral heterogeneity, meaning that there can be a high degree of variability within the tumor tissue. Despite the identification of several subtypes of GBM in recent years, there remains to explore a classification based on genes related to proliferation and growth. METHODS The growth-related genes of GBM were identified by CRISPR-Cas9 and univariate Cox regression analysis. The expression of these genes in the Cancer Genome Atlas cohort (TCGA) was used to construct growth-related genes subtypes (GGSs) via consensus clustering. Validation of this subtyping was performed using the nearest template prediction (NTP) algorithm in two independent Gene Expression Omnibus (GEO) cohorts and the ZZ cohort. Additionally, copy number variations, biological functions, and potential drugs were analyzed for each of the different subtypes separately. RESULTS Our research established multicenter-validated GGSs. GGS1 exhibits the poorest prognosis, with the highest frequency of chr 7 gain & chr 10 loss, and the lowest frequency of chr 19 & 20 co-gain. Additionally, GGS1 displays the highest expression of EGFR. Furthermore, it is significantly enriched in metabolic, stemness, proliferation, and signaling pathways. Besides we showed that Foretinib may be a potential therapeutic agent for GGS1, the worst prognostic subtype, through data screening and in vitro experiments. GGS2 has a moderate prognosis, with a slightly higher proportion of chr 7 gain & chr 10 loss, and the highest proportion of chr 19 & 20 co-gain. The prognosis of GGS3 is the best, with the least chr 7 gain & 10 loss and EGFR expression. CONCLUSIONS These results enhance our understanding of the heterogeneity of GBM and offer insights for stratified management and precise treatment of GBM patients.
Collapse
Affiliation(s)
- Nannan Zhao
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuqin Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunguang Guo
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Yuchen Ji
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
16
|
Liu Z, Xu Y, Wang Y, Weng S, Xu H, Ren Y, Guo C, Liu L, Zhang Z, Han X. Immune-related interaction perturbation networks unravel biological peculiars and clinical significance of glioblastoma. IMETA 2023; 2:e127. [PMID: 38867932 PMCID: PMC10989959 DOI: 10.1002/imt2.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/27/2023] [Accepted: 06/16/2023] [Indexed: 06/14/2024]
Abstract
The immune system is an interacting network of plentiful molecules that could better characterize the relationship between immunity and cancer. This study aims to investigate the behavioral patterns of immune-related interaction perturbation networks in glioblastoma. An immune-related interaction-perturbation framework was introduced to characterize four heterogeneous subtypes using RNA-seq data of TCGA/CGGA glioblastoma tissues and GTEx normal brain tissues. The stability and robustness of the four subtypes were validated in public datasets and our in-house cohort. In the four subtypes, C1 was an inflammatory subtype with high immune infiltration, low tumor purity, and potential response to immunotherapy; C2, an invasive subtype, was featured with dismal prognosis, telomerase reverse transcriptase promoter mutations, moderate levels of immunity, and stromal constituents, as well as sensitivity to receptor tyrosine kinase signaling inhibitors; C3 was a proliferative subtype with high tumor purity, immune-desert microenvironment, sensitivity to phosphatidylinositol 3'-kinase signaling inhibitor and DNA replication inhibitors, and potential resistance to immunotherapy; C4, a synaptogenesis subtype with the best prognosis, exhibited high synaptogenesis-related gene expression, prevalent isocitrate dehydrogenase mutations, and potential sensitivity to radiotherapy and chemotherapy. Overall, this study provided an attractive platform from the perspective of immune-related interaction perturbation networks, which might advance the tailored management of glioblastoma.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Interventional Institute of Zhengzhou UniversityZhengzhouChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouChina
| | - Yudi Xu
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuhui Wang
- Department of Clinical LaboratoryThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Siyuan Weng
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Hui Xu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuqing Ren
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Chunguang Guo
- Department of Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Long Liu
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhenyu Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xinwei Han
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Interventional Institute of Zhengzhou UniversityZhengzhouChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouChina
| |
Collapse
|
17
|
Ntenti C, Lallas K, Papazisis G. Clinical, Histological, and Molecular Prognostic Factors in Childhood Medulloblastoma: Where Do We Stand? Diagnostics (Basel) 2023; 13:diagnostics13111915. [PMID: 37296767 DOI: 10.3390/diagnostics13111915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Medulloblastomas, highly aggressive neoplasms of the central nervous system (CNS) that present significant heterogeneity in clinical presentation, disease course, and treatment outcomes, are common in childhood. Moreover, patients who survive may be diagnosed with subsequent malignancies during their life or could develop treatment-related medical conditions. Genetic and transcriptomic studies have classified MBs into four subgroups: wingless type (WNT), Sonic Hedgehog (SHH), Group 3, and Group 4, with distinct histological and molecular profiles. However, recent molecular findings resulted in the WHO updating their guidelines and stratifying medulloblastomas into further molecular subgroups, changing the clinical stratification and treatment management. In this review, we discuss most of the histological, clinical, and molecular prognostic factors, as well the feasibility of their application, for better characterization, prognostication, and treatment of medulloblastomas.
Collapse
Affiliation(s)
- Charikleia Ntenti
- First Department of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Konstantinos Lallas
- Department of Medical Oncology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Georgios Papazisis
- Clinical Research Unit, Special Unit for Biomedical Research and Education (BRESU), School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| |
Collapse
|
18
|
Radiomic features from dynamic susceptibility contrast perfusion-weighted imaging improve the three-class prediction of molecular subtypes in patients with adult diffuse gliomas. Eur Radiol 2023; 33:3455-3466. [PMID: 36853347 DOI: 10.1007/s00330-023-09459-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/21/2022] [Accepted: 01/20/2023] [Indexed: 03/01/2023]
Abstract
OBJECTIVES To investigate whether radiomic features extracted from dynamic susceptibility contrast perfusion-weighted imaging (DSC-PWI) can improve the prediction of the molecular subtypes of adult diffuse gliomas, and to further develop and validate a multimodal radiomic model by integrating radiomic features from conventional and perfusion MRI. METHODS We extracted 1197 radiomic features from each sequence of conventional MRI and DSC-PWI, respectively. The Boruta algorithm was used for feature selection and combination, and a three-class random forest method was applied to construct the models. We also constructed a combined model by integrating radiomic features and clinical metrics. The models' diagnostic performance for discriminating the molecular subtypes (IDH wild type [IDHwt], IDH mutant and 1p/19q-noncodeleted [IDHmut-noncodel], and IDH mutant and 1p/19q-codeleted [IDHmut-codel]) was compared using AUCs in the validation set. RESULTS We included 272 patients (training set, n = 166; validation set, n = 106) with grade II-IV gliomas (mean age, 48.7 years; range, 19-77 years). The proportions of the molecular subtypes were 66.2% IDHwt, 15.1% IDHmut-noncodel, and 18.8% IDHmut-codel. Nineteen radiomic features (13 from conventional MRI and 6 from DSC-PWI) were selected to build the multimodal radiomic model. In the validation set, the multimodal radiomic model showed better performance than the conventional radiomic model did in predicting the IDHwt and IDHmut-codel subtypes, which was comparable to the conventional radiomic model in predicting the IDHmut-noncodel subtype. The multimodal radiomic model yielded similar performance as the combined model in predicting the three molecular subtypes. CONCLUSIONS Adding DSC-PWI to conventional MRI can improve molecular subtype prediction in patients with diffuse gliomas. KEY POINTS • The multimodal radiomic model outperformed conventional MRI when predicting both the IDH wild type and IDH mutant and 1p/19q-codeleted subtypes of gliomas. • The multimodal radiomic model showed comparable performance to the combined model in the prediction of the three molecular subtypes. • Radiomic features from T1-weighted gadolinium contrast-enhanced and relative cerebral blood volume images played an important role in the prediction of molecular subtypes.
Collapse
|
19
|
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
|
20
|
Li S, Zhou B. A review of radiomics and genomics applications in cancers: the way towards precision medicine. Radiat Oncol 2022; 17:217. [PMID: 36585716 PMCID: PMC9801589 DOI: 10.1186/s13014-022-02192-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
The application of radiogenomics in oncology has great prospects in precision medicine. Radiogenomics combines large volumes of radiomic features from medical digital images, genetic data from high-throughput sequencing, and clinical-epidemiological data into mathematical modelling. The amalgamation of radiomics and genomics provides an approach to better study the molecular mechanism of tumour pathogenesis, as well as new evidence-supporting strategies to identify the characteristics of cancer patients, make clinical decisions by predicting prognosis, and improve the development of individualized treatment guidance. In this review, we summarized recent research on radiogenomics applications in solid cancers and presented the challenges impeding the adoption of radiomics in clinical practice. More standard guidelines are required to normalize radiomics into reproducible and convincible analyses and develop it as a mature field.
Collapse
Affiliation(s)
- Simin Li
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Hospital of China Medical University, Shenyang, 110001 Liaoning People’s Republic of China
| | - Baosen Zhou
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Hospital of China Medical University, Shenyang, 110001 Liaoning People’s Republic of China
| |
Collapse
|
21
|
Iyer S, Ismail M, Tamrazi B, Salloum R, de Blank P, Margol A, Correa R, Chen J, Bera K, Statsevych V, Ho ML, Vaidya P, Verma R, Hawes D, Judkins A, Fu P, Madabhushi A, Tiwari P. Novel MRI deformation-heterogeneity radiomic features are associated with molecular subgroups and overall survival in pediatric medulloblastoma: Preliminary findings from a multi-institutional study. Front Oncol 2022; 12:915143. [PMID: 36620600 PMCID: PMC9811390 DOI: 10.3389/fonc.2022.915143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Medulloblastoma (MB) is a malignant, heterogenous brain tumor. Advances in molecular profiling have led to identifying four molecular subgroups of MB (WNT, SHH, Group 3, Group 4), each with distinct clinical behaviors. We hypothesize that (1) aggressive MB tumors, growing heterogeneously, induce pronounced local structural deformations in the surrounding parenchyma, and (b) these local deformations as captured on Gadolinium (Gd)-enhanced-T1w MRI are independently associated with molecular subgroups, as well as overall survival in MB patients. Methods In this work, a total of 88 MB studies from 2 institutions were analyzed. Following tumor delineation, Gd-T1w scan for every patient was registered to a normal age-specific T1w-MRI template via deformable registration. Following patient-atlas registration, local structural deformations in the brain parenchyma were obtained for every patient by computing statistics from deformation magnitudes obtained from every 5mm annular region, 0 < d < 60 mm, where d is the distance from the tumor infiltrating edge. Results Multi-class comparison via ANOVA yielded significant differences between deformation magnitudes obtained for Group 3, Group 4, and SHH molecular subgroups, observed up to 60-mm outside the tumor edge. Additionally, Kaplan-Meier survival analysis showed that the local deformation statistics, combined with the current clinical risk-stratification approaches (molecular subgroup information and Chang's classification), could identify significant differences between high-risk and low-risk survival groups, achieving better performance results than using any of these approaches individually. Discussion These preliminary findings suggest there exists significant association of our tumor-induced deformation descriptor with overall survival in MB, and that there could be an added value in using the proposed radiomic descriptor along with the current risk classification approaches, towards more reliable risk assessment in pediatric MB.
Collapse
Affiliation(s)
- Sukanya Iyer
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Marwa Ismail
- Department of Radiology and Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Benita Tamrazi
- Department of Pathology, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Ralph Salloum
- Division of Hematology, Oncology & Bone Marrow Transplant, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Peter de Blank
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Ashley Margol
- Department of Pathology, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Ramon Correa
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Jonathan Chen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Kaustav Bera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Volodymyr Statsevych
- Department of Neuroradiology, Imaging Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Mai-Lan Ho
- Department of Radiology, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Pranjal Vaidya
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Ruchika Verma
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Debra Hawes
- Department of Pathology, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Alexander Judkins
- Department of Pathology, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Anant Madabhushi
- Radiology and Imaging Sciences, Biomedical Informatics (BMI) and Pathology, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Pallavi Tiwari
- Department of Radiology and Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
22
|
Feng Z, Li H, Liu Q, Duan J, Zhou W, Yu X, Chen Q, Liu Z, Wang W, Rong P. CT Radiomics to Predict Macrotrabecular-Massive Subtype and Immune Status in Hepatocellular Carcinoma. Radiology 2022; 307:e221291. [PMID: 36511807 DOI: 10.1148/radiol.221291] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Macrotrabecular-massive (MTM) subtype of hepatocellular carcinoma (HCC) is an aggressive variant associated with angiogenesis and immunosuppressive tumor microenvironment, which is expected to be noninvasively identified using radiomics approaches. Purpose To construct a CT radiomics model to predict the MTM subtype and to investigate the underlying immune infiltration patterns. Materials and Methods This study included five retrospective data sets and one prospective data set from three academic medical centers between January 2015 and December 2021. The preoperative liver contrast-enhanced CT studies of 365 adult patients with resected HCC were evaluated. The Third Xiangya Hospital of Central South University provided the training set and internal test set, while Yueyang Central Hospital and Hunan Cancer Hospital provided the external test sets. Radiomic features were extracted and used to develop a radiomics model with machine learning in the training set, and the performance was verified in the two test sets. The outcomes cohort, including 58 adult patients with advanced HCC undergoing transarterial chemoembolization and antiangiogenic therapy, was used to evaluate the predictive value of the radiomics model for progression-free survival (PFS). Bulk RNA sequencing of tumors from 41 patients in The Cancer Genome Atlas (TCGA) and single-cell RNA sequencing from seven prospectively enrolled participants were used to investigate the radiomics-related immune infiltration patterns. Area under the receiver operating characteristics curve of the radiomics model was calculated, and Cox proportional regression was performed to identify predictors of PFS. Results Among 365 patients (mean age, 55 years ± 10 [SD]; 319 men) used for radiomics modeling, 122 (33%) were confirmed to have the MTM subtype. The radiomics model included 11 radiomic features and showed good performance for predicting the MTM subtype, with AUCs of 0.84, 0.80, and 0.74 in the training set, internal test set, and external test set, respectively. A low radiomics model score relative to the median value in the outcomes cohort was independently associated with PFS (hazard ratio, 0.4; 95% CI: 0.2, 0.8; P = .01). The radiomics model was associated with dysregulated humoral immunity involving B-cell infiltration and immunoglobulin synthesis. Conclusion Accurate prediction of the macrotrabecular-massive subtype in patients with hepatocellular carcinoma was achieved using a CT radiomics model, which was also associated with defective humoral immunity. Published under a CC BY 4.0 license. Supplemental material is available for this article. See also the editorial by Yoon and Kim in this issue.
Collapse
Affiliation(s)
- Zhichao Feng
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Huiling Li
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Qianyun Liu
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Junhong Duan
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Wenming Zhou
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Xiaoping Yu
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Qian Chen
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Zhenguo Liu
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Wei Wang
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| | - Pengfei Rong
- From the Departments of Radiology (Z.F., H.L., J.D., W.W., P.R.), Pathology (Q.C.), and Infectious Disease (Z.L.), The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Rd, Changsha 410013, China; Department of Medical Imaging, Yueyang Central Hospital, Yueyang, China (Q.L., W.Z.); and Department of Diagnostic Radiology, Hunan Cancer Hospital, Changsha, China (X.Y.)
| |
Collapse
|
23
|
Zheng H, Li J, Liu H, Ting G, Yin Q, Li R, Liu M, Zhang Y, Duan S, Li Y, Wang D. MRI
Radiomics Signature of Pediatric Medulloblastoma Improves Risk Stratification Beyond Clinical and Conventional
MR
Imaging Features. J Magn Reson Imaging 2022. [DOI: 10.1002/jmri.28537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 11/04/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Hui Zheng
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Jinning Li
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Huanhuan Liu
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Gui Ting
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Qiufeng Yin
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Rui Li
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ming Liu
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yuzhen Zhang
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | | | - Yuhua Li
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Dengbin Wang
- Department of Radiology, Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
24
|
Cui H, Wang KY, Li WY, Zhu HB, Xiao LS, Liu L. CT images-based 3D convolutional neural network to predict early recurrence of solitary hepatocellular carcinoma after radical hepatectomy. Diagn Interv Radiol 2022; 28:524-531. [PMID: 36287132 PMCID: PMC9885724 DOI: 10.5152/dir.2022.201097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE The high rate of recurrence of hepatocellular carcinoma (HCC) after radical hepatectomy is an important factor that affects the long-term survival of patients. This study aimed to develop a computed tomography (CT) images-based 3-dimensional (3D) convolutional neural network (CNN) for the preoperative prediction of early recurrence (ER) (≤2 years) after radical hepatectomy in patients with solitary HCC and to compare the effects of segmentation sampling (SS) and non-segmentation sampling (NSS) on the prediction performance of 3D-CNN. METHODS Contrast-enhanced CT images of 220 HCC patients were used in this study (training group=178 and test group=42). We used SS and NSS to select the volume-of-interest to train SS-3D-CNN and NSS-3D-CNN separately. The prediction accuracy was evaluated using the test group. Finally, gradient-weighted class activation mappings (Grad-CAMs) were plotted to analyze the difference of prediction logic between the SS-3D-CNN and NSS-3D-CNN. RESULTS The areas under the receiver operating characteristic curves (AUCs) of the SS-3D-CNN and NSS3D-CNN in the training group were 0.824 (95% CI: 0.764-0.885) and 0.868 (95% CI: 0.815-0.921). The AUC of the SS-3D-CNN and NSS-3D-CNN in the test group were 0.789 (95% CI: 0.637-0.941) and 0.560 (95% CI: 0.378-0.742). The SS-3D-CNN could stratify patients into low- and high-risk groups, with significant differences in recurrence-free survival (RFS) (P < .001). But NSS-3D-CNN could not effectively stratify them in the test group. According to the Grad-CAMs, compared with SS-3D-CNN, NSS-3D-CNN was obviously interfered by the nearby tissues. CONCLUSION SS-3D-CNN may be of clinical use for identifying high-risk patients and formulating individualized treatment and follow-up strategies. SS is better than NSS in improving the performance of 3D-CNN in our study.
Collapse
Affiliation(s)
- Hao Cui
- Department of Medical Quality Management, Nanfang Hospital, Southern Medical University, Guangzhou, China; Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kun-Yuan Wang
- Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wen-Yuan Li
- Big data center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hong-Bo Zhu
- Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Oncology, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Lu-Shan Xiao
- Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Liu
- Department of Medical Quality Management, Nanfang Hospital, Southern Medical University, Guangzhou, China; Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
25
|
Jian A, Liu S, Di Ieva A. Artificial Intelligence for Survival Prediction in Brain Tumors on Neuroimaging. Neurosurgery 2022; 91:8-26. [PMID: 35348129 DOI: 10.1227/neu.0000000000001938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 01/08/2022] [Indexed: 12/30/2022] Open
Abstract
Survival prediction of patients affected by brain tumors provides essential information to guide surgical planning, adjuvant treatment selection, and patient counseling. Current reliance on clinical factors, such as Karnofsky Performance Status Scale, and simplistic radiological characteristics are, however, inadequate for survival prediction in tumors such as glioma that demonstrate molecular and clinical heterogeneity with variable survival outcomes. Advances in the domain of artificial intelligence have afforded powerful tools to capture a large number of hidden high-dimensional imaging features that reflect abundant information about tumor structure and physiology. Here, we provide an overview of current literature that apply computational analysis tools such as radiomics and machine learning methods to the pipeline of image preprocessing, tumor segmentation, feature extraction, and construction of classifiers to establish survival prediction models based on neuroimaging. We also discuss challenges relating to the development and evaluation of such models and explore ethical issues surrounding the future use of machine learning predictions.
Collapse
Affiliation(s)
- Anne Jian
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Royal Melbourne Hospital, Melbourne, Australia
| | - Sidong Liu
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Centre for Health Informatics, Australian Institute of Health Innovation, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Antonio Di Ieva
- Computational NeuroSurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
26
|
Kalasauskas D, Kosterhon M, Keric N, Korczynski O, Kronfeld A, Ringel F, Othman A, Brockmann MA. Beyond Glioma: The Utility of Radiomic Analysis for Non-Glial Intracranial Tumors. Cancers (Basel) 2022; 14:cancers14030836. [PMID: 35159103 PMCID: PMC8834271 DOI: 10.3390/cancers14030836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/30/2022] [Accepted: 02/04/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Tumor qualities, such as growth rate, firmness, and intrusion into healthy tissue, can be very important for operation planning and further treatment. Radiomics is a promising new method that allows the determination of some of these qualities on images performed before surgery. In this article, we provide a review of the use of radiomics in various tumors of the central nervous system, such as metastases, lymphoma, meningioma, medulloblastoma, and pituitary tumors. Abstract The field of radiomics is rapidly expanding and gaining a valuable role in neuro-oncology. The possibilities related to the use of radiomic analysis, such as distinguishing types of malignancies, predicting tumor grade, determining the presence of particular molecular markers, consistency, therapy response, and prognosis, can considerably influence decision-making in medicine in the near future. Even though the main focus of radiomic analyses has been on glial CNS tumors, studies on other intracranial tumors have shown encouraging results. Therefore, as the main focus of this review, we performed an analysis of publications on PubMed and Web of Science databases, focusing on radiomics in CNS metastases, lymphoma, meningioma, medulloblastoma, and pituitary tumors.
Collapse
Affiliation(s)
- Darius Kalasauskas
- Department of Neurosurgery, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.K.); (M.K.); (N.K.); (F.R.)
| | - Michael Kosterhon
- Department of Neurosurgery, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.K.); (M.K.); (N.K.); (F.R.)
| | - Naureen Keric
- Department of Neurosurgery, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.K.); (M.K.); (N.K.); (F.R.)
| | - Oliver Korczynski
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.K.); (A.K.); (A.O.)
| | - Andrea Kronfeld
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.K.); (A.K.); (A.O.)
| | - Florian Ringel
- Department of Neurosurgery, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.K.); (M.K.); (N.K.); (F.R.)
| | - Ahmed Othman
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.K.); (A.K.); (A.O.)
| | - Marc A. Brockmann
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.K.); (A.K.); (A.O.)
- Correspondence:
| |
Collapse
|
27
|
Che W, Wang Y, Wang X, Lyu J. Midlife brain metastases in the United States: Is male at risk? Cancer Med 2022; 11:1202-1216. [PMID: 35019232 PMCID: PMC8855893 DOI: 10.1002/cam4.4499] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/08/2021] [Accepted: 11/29/2021] [Indexed: 01/07/2023] Open
Abstract
Background Population‐based estimates of the impact of gender throughout the whole course of brain metastases (BMs) at the time of diagnosis of systemic malignancies are insufficient. We aimed to discover the influence of gender on the presence of BMs in newly diagnosed malignancies and the survival of those patients on a population‐based level. Methods Midlife patients (40 years ≤ age ≤60 years) with newly diagnosed malignancies and BMs at the time of diagnosis were abstracted from the Surveillance, Epidemiology, and End Results (SEER) database of the National Cancer Institute. Clinical variables adjusted patient data. The LASSO regression was performed to exclude the possibility of collinearity. Univariable and multivariable logistic regression analyses were applied to find independent predictors for the presence of BMs, while univariable and multivariable Cox proportional hazard regression analyses were used to determine prognosticators of survival. K‐M curves were used to perform the survival analysis. Result 276,327 population‐based samples met inclusion criteria between 2014 and 2016, and 5747 (2.08%) patients were diagnosed with BMs at the time of diagnosis of systematic malignancies. Among all midlife patients with cancer, 44.02% (121,634) were male, while 51.68% (2970) were male among patients with BMs at the time of diagnosis. The most frequent tumor type was breast cancer (23.11%), and lung cancer had the highest incidence proportion of BMs among the entire cohort (19.34%). The multivariable logistic regression model suggested that female (vs. male, odds ratio [OR] 1.07, 95% CI: 1.01–1.14, p < 0.001) was associated with a higher risk of the presence of BMs at the time of diagnosis. Moreover, in the multivariable Cox model for all‐cause mortality in individuals with BMs at diagnosis, female (vs. male, hazard ratio [HR], 0.86, 95% CI, 0.80–0.92, p < 0.001) was shown to have a lower risk of decreased all‐cause mortality. Conclusion The middle‐aged females were at increased risk of developing BMs, while the middle‐aged males with BMs were at higher risk of having poorer survival.
Collapse
Affiliation(s)
- Wenqiang Che
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yujiao Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiangyu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
28
|
Wan Y, Yang P, Xu L, Yang J, Luo C, Wang J, Chen F, Wu Y, Lu Y, Ruan D, Niu T. Radiomics analysis combining unsupervised learning and handcrafted features: A multiple-disease study. Med Phys 2021; 48:7003-7015. [PMID: 34453332 DOI: 10.1002/mp.15199] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
PURPOSE To study and investigate the synergistic benefit of incorporating both conventional handcrafted and learning-based features in disease identification across a wide range of clinical setups. METHODS AND MATERIALS In this retrospective study, we collected 170, 150, 209, and 137 patients with four different disease types associated with identification objectives : Lymph node metastasis status of gastric cancer (GC), 5-year survival status of patients with high-grade osteosarcoma (HOS), early recurrence status of intrahepatic cholangiocarcinoma (ICC), and pathological grades of pancreatic neuroendocrine tumors (pNETs). Computed tomography (CT) and magnetic resonance imaging (MRI) were used to derive image features for GC/HOS/pNETs and ICC, respectively. In each study, 67 universal handcrafted features and study-specific features based on the sparse autoencoder (SAE) method were extracted and fed into the subsequent feature selection and learning model to predict the corresponding disease identification. Models using handcrafted alone, SAE alone, and hybrid features were optimized and their performance was compared. Prominent features were analyzed both qualitatively and quantitatively to generate study-specific and cross-study insight. In addition to direct performance gain assessment, correlation analysis was performed to assess the complementarity between handcrafted features and SAE features. RESULTS On the independent hold-off test, the handcrafted, SAE, and hybrid features based prediction yielded area under the curve of 0.761 versus 0.769 versus 0.829 for GC, 0.629 versus 0.740 versus 0.709 for HOS, 0.717 versus 0.718 versus 0.758 for ICC, and 0.739 versus 0.715 versus 0.771 for pNETs studies, respectively. In three out of the four studies, prediction using the hybrid features yields the best performance, demonstrating the general benefit in using hybrid features. Prediction with SAE features alone had the best performance in the HOS study, which may be explained by the complexity of HOS prognosis and the possibility of a slight overfit due to higher correlation between handcrafted and SAE features. CONCLUSION This study demonstrated the general benefit of combing handcrafted and learning-based features in radiomics modeling. It also clearly illustrates the task-specific and data-specific dependency on the performance gain and suggests that while the common methodology of feature combination may be applied across various studies and tasks, study-specific feature selection and model optimization are still necessary to achieve high accuracy and robustness.
Collapse
Affiliation(s)
- Yidong Wan
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Pengfei Yang
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Lei Xu
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jing Yang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Chen Luo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jing Wang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China.,Department of Radiation Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Chen
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Wu
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Lu
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dan Ruan
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, California, USA
| | - Tianye Niu
- Nuclear & Radiological Engineering and Medical Physics Programs, Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
29
|
Liu X, Zhang D, Liu Z, Li Z, Xie P, Sun K, Wei W, Dai W, Tang Z, Ding Y, Cai G, Tong T, Meng X, Tian J. Deep learning radiomics-based prediction of distant metastasis in patients with locally advanced rectal cancer after neoadjuvant chemoradiotherapy: A multicentre study. EBioMedicine 2021; 69:103442. [PMID: 34157487 PMCID: PMC8237293 DOI: 10.1016/j.ebiom.2021.103442] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/17/2021] [Accepted: 06/01/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Accurate predictions of distant metastasis (DM) in locally advanced rectal cancer (LARC) patients receiving neoadjuvant chemoradiotherapy (nCRT) are helpful in developing appropriate treatment plans. This study aimed to perform DM prediction through deep learning radiomics. METHODS We retrospectively sampled 235 patients receiving nCRT with the minimum 36 months' postoperative follow-up from three hospitals. Through transfer learning, a deep learning radiomic signature (DLRS) based on multiparametric magnetic resonance imaging (MRI) was constructed. A nomogram was established integrating deep MRI information and clinicopathologic factors for better prediction. Harrell's concordance index (C-index) and time-dependent receiver operating characteristic (ROC) were used as performance metrics. Furthermore, the risk of DM in patients with different response to nCRT was evaluated with the nomogram. FINDINGS DLRS performed well in DM prediction, with a C-index of 0·747 and an area under curve (AUC) at three years of 0·894 in the validation cohort. The performance of nomogram was better, with a C-index of 0·775. In addition, the nomogram could stratify patients with different responses to nCRT into high- and low-risk groups of DM (P < 0·05). INTERPRETATION MRI-based deep learning radiomics had potential in predicting the DM of LARC patients receiving nCRT and could help evaluate the risk of DM in patients who have different responses to nCRT. FUNDING The funding bodies that contributed to this study are listed in the Acknowledgements section.
Collapse
Affiliation(s)
- Xiangyu Liu
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Dafu Zhang
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming 650118, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100080, China
| | - Zhenhui Li
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming 650118, China
| | - Peiyi Xie
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| | - Kai Sun
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Wei Wei
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Weixing Dai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Zhenchao Tang
- Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing 100191, China
| | - Yingying Ding
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming 650118, China
| | - Guoxiang Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Tong Tong
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Xiaochun Meng
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China.
| | - Jie Tian
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing 100191, China; Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing 100191, China.
| |
Collapse
|